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










Base de dados
Intervalo de ano de publicação
1.
Eur J Immunol ; 49(9): 1415-1420, 2019 09.
Artigo em Inglês | MEDLINE | ID: mdl-31074841

RESUMO

Protective immunity against intracellular pathogens, including bacteria, usually relies on cellular immunity. However, antibodies are also implicated in mediating protection against intracellular bacteria. In case of airway infection with Legionella pneumophila (Lpn), the causative agent of Legionnaires' disease, pre-existing Lpn-specific antibodies were shown to afford protection within two days of infection. Here we dissected the early kinetics of Ab-mediated protection against airway Lpn infection and observed two kinetically and mechanistically distinct phases of protection by passively administered antibodies. Within the first hour of infection, Lpn-opsonizing antibodies provided almost 10-fold protection in an antibody Fc-dependent, but FcR-independent manner. Later on, by two days post infection, Lpn-specific Ab-mediated protection strictly involved FcγR, Syk kinase activity in alveolar macrophages and induction of reactive oxygen species (ROS). The findings presented here contribute to the understanding of the mechanisms of Ab-mediated control of Lpn infection in actively or passively immunized individuals.


Assuntos
Anticorpos Antibacterianos/imunologia , Legionella pneumophila/imunologia , Doença dos Legionários/imunologia , Animais , Antígenos de Bactérias/imunologia , Proteínas de Bactérias/imunologia , Imunidade Celular/imunologia , Imunização Passiva/métodos , Doença dos Legionários/microbiologia , Macrófagos Alveolares/imunologia , Macrófagos Alveolares/microbiologia , Camundongos , Camundongos Endogâmicos C57BL , Espécies Reativas de Oxigênio/imunologia
2.
Sci Rep ; 6: 18109, 2016 Jan 05.
Artigo em Inglês | MEDLINE | ID: mdl-26728082

RESUMO

To survive antibiotics, bacteria use two different strategies: counteracting antibiotic effects by expression of resistance genes or evading their effects e.g. by persisting inside host cells. Since bacterial adhesins provide access to the shielded, intracellular niche and the adhesin type 1 fimbriae increases bacterial survival chances inside macrophages, we asked if fimbriae also influenced survival by antibiotic evasion. Combined gentamicin survival assays, flow cytometry, single cell microscopy and kinetic modeling of dose response curves showed that type 1 fimbriae increased the adhesion and internalization by macrophages. This was caused by strongly decreased off-rates and affected the number of intracellular bacteria but not the macrophage viability and morphology. Fimbriae thus promote antibiotic evasion which is particularly relevant in the context of chronic infections.


Assuntos
Antibacterianos/farmacologia , Escherichia coli/efeitos dos fármacos , Escherichia coli/fisiologia , Fímbrias Bacterianas , Animais , Aderência Bacteriana , Linhagem Celular , Sobrevivência Celular , Farmacorresistência Bacteriana , Regulação Bacteriana da Expressão Gênica , Lisossomos/imunologia , Lisossomos/microbiologia , Macrófagos/imunologia , Macrófagos/metabolismo , Macrófagos/microbiologia , Camundongos , Viabilidade Microbiana/efeitos dos fármacos , Viabilidade Microbiana/imunologia , Fagocitose , Fenótipo
3.
J Immunol ; 193(8): 4053-9, 2014 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-25210123

RESUMO

Abs are able to mediate local protection from pulmonary infection with Legionella pneumophila, the causative agent of a severe form of pneumonia known as Legionnaires' disease. L. pneumophila is able to infect alveolar macrophages in the lung and replicates intracellularly in a vacuolar compartment with endoplasmic reticulum-like characteristics. However, Abs opsonize the bacteria and confer an FcR-mediated signal to phagocytic host cells that vetoes the bacterial evasion strategies, thereby efficiently targeting the bacteria to intracellular lysosomal degradation. In this study we analyzed the prevalence of pathogen-specific IgG subclasses present in immunized mice and found that the presence of IgG2c and IgG3 correlated with reduced bacterial titers after intranasal infection. We then isolated different IgG subclasses and compared their differential prophylactic potential in restricting airway L. pneumophila replication. We found that all IgG subclasses were effective in restricting pulmonary airway infection in mice when administered at high and equivalent doses. However, at limiting Ab concentrations we found a superior role of IgG2c in restricting L. pneumophila replication in a prophylactic setting. Furthermore, we assessed the therapeutic efficacy of administering an mAb during an established infection and found that bacterial titers could be reduced very efficiently with such a treatment. Thus, we propose the therapeutic use of Abs for the treatment of intracellular bacterial infections in situations where antibiotics might be ineffective.


Assuntos
Anticorpos Antibacterianos/uso terapêutico , Anticorpos Monoclonais/uso terapêutico , Imunoglobulina G/uso terapêutico , Legionella pneumophila , Doença dos Legionários/terapia , Animais , Anticorpos Antibacterianos/administração & dosagem , Anticorpos Antibacterianos/imunologia , Anticorpos Monoclonais/administração & dosagem , Anticorpos Monoclonais/imunologia , Células Cultivadas , Células HEK293 , Humanos , Imunoglobulina G/classificação , Imunoglobulina G/imunologia , Imunoterapia/métodos , Doença dos Legionários/imunologia , Doença dos Legionários/microbiologia , Lipopolissacarídeos/imunologia , Macrófagos Alveolares/imunologia , Macrófagos Alveolares/microbiologia , Camundongos , Camundongos Endogâmicos C57BL
4.
Methods Mol Biol ; 954: 265-77, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23150402

RESUMO

Upon uptake into a host cell, the intracellular bacterium Legionella pneumophila is not degraded on the lysosomal pathway but efficiently establishes a highly specialized replicative vacuole in which it readily multiplies. As many Icm/Dot type 4 secretion translocated bacterial effectors contribute to the establishment of this subcellular compartment in close interaction with host cell trafficking pathways, the analysis of the intracellular localization of this bacterium during infection is of pivotal importance to dissect the cellular and bacterial components of this process. In this chapter we describe a protocol for immunofluorescence microscopy in fixed mammalian and amoebal cells as well as transfection protocols to produce host cells expressing fluorescently labeled proteins as intracellular trafficking markers.


Assuntos
Imunofluorescência/métodos , Legionella pneumophila/metabolismo , Microscopia de Fluorescência , Fagócitos/microbiologia , Animais , Proteínas de Bactérias/metabolismo , Técnicas de Cultura de Células , Linhagem Celular , Fagócitos/ultraestrutura , Transfecção/métodos
5.
Methods Mol Biol ; 954: 505-20, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23150417

RESUMO

Legionella pneumophila is the causative agent of the potentially fatal Legionnaires' disease in humans. Mice have proved to be valuable model organisms to study the pathogenesis of this intracellular bacterium, as well as immune responses against it. In this chapter we describe a selection of mouse infection protocols to study the innate and adaptive immune responses raised after an infection with Legionella. Included are protocols for systemic and pulmonary infections, surgical collection of organs as well as determination of cell composition, cytokines, and antibody titers therein. Furthermore, we describe an immunohistology protocol to analyze lung tissue sections by fluorescence microscopy.


Assuntos
Legionella pneumophila/imunologia , Doença dos Legionários/imunologia , Animais , Líquido da Lavagem Broncoalveolar/citologia , Líquido da Lavagem Broncoalveolar/imunologia , Citocinas/imunologia , Citocinas/metabolismo , Modelos Animais de Doenças , Ensaio de Imunoadsorção Enzimática , Humanos , Doença dos Legionários/microbiologia , Pulmão/imunologia , Pulmão/patologia , Linfonodos/imunologia , Linfonodos/patologia , Camundongos , Baço/imunologia , Baço/patologia
6.
Fungal Genet Biol ; 49(11): 915-21, 2012 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-23010151

RESUMO

The filamentous fungus Penicillium chrysogenum is used for the industrial production of ß-lactam antibiotics. The pathway for ß-lactam biosynthesis has been resolved and involves the enzyme phenylacetic acid CoA ligase that is responsible for the CoA activation of the side chain precursor phenylacetic acid (PAA) that is used for the biosynthesis of penicillin G. To identify ABC transporters related to ß-lactam biosynthesis, we analyzed the expression of all 48 ABC transporters present in the genome of P. chryso-genum when grown in the presence and absence of PAA. ABC40 is significantly upregulated when cells are grown or exposed to high levels of PAA. Although deletion of this transporter did not affect ß-lactam biosynthesis, it resulted in a significant increase in sensitivity to PAA and other weak acids. It is concluded that ABC40 is involved in weak acid detoxification in P. chrysogenum including resistance to phenylacetic acid.


Assuntos
Transportadores de Cassetes de Ligação de ATP/sangue , Transportadores de Cassetes de Ligação de ATP/metabolismo , Proteínas Fúngicas/metabolismo , Penicillium chrysogenum/metabolismo , Fenilacetatos/metabolismo , Transportadores de Cassetes de Ligação de ATP/genética , Transporte Biológico , Proteínas Fúngicas/genética , Regulação Fúngica da Expressão Gênica , Penicillium chrysogenum/genética , Regulação para Cima , beta-Lactamas/metabolismo
7.
Appl Environ Microbiol ; 78(19): 7107-13, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22865068

RESUMO

Intense classical strain improvement has yielded industrial Penicillium chrysogenum strains that produce high titers of penicillin. These strains contain multiple copies of the penicillin biosynthesis cluster encoding the three key enzymes: δ-(l-α-aminoadipyl)-L-cysteinyl-D-valine synthetase (ACVS), isopenicillin N synthase (IPNS), and isopenicillin N acyltransferase (IAT). The phenylacetic acid coenzyme A (CoA) ligase (PCL) gene encoding the enzyme responsible for the activation of the side chain precursor phenylacetic acid is localized elsewhere in the genome in a single copy. Since the protein level of IAT already saturates at low cluster copy numbers, IAT might catalyze a limiting step in high-yielding strains. Here, we show that penicillin production in high-yielding strains can be further improved by the overexpression of IAT while at very high levels of IAT the precursor 6-aminopenicillic acid (6-APA) accumulates. Overproduction of PCL only marginally stimulates penicillin production. These data demonstrate that in high-yielding strains IAT is the limiting factor and that this limitation can be alleviated by a balanced overproduction of this enzyme.


Assuntos
Aciltransferases/biossíntese , Aciltransferases/genética , Dosagem de Genes , Engenharia Metabólica , Proteínas de Ligação às Penicilinas/biossíntese , Proteínas de Ligação às Penicilinas/genética , Penicilinas/biossíntese , Penicillium chrysogenum/genética , Penicillium chrysogenum/metabolismo , Expressão Gênica , Família Multigênica
8.
J Immunol ; 189(2): 841-9, 2012 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-22696443

RESUMO

Abs confer protection from secondary infection with Legionella pneumophila, the causative agent of a severe form of pneumonia known as Legionnaires' disease. In this study, we demonstrate that Ab-mediated protection is effective across L. pneumophila serogroups, suggesting that Abs specific for conserved protein Ags are sufficient to mediate this protective effect. We used two independent methods to identify immunogenic L. pneumophila protein Ags, namely, the screening of a λ phage library representing the complete L. pneumophila genome and two-dimensional gel electrophoresis combined with Western blot analysis and protein spot identification by mass spectrometry. A total of 30 novel L. pneumophila B cell Ags were identified, the majority of which are located in or associated with the bacterial membrane, where they are accessible for Abs and, therefore, likely to be relevant for Ab-mediated protection against L. pneumophila. Selected B cell Ags were recombinantly expressed and tested in a vaccination protocol. Mice immunized with either single-protein Ags or an Ag combination showed reduced bacterial titers in bronchoalveolar lavage and lung after L. pneumophila challenge. To determine the clinical relevance of these findings, we tested Legionnaires' disease patient sera for reactivity with the identified L. pneumophila Ags. The recognized Ags were indeed conserved across host species, because Abs specific for all three selected Ags could be detected in patient sera, rendering the identified protein Ags potential vaccine candidates.


Assuntos
Antígenos de Bactérias/isolamento & purificação , Subpopulações de Linfócitos B/imunologia , Subpopulações de Linfócitos B/microbiologia , Legionella pneumophila/imunologia , Doença dos Legionários/imunologia , Administração Intranasal , Animais , Anticorpos Antibacterianos/biossíntese , Anticorpos Antibacterianos/sangue , Anticorpos Antibacterianos/uso terapêutico , Antígenos de Bactérias/administração & dosagem , Antígenos de Bactérias/imunologia , Subpopulações de Linfócitos B/metabolismo , Vacinas Bacterianas/administração & dosagem , Vacinas Bacterianas/síntese química , Vacinas Bacterianas/imunologia , Bacteriófago lambda/genética , Bacteriófago lambda/imunologia , Sequência Conservada/imunologia , Humanos , Imunoglobulina G/biossíntese , Imunoglobulina G/sangue , Legionella pneumophila/patogenicidade , Doença dos Legionários/sangue , Doença dos Legionários/prevenção & controle , Camundongos , Camundongos Endogâmicos A , Camundongos Endogâmicos C57BL , Vacinas Sintéticas/administração & dosagem , Vacinas Sintéticas/imunologia , Vacinas Sintéticas/uso terapêutico
9.
Methods Mol Biol ; 835: 1-16, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22183644

RESUMO

Fungal cells are highly complex as their metabolism is compartmentalized harboring various types of subcellular organelles that are bordered by one or more membranes. Knowledge about the intracellular localization of transporter proteins is often required for the understanding of their biological function. Among different approaches available, the localization analysis based on the expression of GFP fusions is commonly used as a relatively fast and cost-efficient method that allows visualization of proteins of interest in both live and fixed cells. In addition, inactivation of transporter genes is an important tool to resolve their specific function. Here we provide a detailed protocol for the deletion and localization analysis of ABC transporters in the filamentous fungus Penicillium chrysogenum. It includes construction of expression plasmids, their transformation into fungal strains, cultivation of transformants, microscopy analysis, as well as additional protocols on staining of fungal cells with organelle-specific dyes like Hoechst 33342, MitoTracker DeepRed, and FM4-64.


Assuntos
Transportadores de Cassetes de Ligação de ATP/análise , Transportadores de Cassetes de Ligação de ATP/genética , Proteínas Fúngicas/análise , Proteínas Fúngicas/genética , Penicillium chrysogenum/citologia , Penicillium chrysogenum/genética , Deleção de Genes , Proteínas de Fluorescência Verde/análise , Proteínas de Fluorescência Verde/genética , Microscopia de Fluorescência/métodos , Plasmídeos/genética , Coloração e Rotulagem/métodos , Transformação Genética
10.
Biotechnol J ; 7(2): 225-36, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-22057844

RESUMO

Industrial production of ß-lactam antibiotics by the filamentous fungus Penicillium chrysogenum is based on successive classical strain improvement cycles. This review summarizes our current knowledge on the results of this classical strain improvement process, and discusses avenues to improve ß-lactam biosynthesis and to exploit P. chrysogenum as an industrial host for the production of other antibiotics and peptide products. Genomic and transcriptional analysis of strain lineages has led to the identification of several important alterations in high-yielding strains, including the amplification of the penicillin biosynthetic gene cluster, elevated transcription of genes involved in biosynthesis of penicillin and amino acid precursors, and genes encoding microbody proliferation factors. In recent years, successful metabolic engineering and synthetic biology approaches have resulted in the redirection of the penicillin pathway towards the production of cephalosporins. This sets a new direction in industrial antibiotics productions towards more sustainable methods for the fermentative production of unnatural antibiotics and related compounds.


Assuntos
Penicillium chrysogenum/genética , Penicillium chrysogenum/metabolismo , beta-Lactamas/metabolismo , Engenharia Metabólica/métodos , Biologia Sintética/métodos
11.
Eur J Immunol ; 41(4): 889-97, 2011 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-21413006

RESUMO

Intracellular pathogen-specific antibodies (Abs) can contribute to host protection by a number of different mechanisms. Ab opsonization of pathogens residing outside a host cell can prevent infection of target cells either via neutralization of the critical surface epitopes required for host cell entry, complement-mediated degradation, or via subsequent intracellular degradation. In the case of intracellular localization, Abs can bind to infected cells and thus mark them for destruction by Fc receptor (FcR)-bearing effector cells. This review focuses on the protective role of Abs against intracellular bacteria and parasites involving FcR interactions that modulate the intracellular trafficking of the pathogen, the ability of FcRs to interfere with the establishment of an intracellular replicative niche and the involvement of FcRs to modulate pathogen-specific T-cell responses.


Assuntos
Anticorpos/imunologia , Espaço Intracelular/imunologia , Receptores Fc/imunologia , Animais , Humanos , Lisossomos/imunologia , Linfócitos T/imunologia
12.
Proc Natl Acad Sci U S A ; 107(47): 20441-6, 2010 Nov 23.
Artigo em Inglês | MEDLINE | ID: mdl-21048081

RESUMO

The protective effect of antibodies (Abs) is generally attributed to neutralization or complement activation. Using Legionella pneumophila and Mycobacterium bovis bacillus Calmette-Guérin as a model, we discovered an additional mechanism of Ab-mediated protection effective against intracellular pathogens that normally evade lysosomal fusion. We show that Fc receptor (FcR) engagement by Abs, which can be temporally and spatially separated from bacterial infection, renders the host cell nonpermissive for bacterial replication and targets the pathogens to lysosomes. This process is strictly dependent on kinases involved in FcR signaling but not on host cell protein synthesis or protease activation. Based on these findings, we propose a mechanism whereby Abs and FcR engagement subverts the strategies by which intracellular bacterial pathogens evade lysosomal degradation.


Assuntos
Anticorpos Antibacterianos/imunologia , Infecções Bacterianas/imunologia , Lisossomos/imunologia , Receptores Fc/imunologia , Transdução de Sinais/imunologia , Animais , Carga Bacteriana , Imunização , Legionella pneumophila , Camundongos , Microscopia de Fluorescência , Mycobacterium bovis
13.
Mol Microbiol ; 71(6): 1341-52, 2009 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-19208094

RESUMO

Phosphoinositide (PI) glycerolipids are key regulators of eukaryotic signal transduction, cytoskeleton architecture and membrane dynamics. The host cell PI metabolism is targeted by intracellular bacterial pathogens, which evolved intricate strategies to modulate uptake processes and vesicle trafficking pathways. Upon entering eukaryotic host cells, pathogenic bacteria replicate in distinct vacuoles or in the host cytoplasm. Vacuolar pathogens manipulate PI levels to mimic or modify membranes of subcellular compartments and thereby establish their replicative niche. Legionella pneumophila, Brucella abortus, Mycobacterium tuberculosis and Salmonella enterica translocate effector proteins into the host cell, some of which anchor to the vacuolar membrane via PIs or enzymatically turnover PIs. Cytoplasmic pathogens target PI metabolism at the plasma membrane, thus modulating their uptake and antiapoptotic signalling pathways. Employing this strategy, Shigella flexneri directly injects a PI-modifying effector protein, while Listeria monocytogenes exploits PI metabolism indirectly by binding to transmembrane receptors. Thus, regardless of the intracellular lifestyle of the pathogen, PI metabolism is critically involved in the interactions with host cells.


Assuntos
Bactérias/patogenicidade , Fosfatidilinositóis/metabolismo , Vacúolos/microbiologia , Bactérias/metabolismo , Fagossomos/microbiologia , Fosfatidilinositol 3-Quinases/metabolismo , Monoéster Fosfórico Hidrolases/metabolismo , Transdução de Sinais
14.
J Biol Chem ; 284(8): 4846-56, 2009 Feb 20.
Artigo em Inglês | MEDLINE | ID: mdl-19095644

RESUMO

The causative agent of Legionnaires disease, Legionella pneumophila, forms a replicative vacuole in phagocytes by means of the intracellular multiplication/defective organelle trafficking (Icm/Dot) type IV secretion system and translocated effector proteins, some of which subvert host GTP and phosphoinositide (PI) metabolism. The Icm/Dot substrate SidC anchors to the membrane of Legionella-containing vacuoles (LCVs) by specifically binding to phosphatidylinositol 4-phosphate (PtdIns(4)P). Using a nonbiased screen for novel L. pneumophila PI-binding proteins, we identified the Rab1 guanine nucleotide exchange factor (GEF) SidM/DrrA as the predominant PtdIns(4)P-binding protein. Purified SidM specifically and directly bound to PtdIns(4)P, whereas the SidM-interacting Icm/Dot substrate LidA preferentially bound PtdIns(3)P but also PtdIns(4)P, and the L. pneumophila Arf1 GEF RalF did not bind to any PIs. The PtdIns(4)P-binding domain of SidM was mapped to the 12-kDa C-terminal sequence, termed "P4M" (PtdIns4P binding of SidM/DrrA). The isolated P4M domain is largely helical and displayed higher PtdIns(4)P binding activity in the context of the alpha-helical, monomeric full-length protein. SidM constructs containing P4M were translocated by Icm/Dot-proficient L. pneumophila and localized to the LCV membrane, indicating that SidM anchors to PtdIns(4)P on LCVs via its P4M domain. An L. pneumophila DeltasidM mutant strain displayed significantly higher amounts of SidC on LCVs, suggesting that SidM and SidC compete for limiting amounts of PtdIns(4)P on the vacuole. Finally, RNA interference revealed that PtdIns(4)P on LCVs is specifically formed by host PtdIns 4-kinase IIIbeta. Thus, L. pneumophila exploits PtdIns(4)P produced by PtdIns 4-kinase IIIbeta to anchor the effectors SidC and SidM to LCVs.


Assuntos
Proteínas de Bactérias/química , Proteínas de Transporte/química , Fatores de Troca do Nucleotídeo Guanina/química , Legionella pneumophila/química , Fosfatos de Fosfatidilinositol/química , Proteínas rab1 de Ligação ao GTP/química , Animais , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Proteínas de Transporte/genética , Proteínas de Transporte/metabolismo , Linhagem Celular , Drosophila , Fatores de Troca do Nucleotídeo Guanina/genética , Fatores de Troca do Nucleotídeo Guanina/metabolismo , Humanos , Legionella pneumophila/genética , Legionella pneumophila/metabolismo , Legionella pneumophila/patogenicidade , Doença dos Legionários/genética , Doença dos Legionários/metabolismo , Mutação , Mapeamento de Peptídeos , Fagócitos/metabolismo , Fagócitos/microbiologia , Fosfatos de Fosfatidilinositol/genética , Fosfatos de Fosfatidilinositol/metabolismo , Ligação Proteica/fisiologia , Estrutura Terciária de Proteína/fisiologia , Vacúolos/genética , Vacúolos/metabolismo , Vacúolos/microbiologia , Proteínas rab1 de Ligação ao GTP/genética , Proteínas rab1 de Ligação ao GTP/metabolismo
15.
Cell Microbiol ; 11(3): 442-60, 2009 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-19021631

RESUMO

Legionella pneumophila, the causative agent of Legionnaires' disease, replicates within a specific vacuole in amoebae and macrophages. To form these 'Legionella-containing vacuoles' (LCVs), the bacteria employ the Icm/Dot type IV secretion system and effector proteins, some of which anchor to the LCV membrane via the host glycolipid phosphatidylinositol 4-phosphate [PtdIns(4)P]. Here we analysed the role of inositol polyphosphate 5-phosphatases (IP5Ps) during L. pneumophila infections. Bacterial replication and LCV formation occurred more efficiently in Dictyostelium discoideum amoebae lacking the IP5P Dd5P4, a homologue of human OCRL1 (Oculocerebrorenal syndrome of Lowe), implicated in retrograde endosome to Golgi trafficking. The phenotype was complemented by Dd5P4 but not the catalytically inactive 5-phosphatase. Ectopically expressed Dd5P4 or OCRL1 localized to LCVs in D. discoideum via an N-terminal domain previously not implicated in membrane targeting, and OCRL1 was also identified on LCVs in macrophages. Dd5P4 was catalytically active on LCVs and accumulated on LCVs harbouring wild-type but not DeltaicmT mutant L. pneumophila. The N-terminal domain of OCRL1 bound L. pneumophila LpnE, a Sel1-like repeat protein involved in LCV formation, which localizes to LCVs and selectively binds PtdIns(3)P. Our results indicate that OCRL1 restricts intracellular growth of L. pneumophila and binds to LCVs in association with LpnE.


Assuntos
Proteínas de Bactérias/metabolismo , Dictyostelium/enzimologia , Dictyostelium/microbiologia , Legionella pneumophila/crescimento & desenvolvimento , Monoéster Fosfórico Hidrolases/metabolismo , Vacúolos/microbiologia , Fatores de Virulência/metabolismo , Animais , Linhagem Celular , Deleção de Genes , Teste de Complementação Genética , Inositol Polifosfato 5-Fosfatases , Macrófagos/enzimologia , Macrófagos/microbiologia , Camundongos , Monoéster Fosfórico Hidrolases/análise , Monoéster Fosfórico Hidrolases/genética , Ligação Proteica , Vacúolos/química
16.
Cell Microbiol ; 10(12): 2416-33, 2008 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-18673369

RESUMO

Legionella pneumophila, the causative agent of Legionnaires' disease, uses the intracellular multiplication/defective organelle trafficking (Icm/Dot) type IV secretion system to establish within amoebae and macrophages an endoplasmic reticulum (ER)-derived replication-permissive compartment, the Legionella-containing vacuole (LCV). The Icm/Dot substrate SidC and its paralogue SdcA anchor to LCVs via phosphatidylinositol-4 phosphate [PtdIns(4)P]. Here we identify the unique 20 kDa PtdIns(4)P-binding domain of SidC, which upon heterologous expression in Dictyostelium binds to LCVs and thus is useful as a PtdIns(4)P-specific probe. LCVs harbouring L. pneumophilaDeltasidC-sdcA mutant bacteria recruit ER and ER-derived vesicles less efficiently and carry endosomal but not lysosomal markers. The phenotypes are complemented by supplying sidC on a plasmid. L. pneumophilaDeltasidC-sdcA grows at wild-type rate in calnexin-negative LCVs, suggesting that communication with the ER is dispensable for establishing a replicative compartment. The amount of SidC and calnexin is directly proportional on isolated LCVs, and in a cell-free system, the recruitment of calnexin-positive vesicles to LCVs harbouring DeltasidC-sdcA mutant bacteria is impaired. Beads coated with purified SidC or its 70 kDa N-terminal fragment recruit ER vesicles in Dictyostelium and macrophage lysates. Our results establish SidC as an L. pneumophila effector protein, which anchors to PtdIns(4)P on LCVs and recruits ER vesicles to a replication-permissive vacuole.


Assuntos
Proteínas de Bactérias/metabolismo , Vesículas Citoplasmáticas/metabolismo , Vesículas Citoplasmáticas/microbiologia , Legionella pneumophila/patogenicidade , Fatores de Virulência/metabolismo , Proteínas de Bactérias/genética , Calnexina/análise , Vesículas Citoplasmáticas/química , Vesículas Citoplasmáticas/ultraestrutura , Deleção de Genes , Teste de Complementação Genética , Humanos , Microscopia Confocal , Microscopia Eletrônica de Transmissão , Fatores de Virulência/genética
17.
Cell Microbiol ; 9(12): 2903-20, 2007 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-17614967

RESUMO

Legionella pneumophila is an opportunistic human pathogen that replicates within environmental amoebae including Acanthamoeba castellanii and Dictyostelium discoideum. The Icm/Dot type IV secretion system promotes phagocytosis and intracellular replication of L. pneumophila in an endoplasmic reticulum-derived 'Legionella-containing vacuole' (LCV). L. pneumophila adopts a biphasic life cycle consisting of a replicative growth phase and a transmissive (stationary) phase, the latter of which is characterized by the preferential expression of genes required for motility and virulence. A bioinformatic analysis of the L. pneumophila genome revealed a gene cluster homologous to the Vibrio cholerae cqsAS genes, encoding a putative quorum sensing autoinducer synthase (lqsA) and a sensor kinase (lqsS), which flank a novel response regulator (lqsR). We report here that an L. pneumophila lqsR deletion mutant grew in broth with the same rate as wild-type bacteria, but entered the replicative growth phase earlier. Overexpression of lqsR led to an elongated morphology of the bacteria. The lqsR mutant strain was found to be more salt-resistant and impaired for intracellular growth in A. castellanii, D. discoideum and macrophages, formation of the ER-derived LCV and toxicity. Moreover, L. pneumophila lacking LqsR, as well as strains lacking the stationary sigma factor RpoS or the two-component response regulator LetA, were phagocytosed less efficiently by A. castellanii, D. discoideum or macrophages. The expression of lqsR was dependent on RpoS and, to a lesser extent, also on LetA. DNA microarray experiments revealed that lqsR regulates the expression of genes involved in virulence, motility and cell division, consistent with a role for LqsR in the transition from the replicative to the transmissive (virulent) phase. Our findings indicate that LqsR is a novel pleiotropic regulator involved in RpoS- and LetA-controlled interactions of L. pneumophila with phagocytes.


Assuntos
Proteínas de Bactérias/fisiologia , Legionella pneumophila/crescimento & desenvolvimento , Fator sigma/fisiologia , Fatores de Transcrição/fisiologia , Acanthamoeba castellanii/microbiologia , Animais , Divisão Celular/genética , Linhagem Celular , Dictyostelium/microbiologia , Deleção de Genes , Dosagem de Genes , Perfilação da Expressão Gênica , Regulação Bacteriana da Expressão Gênica , Humanos , Legionella pneumophila/citologia , Legionella pneumophila/genética , Locomoção/genética , Macrófagos/microbiologia , Análise de Sequência com Séries de Oligonucleotídeos , Fatores de Transcrição/genética , Virulência
18.
Environ Microbiol ; 9(3): 563-75, 2007 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-17298357

RESUMO

Environmental bacteria are constantly threatened by bacterivorous predators such as free-living protozoa and nematodes. In the course of their coevolution with environmental predators, some bacteria developed sophisticated defence mechanisms, including the secretion of toxins, or the capacity to avoid lysosomal killing and to replicate intracellularly within protozoa. To analyse the interactions with bacterial pathogens on a molecular, cellular or organismic level, protozoa and other non-mammalian hosts are increasingly used. These include amoebae, as well as genetically tractable hosts, such as the social amoeba Dictyostelium discoideum, the nematode Caenorhabditis elegans and the fruit fly Drosophila melanogaster. Using these hosts, the virulence mechanisms of opportunistic pathogenic bacteria such as Legionella, Mycobacterium, Pseudomonas or Vibrio were found to be not only relevant for the interactions of the bacteria with protozoa, nematodes and insect phagocytes, but also with mammalian hosts including humans. Thus, non-mammalian model hosts provide valuable insight into the pathogenesis of environmental bacteria.


Assuntos
Amoeba/microbiologia , Caenorhabditis elegans/microbiologia , Drosophila melanogaster/microbiologia , Bactérias Gram-Negativas/patogenicidade , Bactérias Gram-Positivas/patogenicidade , Animais , Infecções Bacterianas/microbiologia , Infecções Bacterianas/patologia , Modelos Animais de Doenças , Humanos , Comportamento Predatório
19.
PLoS Pathog ; 2(5): e46, 2006 May.
Artigo em Inglês | MEDLINE | ID: mdl-16710455

RESUMO

The causative agent of Legionnaires' disease, Legionella pneumophila, employs the intracellular multiplication (Icm)/defective organelle trafficking (Dot) type IV secretion system (T4SS) to upregulate phagocytosis and to establish a replicative vacuole in amoebae and macrophages. Legionella-containing vacuoles (LCVs) do not fuse with endosomes but recruit early secretory vesicles. Here we analyze the role of host cell phosphoinositide (PI) metabolism during uptake and intracellular replication of L. pneumophila. Genetic and pharmacological evidence suggests that class I phosphatidylinositol(3) kinases (PI3Ks) are dispensable for phagocytosis of wild-type L. pneumophila but inhibit intracellular replication of the bacteria and participate in the modulation of the LCV. Uptake and degradation of an icmT mutant strain lacking a functional Icm/Dot transporter was promoted by PI3Ks. We identified Icm/Dot-secreted proteins which specifically bind to phosphatidylinositol(4) phosphate (PI(4)P) in vitro and preferentially localize to LCVs in the absence of functional PI3Ks. PI(4)P was found to be present on LCVs using as a probe either an antibody against PI(4)P or the PH domain of the PI(4)P-binding protein FAPP1 (phosphatidylinositol(4) phosphate adaptor protein-1). Moreover, the presence of PI(4)P on LCVs required a functional Icm/Dot T4SS. Our results indicate that L. pneumophila modulates host cell PI metabolism and exploits the Golgi lipid second messenger PI(4)P to anchor secreted effector proteins to the LCV.


Assuntos
Proteínas de Bactérias/metabolismo , Legionella pneumophila/fisiologia , Fosfatos de Fosfatidilinositol/fisiologia , Vacúolos/metabolismo , Animais , Proteínas de Bactérias/genética , Biomarcadores/metabolismo , Linhagem Celular , Dictyostelium/crescimento & desenvolvimento , Humanos , Legionella pneumophila/crescimento & desenvolvimento , Camundongos , Dados de Sequência Molecular , Mutação , Organelas/metabolismo , Fagocitose/fisiologia , Fosfatidilinositol 3-Quinases/deficiência , Fosfatidilinositol 3-Quinases/fisiologia , Fosfatos de Fosfatidilinositol/metabolismo , Transporte Proteico , Distribuição Tecidual , Vacúolos/microbiologia
20.
Appl Environ Microbiol ; 72(4): 2885-95, 2006 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-16597995

RESUMO

Legionella pneumophila persists for a long time in aquatic habitats, where the bacteria associate with biofilms and replicate within protozoan predators. While L. pneumophila serves as a paradigm for intracellular growth within protozoa, it is less clear whether the bacteria form or replicate within biofilms in the absence of protozoa. In this study, we analyzed surface adherence of and biofilm formation by L. pneumophila in a rich medium that supported axenic replication. Biofilm formation by the virulent L. pneumophila strain JR32 and by clinical and environmental isolates was analyzed by confocal microscopy and crystal violet staining. Strain JR32 formed biofilms on glass surfaces and upright polystyrene wells, as well as on pins of "inverse" microtiter plates, indicating that biofilm formation was not simply due to sedimentation of the bacteria. Biofilm formation by an L. pneumophila fliA mutant lacking the alternative sigma factor sigma(28) was reduced, which demonstrated that bacterial factors are required. Accumulation of biomass coincided with an increase in the optical density at 600 nm and ceased when the bacteria reached the stationary growth phase. L. pneumophila neither grew nor formed biofilms in the inverse system if the medium was exchanged twice a day. However, after addition of Acanthamoeba castellanii, the bacteria proliferated and adhered to surfaces. Sessile (surface-attached) and planktonic (free-swimming) L. pneumophila expressed beta-galactosidase activity to similar extents, and therefore, the observed lack of proliferation of surface-attached bacteria was not due to impaired protein synthesis or metabolic activity. Cocultivation of green fluorescent protein (GFP)- and DsRed-labeled L. pneumophila led to randomly interspersed cells on the substratum and in aggregates, and no sizeable patches of clonally growing bacteria were observed. Our findings indicate that biofilm formation by L. pneumophila in a rich medium is due to growth of planktonic bacteria rather than to growth of sessile bacteria. In agreement with this conclusion, GFP-labeled L. pneumophila initially adhered in a continuous-flow chamber system but detached over time; the detachment correlated with the flow rate, and there was no accumulation of biomass. Under these conditions, L. pneumophila persisted in biofilms formed by Empedobacter breve or Microbacterium sp. but not in biofilms formed by Klebsiella pneumoniae or other environmental bacteria, suggesting that specific interactions between the bacteria modulate adherence.


Assuntos
Biofilmes/crescimento & desenvolvimento , Legionella pneumophila/crescimento & desenvolvimento , Animais , Aderência Bacteriana , Meios de Cultura , Violeta Genciana/metabolismo , Legionella pneumophila/genética , Legionella pneumophila/metabolismo , Microscopia Confocal , Plâncton/crescimento & desenvolvimento , Coloração e Rotulagem
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...