Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 444
Filtrar
Más filtros

Intervalo de año de publicación
1.
Proc Natl Acad Sci U S A ; 118(42)2021 10 19.
Artículo en Inglés | MEDLINE | ID: mdl-34625471

RESUMEN

Cellular ionic concentrations are a central factor orchestrating host innate immunity, but no pathogenic mechanism that perturbs host innate immunity by directly targeting metal ions has yet been described. Here, we report a unique virulence strategy of Yersinia pseudotuberculosis (Yptb) involving modulation of the availability of Mn2+, an immunostimulatory metal ion in host cells. We showed that the Yptb type VI secretion system (T6SS) delivered a micropeptide, TssS, into host cells to enhance its virulence. The mutant strain lacking TssS (ΔtssS) showed substantially reduced virulence but induced a significantly stronger host innate immune response, indicating an antagonistic role of this effector in host antimicrobial immunity. Subsequent studies revealed that TssS is a Mn2+-chelating protein and that its Mn2+-chelating ability is essential for the disruption of host innate immunity. Moreover, we showed that Mn2+ enhances the host innate immune response to Yptb infection by activating the stimulator of interferon genes (STING)-mediated immune response. Furthermore, we demonstrated that TssS counteracted the cytoplasmic Mn2+ increase to inhibit the STING-mediated innate immune response by sequestering Mn2+ Finally, TssS-mediated STING inhibition sabotaged bacterial clearance in vivo. These results reveal a previously unrecognized bacterial immune evasion strategy involving modulation of the bioavailability of intracellular metal ions and provide a perspective on the role of the T6SS in pathogenesis.


Asunto(s)
Inmunidad Innata , Manganeso/metabolismo , Proteínas de la Membrana/metabolismo , Sistemas de Secreción Tipo VI , Animales , Ratones , Ratones Endogámicos C57BL , Unión Proteica , Transporte de Proteínas , Yersinia pseudotuberculosis/metabolismo , Yersinia pseudotuberculosis/patogenicidad
2.
PLoS Pathog ; 16(1): e1008184, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31951643

RESUMEN

Frequent transitions of bacterial pathogens between their warm-blooded host and external reservoirs are accompanied by abrupt temperature shifts. A temperature of 37°C serves as reliable signal for ingestion by a mammalian host, which induces a major reprogramming of bacterial gene expression and metabolism. Enteric Yersiniae are Gram-negative pathogens accountable for self-limiting gastrointestinal infections. Among the temperature-regulated virulence genes of Yersinia pseudotuberculosis is cnfY coding for the cytotoxic necrotizing factor (CNFY), a multifunctional secreted toxin that modulates the host's innate immune system and contributes to the decision between acute infection and persistence. We report that the major determinant of temperature-regulated cnfY expression is a thermo-labile RNA structure in the 5'-untranslated region (5'-UTR). Various translational gene fusions demonstrated that this region faithfully regulates translation initiation regardless of the transcription start site, promoter or reporter strain. RNA structure probing revealed a labile stem-loop structure, in which the ribosome binding site is partially occluded at 25°C but liberated at 37°C. Consistent with translational control in bacteria, toeprinting (primer extension inhibition) experiments in vitro showed increased ribosome binding at elevated temperature. Point mutations locking the 5'-UTR in its 25°C structure impaired opening of the stem loop, ribosome access and translation initiation at 37°C. To assess the in vivo relevance of temperature control, we used a mouse infection model. Y. pseudotuberculosis strains carrying stabilized RNA thermometer variants upstream of cnfY were avirulent and attenuated in their ability to disseminate into mesenteric lymph nodes and spleen. We conclude with a model, in which the RNA thermometer acts as translational roadblock in a two-layered regulatory cascade that tightly controls provision of the CNFY toxin during acute infection. Similar RNA structures upstream of various cnfY homologs suggest that RNA thermosensors dictate the production of secreted toxins in a wide range of pathogens.


Asunto(s)
Toxinas Bacterianas/biosíntesis , Toxinas Bacterianas/genética , Regulación Bacteriana de la Expresión Génica , ARN Bacteriano/metabolismo , Infecciones por Yersinia pseudotuberculosis/microbiología , Yersinia pseudotuberculosis/metabolismo , Regiones no Traducidas 5' , Animales , Toxinas Bacterianas/química , Femenino , Humanos , Secuencias Invertidas Repetidas , Ratones , Ratones Endogámicos BALB C , Conformación de Ácido Nucleico , ARN Bacteriano/química , ARN Bacteriano/genética , Temperatura , Virulencia , Yersinia pseudotuberculosis/química , Yersinia pseudotuberculosis/genética , Yersinia pseudotuberculosis/patogenicidad
3.
PLoS Pathog ; 16(5): e1008576, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-32392230

RESUMEN

Yersinia suppress neutrophil responses by using a type 3 secretion system (T3SS) to inject 6-7 Yersinia effector proteins (Yops) effectors into their cytoplasm. YopH is a tyrosine phosphatase that causes dephosphorylation of the adaptor protein SKAP2, among other targets in neutrophils. SKAP2 functions in reactive oxygen species (ROS) production, phagocytosis, and integrin-mediated migration by neutrophils. Here we identify essential neutrophil functions targeted by YopH, and investigate how the interaction between YopH and SKAP2 influence Yersinia pseudotuberculosis (Yptb) survival in tissues. The growth defect of a ΔyopH mutant was restored in mice defective in the NADPH oxidase complex, demonstrating that YopH is critical for protecting Yptb from ROS during infection. The growth of a ΔyopH mutant was partially restored in Skap2-deficient (Skap2KO) mice compared to wild-type (WT) mice, while induction of neutropenia further enhanced the growth of the ΔyopH mutant in both WT and Skap2KO mice. YopH inhibited both ROS production and degranulation triggered via integrin receptor, G-protein coupled receptor (GPCR), and Fcγ receptor (FcγR) stimulation. SKAP2 was required for integrin receptor and GPCR-mediated ROS production, but dispensable for degranulation under all conditions tested. YopH blocked SKAP2-independent FcγR-stimulated phosphorylation of the proximal signaling proteins Syk, SLP-76, and PLCγ2, and the more distal signaling protein ERK1/2, while only ERK1/2 phosphorylation was dependent on SKAP2 following integrin receptor activation. These findings reveal that YopH prevents activation of both SKAP2-dependent and -independent neutrophilic defenses, uncouple integrin- and GPCR-dependent ROS production from FcγR responses based on their SKAP2 dependency, and show that SKAP2 is not required for degranulation.


Asunto(s)
Proteínas de la Membrana Bacteriana Externa/inmunología , Péptidos y Proteínas de Señalización Intracelular/inmunología , Neutrófilos/inmunología , Proteínas Tirosina Fosfatasas/inmunología , Transducción de Señal/inmunología , Infecciones por Yersinia pseudotuberculosis/inmunología , Yersinia pseudotuberculosis/inmunología , Animales , Péptidos y Proteínas de Señalización Intracelular/genética , Sistema de Señalización de MAP Quinasas/genética , Sistema de Señalización de MAP Quinasas/inmunología , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , Neutrófilos/patología , Especies Reactivas de Oxígeno/inmunología , Transducción de Señal/genética , Yersinia pseudotuberculosis/patogenicidad , Infecciones por Yersinia pseudotuberculosis/genética , Infecciones por Yersinia pseudotuberculosis/patología
4.
PLoS Pathog ; 15(12): e1008001, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31869388

RESUMEN

The enteropathogen Yersinia pseudotuberculosis and the related plague agent Y. pestis require the Ysc type III secretion system (T3SS) to subvert phagocyte defense mechanisms and cause disease. Yet type III secretion (T3S) in Yersinia induces growth arrest and innate immune recognition, necessitating tight regulation of the T3SS. Here we show that Y. pseudotuberculosis T3SS expression is kept low under anaerobic, iron-rich conditions, such as those found in the intestinal lumen where the Yersinia T3SS is not required for growth. In contrast, the Yersinia T3SS is expressed under aerobic or anaerobic, iron-poor conditions, such as those encountered by Yersinia once they cross the epithelial barrier and encounter phagocytic cells. We further show that the [2Fe-2S] containing transcription factor, IscR, mediates this oxygen and iron regulation of the T3SS by controlling transcription of the T3SS master regulator LcrF. IscR binds directly to the lcrF promoter and, importantly, a mutation that prevents this binding leads to decreased disseminated infection of Y. pseudotuberculosis but does not perturb intestinal colonization. Similar to E. coli, Y. pseudotuberculosis uses the Fe-S cluster occupancy of IscR as a readout of oxygen and iron conditions that impact cellular Fe-S cluster homeostasis. We propose that Y. pseudotuberculosis has coopted this system to sense entry into deeper tissues and induce T3S where it is required for virulence. The IscR binding site in the lcrF promoter is completely conserved between Y. pseudotuberculosis and Y. pestis. Deletion of iscR in Y. pestis leads to drastic disruption of T3S, suggesting that IscR control of the T3SS evolved before Y. pestis split from Y. pseudotuberculosis.


Asunto(s)
Hierro/metabolismo , Oxígeno/metabolismo , Sistemas de Secreción Tipo III/metabolismo , Infecciones por Yersinia pseudotuberculosis/inmunología , Animales , Proteínas Bacterianas/metabolismo , Sistemas de Secreción Bacterianos/genética , Escherichia coli/metabolismo , Proteínas de Escherichia coli/metabolismo , Regulación Bacteriana de la Expresión Génica/genética , Regiones Promotoras Genéticas/genética , Transcripción Genética , Yersinia/metabolismo , Yersinia pseudotuberculosis/patogenicidad , Infecciones por Yersinia pseudotuberculosis/metabolismo
5.
PLoS Pathog ; 14(2): e1006858, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-29390040

RESUMEN

Gastrointestinal infections caused by enteric yersiniae can become persistent and complicated by relapsing enteritis and severe autoimmune disorders. To establish a persistent infection, the bacteria have to cope with hostile surroundings when they transmigrate through the intestinal epithelium and colonize underlying gut-associated lymphatic tissues. How the bacteria gain a foothold in the face of host immune responses is poorly understood. Here, we show that the CNFY toxin, which enhances translocation of the antiphagocytic Yop effectors, induces inflammatory responses. This results in extensive tissue destruction, alteration of the intestinal microbiota and bacterial clearance. Suppression of CNFY function, however, increases interferon-γ-mediated responses, comprising non-inflammatory antimicrobial activities and tolerogenesis. This process is accompanied by a preterm reprogramming of the pathogen's transcriptional response towards persistence, which gives the bacteria a fitness edge against host responses and facilitates establishment of a commensal-type life style.


Asunto(s)
Toxinas Bacterianas/genética , Eliminación de Gen , Inflamación/genética , Factores de Virulencia/genética , Infecciones por Yersinia pseudotuberculosis/genética , Yersinia pseudotuberculosis/genética , Animales , Ciego/microbiología , Progresión de la Enfermedad , Femenino , Gastroenteritis/genética , Gastroenteritis/microbiología , Enfermedades Gastrointestinales/genética , Enfermedades Gastrointestinales/microbiología , Microbioma Gastrointestinal/fisiología , Inflamación/microbiología , Ratones , Ratones Endogámicos BALB C , Organismos Modificados Genéticamente , Yersinia pseudotuberculosis/patogenicidad , Infecciones por Yersinia pseudotuberculosis/patología
6.
Bull Exp Biol Med ; 170(2): 223-225, 2020 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-33263854

RESUMEN

Electron microscopy study revealed changes in the ultrastructure of bacteria of Yersinia pseudotuberculosis strains characterized by significantly reduced reproductive ability and virulence potential after long-term storage at low temperature of 4-8°C. Most bacterial cells contained dark cytosol with reduced cellular material or empty cytosol, while the cell wall was preserved. The revealed ultrastructural changes in the bacterial cells of the static culture of Y. pseudotuberculosis suggest that storage of strains under low positive temperatures could induce the transition of the majority of bacterial cell population to a dormant, non-cultivated state with a decrease in their virulence. This fact is of great scientific and applied importance in studies of causative agents of saprozoonoses, including pseudotuberculosis, which has the etiopathogenetic background of persistent infection.


Asunto(s)
Infecciones por Yersinia pseudotuberculosis/microbiología , Yersinia pseudotuberculosis/patogenicidad , Yersinia pseudotuberculosis/ultraestructura , Antibacterianos/farmacología , Pared Celular/metabolismo , Cromatina/química , Frío , Citosol/metabolismo , Humanos , Pruebas de Sensibilidad Microbiana , Técnicas Microbiológicas , Microscopía Electrónica de Transmisión , Manejo de Especímenes , Virulencia
7.
J Biol Chem ; 293(4): 1466-1479, 2018 01 26.
Artículo en Inglés | MEDLINE | ID: mdl-29197826

RESUMEN

Yersinia pseudotuberculosis is a foodborne pathogenic bacterium that causes acute gastrointestinal illness, but its mechanisms of infection are incompletely described. We examined how host cell sterol composition affected Y. pseudotuberculosis uptake. To do this, we depleted or substituted cholesterol in human MDA-MB-231 epithelial cells with various alternative sterols. Decreasing host cell cholesterol significantly reduced pathogen internalization. When host cell cholesterol was substituted with various sterols, only desmosterol and 7-dehydrocholesterol supported internalization. This specificity was not due to sterol dependence of bacterial attachment to host cells, which was similar with all sterols studied. Because a key step in Y. pseudotuberculosis internalization is interaction of the bacterial adhesins invasin and YadA with host cell ß1 integrin, we compared the sterol dependence of wildtype Y. pseudotuberculosis internalization with that of Δinv, ΔyadA, and ΔinvΔyadA mutant strains. YadA deletion decreased bacterial adherence to host cells, whereas invasin deletion had no effect. Nevertheless, host cell sterol substitution had a similar effect on internalization of these bacterial deletion strains as on the wildtype bacteria. The ΔinvΔyadA double mutant adhered least to cells and so was not significantly internalized. The sterol structure dependence of Y. pseudotuberculosis internalization differed from that of endocytosis, as monitored using antibody-clustered ß1 integrin and previous studies on other proteins, which had a more permissive sterol dependence. This study suggests that agents could be designed to interfere with internalization of Yersinia without disturbing endocytosis.


Asunto(s)
Adhesión Bacteriana , Deshidrocolesteroles/metabolismo , Integrina beta1/metabolismo , Infecciones por Yersinia pseudotuberculosis/metabolismo , Yersinia pseudotuberculosis/metabolismo , Línea Celular Tumoral , Femenino , Eliminación de Gen , Humanos , Yersinia pseudotuberculosis/genética , Yersinia pseudotuberculosis/patogenicidad , Infecciones por Yersinia pseudotuberculosis/genética , Infecciones por Yersinia pseudotuberculosis/patología
8.
J Biol Chem ; 293(22): 8672-8690, 2018 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-29535184

RESUMEN

Yersinia pseudotuberculosis is a Gram-negative bacterium and zoonotic pathogen responsible for a wide range of diseases, ranging from mild diarrhea, enterocolitis, lymphatic adenitis to persistent local inflammation. The Y. pseudotuberculosis invasin D (InvD) molecule belongs to the invasin (InvA)-type autotransporter proteins, but its structure and function remain unknown. In this study, we present the first crystal structure of InvD, analyzed its expression and function in a murine infection model, and identified its target molecule in the host. We found that InvD is induced at 37 °C and expressed in vivo 2-4 days after infection, indicating that InvD is a virulence factor. During infection, InvD was expressed in all parts of the intestinal tract, but not in deeper lymphoid tissues. The crystal structure of the C-terminal adhesion domain of InvD revealed a distinct Ig-related fold that, apart from the canonical ß-sheets, comprises various modifications of and insertions into the Ig-core structure. We identified the Fab fragment of host-derived IgG/IgA antibodies as the target of the adhesion domain. Phage display panning and flow cytometry data further revealed that InvD exhibits a preferential binding specificity toward antibodies with VH3/VK1 variable domains and that it is specifically recruited to a subset of B cells. This finding suggests that InvD modulates Ig functions in the intestine and affects direct interactions with a subset of cell surface-exposed B-cell receptors. In summary, our results provide extensive insights into the structure of InvD and its specific interaction with the target molecule in the host.


Asunto(s)
Adhesinas Bacterianas/metabolismo , Anticuerpos/metabolismo , Fragmentos Fab de Inmunoglobulinas/metabolismo , Intestinos/microbiología , Infecciones por Yersinia pseudotuberculosis/microbiología , Yersinia pseudotuberculosis/patogenicidad , Adhesinas Bacterianas/química , Secuencia de Aminoácidos , Animales , Anticuerpos/inmunología , Adhesión Bacteriana , Femenino , Fragmentos Fab de Inmunoglobulinas/inmunología , Intestinos/inmunología , Intestinos/patología , Ratones , Ratones Endogámicos BALB C , Modelos Moleculares , Conformación Proteica , Homología de Secuencia , Virulencia , Yersinia pseudotuberculosis/inmunología , Infecciones por Yersinia pseudotuberculosis/metabolismo , Infecciones por Yersinia pseudotuberculosis/patología
9.
J Biol Chem ; 293(22): 8691-8692, 2018 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-29858390

RESUMEN

In a new paper, the protein InvD from Yersinia pseudotuberculosis, a zoonotic pathogen, is shown to assist late-stage invasion of intestinal epithelia. Remarkably, InvD acts by binding the Fab region of IgG or IgA. It straddles adjacent light-chain and heavy-chain variable domains, but its binding is different from that of antigens in that complementarity-determining regions do not participate. Structure determination revealed that its Fab-interacting domain adopts an immunoglobulin-like fold, fused to the preceding immunoglobulin-like domain and carried on a long stalk anchored to the bacterial outer membrane. Possible roles of this unusual host-pathogen interaction include avoidance of clearance from the intestine by secretory IgA.


Asunto(s)
Adhesinas Bacterianas/metabolismo , Anticuerpos/metabolismo , Fragmentos Fab de Inmunoglobulinas/metabolismo , Intestinos/microbiología , Infecciones por Yersinia pseudotuberculosis/microbiología , Yersinia pseudotuberculosis/patogenicidad , Adhesinas Bacterianas/química , Animales , Anticuerpos/inmunología , Humanos , Fragmentos Fab de Inmunoglobulinas/inmunología , Región Variable de Inmunoglobulina , Intestinos/inmunología , Intestinos/patología , Yersinia pseudotuberculosis/inmunología , Infecciones por Yersinia pseudotuberculosis/metabolismo , Infecciones por Yersinia pseudotuberculosis/patología
10.
Infect Immun ; 87(1)2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30348825

RESUMEN

Yersinia pseudotuberculosis is a Gram-negative enteropathogen and causes gastrointestinal infections. It disseminates from gut to mesenteric lymph nodes (MLNs), spleen, and liver of infected humans and animals. Although the molecular mechanisms for dissemination and infection are unclear, many Gram-negative enteropathogens presumably invade the small intestine via Peyer's patches to initiate dissemination. In this study, we demonstrate that Y. pseudotuberculosis utilizes its lipopolysaccharide (LPS) core to interact with CD209 receptors, leading to invasion of human dendritic cells (DCs) and murine macrophages. These Y. pseudotuberculosis-CD209 interactions result in bacterial dissemination to MLNs, spleens, and livers of both wild-type and Peyer's patch-deficient mice. The blocking of the Y. pseudotuberculosis-CD209 interactions by expression of O-antigen and with oligosaccharides reduces infectivity. Based on the well-documented studies in which HIV-CD209 interaction leads to viral dissemination, we therefore propose an infection route for Y. pseudotuberculosis where this pathogen, after penetrating the intestinal mucosal membrane, hijacks the Y. pseudotuberculosis-CD209 interaction antigen-presenting cells to reach their target destinations, MLNs, spleens, and livers.


Asunto(s)
Moléculas de Adhesión Celular/metabolismo , Células Dendríticas/microbiología , Endocitosis , Interacciones Huésped-Patógeno , Lectinas Tipo C/metabolismo , Lipopolisacáridos/metabolismo , Macrófagos/microbiología , Receptores de Superficie Celular/metabolismo , Yersinia pseudotuberculosis/patogenicidad , Animales , Adhesión Bacteriana , Células Cultivadas , Modelos Animales de Enfermedad , Humanos , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Unión Proteica , Yersiniosis/microbiología , Yersiniosis/patología , Yersiniosis/fisiopatología
11.
Infect Immun ; 87(10)2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31331960

RESUMEN

In this study, a novel recombinant attenuated Yersinia pseudotuberculosis PB1+ strain (χ10069) engineered with ΔyopK ΔyopJ Δasd triple mutations was used to deliver a Y. pestis fusion protein, YopE amino acid 1 to 138-LcrV (YopENt138-LcrV), to Swiss Webster mice as a protective antigen against infections by yersiniae. χ10069 bacteria harboring the pYA5199 plasmid constitutively synthesized the YopENt138-LcrV fusion protein and secreted it via the type 3 secretion system (T3SS) at 37°C under calcium-deprived conditions. The attenuated strain χ10069(pYA5199) was manifested by the establishment of controlled infection in different tissues without developing conspicuous signs of disease in histopathological analysis of microtome sections. A single-dose oral immunization of χ10069(pYA5199) induced strong serum antibody titers (log10 mean value, 4.2), secretory IgA in bronchoalveolar lavage (BAL) fluid from immunized mice, and Yersinia-specific CD4+ and CD8+ T cells producing high levels of tumor necrosis factor alpha (TNF-α), gamma interferon (IFN-γ), and interleukin 2 (IL-2), as well as IL-17, in both lungs and spleens of immunized mice, conferring comprehensive Th1- and Th2-mediated immune responses and protection against bubonic and pneumonic plague challenges, with 80% and 90% survival, respectively. Mice immunized with χ10069(pYA5199) also exhibited complete protection against lethal oral infections by Yersinia enterocolitica WA and Y. pseudotuberculosis PB1+. These findings indicated that χ10069(pYA5199) as an oral vaccine induces protective immunity to prevent bubonic and pneumonic plague, as well as yersiniosis, in mice and would be a promising oral vaccine candidate for protection against plague and yersiniosis for human and veterinary applications.


Asunto(s)
Anticuerpos Antibacterianos/biosíntesis , Inmunoglobulina A/biosíntesis , Vacuna contra la Peste/administración & dosificación , Peste/prevención & control , Proteínas Recombinantes de Fusión/administración & dosificación , Yersinia pestis/efectos de los fármacos , Infecciones por Yersinia pseudotuberculosis/prevención & control , Yersinia pseudotuberculosis/efectos de los fármacos , Administración Oral , Animales , Antígenos Bacterianos/genética , Antígenos Bacterianos/inmunología , Proteínas de la Membrana Bacteriana Externa/genética , Proteínas de la Membrana Bacteriana Externa/inmunología , Linfocitos T CD4-Positivos/efectos de los fármacos , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/microbiología , Linfocitos T CD8-positivos/efectos de los fármacos , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/microbiología , Protección Cruzada , Femenino , Expresión Génica , Humanos , Inmunización , Interferón gamma/genética , Interferón gamma/inmunología , Interleucina-2/genética , Interleucina-2/inmunología , Pulmón/efectos de los fármacos , Pulmón/inmunología , Pulmón/microbiología , Masculino , Ratones , Peste/inmunología , Peste/microbiología , Peste/mortalidad , Vacuna contra la Peste/biosíntesis , Vacuna contra la Peste/genética , Vacuna contra la Peste/inmunología , Plásmidos/química , Plásmidos/metabolismo , Proteínas Citotóxicas Formadoras de Poros/genética , Proteínas Citotóxicas Formadoras de Poros/inmunología , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/inmunología , Análisis de Supervivencia , Factor de Necrosis Tumoral alfa/genética , Factor de Necrosis Tumoral alfa/inmunología , Vacunas Sintéticas , Yersinia pestis/inmunología , Yersinia pestis/patogenicidad , Yersinia pseudotuberculosis/inmunología , Yersinia pseudotuberculosis/patogenicidad , Infecciones por Yersinia pseudotuberculosis/inmunología , Infecciones por Yersinia pseudotuberculosis/microbiología , Infecciones por Yersinia pseudotuberculosis/mortalidad
12.
PLoS Pathog ; 12(12): e1006091, 2016 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-28006011

RESUMEN

Different biomolecules have been identified in bacterial pathogens that sense changes in temperature and trigger expression of virulence programs upon host entry. However, the dynamics and quantitative outcome of this response in individual cells of a population, and how this influences pathogenicity are unknown. Here, we address these questions using a thermosensing virulence regulator of an intestinal pathogen (RovA of Yersinia pseudotuberculosis) as a model. We reveal that this regulator is part of a novel thermoresponsive bistable switch, which leads to high- and low-invasive subpopulations within a narrow temperature range. The temperature range in which bistability is observed is defined by the degradation and synthesis rate of the regulator, and is further adjustable via a nutrient-responsive regulator. The thermoresponsive switch is also characterized by a hysteretic behavior in which activation and deactivation occurred on vastly different time scales. Mathematical modeling accurately mirrored the experimental behavior and predicted that the thermoresponsiveness of this sophisticated bistable switch is mainly determined by the thermo-triggered increase of RovA proteolysis. We further observed RovA ON and OFF subpopulations of Y. pseudotuberculosis in the Peyer's patches and caecum of infected mice, and that changes in the RovA ON/OFF cell ratio reduce tissue colonization and overall virulence. This points to a bet-hedging strategy in which the thermoresponsive bistable switch plays a key role in adapting the bacteria to the fluctuating conditions encountered as they pass through the host's intestinal epithelium and suggests novel strategies for the development of antimicrobial therapies.


Asunto(s)
Proteínas Bacterianas/metabolismo , Factores de Transcripción/metabolismo , Factores de Virulencia/metabolismo , Infecciones por Yersinia pseudotuberculosis/parasitología , Yersinia pseudotuberculosis/patogenicidad , Animales , Western Blotting , Modelos Animales de Enfermedad , Ensayo de Cambio de Movilidad Electroforética , Femenino , Citometría de Flujo , Ratones , Ratones Endogámicos BALB C , Temperatura , Imagen de Lapso de Tiempo , Virulencia
13.
J Immunol ; 197(10): 4110-4117, 2016 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-27733552

RESUMEN

Innate immune responses that are crucial for control of infection are often targeted by microbial pathogens. Blockade of NF-κB and MAPK signaling by the Yersinia virulence factor YopJ inhibits cytokine production by innate immune cells but also triggers cell death. This cell death requires RIPK1 kinase activity and caspase-8, which are engaged by TLR4 and the adaptor protein TRIF. Nevertheless, TLR4- and TRIF-deficient cells undergo significant apoptosis, implicating TLR4/TRIF-independent pathways in the death of Yersinia-infected cells. In this article, we report a key role for TNF/TNFR1 in Yersinia-induced cell death of murine macrophages, which occurs despite the blockade of NF-κB and MAPK signaling imposed by Yersinia on infected cells. Intriguingly, direct analysis of YopJ injection revealed a heterogeneous population of injection-high and injection-low cells, and demonstrated that TNF expression came from the injection-low population. Moreover, TNF production by this subpopulation was necessary for maximal apoptosis in the population of highly injected cells, and TNFR-deficient mice displayed enhanced susceptibility to Yersinia infection. These data demonstrate an important role for collaboration between TNF and pattern recognition receptor signals in promoting maximal apoptosis during bacterial infection, and demonstrate that heterogeneity in virulence factor injection and cellular responses play an important role in promoting anti-Yersinia immune defense.


Asunto(s)
Proteínas Adaptadoras del Transporte Vesicular/metabolismo , Apoptosis , Macrófagos/microbiología , Macrófagos/fisiología , Factores de Necrosis Tumoral/metabolismo , Yersiniosis/inmunología , Yersinia pseudotuberculosis/patogenicidad , Animales , Proteínas Bacterianas/genética , Caspasa 1/metabolismo , Muerte Celular , Inmunidad Innata , L-Lactato Deshidrogenasa/metabolismo , Ratones , Plásmidos/genética , Transducción de Señal , Receptor Toll-Like 4/inmunología , Factores de Necrosis Tumoral/deficiencia , Factores de Necrosis Tumoral/inmunología , Yersinia pseudotuberculosis/inmunología
14.
Proc Natl Acad Sci U S A ; 112(21): 6688-93, 2015 May 26.
Artículo en Inglés | MEDLINE | ID: mdl-25964352

RESUMEN

Inflammasomes are critical for host defense against bacterial pathogens. In murine macrophages infected by gram-negative bacteria, the canonical inflammasome activates caspase-1 to mediate pyroptotic cell death and release of IL-1 family cytokines. Additionally, a noncanonical inflammasome controlled by caspase-11 induces cell death and IL-1 release. However, humans do not encode caspase-11. Instead, humans encode two putative orthologs: caspase-4 and caspase-5. Whether either ortholog functions similar to caspase-11 is poorly defined. Therefore, we sought to define the inflammatory caspases in primary human macrophages that regulate inflammasome responses to gram-negative bacteria. We find that human macrophages activate inflammasomes specifically in response to diverse gram-negative bacterial pathogens that introduce bacterial products into the host cytosol using specialized secretion systems. In primary human macrophages, IL-1ß secretion requires the caspase-1 inflammasome, whereas IL-1α release and cell death are caspase-1-independent. Instead, caspase-4 mediates IL-1α release and cell death. Our findings implicate human caspase-4 as a critical regulator of noncanonical inflammasome activation that initiates defense against bacterial pathogens in primary human macrophages.


Asunto(s)
Caspasas Iniciadoras/inmunología , Bacterias Gramnegativas/inmunología , Bacterias Gramnegativas/patogenicidad , Inflamasomas/inmunología , Animales , Caspasa 1/inmunología , Muerte Celular , Células Cultivadas , Humanos , Interleucina-1alfa/metabolismo , Interleucina-1beta/metabolismo , Legionella pneumophila/inmunología , Legionella pneumophila/patogenicidad , Lipopolisacáridos/toxicidad , Macrófagos/enzimología , Macrófagos/inmunología , Macrófagos/microbiología , Ratones , Salmonella typhimurium/inmunología , Salmonella typhimurium/patogenicidad , Yersinia pseudotuberculosis/inmunología , Yersinia pseudotuberculosis/patogenicidad
15.
PLoS Genet ; 11(3): e1005087, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25816203

RESUMEN

One hallmark of pathogenic yersiniae is their ability to rapidly adjust their life-style and pathogenesis upon host entry. In order to capture the range, magnitude and complexity of the underlying gene control mechanisms we used comparative RNA-seq-based transcriptomic profiling of the enteric pathogen Y. pseudotuberculosis under environmental and infection-relevant conditions. We identified 1151 individual transcription start sites, multiple riboswitch-like RNA elements, and a global set of antisense RNAs and previously unrecognized trans-acting RNAs. Taking advantage of these data, we revealed a temperature-induced and growth phase-dependent reprogramming of a large set of catabolic/energy production genes and uncovered the existence of a thermo-regulated 'acetate switch', which appear to prime the bacteria for growth in the digestive tract. To elucidate the regulatory architecture linking nutritional status to virulence we also refined the CRP regulon. We identified a massive remodelling of the CRP-controlled network in response to temperature and discovered CRP as a transcriptional master regulator of numerous conserved and newly identified non-coding RNAs which participate in this process. This finding highlights a novel level of complexity of the regulatory network in which the concerted action of transcriptional regulators and multiple non-coding RNAs under control of CRP adjusts the control of Yersinia fitness and virulence to the requirements of their environmental and virulent life-styles.


Asunto(s)
Proteína Receptora de AMP Cíclico/genética , Proteínas Fúngicas/genética , Perfilación de la Expresión Génica , Regulón/genética , Yersinia pseudotuberculosis/genética , AMP Cíclico/genética , AMP Cíclico/metabolismo , Regulación Fúngica de la Expresión Génica , Interacción Gen-Ambiente , ARN sin Sentido/genética , ARN sin Sentido/aislamiento & purificación , Riboswitch/genética , Temperatura , Sitio de Iniciación de la Transcripción , Yersinia pseudotuberculosis/crecimiento & desarrollo , Yersinia pseudotuberculosis/patogenicidad
16.
Infect Immun ; 85(4)2017 04.
Artículo en Inglés | MEDLINE | ID: mdl-28115509

RESUMEN

The twin arginine translocation (Tat) system targets folded proteins across the inner membrane and is crucial for virulence in many important human-pathogenic bacteria. Tat has been shown to be required for the virulence of Yersinia pseudotuberculosis, and we recently showed that the system is critical for different virulence-related stress responses as well as for iron uptake. In this study, we wanted to address the role of the Tat substrates in in vivo virulence. Therefore, 22 genes encoding potential Tat substrates were mutated, and each mutant was evaluated in a competitive oral infection of mice. Interestingly, a ΔsufI mutant was essentially as attenuated for virulence as the Tat-deficient strain. We also verified that SufI was Tat dependent for membrane/periplasmic localization in Y. pseudotuberculosisIn vivo bioluminescent imaging of orally infected mice revealed that both the ΔsufI and ΔtatC mutants were able to colonize the cecum and Peyer's patches (PPs) and could spread to the mesenteric lymph nodes (MLNs). Importantly, at this point, neither the ΔtatC mutant nor the ΔsufI mutant was able to spread systemically, and they were gradually cleared. Immunostaining of MLNs revealed that both the ΔtatC and ΔsufI mutants were unable to spread from the initial infection foci and appeared to be contained by neutrophils, while wild-type bacteria readily spread to establish multiple foci from day 3 postinfection. Our results show that SufI alone is required for the establishment of systemic infection and is the major cause of the attenuation of the ΔtatC mutant.


Asunto(s)
Proteínas Bacterianas/metabolismo , Sistema de Translocación de Arginina Gemela/metabolismo , Infecciones por Yersinia pseudotuberculosis/microbiología , Yersinia pseudotuberculosis/fisiología , Animales , Carga Bacteriana , Proteínas Bacterianas/genética , Femenino , Expresión Génica , Genes Reporteros , Ratones , Mutagénesis , Neutrófilos/inmunología , Neutrófilos/metabolismo , Especificidad por Sustrato , Sistema de Translocación de Arginina Gemela/genética , Virulencia/genética , Yersinia pseudotuberculosis/patogenicidad
17.
PLoS Pathog ; 11(7): e1005020, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-26134274

RESUMEN

Type VI secretion systems (T6SSs) are widespread multi-component machineries that translocate effectors into either eukaryotic or prokaryotic cells, for virulence or for interbacterial competition. Herein, we report that the T6SS-4 from Yersinia pseudotuberculosis displays an unexpected function in the transportation of Zn2+ to combat diverse stresses and host immunity. Environmental insults such as oxidative stress induce the expression of T6SS-4 via OxyR, the transcriptional factor that also regulates many oxidative response genes. Zinc transportation is achieved by T6SS-4-mediated translocation of a novel Zn2+-binding protein substrate YezP (YPK_3549), which has the capacity to rescue the sensitivity to oxidative stress exhibited by T6SS-4 mutants when added to extracellular milieu. Disruption of the classic zinc transporter ZnuABC together with T6SS-4 or yezP results in mutants that almost completely lost virulence against mice, further highlighting the importance of T6SS-4 in resistance to host immunity. These results assigned an unconventional role to T6SSs, which will lay the foundation for studying novel mechanisms of metal ion uptake by bacteria and the role of this process in their resistance to host immunity and survival in harmful environments.


Asunto(s)
Adaptación Fisiológica/inmunología , Sistemas de Secreción Tipo VI/metabolismo , Infecciones por Yersinia pseudotuberculosis/metabolismo , Yersinia pseudotuberculosis/metabolismo , Yersinia pseudotuberculosis/patogenicidad , Animales , Western Blotting , Ensayo de Cambio de Movilidad Electroforética , Ratones , Reacción en Cadena de la Polimerasa , Reacción en Cadena en Tiempo Real de la Polimerasa , Estrés Fisiológico/inmunología , Sistemas de Secreción Tipo VI/inmunología , Infecciones por Yersinia pseudotuberculosis/inmunología , Zinc/metabolismo
18.
PLoS Pathog ; 11(1): e1004600, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25590628

RESUMEN

We recently found that Yersinia pseudotuberculosis can be used as a model of persistent bacterial infections. We performed in vivo RNA-seq of bacteria in small cecal tissue biopsies at early and persistent stages of infection to determine strategies associated with persistence. Comprehensive analysis of mixed RNA populations from infected tissues revealed that Y. pseudotuberculosis undergoes transcriptional reprogramming with drastic down-regulation of T3SS virulence genes during persistence when the pathogen resides within the cecum. At the persistent stage, the expression pattern in many respects resembles the pattern seen in vitro at 26oC, with for example, up-regulation of flagellar genes and invA. These findings are expected to have impact on future rationales to identify suitable bacterial targets for new antibiotics. Other genes that are up-regulated during persistence are genes involved in anaerobiosis, chemotaxis, and protection against oxidative and acidic stress, which indicates the influence of different environmental cues. We found that the Crp/CsrA/RovA regulatory cascades influence the pattern of bacterial gene expression during persistence. Furthermore, arcA, fnr, frdA, and wrbA play critical roles in persistence. Our findings suggest a model for the life cycle of this enteropathogen with reprogramming from a virulent to an adapted phenotype capable of persisting and spreading by fecal shedding.


Asunto(s)
Análisis de Secuencia de ARN/métodos , Virulencia/genética , Infecciones por Yersinia pseudotuberculosis/genética , Yersinia pseudotuberculosis/genética , Yersinia pseudotuberculosis/patogenicidad , Animales , Ciego/inmunología , Ciego/microbiología , Ciego/patología , Femenino , Regulación Bacteriana de la Expresión Génica , Genes Bacterianos , Ratones , Análisis por Micromatrices , Microbiota/inmunología , ARN Bacteriano/genética , Yersinia pseudotuberculosis/inmunología , Infecciones por Yersinia pseudotuberculosis/inmunología , Infecciones por Yersinia pseudotuberculosis/patología
19.
PLoS Pathog ; 11(10): e1005222, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26484539

RESUMEN

Activation and/or recruitment of the host plasmin, a fibrinolytic enzyme also active on extracellular matrix components, is a common invasive strategy of bacterial pathogens. Yersinia pestis, the bubonic plague agent, expresses the multifunctional surface protease Pla, which activates plasmin and inactivates fibrinolysis inhibitors. Pla is encoded by the pPla plasmid. Following intradermal inoculation, Y. pestis has the capacity to multiply in and cause destruction of the lymph node (LN) draining the entry site. The closely related, pPla-negative, Y. pseudotuberculosis species lacks this capacity. We hypothesized that tissue damage and bacterial multiplication occurring in the LN during bubonic plague were linked and both driven by pPla. Using a set of pPla-positive and pPla-negative Y. pestis and Y. pseudotuberculosis strains in a mouse model of intradermal injection, we found that pPla is not required for bacterial translocation to the LN. We also observed that a pPla-cured Y. pestis caused the same extensive histological lesions as the wild type strain. Furthermore, the Y. pseudotuberculosis histological pattern, characterized by infectious foci limited by inflammatory cell infiltrates with normal tissue density and follicular organization, was unchanged after introduction of pPla. However, the presence of pPla enabled Y. pseudotuberculosis to increase its bacterial load up to that of Y. pestis. Similarly, lack of pPla strongly reduced Y. pestis titers in LNs of infected mice. This pPla-mediated enhancing effect on bacterial load was directly dependent on the proteolytic activity of Pla. Immunohistochemistry of Pla-negative Y. pestis-infected LNs revealed extensive bacterial lysis, unlike the numerous, apparently intact, microorganisms seen in wild type Y. pestis-infected preparations. Therefore, our study demonstrates that tissue destruction and bacterial survival/multiplication are dissociated in the bubo and that the primary action of Pla is to protect bacteria from destruction rather than to alter the tissue environment to favor Y. pestis propagation in the host.


Asunto(s)
Proteínas Bacterianas/metabolismo , Peste/microbiología , Peste/patología , Activadores Plasminogénicos/metabolismo , Yersinia pestis/patogenicidad , Animales , Modelos Animales de Enfermedad , Inmunohistoquímica , Ratones , Mutagénesis Sitio-Dirigida , Peste/enzimología , Virulencia/fisiología , Factores de Virulencia/metabolismo , Yersinia pestis/enzimología , Yersinia pseudotuberculosis/enzimología , Yersinia pseudotuberculosis/patogenicidad , Infecciones por Yersinia pseudotuberculosis/enzimología , Infecciones por Yersinia pseudotuberculosis/microbiología , Infecciones por Yersinia pseudotuberculosis/patología
20.
PLoS Pathog ; 11(10): e1005167, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26468944

RESUMEN

During Yersinia pseudotuberculosis infection of C57BL/6 mice, an exceptionally large CD8+ T cell response to a protective epitope in the type III secretion system effector YopE is produced. At the peak of the response, up to 50% of splenic CD8+ T cells recognize the epitope YopE69-77. The features of the interaction between pathogen and host that result in this large CD8+ T cell response are unknown. Here, we used Y. pseudotuberculosis strains defective for production, secretion and/or translocation of YopE to infect wild-type or mutant mice deficient in specific dendritic cells (DCs). Bacterial colonization of organs and translocation of YopE into spleen cells was measured, and flow cytometry and tetramer staining were used to characterize the cellular immune response. We show that the splenic YopE69-77-specific CD8+ T cells generated during the large response are polyclonal and are produced by a "translocation-dependent" pathway that requires injection of YopE into host cell cytosol. Additionally, a smaller YopE69-77-specific CD8+ T cell response (~10% of the large expansion) can be generated in a "translocation-independent" pathway in which CD8α+ DCs cross present secreted YopE. CCR2-expressing inflammatory DCs were required for the large YopE69-77-specific CD8+ T cell expansion because this response was significantly reduced in Ccr2-/- mice, YopE was translocated into inflammatory DCs in vivo, inflammatory DCs purified from infected spleens activated YopE69-77-specific CD8+ T cells ex vivo and promoted the expansion of YopE69-77-specific CD8+ T cells in infected Ccr2-/- mice after adoptive transfer. A requirement for inflammatory DCs in producing a protective CD8+ T cell response to a bacterial antigen has not previously been demonstrated. Therefore, the production of YopE69-77-specific CD8+ T cells by inflammatory DCs that are injected with YopE during Y. pseudotuberculosis infection represents a novel mechanism for generating a massive and protective adaptive immune response.


Asunto(s)
Proteínas de la Membrana Bacteriana Externa/inmunología , Linfocitos T CD8-positivos/inmunología , Células Dendríticas/inmunología , Sistemas de Secreción Tipo III/inmunología , Infecciones por Yersinia pseudotuberculosis/inmunología , Traslado Adoptivo , Animales , Presentación de Antígeno/inmunología , Modelos Animales de Enfermedad , Epítopos de Linfocito T/inmunología , Femenino , Citometría de Flujo , Immunoblotting , Inflamación/inmunología , Activación de Linfocitos/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Mutantes , Transporte de Proteínas/inmunología , Receptores CCR2/inmunología , Factores de Virulencia/inmunología , Yersinia pseudotuberculosis/inmunología , Yersinia pseudotuberculosis/patogenicidad
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA