Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 20
Filtrar
1.
Eur J Immunol ; 49(9): 1415-1420, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31074841

RESUMEN

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.


Asunto(s)
Anticuerpos Antibacterianos/inmunología , Legionella pneumophila/inmunología , Enfermedad de los Legionarios/inmunología , Animales , Antígenos Bacterianos/inmunología , Proteínas Bacterianas/inmunología , Inmunidad Celular/inmunología , Inmunización Pasiva/métodos , Enfermedad de los Legionarios/microbiología , Macrófagos Alveolares/inmunología , Macrófagos Alveolares/microbiología , Ratones , Ratones Endogámicos C57BL , Especies Reactivas de Oxígeno/inmunología
2.
Sci Rep ; 6: 18109, 2016 Jan 05.
Artículo en Inglés | MEDLINE | ID: mdl-26728082

RESUMEN

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.


Asunto(s)
Antibacterianos/farmacología , Escherichia coli/efectos de los fármacos , Escherichia coli/fisiología , Fimbrias Bacterianas , Animales , Adhesión Bacteriana , Línea Celular , Supervivencia Celular , Farmacorresistencia Bacteriana , Regulación Bacteriana de la Expresión Génica , Lisosomas/inmunología , Lisosomas/microbiología , Macrófagos/inmunología , Macrófagos/metabolismo , Macrófagos/microbiología , Ratones , Viabilidad Microbiana/efectos de los fármacos , Viabilidad Microbiana/inmunología , Fagocitosis , Fenotipo
3.
J Immunol ; 193(8): 4053-9, 2014 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-25210123

RESUMEN

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.


Asunto(s)
Anticuerpos Antibacterianos/uso terapéutico , Anticuerpos Monoclonales/uso terapéutico , Inmunoglobulina G/uso terapéutico , Legionella pneumophila , Enfermedad de los Legionarios/terapia , Animales , Anticuerpos Antibacterianos/administración & dosificación , Anticuerpos Antibacterianos/inmunología , Anticuerpos Monoclonales/administración & dosificación , Anticuerpos Monoclonales/inmunología , Células Cultivadas , Células HEK293 , Humanos , Inmunoglobulina G/clasificación , Inmunoglobulina G/inmunología , Inmunoterapia/métodos , Enfermedad de los Legionarios/inmunología , Enfermedad de los Legionarios/microbiología , Lipopolisacáridos/inmunología , Macrófagos Alveolares/inmunología , Macrófagos Alveolares/microbiología , Ratones , Ratones Endogámicos C57BL
4.
Methods Mol Biol ; 954: 265-77, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23150402

RESUMEN

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.


Asunto(s)
Técnica del Anticuerpo Fluorescente/métodos , Legionella pneumophila/metabolismo , Microscopía Fluorescente , Fagocitos/microbiología , Animales , Proteínas Bacterianas/metabolismo , Técnicas de Cultivo de Célula , Línea Celular , Fagocitos/ultraestructura , Transfección/métodos
5.
Methods Mol Biol ; 954: 505-20, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23150417

RESUMEN

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.


Asunto(s)
Legionella pneumophila/inmunología , Enfermedad de los Legionarios/inmunología , Animales , Líquido del Lavado Bronquioalveolar/citología , Líquido del Lavado Bronquioalveolar/inmunología , Citocinas/inmunología , Citocinas/metabolismo , Modelos Animales de Enfermedad , Ensayo de Inmunoadsorción Enzimática , Humanos , Enfermedad de los Legionarios/microbiología , Pulmón/inmunología , Pulmón/patología , Ganglios Linfáticos/inmunología , Ganglios Linfáticos/patología , Ratones , Bazo/inmunología , Bazo/patología
6.
Fungal Genet Biol ; 49(11): 915-21, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23010151

RESUMEN

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.


Asunto(s)
Transportadoras de Casetes de Unión a ATP/sangre , Transportadoras de Casetes de Unión a ATP/metabolismo , Proteínas Fúngicas/metabolismo , Penicillium chrysogenum/metabolismo , Fenilacetatos/metabolismo , Transportadoras de Casetes de Unión a ATP/genética , Transporte Biológico , Proteínas Fúngicas/genética , Regulación Fúngica de la Expresión Génica , Penicillium chrysogenum/genética , Regulación hacia Arriba , beta-Lactamas/metabolismo
7.
Appl Environ Microbiol ; 78(19): 7107-13, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22865068

RESUMEN

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.


Asunto(s)
Aciltransferasas/biosíntesis , Aciltransferasas/genética , Dosificación de Gen , Ingeniería Metabólica , Proteínas de Unión a las Penicilinas/biosíntesis , Proteínas de Unión a las Penicilinas/genética , Penicilinas/biosíntesis , Penicillium chrysogenum/genética , Penicillium chrysogenum/metabolismo , Expresión Génica , Familia de Multigenes
8.
J Immunol ; 189(2): 841-9, 2012 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-22696443

RESUMEN

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.


Asunto(s)
Antígenos Bacterianos/aislamiento & purificación , Subgrupos de Linfocitos B/inmunología , Subgrupos de Linfocitos B/microbiología , Legionella pneumophila/inmunología , Enfermedad de los Legionarios/inmunología , Administración Intranasal , Animales , Anticuerpos Antibacterianos/biosíntesis , Anticuerpos Antibacterianos/sangre , Anticuerpos Antibacterianos/uso terapéutico , Antígenos Bacterianos/administración & dosificación , Antígenos Bacterianos/inmunología , Subgrupos de Linfocitos B/metabolismo , Vacunas Bacterianas/administración & dosificación , Vacunas Bacterianas/síntesis química , Vacunas Bacterianas/inmunología , Bacteriófago lambda/genética , Bacteriófago lambda/inmunología , Secuencia Conservada/inmunología , Humanos , Inmunoglobulina G/biosíntesis , Inmunoglobulina G/sangre , Legionella pneumophila/patogenicidad , Enfermedad de los Legionarios/sangre , Enfermedad de los Legionarios/prevención & control , Ratones , Ratones Endogámicos A , Ratones Endogámicos C57BL , Vacunas Sintéticas/administración & dosificación , Vacunas Sintéticas/inmunología , Vacunas Sintéticas/uso terapéutico
9.
Methods Mol Biol ; 835: 1-16, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22183644

RESUMEN

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.


Asunto(s)
Transportadoras de Casetes de Unión a ATP/análisis , Transportadoras de Casetes de Unión a ATP/genética , Proteínas Fúngicas/análisis , Proteínas Fúngicas/genética , Penicillium chrysogenum/citología , Penicillium chrysogenum/genética , Eliminación de Gen , Proteínas Fluorescentes Verdes/análisis , Proteínas Fluorescentes Verdes/genética , Microscopía Fluorescente/métodos , Plásmidos/genética , Coloración y Etiquetado/métodos , Transformación Genética
10.
Biotechnol J ; 7(2): 225-36, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22057844

RESUMEN

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.


Asunto(s)
Penicillium chrysogenum/genética , Penicillium chrysogenum/metabolismo , beta-Lactamas/metabolismo , Ingeniería Metabólica/métodos , Biología Sintética/métodos
11.
Eur J Immunol ; 41(4): 889-97, 2011 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21413006

RESUMEN

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.


Asunto(s)
Anticuerpos/inmunología , Espacio Intracelular/inmunología , Receptores Fc/inmunología , Animales , Humanos , Lisosomas/inmunología , Linfocitos T/inmunología
12.
Proc Natl Acad Sci U S A ; 107(47): 20441-6, 2010 Nov 23.
Artículo en Inglés | MEDLINE | ID: mdl-21048081

RESUMEN

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.


Asunto(s)
Anticuerpos Antibacterianos/inmunología , Infecciones Bacterianas/inmunología , Lisosomas/inmunología , Receptores Fc/inmunología , Transducción de Señal/inmunología , Animales , Carga Bacteriana , Inmunización , Legionella pneumophila , Ratones , Microscopía Fluorescente , Mycobacterium bovis
13.
Mol Microbiol ; 71(6): 1341-52, 2009 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-19208094

RESUMEN

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.


Asunto(s)
Bacterias/patogenicidad , Fosfatidilinositoles/metabolismo , Vacuolas/microbiología , Bacterias/metabolismo , Fagosomas/microbiología , Fosfatidilinositol 3-Quinasas/metabolismo , Monoéster Fosfórico Hidrolasas/metabolismo , Transducción de Señal
14.
J Biol Chem ; 284(8): 4846-56, 2009 Feb 20.
Artículo en Inglés | MEDLINE | ID: mdl-19095644

RESUMEN

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.


Asunto(s)
Proteínas Bacterianas/química , Proteínas Portadoras/química , Factores de Intercambio de Guanina Nucleótido/química , Legionella pneumophila/química , Fosfatos de Fosfatidilinositol/química , Proteínas de Unión al GTP rab1/química , Animales , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Proteínas Portadoras/genética , Proteínas Portadoras/metabolismo , Línea Celular , Drosophila , Factores de Intercambio de Guanina Nucleótido/genética , Factores de Intercambio de Guanina Nucleótido/metabolismo , Humanos , Legionella pneumophila/genética , Legionella pneumophila/metabolismo , Legionella pneumophila/patogenicidad , Enfermedad de los Legionarios/genética , Enfermedad de los Legionarios/metabolismo , Mutación , Mapeo Peptídico , Fagocitos/metabolismo , Fagocitos/microbiología , Fosfatos de Fosfatidilinositol/genética , Fosfatos de Fosfatidilinositol/metabolismo , Unión Proteica/fisiología , Estructura Terciaria de Proteína/fisiología , Vacuolas/genética , Vacuolas/metabolismo , Vacuolas/microbiología , Proteínas de Unión al GTP rab1/genética , Proteínas de Unión al GTP rab1/metabolismo
15.
Cell Microbiol ; 11(3): 442-60, 2009 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-19021631

RESUMEN

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.


Asunto(s)
Proteínas Bacterianas/metabolismo , Dictyostelium/enzimología , Dictyostelium/microbiología , Legionella pneumophila/crecimiento & desarrollo , Monoéster Fosfórico Hidrolasas/metabolismo , Vacuolas/microbiología , Factores de Virulencia/metabolismo , Animales , Línea Celular , Eliminación de Gen , Prueba de Complementación Genética , Inositol Polifosfato 5-Fosfatasas , Macrófagos/enzimología , Macrófagos/microbiología , Ratones , Monoéster Fosfórico Hidrolasas/análisis , Monoéster Fosfórico Hidrolasas/genética , Unión Proteica , Vacuolas/química
16.
Cell Microbiol ; 10(12): 2416-33, 2008 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-18673369

RESUMEN

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.


Asunto(s)
Proteínas Bacterianas/metabolismo , Vesículas Citoplasmáticas/metabolismo , Vesículas Citoplasmáticas/microbiología , Legionella pneumophila/patogenicidad , Factores de Virulencia/metabolismo , Proteínas Bacterianas/genética , Calnexina/análisis , Vesículas Citoplasmáticas/química , Vesículas Citoplasmáticas/ultraestructura , Eliminación de Gen , Prueba de Complementación Genética , Humanos , Microscopía Confocal , Microscopía Electrónica de Transmisión , Factores de Virulencia/genética
17.
Cell Microbiol ; 9(12): 2903-20, 2007 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-17614967

RESUMEN

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.


Asunto(s)
Proteínas Bacterianas/fisiología , Legionella pneumophila/crecimiento & desarrollo , Factor sigma/fisiología , Factores de Transcripción/fisiología , Acanthamoeba castellanii/microbiología , Animales , División Celular/genética , Línea Celular , Dictyostelium/microbiología , Eliminación de Gen , Dosificación de Gen , Perfilación de la Expresión Génica , Regulación Bacteriana de la Expresión Génica , Humanos , Legionella pneumophila/citología , Legionella pneumophila/genética , Locomoción/genética , Macrófagos/microbiología , Análisis de Secuencia por Matrices de Oligonucleótidos , Factores de Transcripción/genética , Virulencia
18.
Environ Microbiol ; 9(3): 563-75, 2007 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-17298357

RESUMEN

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.


Asunto(s)
Amoeba/microbiología , Caenorhabditis elegans/microbiología , Drosophila melanogaster/microbiología , Bacterias Gramnegativas/patogenicidad , Bacterias Grampositivas/patogenicidad , Animales , Infecciones Bacterianas/microbiología , Infecciones Bacterianas/patología , Modelos Animales de Enfermedad , Humanos , Conducta Predatoria
19.
PLoS Pathog ; 2(5): e46, 2006 May.
Artículo en Inglés | MEDLINE | ID: mdl-16710455

RESUMEN

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.


Asunto(s)
Proteínas Bacterianas/metabolismo , Legionella pneumophila/fisiología , Fosfatos de Fosfatidilinositol/fisiología , Vacuolas/metabolismo , Animales , Proteínas Bacterianas/genética , Biomarcadores/metabolismo , Línea Celular , Dictyostelium/crecimiento & desarrollo , Humanos , Legionella pneumophila/crecimiento & desarrollo , Ratones , Datos de Secuencia Molecular , Mutación , Orgánulos/metabolismo , Fagocitosis/fisiología , Fosfatidilinositol 3-Quinasas/deficiencia , Fosfatidilinositol 3-Quinasas/fisiología , Fosfatos de Fosfatidilinositol/metabolismo , Transporte de Proteínas , Distribución Tisular , Vacuolas/microbiología
20.
Appl Environ Microbiol ; 72(4): 2885-95, 2006 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-16597995

RESUMEN

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.


Asunto(s)
Biopelículas/crecimiento & desarrollo , Legionella pneumophila/crecimiento & desarrollo , Animales , Adhesión Bacteriana , Medios de Cultivo , Violeta de Genciana/metabolismo , Legionella pneumophila/genética , Legionella pneumophila/metabolismo , Microscopía Confocal , Plancton/crecimiento & desarrollo , Coloración y Etiquetado
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...