Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
J Cell Sci ; 123(Pt 24): 4280-91, 2010 Dec 15.
Article in English | MEDLINE | ID: mdl-21098633

ABSTRACT

Staphylococcus aureus, which is a leading cause of hospital-acquired infections, binds via fibronectin to integrin α5ß1, a process that can promote host colonization in vivo. Integrin engagement induces actin cytoskeleton rearrangements that result in the uptake of S. aureus by non-professional phagocytic cells. Interestingly, we found that fibronectin-binding S. aureus trigger the redistribution of membrane microdomain components. In particular, ganglioside GM1 and GPI-linked proteins were recruited upon integrin ß1 engagement, and disruption of membrane microdomains blocked bacterial internalization. Several membrane-microdomain-associated proteins, such as flotillin-1 and flotillin-2, as well as caveolin, were recruited to sites of bacterial attachment. Whereas dominant-negative versions of flotillin-2 did not affect bacterial attachment or internalization, cells deficient for caveolin-1 (Cav1(-/-)) showed increased uptake of S. aureus and other Fn-binding pathogens. Recruitment of membrane microdomains to cell-associated bacteria was unaltered in Cav1(-/-) cells. However, fluorescence recovery after photobleaching (FRAP) revealed an enhanced mobility of membrane-microdomain-associated proteins in the absence of caveolin-1. Enhanced membrane microdomain mobility and increased uptake of S. aureus was repressed by expression of wild-type caveolin-1, but not caveolin-1 G83S, which harbors a point mutation in the caveolin scaffolding domain. Similarly, chemical or physical stimulation of membrane fluidity led to increased uptake of S. aureus. These results highlight a crucial role for caveolin-1 in negative regulation of membrane microdomain mobility, thereby affecting endocytosis of bacteria-engaged integrins. This process might not only limit host cell invasion by integrin-binding bacterial pathogens, but might also be physiologically relevant for integrin-mediated cell adhesion.


Subject(s)
Caveolin 1/metabolism , Fibronectins/metabolism , Integrin beta1/metabolism , Membrane Microdomains/metabolism , Staphylococcus aureus/metabolism , Amino Acid Sequence , Animals , Bacterial Adhesion , Bacterial Proteins/metabolism , Blood Proteins/metabolism , Cattle , Caveolin 1/chemistry , Caveolin 1/deficiency , Endocytosis , Fibroblasts/cytology , Fibroblasts/enzymology , Fibroblasts/microbiology , Fibroblasts/ultrastructure , HEK293 Cells , Humans , Membrane Proteins/metabolism , Mice , Molecular Sequence Data , Protein Transport , Staphylococcal Infections/metabolism , Staphylococcus aureus/cytology , src-Family Kinases/metabolism
2.
Infect Immun ; 75(8): 4116-26, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17517873

ABSTRACT

Several pathogenic bacteria exploit human carcinoembryonic antigen-related cell adhesion molecules (CEACAMs) for adhesion to and invasion into their host cells. CEACAM isoforms have characteristic expression patterns on epithelial, endothelial, or hematopoietic cells, providing bacteria with distinct sets of receptors on particular tissues. For example, while CEACAM1 and CEACAM6 have a wide tissue distribution, CEACAM3, CEACAM4, and CEACAM8 are uniquely expressed on primary human granulocytes, whereas CEA and CEACAM7 are limited to epithelia. By reconstitution of a CEACAM-deficient cell line with individual CEACAMs, we have analyzed the requirements for CEACAM-mediated internalization of Neisseria gonorrhoeae. Our results point to two mechanistically different uptake pathways triggered by either epithelial CEACAMs (CEACAM1, CEA, and CEACAM6) or the granulocyte-specific CEACAM3. In particular, CEACAM3-mediated uptake critically depends on Src family protein tyrosine kinase (PTK) activity, and CEACAM3 associates with the SH2 domains of several Src PTKs. In contrast, epithelial CEACAMs require the integrity of cholesterol-rich membrane microdomains and are affected by cholesterol depletion, whereas CEACAM3-mediated uptake by transfected cells or the opsonin-independent phagocytosis by human granulocytes is not altered in the presence of cholesterol chelators. These results allow the subdivision of all human CEACAMs known to be utilized as pathogen receptors into functional groups and point to important consequences for bacterial engagement of distinct CEACAM isoforms.


Subject(s)
Bacterial Adhesion/physiology , Bacterial Outer Membrane Proteins/metabolism , Cell Adhesion Molecules/metabolism , Membrane Microdomains/metabolism , Neisseria gonorrhoeae/physiology , src-Family Kinases/metabolism , Cell Adhesion Molecules/genetics , Cell Line , Colony Count, Microbial , Cytosol/microbiology , Humans , Protein Isoforms/metabolism
3.
J Bacteriol ; 188(16): 5783-96, 2006 Aug.
Article in English | MEDLINE | ID: mdl-16885446

ABSTRACT

Staphylococcus aureus is an important pathogen, causing a wide range of infections including sepsis, wound infections, pneumonia, and catheter-related infections. In several pathogens ClpP proteases were identified by in vivo expression technologies to be important for virulence. Clp proteolytic complexes are responsible for adaptation to multiple stresses by degrading accumulated and misfolded proteins. In this report clpP, encoding the proteolytic subunit of the ATP-dependent Clp protease, was deleted, and gene expression of DeltaclpP was determined by global transcriptional analysis using DNA-microarray technology. The transcriptional profile reveals a strong regulatory impact of ClpP on the expression of genes encoding proteins that are involved in the pathogenicity of S. aureus and adaptation of the pathogen to several stresses. Expression of the agr system and agr-dependent extracellular virulence factors was diminished. Moreover, the loss of clpP leads to a complete transcriptional derepression of genes of the CtsR- and HrcA-controlled heat shock regulon and a partial derepression of genes involved in oxidative stress response, metal homeostasis, and SOS DNA repair controlled by PerR, Fur, MntR, and LexA. The levels of transcription of genes encoding proteins involved in adaptation to anaerobic conditions potentially regulated by an Fnr-like regulator were decreased. Furthermore, the expression of genes whose products are involved in autolysis was deregulated, leading to enhanced autolysis in the mutant. Our results indicate a strong impact of ClpP proteolytic activity on virulence, stress response, and physiology in S. aureus.


Subject(s)
Bacteriolysis/physiology , DNA Repair/physiology , Endopeptidase Clp/metabolism , Oxidative Stress/physiology , Regulon/physiology , Staphylococcus aureus/enzymology , Virulence Factors/metabolism , Anaerobiosis , Bacterial Adhesion , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Cell Line , DNA Repair/genetics , DNA, Bacterial/metabolism , Down-Regulation , Endopeptidase Clp/genetics , Gene Expression Regulation, Bacterial , Gene Expression Regulation, Enzymologic , Humans , Regulon/genetics , Serine Endopeptidases/genetics , Serine Endopeptidases/metabolism , Staphylococcus aureus/genetics , Up-Regulation , Urease/metabolism , Virulence Factors/genetics
4.
Eur J Cell Biol ; 85(3-4): 235-42, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16546567

ABSTRACT

A large number of bacterial pathogens targets cell adhesion molecules to establish an intimate contact with host cells and tissues. Members of the integrin, cadherin and immunoglobulin-related cell adhesion molecule (IgCAM) families are frequently recognized by specific bacterial surface proteins. Binding can trigger bacterial internalization following cytoskeletal rearrangements that are initiated upon receptor clustering. Moreover, signals emanating from the occupied receptors can result in cellular responses such as gene expression events that influence the phenotype of the infected cell. This review will address recent advances in our understanding of bacterial engagement of cellular adhesion molecules by discussing the binding of integrins by Staphylococcus aureus as well as the exploitation of IgCAMs by pathogenic Neisseria species.


Subject(s)
Cell Adhesion Molecules/metabolism , Gram-Negative Bacterial Infections/microbiology , Neisseria/pathogenicity , Staphylococcal Infections/microbiology , Staphylococcus aureus/pathogenicity , Epithelial Cells/cytology , Epithelial Cells/metabolism , Epithelial Cells/ultrastructure , Humans , Immunoglobulins/immunology , Immunoglobulins/metabolism , Integrins/metabolism , Neisseria/cytology , Neisseria/metabolism , Staphylococcus aureus/cytology , Staphylococcus aureus/metabolism
5.
J Cell Sci ; 118(Pt 10): 2189-200, 2005 May 15.
Article in English | MEDLINE | ID: mdl-15855238

ABSTRACT

Nosocomial infections by Staphylococcus aureus, a Gram-positive pathogen colonising human skin and mucosal surfaces, are an increasing health care problem. Clinical isolates almost invariably express fibronectin-binding proteins that, by indirectly linking the bacteria with host integrin alpha5beta1, can promote uptake of the microorganisms by eukaryotic cells. Integrin engagement by pathogenic fibronectin-binding S. aureus, but not by non-pathogenic S. carnosus, triggered the recruitment of focal contact-associated proteins vinculin, tensin, zyxin and FAK to the sites of bacterial attachment. Moreover, dominant-negative versions of FAK-blocked integrin-mediated internalisation and FAK-deficient cells were severely impaired in their ability to internalise S. aureus. Pathogen binding induced tyrosine phosphorylation of several host proteins associated with bacterial attachment sites, including FAK and the Src substrate cortactin. In FAK-deficient cells, local recruitment of cortactin still occurred, whereas the integrin- and Src-dependent tyrosine phosphorylation of cortactin was abolished. As siRNA-mediated gene silencing of cortactin or mutation of critical amino acid residues within cortactin interfered with uptake of S. aureus, our results reveal a novel functional connection between integrin engagement, FAK activation and Src-mediated cortactin phosphorylation. Cooperation between FAK, Src and cortactin in integrin-mediated internalisation of bacteria also suggests a molecular scenario of how engagement of integrins could be coupled to membrane endocytosis.


Subject(s)
Cortactin/metabolism , Focal Adhesion Protein-Tyrosine Kinases/physiology , Integrin alpha5beta1/physiology , Staphylococcus aureus/physiology , Actins/metabolism , Animals , Cells, Cultured , Cortactin/genetics , Cytoskeletal Proteins , Endocytosis , Fibronectins/physiology , Focal Adhesion Protein-Tyrosine Kinases/genetics , Glycoproteins/metabolism , Humans , Mice , Mice, Knockout , Microfilament Proteins/metabolism , Microscopy, Electron, Scanning , Mutation , Phosphorylation , Staphylococcus/physiology , Staphylococcus/ultrastructure , Staphylococcus aureus/ultrastructure , Tensins , Tyrosine/metabolism , Vinculin/metabolism , Zyxin , src-Family Kinases/antagonists & inhibitors , src-Family Kinases/metabolism
6.
J Microbiol Methods ; 59(1): 23-32, 2004 Oct.
Article in English | MEDLINE | ID: mdl-15325750

ABSTRACT

Microscopic discrimination between extracellular and invasive, intracellular bacteria is a valuable technique in microbiology and immunology. We describe a novel fluorescence staining protocol, called FITC-biotin-avidin (FBA) staining, which allows the differentiation between extracellular and intracellular bacteria and is independent of specific antibodies directed against the microorganisms. FBA staining of eukaryotic cells infected with Gram-negative bacteria of the genus Neisseria or the Gram-positive pathogen Staphylococcus aureus are employed to validate the novel technique. The quantitative evaluation of intracellular pathogens by the FBA staining protocol yields identical results compared to parallel samples stained with conventional, antibody-dependent methods. FBA staining eliminates the need for cell permeabilization resulting in robust and rapid detection of invasive microbes. Taken together, FBA staining provides a reliable and convenient alternative for the differential detection of intracellular and extracellular bacteria and should be a valuable technical tool for the quantitative analysis of the invasive properties of pathogenic bacteria and other microorganisms.


Subject(s)
Bacterial Proteins/chemistry , Biotin/chemistry , Fluorescein-5-isothiocyanate/chemistry , Fluorescent Dyes/chemistry , Neisseria gonorrhoeae/growth & development , Staining and Labeling/methods , Staphylococcus aureus/growth & development , Streptavidin/chemistry , Biotin/analogs & derivatives , Cell Line , Humans , Image Processing, Computer-Assisted , Microscopy, Fluorescence , Neisseria gonorrhoeae/metabolism , Staphylococcus aureus/metabolism
7.
J Exp Med ; 199(1): 35-46, 2004 Jan 05.
Article in English | MEDLINE | ID: mdl-14707113

ABSTRACT

Carcinoembryonic antigen-related cell adhesion molecules (CEACAMs) are used by several human pathogens to anchor themselves to or invade host cells. Interestingly, human granulocytes express a specific isoform, CEACAM3, that participates together with CEACAM1 and CEACAM6 in the recognition of CEACAM-binding microorganisms. Here we show that CEACAM3 can direct efficient, opsonin-independent phagocytosis of CEACAM-binding Neisseria, Moraxella, and Haemophilus species. CEACAM3- but not CEACAM6-mediated uptake is blocked by dominant-negative versions of the small GTPase Rac. Moreover, CEACAM3 engagement triggers membrane recruitment and increased GTP loading of Rac that are not observed upon bacterial binding to CEACAM6. Internalization and Rac stimulation are also inhibited by compromising the integrity of an immunoreceptor tyrosine-based activation motif (ITAM)-like sequence in the cytoplasmic tail of CEACAM3 or by interference with Src family protein tyrosine kinases that phosphorylate CEACAM3. In contrast to interfering with CEACAM6, blockage of CEACAM3-mediated events reduces the ability of primary human granulocytes to internalize and eliminate CEACAM-binding bacteria, indicating an important role of CEACAM3 in the control of human-specific pathogens by the innate immune system.


Subject(s)
Carcinoembryonic Antigen/physiology , Granulocytes/microbiology , Phagocytosis/physiology , Base Sequence , Carcinoembryonic Antigen/genetics , Cell Adhesion Molecules/physiology , DNA Primers , Granulocytes/immunology , Humans , Immunity, Innate , Molecular Sequence Data , Neisseria gonorrhoeae/pathogenicity , Neisseria gonorrhoeae/ultrastructure , Plasmids/genetics
8.
J Biol Chem ; 278(43): 42524-31, 2003 Oct 24.
Article in English | MEDLINE | ID: mdl-12893831

ABSTRACT

Staphylococcus aureus, a common cause of nosocomial infections, is able to invade eukaryotic cells by indirectly engaging beta1 integrin-containing host receptors, whereas non-pathogenic Staphylococcus carnosus is not invasive. Here, we identify intracellular signals involved in integrin-initiated internalization of S. aureus. In particular, the host cell actin cytoskeleton and Src family protein-tyrosine kinases (PTKs) are essential to mediate S. aureus invasion. Src PTKs are activated in response to pathogenic S. aureus, but not S. carnosus. In addition, pharmacological and genetic interference with Src PTK function reduces bacterial internalization. Importantly, Src PTK-deficient cells are resistant to S. aureus invasion, demonstrating the essentiality of host Src PTKs in integrin-mediated uptake of this pathogen.


Subject(s)
Integrin beta1/physiology , Staphylococcus aureus/pathogenicity , src-Family Kinases/physiology , Actins/physiology , Cell Line , Endocytosis , Humans , Signal Transduction , Staphylococcal Infections/etiology , Staphylococcal Infections/microbiology , Staphylococcus aureus/physiology , src-Family Kinases/metabolism
9.
J Bacteriol ; 185(6): 1831-40, 2003 Mar.
Article in English | MEDLINE | ID: mdl-12618447

ABSTRACT

Genomes of prokaryotes differ significantly in size and DNA composition. Escherichia coli is considered a model organism to analyze the processes involved in bacterial genome evolution, as the species comprises numerous pathogenic and commensal variants. Pathogenic and nonpathogenic E. coli strains differ in the presence and absence of additional DNA elements contributing to specific virulence traits and also in the presence and absence of additional genetic information. To analyze the genetic diversity of pathogenic and commensal E. coli isolates, a whole-genome approach was applied. Using DNA arrays, the presence of all translatable open reading frames (ORFs) of nonpathogenic E. coli K-12 strain MG1655 was investigated in 26 E. coli isolates, including various extraintestinal and intestinal pathogenic E. coli isolates, 3 pathogenicity island deletion mutants, and commensal and laboratory strains. Additionally, the presence of virulence-associated genes of E. coli was determined using a DNA "pathoarray" developed in our laboratory. The frequency and distributional pattern of genomic variations vary widely in different E. coli strains. Up to 10% of the E. coli K-12-specific ORFs were not detectable in the genomes of the different strains. DNA sequences described for extraintestinal or intestinal pathogenic E. coli are more frequently detectable in isolates of the same origin than in other pathotypes. Several genes coding for virulence or fitness factors are also present in commensal E. coli isolates. Based on these results, the conserved E. coli core genome is estimated to consist of at least 3,100 translatable ORFs. The absence of K-12-specific ORFs was detectable in all chromosomal regions. These data demonstrate the great genome heterogeneity and genetic diversity among E. coli strains and underline the fact that both the acquisition and deletion of DNA elements are important processes involved in the evolution of prokaryotes.


Subject(s)
Escherichia coli/pathogenicity , Evolution, Molecular , Genetic Variation , Genome, Bacterial , Oligonucleotide Array Sequence Analysis/methods , Electrophoresis, Gel, Pulsed-Field , Escherichia coli/genetics , Escherichia coli Infections/microbiology , Escherichia coli Proteins/genetics , Humans , Molecular Sequence Data , Open Reading Frames/genetics , Polymerase Chain Reaction , Sequence Analysis, DNA , Virulence
SELECTION OF CITATIONS
SEARCH DETAIL
...