Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 2.061
Filtrar
Más filtros

Intervalo de año de publicación
1.
Annu Rev Immunol ; 31: 73-106, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23215645

RESUMEN

Inflammasomes are cytosolic multiprotein complexes that assemble in response to a variety of infectious and noxious insults. Inflammasomes play a critical role in the initiation of innate immune responses, primarily by serving as platforms for the activation of inflammatory caspase proteases. One such caspase, CASPASE-1 (CASP1), initiates innate immune responses by cleaving pro-IL-1ß and pro-IL-18, leading to their activation and release. CASP1 and another inflammatory caspase termed CASP11 can also initiate a rapid and inflammatory form of cell death termed pyroptosis. Several distinct inflammasomes have been described, each of which contains a unique sensor protein of the NLR (nucleotide-binding domain, leucine-rich repeat-containing) superfamily or the PYHIN (PYRIN and HIN-200 domain-containing) superfamily. Here we describe the surprisingly diverse mechanisms by which NLR/PYHIN proteins sense bacteria and initiate innate immune responses. We conclude that inflammasomes represent a highly adaptable scaffold ideally suited for detecting and initiating rapid innate responses to diverse and rapidly evolving bacteria.


Asunto(s)
Bacterias/patogenicidad , Inflamasomas/metabolismo , Animales , Bacillus anthracis/patogenicidad , Proteínas Adaptadoras de Señalización CARD/metabolismo , Proteínas Adaptadoras de Señalización CARD/fisiología , Proteínas de Unión al Calcio/metabolismo , Proteínas de Unión al Calcio/fisiología , Flagelos/metabolismo , Flagelos/fisiología , Humanos , Inflamasomas/genética , Inflamasomas/fisiología , Legionella pneumophila/patogenicidad , Listeria monocytogenes/patogenicidad , Salmonella typhimurium/patogenicidad
2.
Nat Immunol ; 22(11): 1382-1390, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34663978

RESUMEN

Intergenerational inheritance of immune traits linked to epigenetic modifications has been demonstrated in plants and invertebrates. Here we provide evidence for transmission of trained immunity across generations to murine progeny that survived a sublethal systemic infection with Candida albicans or a zymosan challenge. The progeny of trained mice exhibited cellular, developmental, transcriptional and epigenetic changes associated with the bone marrow-resident myeloid effector and progenitor cell compartment. Moreover, the progeny of trained mice showed enhanced responsiveness to endotoxin challenge, alongside improved protection against systemic heterologous Escherichia coli and Listeria monocytogenes infections. Sperm DNA of parental male mice intravenously infected with the fungus C. albicans showed DNA methylation differences linked to immune gene loci. These results provide evidence for inheritance of trained immunity in mammals, enhancing protection against infections.


Asunto(s)
Candida albicans/inmunología , Candidiasis/inmunología , Infecciones por Escherichia coli/inmunología , Escherichia coli/inmunología , Herencia , Inmunidad Innata/genética , Listeria monocytogenes/inmunología , Listeriosis/inmunología , Células Mieloides/inmunología , Animales , Candida albicans/patogenicidad , Candidiasis/genética , Candidiasis/metabolismo , Candidiasis/microbiología , Células Cultivadas , Metilación de ADN , Modelos Animales de Enfermedad , Epigénesis Genética , Escherichia coli/patogenicidad , Infecciones por Escherichia coli/genética , Infecciones por Escherichia coli/metabolismo , Infecciones por Escherichia coli/microbiología , Interacciones Huésped-Patógeno , Listeria monocytogenes/patogenicidad , Listeriosis/genética , Listeriosis/metabolismo , Listeriosis/microbiología , Masculino , Ratones Transgénicos , Células Mieloides/metabolismo , Células Mieloides/microbiología , Espermatozoides/inmunología , Espermatozoides/metabolismo , Transcripción Genética
3.
Cell ; 174(1): 117-130.e14, 2018 06 28.
Artículo en Inglés | MEDLINE | ID: mdl-29909981

RESUMEN

Heterogeneity is a hallmark feature of the adaptive immune system in vertebrates. Following infection, naive T cells differentiate into various subsets of effector and memory T cells, which help to eliminate pathogens and maintain long-term immunity. The current model suggests there is a single lineage of naive T cells that give rise to different populations of effector and memory T cells depending on the type and amounts of stimulation they encounter during infection. Here, we have discovered that multiple sub-populations of cells exist in the naive CD8+ T cell pool that are distinguished by their developmental origin, unique transcriptional profiles, distinct chromatin landscapes, and different kinetics and phenotypes after microbial challenge. These data demonstrate that the naive CD8+ T cell pool is not as homogeneous as previously thought and offers a new framework for explaining the remarkable heterogeneity in the effector and memory T cell subsets that arise after infection.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Genes del Desarrollo , Listeria monocytogenes/patogenicidad , Animales , Linfocitos T CD8-positivos/efectos de los fármacos , Linfocitos T CD8-positivos/metabolismo , Diferenciación Celular , Línea Celular Tumoral , Cromatina/metabolismo , Citocinas/farmacología , Epítopos de Linfocito T/genética , Epítopos de Linfocito T/metabolismo , Memoria Inmunológica , Interferón gamma/metabolismo , Células Asesinas Naturales/citología , Células Asesinas Naturales/metabolismo , Listeria monocytogenes/metabolismo , Ratones , Ratones Endogámicos C57BL , Análisis de Componente Principal , Subgrupos de Linfocitos T/citología , Subgrupos de Linfocitos T/inmunología , Subgrupos de Linfocitos T/metabolismo , Timo/trasplante , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Transcriptoma
4.
Cell ; 170(4): 787-799.e18, 2017 Aug 10.
Artículo en Inglés | MEDLINE | ID: mdl-28802046

RESUMEN

Replication-transcription collisions shape genomes, influence evolution, and promote genetic diseases. Although unclear why, head-on transcription (lagging strand genes) is especially disruptive to replication and promotes genomic instability. Here, we find that head-on collisions promote R-loop formation in Bacillus subtilis. We show that pervasive R-loop formation at head-on collision regions completely blocks replication, elevates mutagenesis, and inhibits gene expression. Accordingly, the activity of the R-loop processing enzyme RNase HIII at collision regions is crucial for stress survival in B. subtilis, as many stress response genes are head-on to replication. Remarkably, without RNase HIII, the ability of the intracellular pathogen Listeria monocytogenes to infect and replicate in hosts is weakened significantly, most likely because many virulence genes are head-on to replication. We conclude that the detrimental effects of head-on collisions stem primarily from excessive R-loop formation and that the resolution of these structures is critical for bacterial stress survival and pathogenesis.


Asunto(s)
Bacillus subtilis/fisiología , Replicación del ADN , Listeria monocytogenes/fisiología , Transcripción Genética , Animales , Momento de Replicación del ADN , Femenino , Expresión Génica , Técnicas de Inactivación de Genes , Listeria monocytogenes/genética , Listeria monocytogenes/patogenicidad , Listeriosis/microbiología , Ratones , Estrés Fisiológico , Virulencia
5.
Nat Immunol ; 20(4): 433-446, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30804553

RESUMEN

Cells use mitophagy to remove damaged or unwanted mitochondria to maintain homeostasis. Here we report that the intracellular bacterial pathogen Listeria monocytogenes exploits host mitophagy to evade killing. We found that L. monocytogenes induced mitophagy in macrophages through the virulence factor listeriolysin O (LLO). We discovered that NLRX1, the only Nod-like receptor (NLR) family member with a mitochondrial targeting sequence, contains an LC3-interacting region (LIR) and directly associated with LC3 through the LIR. NLRX1 and its LIR motif were essential for L. monocytogenes-induced mitophagy. NLRX1 deficiency and use of a mitophagy inhibitor both increased mitochondrial production of reactive oxygen species and thereby suppressed the survival of L. monocytogenes. Mechanistically, L. monocytogenes and LLO induced oligomerization of NLRX1 to promote binding of its LIR motif to LC3 for induction of mitophagy. Our study identifies NLRX1 as a novel mitophagy receptor and discovers a previously unappreciated strategy used by pathogens to hijack a host cell homeostasis system for their survival.


Asunto(s)
Listeria monocytogenes/fisiología , Proteínas Mitocondriales/fisiología , Mitofagia , Animales , Autofagia , Toxinas Bacterianas/metabolismo , Línea Celular , Femenino , Proteínas de Choque Térmico/metabolismo , Proteínas Hemolisinas/metabolismo , Humanos , Listeria monocytogenes/patogenicidad , Listeriosis/metabolismo , Listeriosis/microbiología , Macrófagos/microbiología , Macrófagos/ultraestructura , Masculino , Ratones , Ratones Noqueados , Viabilidad Microbiana , Mitocondrias/metabolismo , Mitocondrias/ultraestructura , Proteínas Mitocondriales/química , Proteínas Mitocondriales/genética , Proteínas Mitocondriales/metabolismo , Dominios Proteicos , Especies Reactivas de Oxígeno/metabolismo , Factores de Virulencia/metabolismo
6.
Nature ; 603(7903): 900-906, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-35296858

RESUMEN

Infections of the central nervous system are among the most serious infections1,2, but the mechanisms by which pathogens access the brain remain poorly understood. The model microorganism Listeria monocytogenes (Lm) is a major foodborne pathogen that causes neurolisteriosis, one of the deadliest infections of the central nervous system3,4. Although immunosuppression is a well-established host risk factor for neurolisteriosis3,5, little is known about the bacterial factors that underlie the neuroinvasion of Lm. Here we develop a clinically relevant experimental model of neurolisteriosis, using hypervirulent neuroinvasive strains6 inoculated in a humanized mouse model of infection7, and we show that the bacterial surface protein InlB protects infected monocytes from Fas-mediated cell death by CD8+ T cells in a manner that depends on c-Met, PI3 kinase and FLIP. This blockade of specific anti-Lm cellular immune killing lengthens the lifespan of infected monocytes, and thereby favours the transfer of Lm from infected monocytes to the brain. The intracellular niche that is created by InlB-mediated cell-autonomous immune resistance also promotes Lm faecal shedding, which accounts for the selection of InlB as a core virulence gene of Lm. We have uncovered a specific mechanism by which a bacterial pathogen confers an increased lifespan to the cells it infects by rendering them resistant to cell-mediated immunity. This promotes the persistence of Lm within the host, its dissemination to the central nervous system and its transmission.


Asunto(s)
Enfermedades del Sistema Nervioso Central , Listeria monocytogenes , Listeriosis , Animales , Proteínas Bacterianas/metabolismo , Linfocitos T CD8-positivos/metabolismo , Enfermedades del Sistema Nervioso Central/microbiología , Modelos Animales de Enfermedad , Listeria monocytogenes/patogenicidad , Listeriosis/microbiología , Ratones , Monocitos , Virulencia
7.
Nature ; 590(7846): 457-462, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33568812

RESUMEN

In contrast to nearly all other tissues, the anatomy of cell differentiation in the bone marrow remains unknown. This is owing to a lack of strategies for examining myelopoiesis-the differentiation of myeloid progenitors into a large variety of innate immune cells-in situ in the bone marrow. Such strategies are required to understand differentiation and lineage-commitment decisions, and to define how spatial organizing cues inform tissue function. Here we develop approaches for imaging myelopoiesis in mice, and generate atlases showing the differentiation of granulocytes, monocytes and dendritic cells. The generation of granulocytes and dendritic cells-monocytes localizes to different blood-vessel structures known as sinusoids, and displays lineage-specific spatial and clonal architectures. Acute systemic infection with Listeria monocytogenes induces lineage-specific progenitor clusters to undergo increased self-renewal of progenitors, but the different lineages remain spatially separated. Monocyte-dendritic cell progenitors (MDPs) map with nonclassical monocytes and conventional dendritic cells; these localize to a subset of blood vessels expressing a major regulator of myelopoiesis, colony-stimulating factor 1 (CSF1, also known as M-CSF)1. Specific deletion of Csf1 in endothelium disrupts the architecture around MDPs and their localization to sinusoids. Subsequently, there are fewer MDPs and their ability to differentiate is reduced, leading to a loss of nonclassical monocytes and dendritic cells during both homeostasis and infection. These data indicate that local cues produced by distinct blood vessels are responsible for the spatial organization of definitive blood cell differentiation.


Asunto(s)
Rastreo Celular/métodos , Células Mieloides/citología , Mielopoyesis , Coloración y Etiquetado/métodos , Animales , Atlas como Asunto , Vasos Sanguíneos/citología , Vasos Sanguíneos/metabolismo , Linaje de la Célula , Autorrenovación de las Células , Células Dendríticas/citología , Endotelio Vascular/citología , Endotelio Vascular/metabolismo , Femenino , Granulocitos/citología , Listeria monocytogenes/patogenicidad , Listeriosis/microbiología , Factor Estimulante de Colonias de Macrófagos/deficiencia , Factor Estimulante de Colonias de Macrófagos/genética , Factor Estimulante de Colonias de Macrófagos/metabolismo , Masculino , Ratones , Monocitos/citología , Células Mieloides/metabolismo
8.
Proc Natl Acad Sci U S A ; 119(13): e2122173119, 2022 03 29.
Artículo en Inglés | MEDLINE | ID: mdl-35316134

RESUMEN

Flavin mononucleotide (FMN) and flavin adenine dinucleotide (FAD) are essential riboflavin-derived cofactors involved in a myriad of redox reactions across all forms of life. Nevertheless, the basis of flavin acquisition strategies by riboflavin auxotrophic pathogens remains poorly defined. In this study, we examined how the facultative intracellular pathogen Listeria monocytogenes, a riboflavin auxotroph, acquires flavins during infection. A L. monocytogenes mutant lacking the putative riboflavin transporter (RibU) was completely avirulent in mice but had no detectable growth defect in nutrient-rich media. However, unlike wild type, the RibU mutant was unable to grow in defined media supplemented with FMN or FAD or to replicate in macrophages starved for riboflavin. Consistent with RibU functioning to scavenge FMN and FAD inside host cells, a mutant unable to convert riboflavin to FMN or FAD retained virulence and grew in cultured macrophages and in spleens and livers of infected mice. However, this FMN- and FAD-requiring strain was unable to grow in the gallbladder or intestines, where L. monocytogenes normally grows extracellularly, suggesting that these sites do not contain sufficient flavin cofactors to promote replication. Thus, by deleting genes required to synthesize FMN and FAD, we converted L. monocytogenes from a facultative to an obligate intracellular pathogen. Collectively, these data indicate that L. monocytogenes requires riboflavin to grow extracellularly in vivo but scavenges FMN and FAD to grow in host cells.


Asunto(s)
Proteínas Bacterianas , Mononucleótido de Flavina , Flavina-Adenina Dinucleótido , Listeria monocytogenes , Proteínas de Transporte de Membrana , Riboflavina , Proteínas Bacterianas/metabolismo , Mononucleótido de Flavina/metabolismo , Flavina-Adenina Dinucleótido/metabolismo , Listeria monocytogenes/crecimiento & desarrollo , Listeria monocytogenes/metabolismo , Listeria monocytogenes/patogenicidad , Proteínas de Transporte de Membrana/metabolismo , Riboflavina/metabolismo
9.
Infect Immun ; 92(6): e0016224, 2024 Jun 11.
Artículo en Inglés | MEDLINE | ID: mdl-38752742

RESUMEN

Ethanolamine (EA) affects the colonization and pathogenicity of certain human bacterial pathogens in the gastrointestinal tract. However, EA can also affect the intracellular survival and replication of host cell invasive bacteria such as Listeria monocytogenes (LMO) and Salmonella enterica serovar Typhimurium (S. Typhimurium). The EA utilization (eut) genes can be categorized as regulatory, enzymatic, or structural, and previous work in LMO showed that loss of genes encoding functions for the enzymatic breakdown of EA inhibited LMO intracellular replication. In this work, we sought to further characterize the role of EA utilization during LMO infection of host cells. Unlike what was previously observed for S. Typhimurium, in LMO, an EA regulator mutant (ΔeutV) was equally deficient in intracellular replication compared to an EA metabolism mutant (ΔeutB), and this was consistent across Caco-2, RAW 264.7, and THP-1 cell lines. The structural genes encode proteins that self-assemble into bacterial microcompartments (BMCs) that encase the enzymes necessary for EA metabolism. For the first time, native EUT BMCs were fluorescently tagged, and EUT BMC formation was observed in vitro and in vivo. Interestingly, BMC formation was observed in bacteria infecting Caco-2 cells, but not the macrophage cell lines. Finally, the cellular immune response of Caco-2 cells to infection with eut mutants was examined, and it was discovered that ΔeutB and ΔeutV mutants similarly elevated the expression of inflammatory cytokines. In conclusion, EA sensing and utilization during LMO intracellular infection are important for optimal LMO replication and immune evasion but are not always concomitant with BMC formation.IMPORTANCEListeria monocytogenes (LMO) is a bacterial pathogen that can cause severe disease in immunocompromised individuals when consumed in contaminated food. It can replicate inside of mammalian cells, escaping detection by the immune system. Therefore, understanding the features of this human pathogen that contribute to its infectiousness and intracellular lifestyle is important. In this work we demonstrate that genes encoding both regulators and enzymes of EA metabolism are important for optimal growth inside mammalian cells. Moreover, the formation of specialized compartments to enable EA metabolism were visualized by tagging with a fluorescent protein and found to form when LMO infects some mammalian cell types, but not others. Interestingly, the formation of the compartments was associated with features consistent with an early stage of the intracellular infection. By characterizing bacterial metabolic pathways that contribute to survival in host environments, we hope to positively impact knowledge and facilitate new treatment strategies.


Asunto(s)
Etanolamina , Listeria monocytogenes , Listeriosis , Listeria monocytogenes/metabolismo , Listeria monocytogenes/crecimiento & desarrollo , Listeria monocytogenes/genética , Listeria monocytogenes/patogenicidad , Listeriosis/microbiología , Humanos , Etanolamina/metabolismo , Ratones , Animales , Células RAW 264.7 , Células CACO-2 , Células THP-1 , Proteínas Bacterianas/metabolismo , Proteínas Bacterianas/genética , Macrófagos/microbiología , Macrófagos/metabolismo
10.
PLoS Pathog ; 18(1): e1010166, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-35007292

RESUMEN

A hallmark of Listeria (L.) monocytogenes pathogenesis is bacterial escape from maturing entry vacuoles, which is required for rapid bacterial replication in the host cell cytoplasm and cell-to-cell spread. The bacterial transcriptional activator PrfA controls expression of key virulence factors that enable exploitation of this intracellular niche. The transcriptional activity of PrfA within infected host cells is controlled by allosteric coactivation. Inhibitory occupation of the coactivator site has been shown to impair PrfA functions, but consequences of PrfA inhibition for L. monocytogenes infection and pathogenesis are unknown. Here we report the crystal structure of PrfA with a small molecule inhibitor occupying the coactivator site at 2.0 Å resolution. Using molecular imaging and infection studies in macrophages, we demonstrate that PrfA inhibition prevents the vacuolar escape of L. monocytogenes and enables extensive bacterial replication inside spacious vacuoles. In contrast to previously described spacious Listeria-containing vacuoles, which have been implicated in supporting chronic infection, PrfA inhibition facilitated progressive clearance of intracellular L. monocytogenes from spacious vacuoles through lysosomal degradation. Thus, inhibitory occupation of the PrfA coactivator site facilitates formation of a transient intravacuolar L. monocytogenes replication niche that licenses macrophages to effectively eliminate intracellular bacteria. Our findings encourage further exploration of PrfA as a potential target for antimicrobials and highlight that intra-vacuolar residence of L. monocytogenes in macrophages is not inevitably tied to bacterial persistence.


Asunto(s)
Listeria monocytogenes/patogenicidad , Listeriosis/microbiología , Macrófagos/microbiología , Vacuolas/microbiología , Virulencia/fisiología , Animales , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL
11.
Cell ; 139(4): 770-9, 2009 Nov 13.
Artículo en Inglés | MEDLINE | ID: mdl-19914169

RESUMEN

Riboswitches are RNA elements acting in cis, controlling expression of their downstream genes through a metabolite-induced alteration of their secondary structure. Here, we demonstrate that two S-adenosylmethionine (SAM) riboswitches, SreA and SreB, can also function in trans and act as noncoding RNAs in Listeria monocytogenes. SreA and SreB control expression of the virulence regulator PrfA by binding to the 5'-untranslated region of its mRNA. Absence of the SAM riboswitches SreA and SreB increases the level of PrfA and virulence gene expression in L. monocytogenes. Thus, the impact of the SAM riboswitches on PrfA expression highlights a link between bacterial virulence and nutrient availability. Together, our results uncover an unexpected role for riboswitches and a distinct class of regulatory noncoding RNAs in bacteria.


Asunto(s)
Proteínas Bacterianas/genética , Listeria monocytogenes/genética , Factores de Terminación de Péptidos/genética , Secuencias Reguladoras de Ácido Ribonucleico , Regiones no Traducidas 5' , Secuencia de Bases , Escherichia coli/genética , Escherichia coli/metabolismo , Listeria monocytogenes/metabolismo , Listeria monocytogenes/patogenicidad , Datos de Secuencia Molecular , Temperatura , Virulencia
12.
Proc Natl Acad Sci U S A ; 118(16)2021 04 20.
Artículo en Inglés | MEDLINE | ID: mdl-33859042

RESUMEN

The transcription factor IRF4 is required for CD8+ T cell activation, proliferation, and differentiation to effector cells and thus is essential for robust CD8+ T cell responses. The function of IRF4 in memory CD8+ T cells yet needs to be explored. To investigate the role of IRF4 for maintaining differentiation state and survival of CD8+ memory T cells, we used a mouse model with tamoxifen-inducible Irf4 knockout to preclude effects due to inefficient memory cell differentiation in absence of IRF4. We infected mice with ovalbumin-recombinant listeria and induced Irf4 knockout after clearance of the pathogen. Loss of IRF4 resulted in phenotypical changes of CD8+ memory T cells but did not cause a reduction of the total memory T cell population. However, upon reencounter of the pathogen, CD8+ memory T cells showed impaired expansion and acquisition of effector functions. When compared to CD8+ effector memory T cells, CD8+ tissue-resident memory T cells (TRM cells) expressed higher IRF4 levels. Mice with constitutive Irf4 knockout had diminished CD8+ TRM-cell populations, and tamoxifen-induced Irf4 deletion caused a reduction of this cell population. In conclusion, our results demonstrate that IRF4 is required for effective reactivation but not for general survival of CD8+ memory T cells. Formation and maintenance of CD8+ TRM cells, in contrast, appear to depend on IRF4.


Asunto(s)
Linfocitos T CD8-positivos/fisiología , Memoria Inmunológica/fisiología , Factores Reguladores del Interferón/metabolismo , Animales , Linfocitos T CD8-positivos/metabolismo , Diferenciación Celular , Proliferación Celular , Femenino , Memoria Inmunológica/genética , Factores Reguladores del Interferón/genética , Factores Reguladores del Interferón/fisiología , Listeria monocytogenes/patogenicidad , Activación de Linfocitos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados
13.
Foodborne Pathog Dis ; 21(6): 386-394, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38346310

RESUMEN

Listeria monocytogenes is a critical foodborne pathogen that causes severe invasive and noninvasive diseases and is associated with high mortality. Information on the prevalence of L. monocytogenes infections in Taiwan is very limited. This study aimed to analyze the molecular epidemiological surveillance and virulence gene distribution of 176 human clinical L. monocytogenes isolates collected between 2009 and 2019 in northern Taiwan. Our results showed that the isolates belonged to 4 serogroups (IIa, IIb, IVb, and IIc), with most isolates in serogroups IIa (81/176, 46%) and IIb (71/176, 40.3%). Multilocus sequence typing analysis revealed 18 sequence types (STs) and 13 clonal complexes (CCs). Eighty-four percent of all isolates belonged to six STs: CC87-ST87 (40/176, 22.7%), CC19-ST378 (36/176, 19.9%), CC155-ST155 (28/176, 15.5%), CC1-ST710 (16/176, 8.8%), CC5-ST5 (16/176, 8.8%), and CC101-ST101 (11/176, 6.1%). Furthermore, our analysis showed the distributions of four Listeria pathogenicity islands (LIPI) among all isolates. LIPI-1 and LIPI-2 existed in all isolates, whereas LIPI-3 and LIPI-4 only existed in specific STs and CCs. LIPI-3 existed in the STs, CC1-ST710, CC3-ST3, CC288-ST295, and CC191-ST1458, whereas LIPI-4 could be found in the STs, CC87-ST87 and CC87-ST1459. Strains containing LIPI-3 and LIPI-4 are potentially hypervirulent; thus, 68/176 isolates (39.1%) collected in this study were potentially hypervirulent. Since L. monocytogenes infections are considered highly correlated with diet, molecular epidemiological surveillance of Listeria in food is important; continued surveillance will provide critical information to prevent foodborne diseases.


Asunto(s)
Listeria monocytogenes , Listeriosis , Tipificación de Secuencias Multilocus , Listeria monocytogenes/genética , Listeria monocytogenes/patogenicidad , Listeria monocytogenes/aislamiento & purificación , Listeria monocytogenes/clasificación , Taiwán/epidemiología , Humanos , Listeriosis/microbiología , Listeriosis/epidemiología , Virulencia/genética , Serogrupo , Factores de Virulencia/genética , Islas Genómicas , Enfermedades Transmitidas por los Alimentos/microbiología , Enfermedades Transmitidas por los Alimentos/epidemiología , Epidemiología Molecular
14.
PLoS Pathog ; 17(12): e1010173, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34929015

RESUMEN

For many intracellular bacterial pathogens manipulating host cell survival is essential for maintaining their replicative niche, and is a common strategy used to promote infection. The bacterial pathogen Listeria monocytogenes is well known to hijack host machinery for its own benefit, such as targeting the host histone H3 for modification by SIRT2. However, by what means this modification benefits infection, as well as the molecular players involved, were unknown. Here we show that SIRT2 activity supports Listeria intracellular survival by maintaining genome integrity and host cell viability. This protective effect is dependent on H3K18 deacetylation, which safeguards the host genome by counteracting infection-induced DNA damage. Mechanistically, infection causes SIRT2 to interact with the nucleic acid binding protein TDP-43 and localise to genomic R-loops, where H3K18 deacetylation occurs. This work highlights novel functions of TDP-43 and R-loops during bacterial infection and identifies the mechanism through which L. monocytogenes co-opts SIRT2 to allow efficient infection.


Asunto(s)
Histonas/metabolismo , Interacciones Huésped-Patógeno/fisiología , Listeria monocytogenes/patogenicidad , Listeriosis/metabolismo , Sirtuina 3/metabolismo , Animales , Supervivencia Celular/fisiología , Humanos , Listeria monocytogenes/metabolismo
15.
PLoS Pathog ; 17(10): e1009881, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34624065

RESUMEN

Pathogenic bacteria rely on protein phosphorylation to adapt quickly to stress, including that imposed by the host during infection. Penicillin-binding protein and serine/threonine-associated (PASTA) kinases are signal transduction systems that sense cell wall integrity and modulate multiple facets of bacterial physiology in response to cell envelope stress. The PASTA kinase in the cytosolic pathogen Listeria monocytogenes, PrkA, is required for cell wall stress responses, cytosolic survival, and virulence, yet its substrates and downstream signaling pathways remain incompletely defined. We combined orthogonal phosphoproteomic and genetic analyses in the presence of a ß-lactam antibiotic to define PrkA phosphotargets and pathways modulated by PrkA. These analyses synergistically highlighted ReoM, which was recently identified as a PrkA target that influences peptidoglycan (PG) synthesis, as an important phosphosubstrate during cell wall stress. We find that deletion of reoM restores cell wall stress sensitivities and cytosolic survival defects of a ΔprkA mutant to nearly wild-type levels. While a ΔprkA mutant is defective for PG synthesis during cell wall stress, a double ΔreoM ΔprkA mutant synthesizes PG at rates similar to wild type. In a mouse model of systemic listeriosis, deletion of reoM in a ΔprkA background almost fully restored virulence to wild-type levels. However, loss of reoM alone also resulted in attenuated virulence, suggesting ReoM is critical at some points during pathogenesis. Finally, we demonstrate that the PASTA kinase/ReoM cell wall stress response pathway is conserved in a related pathogen, methicillin-resistant Staphylococcus aureus. Taken together, our phosphoproteomic analysis provides a comprehensive overview of the PASTA kinase targets of an important model pathogen and suggests that a critical role of PrkA in vivo is modulating PG synthesis through regulation of ReoM to facilitate cytosolic survival and virulence.


Asunto(s)
Pared Celular/fisiología , Listeria monocytogenes/metabolismo , Listeria monocytogenes/patogenicidad , Peptidoglicano/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Animales , Listeriosis/metabolismo , Ratones , Ratones Endogámicos C57BL , Virulencia
16.
PLoS Pathog ; 17(8): e1009379, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-34398937

RESUMEN

The Gram-positive bacterium Listeria monocytogenes is the causative agent of the foodborne disease listeriosis, one of the deadliest bacterial infections known. In order to cause disease, L. monocytogenes must properly coordinate its metabolic and virulence programs in response to rapidly changing environments within the host. However, the mechanisms by which L. monocytogenes senses and adapts to the many stressors encountered as it transits through the gastrointestinal (GI) tract and disseminates to peripheral organs are not well understood. In this study, we investigated the role of the redox-responsive transcriptional regulator Rex in L. monocytogenes growth and pathogenesis. Rex is a conserved canonical transcriptional repressor that monitors the intracellular redox state of the cell by sensing the ratio of reduced and oxidized nicotinamide adenine dinucleotides (NADH and NAD+, respectively). Here, we demonstrated that L. monocytogenes Rex represses fermentative metabolism and is therefore required for optimal growth in the presence of oxygen. We also show that in vitro, Rex represses the production of virulence factors required for survival and invasion of the GI tract, as a strain lacking rex was more resistant to acidified bile and invaded host cells better than wild type. Consistent with these results, Rex was dispensable for colonizing the GI tract and disseminating to peripheral organs in an oral listeriosis model of infection. However, Rex-dependent regulation was required for colonizing the spleen and liver, and L. monocytogenes lacking the Rex repressor were nearly sterilized from the gallbladder. Taken together, these results demonstrated that Rex functions as a repressor of fermentative metabolism and suggests a role for Rex-dependent regulation in L. monocytogenes pathogenesis. Importantly, the gallbladder is the bacterial reservoir during listeriosis, and our data suggest redox sensing and Rex-dependent regulation are necessary for bacterial survival and replication in this organ.


Asunto(s)
Proteínas Bacterianas/metabolismo , Fermentación , Productos del Gen rex/metabolismo , Listeria monocytogenes/patogenicidad , Listeriosis/microbiología , Factores de Virulencia/metabolismo , Virulencia , Animales , Proteínas Bacterianas/genética , Femenino , Regulación Bacteriana de la Expresión Génica , Productos del Gen rex/genética , Listeriosis/metabolismo , Listeriosis/patología , Ratones , Ratones Endogámicos BALB C , Oxidación-Reducción , Transcriptoma , Factores de Virulencia/genética
17.
PLoS Pathog ; 17(4): e1009531, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33878120

RESUMEN

Most individuals who consume foods contaminated with the bacterial pathogen Listeria monocytogenes (Lm) develop mild symptoms, while others are susceptible to life-threatening systemic infections (listeriosis). Although it is known that the risk of severe disease is increased in certain human populations, including the elderly, it remains unclear why others who consume contaminated food develop listeriosis. Here, we used a murine model to discover that pulmonary coinfections can impair the host's ability to adequately control and eradicate systemic Lm that cross from the intestines to the bloodstream. We found that the resistance of mice to oral Lm infection was dramatically reduced by coinfection with Streptococcus pneumoniae (Spn), a bacterium that colonizes the respiratory tract and can also cause severe infections in the elderly. Exposure to Spn or microbial products, including a recombinant Lm protein (L1S) and lipopolysaccharide (LPS), rendered otherwise resistant hosts susceptible to severe systemic Lm infection. In addition, we show that this increase in susceptibility was dependent on an increase in the production of interleukin-10 (IL-10) from Ncr1+ cells, including natural killer (NK) cells. Lastly, the ability of Ncr1+ cell derived IL-10 to increase disease susceptibility correlated with a dampening of both myeloid cell accumulation and myeloid cell phagocytic capacity in infected tissues. These data suggest that efforts to minimize inflammation in response to an insult at the respiratory mucosa render the host more susceptible to infections by Lm and possibly other pathogens that access the oral mucosa.


Asunto(s)
Listeria monocytogenes/inmunología , Listeriosis/inmunología , Neumonía/inmunología , Animales , Progresión de la Enfermedad , Susceptibilidad a Enfermedades , Femenino , Interleucina-10/metabolismo , Células Asesinas Naturales/metabolismo , Células Asesinas Naturales/fisiología , Lipopolisacáridos , Listeria monocytogenes/patogenicidad , Listeriosis/complicaciones , Listeriosis/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Enfermedades de la Boca/complicaciones , Enfermedades de la Boca/inmunología , Enfermedades de la Boca/microbiología , Enfermedades de la Boca/patología , Neumonía/complicaciones , Neumonía/etiología , Neumonía/patología
18.
Proc Natl Acad Sci U S A ; 117(38): 23774-23781, 2020 09 22.
Artículo en Inglés | MEDLINE | ID: mdl-32878997

RESUMEN

Intracellular pathogens are responsible for an enormous amount of worldwide morbidity and mortality, and each has evolved specialized strategies to establish and maintain their replicative niche. Listeria monocytogenes is a facultative intracellular pathogen that secretes a pore-forming cytolysin called listeriolysin O (LLO), which disrupts the phagosomal membrane and, thereby, allows the bacteria access to their replicative niche in the cytosol. Nonsynonymous and synonymous mutations in a PEST-like domain near the LLO N terminus cause enhanced LLO translation during intracellular growth, leading to host cell death and loss of virulence. Here, we explore the mechanism of translational control and show that there is extensive codon restriction within the PEST-encoding region of the LLO messenger RNA (mRNA) (hly). This region has considerable complementarity with the 5' UTR and is predicted to form an extensive secondary structure that overlaps the ribosome binding site. Analysis of both 5' UTR and synonymous mutations in the PEST-like domain that are predicted to disrupt the secondary structure resulted in up to a 10,000-fold drop in virulence during mouse infection, while compensatory double mutants restored virulence to WT levels. We showed by dynamic protein radiolabeling that LLO synthesis was growth phase-dependent. These data provide a mechanism to explain how the bacteria regulate translation of LLO to promote translation during starvation in a phagosome while repressing it during growth in the cytosol. These studies also provide a molecular explanation for codon bias at the 5' end of this essential determinant of pathogenesis.


Asunto(s)
Toxinas Bacterianas , Proteínas de Choque Térmico , Proteínas Hemolisinas , Listeria monocytogenes , ARN Bacteriano/química , ARN Mensajero/química , Regiones no Traducidas 5'/genética , Animales , Toxinas Bacterianas/química , Toxinas Bacterianas/genética , Toxinas Bacterianas/metabolismo , Replicación del ADN/genética , Proteínas de Choque Térmico/química , Proteínas de Choque Térmico/genética , Proteínas de Choque Térmico/metabolismo , Proteínas Hemolisinas/química , Proteínas Hemolisinas/genética , Proteínas Hemolisinas/metabolismo , Listeria monocytogenes/genética , Listeria monocytogenes/patogenicidad , Listeriosis , Ratones , Conformación de Ácido Nucleico , ARN Bacteriano/genética , ARN Mensajero/genética
19.
Int J Mol Sci ; 24(5)2023 Feb 28.
Artículo en Inglés | MEDLINE | ID: mdl-36902108

RESUMEN

We explored the antimicrobial activity of sertraline on Listeria monocytogenes and further investigated the effects of sertraline on biofilm formation and the virulence gene expression of L. monocytogenes. The minimum inhibitory concentration and minimum bactericidal concentration for sertraline against L. monocytogenes were in the range of 16-32 µg/mL and 64 µg/mL, respectively. Sertraline-dependent damage of the cell membrane and a decrease in intracellular ATP and pHin in L. monocytogenes were observed. In addition, sertraline reduced the biofilm formation efficiency of the L. monocytogenes strains. Importantly, low concentrations (0.1 µg/mL and 1 µg/mL) of sertraline significantly down-regulated the expression levels of various L. monocytogens virulence genes (prfA, actA, degU, flaA, sigB, ltrC and sufS). These results collectively suggest a role of sertraline for the control of L. monocytogenes in the food industry.


Asunto(s)
Antiinfecciosos , Proteínas Bacterianas , Listeria monocytogenes , Sertralina , Factores de Virulencia , Antiinfecciosos/farmacología , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Regulación Bacteriana de la Expresión Génica/efectos de los fármacos , Listeria monocytogenes/efectos de los fármacos , Listeria monocytogenes/genética , Listeria monocytogenes/patogenicidad , Sertralina/farmacología , Virulencia/efectos de los fármacos , Virulencia/genética , Factores de Virulencia/genética , Factores de Virulencia/metabolismo
20.
Can Vet J ; 64(4): 363-366, 2023 04.
Artículo en Inglés | MEDLINE | ID: mdl-37008639

RESUMEN

Clinical disease caused by infection with Listeria monocytogenes is rare in adult horses, and there is a paucity of ante-mortem clinicopathologic changes for this species reported in the literature. Confirmatory diagnosis is difficult and often requires post-mortem sampling of the brainstem. This report details a case of meningoencephalitis caused by Listeria monocytogenes in an adult American quarter horse gelding presenting with central neurologic signs. Pre-mortem analysis of the cerebrospinal fluid revealed a mononuclear, primarily lymphocytic, pleocytosis, which is a reported finding in other species with listeriosis. Post-mortem histopathologic changes of the brainstem were characteristic of listeriosis, and infection was confirmed with immunohistochemical labeling and bacterial culture. Key clinical message: Listeriosis should be included as a differential diagnosis in neurologic horses with mononuclear pleocytosis identified on cerebrospinal fluid analysis.


Pléocytose mononucléaire et méningo-encéphalite causées par Listeria monocytogenes chez un cheval adulte. La maladie clinique causée par une infection à L. monocytogenes est rare chez les chevaux adultes, et il y a peu de changements clinico-pathologiques ante-mortem rapportés dans la littérature pour cette espèce. Le diagnostic de confirmation est difficile et nécessite souvent un prélèvement post-mortem du tronc cérébral. Ce rapport détaille un cas de méningo-encéphalite causée par L. monocytogenes chez un hongre quarter horse américain adulte présentant des signes neurologiques centraux. L'analyse pré-mortem du liquide céphalo-rachidien a révélé une pléocytose mononucléaire, principalement lymphocytaire, qui est une trouvaille rapportée chez d'autres espèces atteintes de listériose. Les modifications histopathologiques post-mortem du tronc cérébral étaient caractéristiques de la listériose et l'infection a été confirmée par un marquage immunohistochimique et une culture bactérienne.Message clinique clé :La listériose doit être incluse comme diagnostic différentiel chez les chevaux avec signes neurologiques présentant une pléocytose mononucléaire identifiée lors de l'analyse du liquide céphalo-rachidien.(Traduit par Dr Serge Messier).


Asunto(s)
Enfermedades de los Caballos , Listeriosis , Meningoencefalitis , Animales , Masculino , Diagnóstico Diferencial , Enfermedades de los Caballos/diagnóstico , Enfermedades de los Caballos/microbiología , Caballos , Leucocitosis/diagnóstico , Leucocitosis/veterinaria , Listeria monocytogenes/patogenicidad , Listeriosis/diagnóstico , Listeriosis/veterinaria , Meningoencefalitis/diagnóstico , Meningoencefalitis/microbiología , Meningoencefalitis/veterinaria , Líquido Cefalorraquídeo/citología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA