Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Small ; 19(29): e2300282, 2023 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-37026659

RESUMO

Molecular thin carbon nanomembranes (CNMs) synthesized by electron irradiation induced cross-linking of aromatic self-assembled monolayers (SAMs) are promising 2D materials for the next generation of filtration technologies. Their unique properties including ultimately low thickness of ≈1 nm, sub-nanometer porosity, mechanical and chemical stability are attractive for the development of innovative filters with low energy consumption, improved selectivity, and robustness. However, the permeation mechanisms through CNMs resulting in, e.g., an ≈1000 times higher fluxes of water in comparison to helium have not been yet understood. Here, a study of the permeation of He, Ne, D2 , CO2 , Ar, O2 and D2 O using mass spectrometry in the temperature range from room temperature to ≈120 °C is studied. As a model system, CNMs made from [1″,4',1',1]-terphenyl-4-thiol SAMs are investigated. It is found out that all studied gases experience an activation energy barrier upon the permeation which scales with their kinetic diameters. Moreover, their permeation rates are dependent on the adsorption on the nanomembrane surface. These findings enable to rationalize the permeation mechanisms and establish a model, which paves the way toward the rational design not only of CNMs but also of other organic and inorganic 2D materials for energy-efficient and highly selective filtration applications.

2.
Infect Immun ; 90(11): e0027622, 2022 11 17.
Artigo em Inglês | MEDLINE | ID: mdl-36314784

RESUMO

The peptidyl-prolyl-cis/trans-isomerase (PPIase) macrophage infectivity potentiator (Mip) contributes to the pathogenicity and fitness of L. pneumophila, the causative agent of Legionnaires' disease. Here, we identified the stringent starvation protein SspB, hypothetical protein Lpc2061, and flagellin FlaA as bacterial interaction partners of Mip. The macrolide FK506, which inhibits the PPIase activity of Mip, interfered with the binding of Lpc2061. Moreover, we demonstrated that the N-terminal dimerization region and amino acid Y185 in the C-terminal PPIase domain of Mip are required for the binding of Lpc2061 and FlaA. The modeling of the interaction partners and global docking with Mip suggested nonoverlapping binding interfaces, and a molecular dynamic simulation predicted an increased stability for the tripartite interaction of Lpc2061, Mip, and FlaA. On the functional level, we demonstrated that Mip promotes L. pneumophila flagellation, which is positively influenced by the binding of Lpc2061 and reduced by FK506. Also, L. pneumophila mutants expressing the Y185A or the monomeric Mip variant, which bind less Lpc2061, were nonmotile, were less flagellated, and yielded less FlaA when quantified. To our knowledge, this is the first report in which a PPIase and its bacterial interaction partners were demonstrated to influence flagellation.


Assuntos
Proteínas de Bactérias , Flagelos , Legionella pneumophila , Macrófagos , Peptidilprolil Isomerase , Humanos , Proteínas de Bactérias/metabolismo , Legionella pneumophila/metabolismo , Doença dos Legionários/microbiologia , Macrófagos/microbiologia , Peptidilprolil Isomerase/metabolismo , Tacrolimo , Flagelos/metabolismo
3.
Antonie Van Leeuwenhoek ; 114(10): 1483-1496, 2021 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-34355285

RESUMO

Strain M2T was isolated from the beach of Cuxhaven, Wadden Sea, Germany, in course of a program to attain new producers of bioactive natural products. Strain M2T produces litoralimycin and sulfomycin-type thiopeptides. Bioinformatic analysis revealed a potential biosynthetic gene cluster encoding for the M2T thiopeptides. The strain is Gram-stain-positive, rod shaped, non-motile, spore forming, showing a yellow colony color and forms extensively branched substrate mycelium and aerial hyphae. Inferred from the 16S rRNA gene phylogeny strain M2T affiliates with the genus Streptomonospora. It shows 96.6% 16S rRNA gene sequence similarity to the type species Streptomonospora salina DSM 44593 T and forms a distinct branch with Streptomonospora sediminis DSM 45723 T with 97.0% 16S rRNA gene sequence similarity. Genome-based phylogenetic analysis revealed that M2T is closely related to Streptomonospora alba YIM 90003 T with a digital DNA-DNA hybridisation (dDDH) value of 26.6%. The predominant menaquinones of M2T are MK-10(H6), MK-10(H8), and MK-11(H6) (> 10%). Major cellular fatty acids are iso-C16:0, anteiso C17:0 and C18:0 10-methyl. The polar lipid profile consisted of diphosphatidylglycerol phosphatidyl glycerol, phosphatidylinositol, phosphatidylcholine, phosphatidylethanolamine, three glycolipids, two unknown phospholipids, and two unknown lipids. The genome size of type strain M2T is 5,878,427 bp with 72.1 mol % G + C content. Based on the results obtained from phylogenetic and chemotaxonomic studies, strain M2T (= DSM 106425 T = NCCB 100650 T) is considered to represent a novel species within the genus Streptomonospora for which the name Streptomonospora litoralis sp. nov. is proposed.


Assuntos
Areia , Actinobacteria , DNA Bacteriano/genética , Filogenia , RNA Ribossômico 16S/genética
4.
Front Pharmacol ; 10: 340, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31024308

RESUMO

The Gram-positive pathogen Clostridioides difficile is the main bacterial agent of nosocomial antibiotic associated diarrhea. Bacterial peptidyl-prolyl-cis/trans-isomerases (PPIases) are well established modulators of virulence that influence the outcome of human pathologies during infections. Here, we present the first interactomic network of the sole cyclophilin-type PPIase of C. difficile (CdPpiB) and show that it has diverse interaction partners including major enzymes of the amino acid-dependent energy (LdhA, EtfAB, Had, Acd) and the glucose-derived (Fba, GapA, Pfo, Pyk, Pyc) central metabolism. Proteins of the general (UspA), oxidative (Rbr1,2,3, Dsr), alkaline (YloU, YphY) and cold shock (CspB) response were found bound to CdPpiB. The transcriptional (Lrp), translational (InfC, RFF) and folding (GroS, DnaK) control proteins were also found attached. For a crucial enzyme of cysteine metabolism, O-acetylserine sulfhydrylase (CysK), the global transcription regulator Lrp and the flagellar subunit FliC, these interactions were independently confirmed using a bacterial two hybrid system. The active site residues F50, F109, and F110 of CdPpiB were shown to be important for the interaction with the residue P87 of Lrp. CysK activity after heat denaturation was restored by interaction with CdPpiB. In accordance, tolerance toward cell wall stress caused by the exposure to amoxicillin was reduced. In the absence of CdPpiB, C. difficile was more susceptible toward L-cysteine. At the same time, the cysteine-mediated suppression of toxin production ceased resulting in higher cytotoxicity. In summary, the cyclophilin-type PPIase of C. difficile (CdPpiB) coordinates major cellular processes via its interaction with major regulators of transcription, translation, protein folding, stress response and the central metabolism.

5.
Methods Mol Biol ; 1921: 323-331, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30694502

RESUMO

Legionnaires' disease is a severe pneumonia caused by inhalation of Legionella pneumophila. Although powerful infection models ranging from monocellular host systems to mammals were developed, numerous intra- and extracellular interactions of L. pneumophila factors with human lung tissue structures remain unknown. Therefore, we developed and applied a novel infection model for Legionnaires' disease comprising living human lung tissue explants (HLTEs). This model allows analyzing Legionella infections at a unique level of complexity and narrows the gap between current infection models and postmortem histopathology analyses of infected patients. Here we describe the infection of tumor-free pulmonary tissue samples from patients undergoing lobe- or pneumectomy because of lung cancer. The method comprises bacterial cultivation, preparation of HLTEs, and infection of HLTEs. The infected tissue samples allow to characterize tissue damage, bacterial localization, dissemination and growth kinetics, and the host's molecular response.


Assuntos
Legionella pneumophila/fisiologia , Doença dos Legionários/microbiologia , Pulmão/microbiologia , Interações Hospedeiro-Patógeno , Humanos , Doença dos Legionários/patologia , Pulmão/patologia , Técnicas de Cultura de Tecidos
6.
Artigo em Inglês | MEDLINE | ID: mdl-29552544

RESUMO

Environmental bacteria of the genus Legionella naturally parasitize free-living amoebae. Upon inhalation of bacteria-laden aerosols, the opportunistic pathogens grow intracellularly in alveolar macrophages and can cause a life-threatening pneumonia termed Legionnaires' disease. Intracellular replication in amoebae and macrophages takes place in a unique membrane-bound compartment, the Legionella-containing vacuole (LCV). LCV formation requires the bacterial Icm/Dot type IV secretion system, which translocates literally hundreds of "effector" proteins into host cells, where they modulate crucial cellular processes for the pathogen's benefit. The mechanism of LCV formation appears to be evolutionarily conserved, and therefore, amoebae are not only ecologically significant niches for Legionella spp., but also useful cellular models for eukaryotic phagocytes. In particular, Acanthamoeba castellanii and Dictyostelium discoideum emerged over the last years as versatile and powerful models. Using genetic, biochemical and cell biological approaches, molecular interactions between amoebae and Legionella pneumophila have recently been investigated in detail with a focus on the role of phosphoinositide lipids, small and large GTPases, autophagy components and the retromer complex, as well as on bacterial effectors targeting these host factors.


Assuntos
Acanthamoeba/microbiologia , Dictyostelium/microbiologia , Modelos Animais de Doenças , Legionella/metabolismo , Doença dos Legionários/microbiologia , Doença dos Legionários/veterinária , Acanthamoeba castellanii/microbiologia , Amoeba/microbiologia , Animais , Autofagia , Proteínas de Bactérias/metabolismo , Avaliação Pré-Clínica de Medicamentos , Evolução Molecular , GTP Fosfo-Hidrolases , Interações Hospedeiro-Patógeno/fisiologia , Legionella/patogenicidade , Legionella pneumophila/metabolismo , Macrófagos/microbiologia , Fosfatidilinositóis/metabolismo , Proteômica , Sistemas de Secreção Tipo IV/metabolismo , Vacúolos/metabolismo , Vacúolos/microbiologia
7.
Artigo em Inglês | MEDLINE | ID: mdl-28725638

RESUMO

Francisella is a gram-negative bacterial pathogen, which causes tularemia in humans and animals. A crucial step of Francisella infection is its invasion of macrophage cells. Biogenesis of the Francisella-containing phagosome (FCP) is arrested for ~15 min at the endosomal stage, followed by gradual bacterial escape into the cytosol, where the microbe proliferates. The crucial step in pathogenesis of tularemia is short and transient presence of the bacterium within phagosome. Isolation of FCPs for further studies has been challenging due to the short period of time of bacterial residence in it and the characteristics of the FCP. Here, we will for the first time present the method for isolation of the FCPs from infected human monocytes-derived macrophages (hMDMs). For elimination of lysosomal compartment these organelles were pre-loaded with dextran coated colloidal iron particles prior infection and eliminated by magnetic separation of the post-nuclear supernatant (PNS). We encountered the challenge that mitochondria has similar density to the FCP. To separate the FCP in the PNS from mitochondria, we utilized iodophenylnitrophenyltetrazolium, which is converted by the mitochondrial succinate dehydrogenase into formazan, leading to increased density of the mitochondria and allowing separation by the discontinuous sucrose density gradient ultracentrifugation. The purity of the FCP preparation and its acquisition of early endosomal markers was confirmed by Western blots, confocal and transmission electron microscopy. Our strategy to isolate highly pure FCPs from macrophages should facilitate studies on the FCP and its biogenesis.


Assuntos
Francisella/fisiologia , Monócitos/microbiologia , Fagossomos/microbiologia , Tularemia/microbiologia , Células Cultivadas , Francisella/isolamento & purificação , Humanos , Macrófagos/química , Macrófagos/microbiologia , Monócitos/química , Fagossomos/química
8.
Cell Microbiol ; 17(5): 607-20, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25363599

RESUMO

The formation and release of outer membrane vesicles (OMVs) is a phenomenon observed in many bacteria, including Legionella pneumophila. During infection, this human pathogen primarily invades alveolar macrophages and replicates within a unique membrane-bound compartment termed Legionella-containing vacuole. In the current study, we analysed the membrane architecture of L. pneumophila OMVs by small-angle X-ray scattering and biophysically characterized OMV membranes. We investigated the interaction of L. pneumophila OMVs with model membranes by Förster resonance energy transfer and Fourier transform infrared spectroscopy. These experiments demonstrated the incorporation of OMV membrane material into liposomes composed of different eukaryotic phospholipids, revealing an endogenous property of OMVs to fuse with eukaryotic membranes. Cellular co-incubation experiments showed a dose- and time-dependent binding of fluorophore-labelled OMVs to macrophages. Trypan blue quenching experiments disclosed a rapid internalization of OMVs into macrophages at 37 and 4 °C. Purified OMVs induced tumour necrosis factor-α production in human macrophages at concentrations starting at 300 ng ml(-1). Experiments on HEK293-TLR2 and TLR4/MD-2 cell lines demonstrated a dominance of TLR2-dependent signalling pathways. In summary, we demonstrate binding, internalization and biological activity of L. pneumophila OMVs on human macrophages. Our data support OMV membrane fusion as a mechanism for the remote delivery of virulence factors to host cells.


Assuntos
Membrana Celular/metabolismo , Exossomos/metabolismo , Interações Hospedeiro-Patógeno , Legionella pneumophila/fisiologia , Fatores de Virulência/metabolismo , Fenômenos Biofísicos , Células Cultivadas , Endocitose , Células Epiteliais/metabolismo , Exossomos/química , Transferência Ressonante de Energia de Fluorescência , Humanos , Legionella pneumophila/citologia , Macrófagos/imunologia , Macrófagos/metabolismo , Espalhamento a Baixo Ângulo , Espectroscopia de Infravermelho com Transformada de Fourier , Temperatura , Fator de Necrose Tumoral alfa/metabolismo
9.
Genome Biol ; 15(11): 505, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25370836

RESUMO

BACKGROUND: The genus Legionella comprises over 60 species. However, L. pneumophila and L. longbeachae alone cause over 95% of Legionnaires' disease. To identify the genetic bases underlying the different capacities to cause disease we sequenced and compared the genomes of L. micdadei, L. hackeliae and L. fallonii (LLAP10), which are all rarely isolated from humans. RESULTS: We show that these Legionella species possess different virulence capacities in amoeba and macrophages, correlating with their occurrence in humans. Our comparative analysis of 11 Legionella genomes belonging to five species reveals highly heterogeneous genome content with over 60% representing species-specific genes; these comprise a complete prophage in L. micdadei, the first ever identified in a Legionella genome. Mobile elements are abundant in Legionella genomes; many encode type IV secretion systems for conjugative transfer, pointing to their importance for adaptation of the genus. The Dot/Icm secretion system is conserved, although the core set of substrates is small, as only 24 out of over 300 described Dot/Icm effector genes are present in all Legionella species. We also identified new eukaryotic motifs including thaumatin, synaptobrevin or clathrin/coatomer adaptine like domains. CONCLUSIONS: Legionella genomes are highly dynamic due to a large mobilome mainly comprising type IV secretion systems, while a minority of core substrates is shared among the diverse species. Eukaryotic like proteins and motifs remain a hallmark of the genus Legionella. Key factors such as proteins involved in oxygen binding, iron storage, host membrane transport and certain Dot/Icm substrates are specific features of disease-related strains.


Assuntos
Genoma Bacteriano , Sequências Repetitivas Dispersas/genética , Legionella pneumophila/genética , Doença dos Legionários/genética , Amoeba/microbiologia , Sequência de Bases , Linhagem Celular , Surtos de Doenças , Sequenciamento de Nucleotídeos em Larga Escala , Humanos , Legionella pneumophila/patogenicidade , Doença dos Legionários/microbiologia , Macrófagos/microbiologia , Anotação de Sequência Molecular , Especificidade da Espécie
10.
Cell Microbiol ; 13(10): 1558-72, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-21794054

RESUMO

The Legionella virulence factor Mip (macrophage infectivity potentiator) contributes to bacterial dissemination within infected lung tissue. The Mip protein, which belongs to the enzyme family of FK506-binding proteins (FKBP), binds specifically to collagen IV. We identified a surface-exposed Mip-binding sequence in the NC1 domain of human collagen IV α1. The corresponding collagen IV-derived peptide (P290) co-precipitated with Mip and competitively inhibited the Mip-collagen IV binding. Transmigration of Legionella pneumophila across a barrier of NCI-H292 lung epithelial cells and extracellular matrix was efficiently inhibited by P290. This significantly reduced transmigration was comparable to the inefficient transmigration of PPIase-negative Mip mutant or rapamycin-treated L. pneumophila. Based on NMR data and docking studies a model for the mode of interaction of P290 and Mip was developed. The amino acids of the hydrophobic cavity of Mip, D142 and to a lesser extent Y185 were identified to be part of the interaction surface. In the complex structure of Mip(77-213) and P290, both amino acid residues form hydrogen bonds to P290. Utilizing modelling, molecular dynamics (MD) simulations and structural data of human PPIase FKBP12, the most related human orthologue of Mip, we were able to propose optimized P290 variants with increased binding specificity and selectivity for the putative bacterial drug target Mip.


Assuntos
Proteínas de Bactérias/metabolismo , Colágeno Tipo IV/metabolismo , Interações Hospedeiro-Patógeno , Legionella pneumophila/patogenicidade , Peptidilprolil Isomerase/metabolismo , Migração Transendotelial e Transepitelial , Linhagem Celular , Células Epiteliais/microbiologia , Humanos , Imunoprecipitação , Espectroscopia de Ressonância Magnética , Modelos Moleculares , Simulação de Dinâmica Molecular , Ressonância Magnética Nuclear Biomolecular , Ligação Proteica , Mapeamento de Interação de Proteínas
11.
J Biol Chem ; 285(12): 9249-61, 2010 Mar 19.
Artigo em Inglês | MEDLINE | ID: mdl-20071332

RESUMO

Dictyostelium discoideum Coronin7 (DdCRN7) together with human Coronin7 (CRN7) and Pod-1 of Drosophila melanogaster and Caenorhabditis elegans belong to the coronin family of WD-repeat domain-containing proteins. Coronin7 proteins are characterized by two WD-repeat domains that presumably fold into two beta-propeller structures. DdCRN7 shares highest homology with human CRN7, a protein with roles in membrane trafficking. DdCRN7 is present in the cytosol and accumulates in cell surface projections during movement and phago- and pinocytosis. Cells lacking CRN7 have altered chemotaxis and phagocytosis. Furthermore, loss of CRN7 affects the infection process by the pathogen Legionella pneumophila and allows a more efficient internalization of bacteria. To provide a mechanism for CNR7 action, we studied actin-related aspects. We could show that CRN7 binds directly to F-actin and protects actin filaments from depolymerization. CRN7 also associated with F-actin in vivo. It was present in the Triton X-100-insoluble cytoskeleton, colocalized with F-actin, and its distribution was sensitive to drugs affecting the actin cytoskeleton. We propose that the CRN7 role in chemotaxis and phagocytosis is through its effect on the actin cytoskeleton.


Assuntos
Actinas/química , Dictyostelium/metabolismo , Proteínas dos Microfilamentos/química , Adesão Celular , Movimento Celular , Quimiotaxia , AMP Cíclico/metabolismo , Citoesqueleto/metabolismo , Glutationa Transferase/metabolismo , Humanos , Legionella pneumophila/metabolismo , Modelos Biológicos , Mutação , Fagocitose , Pinocitose , Estrutura Terciária de Proteína
12.
Int J Med Microbiol ; 299(7): 489-508, 2009 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-19482547

RESUMO

Legionella pneumophila, the agent of Legionnaires' disease, replicates intracellularly within specialized phagosomes of human macrophages and amoebae. In this study, we have developed a protocol for the isolation of Legionella-containing phagosomes from Dictyostelium discoideum. Cell fractionation, two-dimensional gel electrophoresis and MALDI-TOF MS combined with genomic data identified 157 phagosome host proteins. In addition to proteins with an evident role in phagosome maturation, we identified proteins for which a function remains to be elucidated. Possible interactions of coronin with cytosolic NADPH oxidase components and protein kinase C inhibitors which together may lead to an inhibition of phagosomal superoxide generation are discussed. Comparative proteomics of phagosomes containing highly virulent L. pneumophila Corby versus less virulent L. hackeliae revealed distinctive protein expression patterns, e.g., an abundance of RhoGDI in L. hackeliae degrading phagosomes versus little RhoGDI in L. pneumophila Corby replicative phagosomes. We present a kinetic dissection of phagosome maturation including the complex alterations of the phagosome protein composition. A reference flow chart suggests so far unrecognized consequences of infection for host cell physiology, actin degradation on phagosomes, and a putative cysteine proteinase inhibitor interference with lysosomal enzyme sorting and activation processes.


Assuntos
Dictyostelium/química , Dictyostelium/microbiologia , Legionella/crescimento & desenvolvimento , Fagossomos/química , Fagossomos/microbiologia , Proteoma/análise , Proteínas de Protozoários/análise , Actinas/metabolismo , Animais , Fracionamento Celular , Inibidores de Cisteína Proteinase/metabolismo , Dictyostelium/fisiologia , Dictyostelium/ultraestrutura , Eletroforese em Gel Bidimensional , Genômica/métodos , Lisossomos/enzimologia , Lisossomos/metabolismo , Proteínas dos Microfilamentos/metabolismo , Microscopia Eletrônica de Transmissão , Modelos Biológicos , NADPH Oxidases/farmacologia , Fagossomos/diagnóstico por imagem , Fagossomos/metabolismo , Inibidores de Proteínas Quinases/metabolismo , Proteoma/isolamento & purificação , Proteômica/métodos , Proteínas de Protozoários/isolamento & purificação , Proteínas de Protozoários/metabolismo , Espectrometria de Massas por Ionização e Dessorção a Laser Assistida por Matriz , Superóxidos/metabolismo , Ultrassonografia
13.
Int Rev Cell Mol Biol ; 271: 253-300, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-19081545

RESUMO

Research into phagocytosis and host-pathogen interactions in the lower eukaryote Dictyostelium discoideum has flourished in recent years. This chapter presents a glimpse of where this research stands, with emphasis on the cell biology of the phagocytic process and on the wealth of molecular genetic data that have been gathered. The basic mechanistic machinery and most of the underlying genes appear to be evolutionarily conserved, reflecting the fact that phagocytosis arose as an efficient way to ingest food in single protozoan cells devoid of a rigid cell wall. In spite of some differences, the signal transduction pathways regulating phagosome biogenesis are also emerging as ultimately similar between Dictyostelium and macrophages. Both cell types are hosts for many pathogenic invasive bacteria, which exploit phagocytosis to grow intracellularly. We present an overwiew, based on the analysis of mutants, on how Dictyostelium contributes as a genetic model system to decipher the complexity of host-pathogen interactions.


Assuntos
Dictyostelium/fisiologia , Interações Hospedeiro-Patógeno/fisiologia , Macrófagos/fisiologia , Fagocitose/fisiologia , Animais
14.
Infect Immun ; 76(5): 1825-36, 2008 May.
Artigo em Inglês | MEDLINE | ID: mdl-18250176

RESUMO

Secretion of effector molecules is one of the major mechanisms by which the intracellular human pathogen Legionella pneumophila interacts with host cells during infection. Specific secretion machineries which are responsible for the subfraction of secreted proteins (soluble supernatant proteins [SSPs]) and the production of bacterial outer membrane vesicles (OMVs) both contribute to the protein composition of the extracellular milieu of this lung pathogen. Here we present comprehensive proteome reference maps for both SSPs and OMVs. Protein identification and assignment analyses revealed a total of 181 supernatant proteins, 107 of which were specific to the SSP fraction and 33 of which were specific to OMVs. A functional classification showed that a large proportion of the identified OMV proteins are involved in the pathogenesis of Legionnaires' disease. Zymography and enzyme assays demonstrated that the SSP and OMV fractions possess proteolytic and lipolytic enzyme activities which may contribute to the destruction of the alveolar lining during infection. Furthermore, it was shown that OMVs do not kill host cells but specifically modulate their cytokine response. Binding of immunofluorescently stained OMVs to alveolar epithelial cells, as visualized by confocal laser scanning microscopy, suggested that there is delivery of a large and complex group of proteins and lipids in the infected tissue in association with OMVs. On the basis of these new findings, we discuss the relevance of protein sorting and compartmentalization of virulence factors, as well as environmental aspects of the vesicle-mediated secretion.


Assuntos
Proteínas de Bactérias/análise , Legionella pneumophila/química , Legionella pneumophila/metabolismo , Proteoma/análise , Lipossomas Unilamelares/química , Fatores de Virulência/análise , Acanthamoeba castellanii/parasitologia , Animais , Linhagem Celular , Sobrevivência Celular , Citocinas/biossíntese , Eletroforese em Gel Bidimensional , Células Epiteliais/parasitologia , Humanos , Legionella pneumophila/ultraestrutura , Lipase/análise , Microscopia de Força Atômica , Microscopia Eletrônica de Transmissão , Peptídeo Hidrolases/análise , Espectrometria de Massas por Ionização e Dessorção a Laser Assistida por Matriz , Lipossomas Unilamelares/imunologia , Lipossomas Unilamelares/metabolismo
15.
Cell Microbiol ; 9(2): 450-62, 2007 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-16953800

RESUMO

Guinea pigs are highly susceptible to Legionella pneumophila infection and therefore have been the preferred animal model for studies of legionellosis. In this study guinea pig infections revealed that the Legionella virulence factor Mip (macrophage infectivity potentiator) contributes to the bacterial dissemination within the lung tissue and the spread of Legionella to the spleen. Histopathology of infected animals, binding assays with components of the extracellular matrix (ECM), bacterial transmigration experiments across an artificial lung epithelium barrier, inhibitor studies and ECM degradation assays were used to elucidate the underlying mechanism of the in vivo observation. The Mip protein, which belongs to the enzyme family of FK506-binding proteins (FKBP), was shown to bind to the ECM protein collagen (type I, II, III, IV, V, VI). Transwell assays with L. pneumophila and recombinant Escherichia coli HB101 strains revealed that Mip enables these bacteria to transmigrate across a barrier of NCI-H292 lung epithelial cells and ECM (NCI-H292/ECM barrier). Mip-specific monoclonal antibodies and the immunosuppressants rapamycin and FK506, which inhibit the peptidyl prolyl cis/trans isomerase (PPIase) activity of Mip, were able to inhibit this transmigration. By using protease inhibitors we found that the penetration of the NCI-H292/ECM barrier additionally requires a serine protease activity. Degradation assays with (35)S-labelled ECM proteins supported the finding of a concerted action of Mip and a serine protease. The described synergism between the activity of the collagen binding Mip protein and the serine protease activity represents an entirely new mechanism for bacterial penetration of the lung epithelial barrier and has implications for other prokaryotic and eukaryotic pathogens.


Assuntos
Proteínas de Bactérias/metabolismo , Células Epiteliais/microbiologia , Matriz Extracelular/microbiologia , Legionella pneumophila/fisiologia , Peptidilprolil Isomerase/metabolismo , Animais , Células Cultivadas , Cobaias , Legionella pneumophila/genética , Legionella pneumophila/patogenicidade , Pulmão/citologia , Pulmão/microbiologia
16.
FEMS Microbiol Lett ; 252(1): 19-23, 2005 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-16227110

RESUMO

Legionella pneumophila is a pathogenic bacterium found in freshwater environments that is responsible for pneumonia. People become infected through inhalation of contaminated droplets from water devices, such as cooling towers and showers. It is important to find new treatments that decrease the development of Legionella. We found a Staphylococcus warneri strain that inhibits Legionella growth. This activity is due to a molecule secreted by S. warneri. This molecule displayed a high heat-stability and its activity was lost after protease treatments, suggesting that it might be a bacteriocin. Its purification led us to conclude that this anti-Legionella molecule is an highly hydrophobic peptide. It has an original and very specific spectrum of activity, directed only toward the Legionella genus. This is the first description of an antibacterial peptide active against Legionella.


Assuntos
Bacteriocinas/metabolismo , Legionella pneumophila/efeitos dos fármacos , Peptídeos/metabolismo , Staphylococcus , Antibacterianos/farmacologia , Antibiose , Bacteriocinas/isolamento & purificação , Bacteriocinas/farmacologia , Bactérias Gram-Negativas/efeitos dos fármacos , Bactérias Gram-Positivas/efeitos dos fármacos , Humanos , Legionella pneumophila/crescimento & desenvolvimento , Testes de Sensibilidade Microbiana , Peptídeos/isolamento & purificação , Peptídeos/farmacologia , Staphylococcus/classificação , Staphylococcus/crescimento & desenvolvimento , Staphylococcus/isolamento & purificação , Staphylococcus/metabolismo
17.
Int J Med Microbiol ; 293(2-3): 133-43, 2003 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-12868650

RESUMO

Legionalla pneumophila is a human pathogen causing atypical pneumonia. It is a monopolar flagellated gram-negative bacterium. Flagellation of L. pneumophila is life cycle dependent and the expression of flagella is genetically linked to the virulence phenotype. Non-flagellated mutants of L. pneumophila are less infectious for macrophages and amoebae compared to the wild type. The flagellar operon is expressed in a hierarchical manner, and different sigma factors and transcriptional regulators are involved in this cascade of gene regulation. The genome sequence of L. pneumophila was used to identify putative regulatory elements of various flagellar operons. Preliminary reports about regulators which are involved in the link between virulence gene regulation and flagellation are discussed.


Assuntos
Regulação Bacteriana da Expressão Gênica , Legionella pneumophila/genética , Animais , Flagelos/genética , Flagelina/biossíntese , Flagelina/genética , Genes Reguladores , Genoma Bacteriano , Humanos , Legionella pneumophila/patogenicidade , Macrófagos/microbiologia , Óperon , Virulência/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA