Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
1.
Infect Immun ; 78(12): 5138-50, 2010 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-20876292

RESUMEN

Extracellular Yersinia pseudotuberculosis employs a type III secretion system (T3SS) for translocating virulence factors (Yersinia outer proteins [Yops]) directly into the cytosol of eukaryotic cells. Recently, we used YopE as a carrier molecule for T3SS-dependent secretion and translocation of listeriolysin O (LLO) from Listeria monocytogenes. We demonstrated that translocation of chimeric YopE/LLO into the cytosol of macrophages by Yersinia results in the induction of a codominant antigen-specific CD4 and CD8 T-cell response in orally immunized mice. In this study, we addressed the requirements for processing and major histocompatibility complex (MHC) class II presentation of chimeric YopE proteins translocated into the cytosol of macrophages by the Yersinia T3SS. Our data demonstrate the ability of Yersinia to counteract exogenous MHC class II antigen presentation of secreted hybrid YopE by the action of wild-type YopE and YopH. In the absence of exogenous MHC class II antigen presentation, an alternative pathway was identified for YopE fusion proteins originating in the cytosol. This endogenous antigen-processing pathway was sensitive to inhibitors of phagolysosomal acidification and macroautophagy, but it did not require the function either of the proteasome or of transporters associated with antigen processing. Thus, by an autophagy-dependent mechanism, macrophages are able to compensate for the YopE/YopH-mediated inhibition of the endosomal MHC class II antigen presentation pathway for exogenous antigens. This is the first report demonstrating that autophagy might enable the host to mount an MHC class II-restricted CD4 T-cell response against translocated bacterial virulence factors. We provide critical new insights into the interaction between the mammalian immune system and a human pathogen.


Asunto(s)
Presentación de Antígeno/inmunología , Autofagia/inmunología , Endosomas/inmunología , Infecciones por Yersinia pseudotuberculosis/inmunología , Yersinia pseudotuberculosis/inmunología , Animales , Presentación de Antígeno/fisiología , Autofagia/fisiología , Proteínas de la Membrana Bacteriana Externa/fisiología , Sistemas de Secreción Bacterianos/inmunología , Sistemas de Secreción Bacterianos/fisiología , Western Blotting , Línea Celular , Endosomas/fisiología , Técnica del Anticuerpo Fluorescente , Antígenos de Histocompatibilidad Clase II/inmunología , Macrófagos/inmunología , Macrófagos/fisiología , Ratones , Yersiniosis/inmunología , Yersinia pseudotuberculosis/fisiología , Infecciones por Yersinia pseudotuberculosis/fisiopatología
3.
Genes Dev ; 22(8): 1051-68, 2008 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-18367645

RESUMEN

The cellular and molecular mechanisms that underlie species-specific membrane fusion between male and female gametes remain largely unknown. Here, by use of gene discovery methods in the green alga Chlamydomonas, gene disruption in the rodent malaria parasite Plasmodium berghei, and distinctive features of fertilization in both organisms, we report discovery of a mechanism that accounts for a conserved protein required for gamete fusion. A screen for fusion mutants in Chlamydomonas identified a homolog of HAP2, an Arabidopsis sterility gene. Moreover, HAP2 disruption in Plasmodium blocked fertilization and thereby mosquito transmission of malaria. HAP2 localizes at the fusion site of Chlamydomonas minus gametes, yet Chlamydomonas minus and Plasmodium hap2 male gametes retain the ability, using other, species-limited proteins, to form tight prefusion membrane attachments with their respective gamete partners. Membrane dye experiments show that HAP2 is essential for membrane merger. Thus, in two distantly related eukaryotes, species-limited proteins govern access to a conserved protein essential for membrane fusion.


Asunto(s)
Proteínas Algáceas/genética , Chlamydomonas/genética , Células Germinativas/fisiología , Plasmodium berghei/genética , Proteínas Protozoarias/genética , Interacciones Espermatozoide-Óvulo/genética , Animales , Femenino , Masculino , Fusión de Membrana/genética
4.
Microbiology (Reading) ; 153(Pt 5): 1464-1473, 2007 May.
Artículo en Inglés | MEDLINE | ID: mdl-17464060

RESUMEN

Recently, a number of attenuated mutants of Yersinia pseudotuberculosis have been identified using a bioinformatics approach. One of the target genes identified in that study was vagH, which the authors now characterized further. VagH shows homology to HemK of Escherichia coli, possessing methyltransferase activity similar to that of HemK, and targeting release factors 1 and 2. Microarray studies comparing the wild-type and the vagH mutant revealed that the mRNA levels of only a few genes were altered in the mutant. By proteome analysis, expression of the virulence determinant YopD was found to be increased, indicating a possible connection between VagH and the virulence plasmid-encoded type III secretion system (T3SS). Further analysis showed that Yop expression and secretion were repressed in a vagH mutant. This phenotype could be suppressed by trans-complementation with the wild-type vagH gene or by deletion of the negative regulator yopD. Also, in a similar manner to a T3SS-negative mutant, the avirulent vagH mutant was rapidly cleared from Peyer's patches and could not reach the spleen after oral infection of mice. In a manner analogous to that of T3SS mutants, the vagH mutant could not block phagocytosis by macrophages. However, a vagH mutant showed no defects in the T3SS-independent ability to proliferate intracellularly and replicated to levels similar to those of the wild-type in macrophages. In conclusion, the vagH mutant exhibits a virulence phenotype similar to that of a T3SS-negative mutant, indicating a tight link between VagH and type III secretion in Y. pseudotuberculosis.


Asunto(s)
Proteínas Bacterianas/metabolismo , Metiltransferasas/fisiología , Factores de Virulencia/metabolismo , Yersinia pseudotuberculosis/metabolismo , Animales , Proteínas Bacterianas/genética , Recuento de Colonia Microbiana , Modelos Animales de Enfermedad , Proteínas de Escherichia coli/genética , Femenino , Eliminación de Gen , Perfilación de la Expresión Génica , Macrófagos/microbiología , Metiltransferasas/genética , Metiltransferasas/metabolismo , Ratones , Ratones Endogámicos C57BL , Análisis de Secuencia por Matrices de Oligonucleótidos , Factores de Terminación de Péptidos/metabolismo , Proteína Metiltransferasas/genética , Transporte de Proteínas , Proteoma/análisis , ARN Bacteriano/análisis , ARN Bacteriano/genética , ARN Mensajero/análisis , ARN Mensajero/genética , Factores de Virulencia/genética , Infecciones por Yersinia pseudotuberculosis/microbiología
5.
Vaccine ; 23(42): 4984-98, 2005 Oct 10.
Artículo en Inglés | MEDLINE | ID: mdl-15985316

RESUMEN

Yersinia enterocolitica employs a type III secretion system (TTSS) to target virulence factors (e.g. YopE) into the cytosol of the host cells. We utilized the TTSS to introduce a recombinant antigen directly into the cytosol of host cells and to investigate the potential of Y. enterocolitica and Y. pseudotuberculosis as live carrier for vaccines. The model antigen ovalbumin (Ova) was fused to defined secretion or translocation domains of the Yersinia effector protein YopE and introduced into attenuated mutant strains of Y. enterocolitica and Y. pseudotuberculosis. In vitro experiments showed secretion and translocation of YopE-Ova hybrid proteins into host cells. To investigate the resulting immune responses, mice expressing transgenic Ova-specific T cell receptors were used. Both Y. enterocolitica and Y. pseudotuberculosis mutants induced efficaciously Ova-specific CD8+ T cell responses. The translocation domain of YopE was required for induction of CD8+ T cell responses in vivo, but not for T cell responses induced in vitro. The in vivo frequency of Ova-specific splenic T cells was up to six-fold higher in mice immunized with YopE-Ova-translocating Y. enterocolitica/Y. pseudotuberculosis mutants than in control mice. The Ova-specific T cells were shown to produce high amounts of IFN-gamma. We did not observe significant Ova-specific CD4+ T cell or antibody responses upon vaccination with either of the strains. In conclusion, Yersinia live carrier vaccine strains are suitable to target antigens into the MHC class I pathway and stimulate CD8+ T cell responses and thus, might be useful in vaccine approaches against intracellular pathogens.


Asunto(s)
Vacunas Bacterianas/administración & dosificación , Linfocitos T CD8-positivos/inmunología , Vectores Genéticos/inmunología , Yersinia/genética , Administración Oral , Animales , Anticuerpos/análisis , Proteínas de la Membrana Bacteriana Externa/genética , Vacunas Bacterianas/inmunología , Linfocitos T CD4-Positivos/inmunología , Femenino , Inmunidad Celular , Interferón gamma/análisis , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Modelos Animales , Ovalbúmina/genética , Ovalbúmina/inmunología , Proteínas Recombinantes de Fusión/inmunología , Vacunas Sintéticas/administración & dosificación , Vacunas Sintéticas/inmunología , Yersinia/inmunología , Yersinia enterocolitica , Yersinia pseudotuberculosis
6.
Infect Immun ; 72(3): 1333-40, 2004 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-14977936

RESUMEN

The sequencing of bacterial genomes has opened new perspectives for identification of targets for treatment of infectious diseases. We have identified a set of novel virulence-associated genes (vag genes) by comparing the genome sequences of six human pathogens that are known to cause persistent or chronic infections in humans: Yersinia pestis, Neisseria gonorrhoeae, Helicobacter pylori, Borrelia burgdorferi, Streptococcus pneumoniae, and Treponema pallidum. This comparison was limited to genes annotated as hypothetical in the T. pallidum genome project. Seventeen genes with unknown functions were found to be conserved among these pathogens. Insertional inactivation of 14 of these genes generated nine mutants that were attenuated for virulence in a mouse infection model. Out of these nine genes, five were found to be specifically associated with virulence in mice as demonstrated by infection with Yersinia pseudotuberculosis in-frame deletion mutants. In addition, these five vag genes were essential only in vivo, since all the mutants were able to grow in vitro. These genes are broadly conserved among bacteria. Therefore, we propose that the corresponding vag gene products may constitute novel targets for antimicrobial therapy and that some vag mutants could serve as carrier strains for live vaccines.


Asunto(s)
Bacterias/genética , Bacterias/patogenicidad , Genes Bacterianos , Animales , Antibacterianos/uso terapéutico , Bacterias/crecimiento & desarrollo , Infecciones Bacterianas/tratamiento farmacológico , Infecciones Bacterianas/microbiología , Proteínas Bacterianas/genética , Proteínas Bacterianas/fisiología , Biología Computacional , Farmacorresistencia Bacteriana Múltiple , Femenino , Genoma Bacteriano , Células HeLa , Humanos , Ratones , Ratones Endogámicos C57BL , Mutagénesis , Fenotipo , Virulencia/genética
7.
Infect Immun ; 71(6): 3463-72, 2003 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-12761131

RESUMEN

Yersinia pseudotuberculosis employs a type III secretion system for targeting of several virulence factors directly to the cytosol of eukaryotic cells. This protein translocation mechanism mediates the ability of Yersinia to resist phagocytosis and is required for sustained extracellular bacterial replication. In the present study, the Yersinia outer protein E (YopE) was used as a carrier molecule for type III-dependent secretion and translocation of listeriolysin O (LLO) from Listeria monocytogenes. In comparison to wild-type Yersinia, an attenuated Y. pseudotuberculosis yopK-null mutant strain hypertranslocates chimeric YopE/LLO into the cytosol of macrophages, resulting in enhanced major histocompatibility complex (MHC) class I-restricted antigen presentation of an LLO-derived CD8 T-cell epitope. Remarkably, T-cell activation assays also revealed a superior ability of translocated over secreted LLO to induce MHC class II-restricted antigen presentation. These in vitro observations were confirmed after immunization of mice with a single dose of the yopK-null mutant strain. Animals orally inoculated with recombinant Yersinia expressing translocated chimeric YopE/LLO revealed high numbers of gamma interferon-producing LLO-specific CD4 and CD8 T cells. For the first time, it is shown that cytosolic antigen display mediated by an extracellular bacterial carrier vaccine results in simultaneous CD4 and CD8 T-cell priming, conferring protection against an intracellular pathogen.


Asunto(s)
Antígenos Bacterianos/inmunología , Proteínas de la Membrana Bacteriana Externa/inmunología , Toxinas Bacterianas , Vacunas Bacterianas/inmunología , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Proteínas de Choque Térmico/inmunología , Listeriosis/prevención & control , Proteínas Recombinantes de Fusión/inmunología , Vacunas Sintéticas/inmunología , Yersinia pseudotuberculosis/inmunología , Animales , Proteínas Hemolisinas , Macrófagos/metabolismo , Ratones , Ratones Endogámicos BALB C , Fragmentos de Péptidos/inmunología , Transporte de Proteínas , Vacunas Atenuadas/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA