Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Biomolecules ; 12(5)2022 04 22.
Artigo em Inglês | MEDLINE | ID: mdl-35625552

RESUMO

The environmental bacterium Legionella pneumophila is an intracellular pathogen of various protozoan hosts and able to cause Legionnaires' disease, a severe pneumonia in humans. By encoding a wide selection of virulence factors, the infectious agent possesses several strategies to manipulate its host cells and evade immune detection. In the present study, we demonstrate that the L. pneumophila zinc metalloprotease ProA functions as a modulator of flagellin-mediated TLR5 stimulation and subsequent activation of the pro-inflammatory NF-κB pathway. We found ProA to be capable of directly degrading immunogenic FlaA monomers but not the polymeric form of bacterial flagella. These results indicate a role of the protease in antagonizing immune stimulation, which was further substantiated in HEK-BlueTM hTLR5 Detection assays. Addition of purified proteins, bacterial suspensions of L. pneumophila mutant strains as well as supernatants of human lung tissue explant infection to this reporter cell line demonstrated that ProA specifically decreases the TLR5 response via FlaA degradation. Conclusively, the zinc metalloprotease ProA serves as a powerful regulator of exogenous flagellin and presumably creates an important advantage for L. pneumophila proliferation in mammalian hosts by promoting immune evasion.


Assuntos
Legionella pneumophila , Doença dos Legionários , Animais , Flagelina , Humanos , Legionella pneumophila/fisiologia , Doença dos Legionários/microbiologia , Mamíferos , Metaloproteases , Receptor 5 Toll-Like/genética , Zinco/farmacologia
2.
Cell Microbiol ; 23(5): e13313, 2021 05.
Artigo em Inglês | MEDLINE | ID: mdl-33491325

RESUMO

ProA is a secreted zinc metalloprotease of Legionella pneumophila causing lung damage in animal models of Legionnaires' disease. Here we demonstrate that ProA promotes infection of human lung tissue explants (HLTEs) and dissect the contribution to cell type specific replication and extracellular virulence mechanisms. For the first time, we reveal that co-incubation of HLTEs with purified ProA causes a significant increase of the alveolar septal thickness. This destruction of connective tissue fibres was further substantiated by collagen IV degradation assays. The moderate attenuation of a proA-negative mutant in A549 epithelial cells and THP-1 macrophages suggests that effects of ProA in tissue mainly result from extracellular activity. Correspondingly, ProA contributes to dissemination and serum resistance of the pathogen, which further expands the versatile substrate spectrum of this thermolysin-like protease. The crystal structure of ProA at 1.48 Å resolution showed high congruence to pseudolysin of Pseudomonas aeruginosa, but revealed deviations in flexible loops, the substrate binding pocket S1 ' and the repertoire of cofactors, by which ProA can be distinguished from respective homologues. In sum, this work specified virulence features of ProA at different organisational levels by zooming in from histopathological effects in human lung tissue to atomic details of the protease substrate determination.


Assuntos
Proteínas de Bactérias/metabolismo , Colágeno Tipo IV/metabolismo , Legionella pneumophila/enzimologia , Legionella pneumophila/patogenicidade , Pulmão/microbiologia , Metaloendopeptidases/metabolismo , Alvéolos Pulmonares/patologia , Fatores de Virulência/metabolismo , Células A549 , Proteínas de Bactérias/química , Atividade Bactericida do Sangue , Humanos , Legionella pneumophila/crescimento & desenvolvimento , Pulmão/patologia , Metaloendopeptidases/química , Proteólise , Alvéolos Pulmonares/metabolismo , Células THP-1 , Virulência , Fatores de Virulência/química
3.
J Vis Exp ; (156)2020 02 14.
Artigo em Inglês | MEDLINE | ID: mdl-32116293

RESUMO

Clostridioides difficile infection (CDI) is considered to be one of the most common healthcare-associated gastrointestinal infections in the United States. The innate immune response against C. difficile has been described, but the exact roles of neutrophils and macrophages in CDI are less understood. In the current study, Danio rerio (zebrafish) larvae are used to establish a C. difficile infection model for imaging the behavior and cooperation of these innate immune cells in vivo. To monitor C. difficile, a labeling protocol using a fluorescent dye has been established. A localized infection is achieved by microinjecting labeled C. difficile, which actively grows in the zebrafish intestinal tract and mimics the intestinal epithelial damage in CDI. However, this direct infection protocol is invasive and causes microscopic wounds, which can affect experimental results. Hence, a more noninvasive microgavage protocol is described here. The method involves delivery of C. difficile cells directly into the intestine of zebrafish larvae by intubation through the open mouth. This infection method closely mimics the natural infection route of C. difficile.


Assuntos
Clostridioides difficile/fisiologia , Peixe-Zebra/microbiologia , Animais , Clostridioides difficile/crescimento & desenvolvimento , Infecções por Clostridium/microbiologia , Modelos Animais de Doenças , Corantes Fluorescentes/química , Vida Livre de Germes , Intestinos/microbiologia , Intestinos/patologia , Larva/microbiologia , Esporos Bacterianos/fisiologia
4.
Artigo em Inglês | MEDLINE | ID: mdl-28326293

RESUMO

Legionnaires' disease is an acute fibrinopurulent pneumonia. During infection Legionella pneumophila adheres to the alveolar lining and replicates intracellularly within recruited macrophages. Here we provide a sequence and domain composition analysis of the L. pneumophila PilY1 protein, which has a high homology to PilY1 of Pseudomonas aeruginosa. PilY1 proteins of both pathogens contain a von Willebrand factor A (vWFa) and a C-terminal PilY domain. Using cellular fractionation, we assigned the L. pneumophila PilY1 as an outer membrane protein that is only expressed during the transmissive stationary growth phase. PilY1 contributes to infection of human lung tissue explants (HLTEs). A detailed analysis using THP-1 macrophages and A549 lung epithelial cells revealed that this contribution is due to multiple effects depending on host cell type. Deletion of PilY1 resulted in a lower replication rate in THP-1 macrophages but not in A549 cells. Further on, adhesion to THP-1 macrophages and A549 epithelial cells was decreased. Additionally, the invasion into non-phagocytic A549 epithelial cells was drastically reduced when PilY1 was absent. Complementation variants of a PilY1-negative mutant revealed that the C-terminal PilY domain is essential for restoring the wild type phenotype in adhesion, while the putatively mechanosensitive vWFa domain facilitates invasion into non-phagocytic cells. Since PilY1 also promotes twitching motility of L. pneumophila, we discuss the putative contribution of this newly described virulence factor for bacterial dissemination within infected lung tissue.


Assuntos
Aderência Bacteriana/genética , Proteínas de Fímbrias/genética , Legionella pneumophila/fisiologia , Doença dos Legionários/microbiologia , Pulmão/microbiologia , Proteínas de Fímbrias/química , Proteínas de Fímbrias/metabolismo , Regulação Bacteriana da Expressão Gênica , Interações Hospedeiro-Patógeno , Humanos , Doença dos Legionários/patologia , Pulmão/patologia , Mutação , Domínios Proteicos , Transporte Proteico , Fatores de Virulência/química , Fatores de Virulência/genética , Fatores de Virulência/metabolismo
5.
Int J Med Microbiol ; 306(6): 443-51, 2016 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-27288243

RESUMO

Legionella pneumophila, the causative agent of Legionnaires disease, is naturally found in aquatic habitats. The intracellular life cycle within protozoa pre-adapted the "accidental" human pathogen to also infect human professional phagocytes like alveolar macrophages. Previous studies employing the model organism Caenorhabditis elegans suggest that also nematodes might serve as a natural host for L. pneumophila. Here, we report for the first time from a natural co-habitation of L. pneumophila and environmental nematode species within biofilms of a warm water spring. In addition, we identified the protozoan species Oxytricha bifaria, Stylonychia mytilus, Ciliophrya sp. which have never been described as potential interaction partners of L. pneumophila before. Modeling and dissection of the Legionella-protozoa-nematode interaction revealed that C. elegans ruptures Legionella-infected amoebal cells and by this means incorporate the pathogen. Further infection studies revealed that the macrophage infectivity potentiator (Mip) protein of L. pneumophila, which is known to bind collagen IV during human lung infection, promotes the colonization of the intestinal tract of L4 larvae of C. elegans and negatively influences the life span of the worms. The Mip-negative L. pneumophila mutant exhibited a 32-fold reduced colonization rate of the nematodes after 48h when compared to the wild-type strain. Taken together, these studies suggest that nematodes may serve as natural hosts for L. pneumophila, promote their persistence and dissemination in the environment, and co-evolutionarily pre-adapt the pathogen for interactions with extracellular constituents of human lung tissue.


Assuntos
Biofilmes/crescimento & desenvolvimento , Cilióforos/fisiologia , Fontes Termais/microbiologia , Fontes Termais/parasitologia , Legionella/fisiologia , Nematoides/fisiologia , Animais , Cilióforos/crescimento & desenvolvimento , Cilióforos/microbiologia , Interações Hospedeiro-Parasita , Legionella/crescimento & desenvolvimento , Interações Microbianas , Nematoides/crescimento & desenvolvimento , Nematoides/microbiologia
6.
Expert Opin Ther Targets ; 20(3): 269-85, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26565670

RESUMO

INTRODUCTION: In recent years, Clostridium difficile has become the primary cause of antibiotic-associated diarrhea and pseudomembranous colitis, resulting in long and complicated hospital stays that represent a serious burden for patients as well as health care systems. Currently, conservative treatment of C. difficile infection (CDI) relies on the antibiotics vancomycin, metronidazole or fidaxomicin, or in case of multiple recurrences, fecal microbiota transplantation (FMT). AREAS COVERED: The fast-spreading, epidemic nature of this pathogen urgently necessitates the search for alternative treatment strategies as well as antibiotic targets. Accordingly, in this review, we highlight the recent findings regarding virulence associated traits of C. difficile, evaluate their potential as alternative drug targets, and present current efforts in designing inhibitory compounds, with the aim of pointing out possibilities for future treatment strategies. EXPERT OPINION: Increased attention on systematic analysis of the virulence mechanisms of C. difficile has already led to the identification of several alternative drug targets. In the future, applying state of the art 'omics' and the development of novel infection models that mimic the human gut, a highly complex ecological niche, will unveil the genomic and metabolic plasticity of this pathogen and will certainly help dealing with future challenges.


Assuntos
Antibacterianos/farmacologia , Clostridioides difficile/efeitos dos fármacos , Infecções por Clostridium/tratamento farmacológico , Animais , Clostridioides difficile/isolamento & purificação , Clostridioides difficile/patogenicidade , Infecções por Clostridium/microbiologia , Desenho de Fármacos , Hospitalização , Humanos , Tempo de Internação
7.
Biochim Biophys Acta ; 1850(10): 2096-102, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25529296

RESUMO

BACKGROUND: FK506-binding proteins (FKBPs) contain a domain with peptidyl-prolyl-cis/trans-isomerase (PPIase) activity and bind the immunosuppressive drugs FK506 and rapamycin. FKBPs belong to the immunophilin family and are found in eukaryotes and bacteria. SCOPE OF REVIEW: In this review we describe two major groups of bacterial virulence-associated FKBPs, the trigger factor and Mip-like PPIases. Moreover, we discuss the contribution of host FKBPs in bacterial infection processes. MAJOR CONCLUSIONS: Since PPIases are regarded as alternative antiinfective drug targets we highlight current research strategies utilizing pipecolinic acid and cycloheximide derivatives as well as substrate based inhibitors. GENERAL SIGNIFICANCE: The current research strategies suggest a beneficial synergism of drug development and basic research. This article is part of a Special Issue entitled Proline-directed Foldases: Cell Signaling Catalysts and Drug Targets.


Assuntos
Bactérias/enzimologia , Infecções Bacterianas/enzimologia , Proteínas de Bactérias/metabolismo , Peptidilprolil Isomerase/metabolismo , Proteínas de Ligação a Tacrolimo/metabolismo , Fatores de Virulência/metabolismo , Animais , Bactérias/genética , Bactérias/patogenicidade , Infecções Bacterianas/tratamento farmacológico , Infecções Bacterianas/genética , Proteínas de Bactérias/química , Proteínas de Bactérias/genética , Cicloeximida/análogos & derivados , Cicloeximida/uso terapêutico , Humanos , Peptidilprolil Isomerase/química , Peptidilprolil Isomerase/genética , Sirolimo/química , Tacrolimo/química , Proteínas de Ligação a Tacrolimo/química , Proteínas de Ligação a Tacrolimo/genética , Fatores de Virulência/química , Fatores de Virulência/genética
8.
Biochem Soc Trans ; 42(6): 1728-33, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25399597

RESUMO

Legionella pneumophila, typically a parasite of free-living protozoa, can also replicate in human alveolar macrophages and lung epithelial cells causing Legionnaires' disease in humans, a severe atypical pneumonia. The pathogen encodes six peptidylprolyl cis-trans isomerases (PPIases), which generally accelerate folding of prolyl peptide bonds, and influence protein folding. PPIases can be divided into three classes, cyclophilins, parvulins and FK506-binding proteins (FKBPs). They contribute to a multitude of cellular functions including bacterial virulence. In the present review, we provide an overview of L. pneumophila PPIases, discussing their known and anticipated functions as well as moonlighting phenomena. By taking the example of the macrophage infectivity potentiator (Mip) of L. pneumophila, we highlight the potential of PPIases as promising drug targets.


Assuntos
Legionella pneumophila/enzimologia , Peptidilprolil Isomerase/metabolismo , Virulência , Antibacterianos/farmacologia , Sítios de Ligação , Ciclofilinas/metabolismo , Legionella pneumophila/efeitos dos fármacos , Legionella pneumophila/patogenicidade
9.
Microbiol Mol Biol Rev ; 78(3): 544-71, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-25184565

RESUMO

Initially discovered in the context of immunomodulation, peptidyl-prolyl cis/trans isomerases (PPIases) were soon identified as enzymes catalyzing the rate-limiting protein folding step at peptidyl bonds preceding proline residues. Intense searches revealed that PPIases are a superfamily of proteins consisting of three structurally distinguishable families with representatives in every described species of prokaryote and eukaryote and, recently, even in some giant viruses. Despite the clear-cut enzymatic activity and ubiquitous distribution of PPIases, reports on solely PPIase-dependent biological roles remain scarce. Nevertheless, they have been found to be involved in a plethora of biological processes, such as gene expression, signal transduction, protein secretion, development, and tissue regeneration, underscoring their general importance. Hence, it is not surprising that PPIases have also been identified as virulence-associated proteins. The extent of contribution to virulence is highly variable and dependent on the pleiotropic roles of a single PPIase in the respective pathogen. The main objective of this review is to discuss this variety in virulence-related bacterial and protozoan PPIases as well as the involvement of host PPIases in infectious processes. Moreover, a special focus is given to Legionella pneumophila macrophage infectivity potentiator (Mip) and Mip-like PPIases of other pathogens, as the best-characterized virulence-related representatives of this family. Finally, the potential of PPIases as alternative drug targets and first tangible results are highlighted.


Assuntos
Peptidilprolil Isomerase/genética , Fatores de Virulência/genética , Animais , Antibacterianos/farmacologia , Antiparasitários/farmacologia , Infecções Bacterianas/tratamento farmacológico , Infecções Bacterianas/microbiologia , Proteínas de Bactérias/antagonistas & inibidores , Proteínas de Bactérias/genética , Inibidores Enzimáticos/farmacologia , Humanos , Doenças Parasitárias/tratamento farmacológico , Doenças Parasitárias/parasitologia , Peptidilprolil Isomerase/antagonistas & inibidores , Proteínas de Protozoários/antagonistas & inibidores , Proteínas de Protozoários/genética , Fatores de Virulência/antagonistas & inibidores
10.
Int J Med Microbiol ; 302(6): 261-9, 2012 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-22954413

RESUMO

Non-typeable Haemophilus influenzae (NTHi) is a common pathogen associated with diseases such as acute otitis media or exacerbations in patients with chronic obstructive pulmonary disease. The biofilm-forming capability substantially contributes to the persistence of NTHi. However, the regulation of biofilm formation is not completely understood. Quorum sensing regulated by autoinducer-2 produced by luxS is until now the only described regulatory mechanism. In this study, we show that the two-component signalling system QseB/C is involved in the biofilm formation of NTHi in vitro. An isogenic NTHi mutant of qseC (Hi3655KR2) showed a significant decrease in biofilm formation under static and semi-static conditions as assessed by crystal violet staining. In addition, under constant flow conditions, Hi3655KR2 formed less biofilm after 48 h. The biofilm defects were irrespective of autoinducer-2 levels. Hence, here we suggest for the first time a regulatory circuit in NTHi, which controls biofilm formation by mechanisms other than or in addition to luxS-dependent factors.


Assuntos
Proteínas de Bactérias/metabolismo , Biofilmes , Haemophilus influenzae/fisiologia , Homosserina/análogos & derivados , Lactonas/metabolismo , Percepção de Quorum , Sequência de Aminoácidos , Proteínas de Bactérias/genética , Liases de Carbono-Enxofre/genética , Liases de Carbono-Enxofre/metabolismo , Meios de Cultura/metabolismo , Escherichia coli/genética , Escherichia coli/fisiologia , Proteínas de Escherichia coli/genética , Proteínas de Escherichia coli/metabolismo , Regulação Bacteriana da Expressão Gênica , Técnicas de Inativação de Genes , Genes Bacterianos , Teste de Complementação Genética , Violeta Genciana , Haemophilus influenzae/genética , Recombinação Homóloga , Homosserina/metabolismo , Medições Luminescentes , Viabilidade Microbiana , Dados de Sequência Molecular , Estrutura Terciária de Proteína , Homologia de Sequência de Aminoácidos , Fatores de Tempo
11.
Semin Immunopathol ; 33(5): 395-408, 2011 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-21153593

RESUMO

Gram-negative bacteria have the ability to produce outer membrane-derived vesicles (OMVs) that are released into the extracellular milieu. Even though this intriguing phenomenon is well-known since many years, various aspects of bacterial OMVs are not fully described and are still in the process of being characterized in detail. One major reason for this is that depending on the bacterial species and its respective ecological niche, OMVs exhibit an enormous functional diversity. Research of the past years has clearly shown that OMVs of many pathogenic bacteria contribute to the virulence potential by enriching virulence factors and delivering them over long distances, superseding direct bacterial contact with their host. The subsequent interaction of OMVs with the host can occur at different levels regarding the type of immune response or the target cell type and may lead to different outcomes ranging from non-immunogenic activation or a pro-inflammatory response to cytotoxicity. In contrast to being virulence factors, OMVs are used for vaccination purposes in the combat against bacterial pathogens, and recent research thus is focused on to indirectly aim these versatile bacterial weapons against themselves.


Assuntos
Bactérias Gram-Negativas/metabolismo , Infecções por Bactérias Gram-Negativas/microbiologia , Infecções por Bactérias Gram-Negativas/prevenção & controle , Vesículas Transportadoras/metabolismo , Imunidade Adaptativa/imunologia , Animais , Vacinas Bacterianas/imunologia , Comunicação Celular , Células Epiteliais/imunologia , Células Epiteliais/metabolismo , Células Epiteliais/microbiologia , Bactérias Gram-Negativas/imunologia , Infecções por Bactérias Gram-Negativas/imunologia , Humanos , Imunidade Inata/imunologia , Mucosa/imunologia , Mucosa/metabolismo , Mucosa/microbiologia , Fatores de Virulência/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...