Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
2.
Gut Microbes ; 14(1): 2058851, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35373699

RESUMEN

Enterococcus faecalis is a bacterial species present at a subdominant level in the human gut microbiota. This commensal turns into an opportunistic pathogen under specific conditions involving dysbiosis and host immune deficiency. E. faecalis is one of the rare pathobionts identified to date as contributing to liver damage in alcoholic liver disease. We have previously observed that E. faecalis is internalized in hepatocytes. Here, the survival and fate of E. faecalis was examined in hepatocytes, the main epithelial cell type in the liver. Although referred to as an extracellular pathogen, we demonstrate that E. faecalis is able to survive and divide in hepatocytes, and form intracellular clusters in two distinct hepatocyte cell lines, in primary mouse hepatocytes, as well as in vivo. This novel process extends to kidney cells. Unraveling the intracellular lifestyle of E. faecalis, our findings contribute to the understanding of pathobiont-driven diseases.


Asunto(s)
Enterococcus faecalis , Microbioma Gastrointestinal , Animales , Disbiosis , Hepatocitos , Estilo de Vida , Ratones
3.
Front Cell Infect Microbiol ; 11: 761945, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34858876

RESUMEN

Listeria monocytogenes causes severe foodborne illness in pregnant women and immunocompromised individuals. After the intestinal phase of infection, the liver plays a central role in the clearance of this pathogen through its important functions in immunity. However, recent evidence suggests that during long-term infection of hepatocytes, a subpopulation of Listeria may escape eradication by entering a persistence phase in intracellular vacuoles. Here, we examine whether this long-term infection alters hepatocyte defense pathways, which may be instrumental for bacterial persistence. We first optimized cell models of persistent infection in human hepatocyte cell lines HepG2 and Huh7 and primary mouse hepatocytes (PMH). In these cells, Listeria efficiently entered the persistence phase after three days of infection, while inducing a potent interferon response, of type I in PMH and type III in HepG2, while Huh7 remained unresponsive. RNA-sequencing analysis identified a common signature of long-term Listeria infection characterized by the overexpression of a set of genes involved in antiviral immunity and the under-expression of many acute phase protein (APP) genes, particularly involved in the complement and coagulation systems. Infection also altered the expression of cholesterol metabolism-associated genes in HepG2 and Huh7 cells. The decrease in APP transcripts was correlated with lower protein abundance in the secretome of infected cells, as shown by proteomics, and also occurred in the presence of APP inducers (IL-6 or IL-1ß). Collectively, these results reveal that long-term infection with Listeria profoundly deregulates the innate immune functions of hepatocytes, which could generate an environment favorable to the establishment of persistent infection.


Asunto(s)
Listeria monocytogenes , Listeria , Listeriosis , Animales , Femenino , Hepatocitos , Humanos , Listeria monocytogenes/genética , Ratones , Infección Persistente , Embarazo , Secretoma
4.
PLoS Pathog ; 17(9): e1009880, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34529737

RESUMEN

Staphylococcus aureus is a human commensal organism and opportunist pathogen, causing potentially fatal disease. The presence of non-pathogenic microflora or their components, at the point of infection, dramatically increases S. aureus pathogenicity, a process termed augmentation. Augmentation is associated with macrophage interaction but by a hitherto unknown mechanism. Here, we demonstrate a breadth of cross-kingdom microorganisms can augment S. aureus disease and that pathogenesis of Enterococcus faecalis can also be augmented. Co-administration of augmenting material also forms an efficacious vaccine model for S. aureus. In vitro, augmenting material protects S. aureus directly from reactive oxygen species (ROS), which correlates with in vivo studies where augmentation restores full virulence to the ROS-susceptible, attenuated mutant katA ahpC. At the cellular level, augmentation increases bacterial survival within macrophages via amelioration of ROS, leading to proliferation and escape. We have defined the molecular basis for augmentation that represents an important aspect of the initiation of infection.


Asunto(s)
Interacciones Huésped-Patógeno/inmunología , Macrófagos/inmunología , Infecciones Estafilocócicas/microbiología , Staphylococcus aureus/inmunología , Simbiosis/fisiología , Animales , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Especies Reactivas de Oxígeno/metabolismo , Sepsis/inmunología , Sepsis/microbiología , Infecciones Estafilocócicas/inmunología , Pez Cebra
5.
Gut Microbes ; 11(4): 868-881, 2020 07 03.
Artículo en Inglés | MEDLINE | ID: mdl-31955655

RESUMEN

The capacity of bacterial pathogens to infect their hosts depends on the tight spatiotemporal regulation of virulence genes. The Listeria monocytogenes (Lm) metal efflux pump repressor CadC is highly expressed during late infection stages, modulating lipoprotein processing and host immune response. Here we investigate the potential of CadC as broad repressor of virulence genes. We show that CadC represses the expression of the bile salt hydrolase impairing Lm resistance to bile. During late infection, in absence of CadC-dependent repression, the constitutive bile salt hydrolase expression induces the overexpression of the cholic acid efflux pump MdrT that is unfavorable to Lm virulence. We establish the CadC regulon and show that CadC represses additional virulence factors activated by σB during colonization of the intestinal lumen. CadC is thus a general repressor that promotes Lm virulence by down-regulating, at late infection stages, genes required for survival in the gastrointestinal tract. This demonstrates for the first time how bacterial pathogens can repurpose regulators to spatiotemporally repress virulence genes and optimize their infectious capacity.


Asunto(s)
Proteínas Bacterianas/metabolismo , Listeria monocytogenes/genética , Listeria monocytogenes/patogenicidad , Listeriosis/microbiología , Factores de Virulencia/metabolismo , Amidohidrolasas/genética , Amidohidrolasas/metabolismo , Animales , Proteínas Bacterianas/genética , Ácidos y Sales Biliares/metabolismo , Ácidos y Sales Biliares/farmacología , Ácido Cólico/metabolismo , Farmacorresistencia Bacteriana , Femenino , Tracto Gastrointestinal/microbiología , Regulación Bacteriana de la Expresión Génica , Genes Bacterianos , Genes MDR , Listeria monocytogenes/efectos de los fármacos , Listeria monocytogenes/fisiología , Ratones , Ratones Endogámicos C57BL , Regulón , Virulencia/genética , Factores de Virulencia/genética
6.
Sci Rep ; 9(1): 8926, 2019 06 20.
Artículo en Inglés | MEDLINE | ID: mdl-31222056

RESUMEN

Enterococci are subdominant members of the human gastrointestinal microbiota. Enterococcus faecalis is generally harmless for healthy individuals, but it can cause a diverse range of infections in immunodeficient or elderly patients with severe underlying diseases. In this study, we analysed the levels of intestinal translocation of indigenous enterococci in C57BL/6, CF-1 and CX3CR1-/- mice upon clindamycin antibiotic-induced dysbiosis. We found that C57BL/6 was the most permissive model for enterococcal translocation and that initiation of E. faecalis translocation coincided with a threshold of enterococcal colonisation in the gut lumen, which once reached, triggered E. faecalis dissemination to deeper organs. We showed that the extent to which E. faecalis clinical strain VE14821 competed with indigenous enterococci differed between the C57BL/6 and CX3CR1-/- models. Finally, using a simplified gnotobiotic model, we observed E. faecalis crossing an intact intestinal tract using intestinal epithelial cells as one route to reach the lamina propria. Our study opens new perspectives for assessing the effect of various immunodeficiencies and for investigating mechanisms underlying enterococcal translocation.


Asunto(s)
Enterococcus/crecimiento & desarrollo , Microbioma Gastrointestinal , Animales , Transporte Biológico , Receptor 1 de Quimiocinas CX3C/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados
7.
Sci Rep ; 8(1): 1749, 2018 01 29.
Artículo en Inglés | MEDLINE | ID: mdl-29379180

RESUMEN

Enterococcus faecalis, an organism generally not pathogenic for healthy humans, has the potential to cause disease in susceptible hosts. While it seems to be equipped to interact with and circumvent host immune defense, most of the molecular and cellular mechanisms underlying the enterococcal infectious process remain elusive. Here, we investigated the role of the Enterococcal Leucine Rich protein A (ElrA), an internalin-like protein of E. faecalis also known as a virulence factor. ElrA was previously shown to prevent adhesion to macrophages. We show that ElrA does not inhibit the basic phagocytic process, but is able to prevent sensing and migration of macrophages toward E. faecalis. Presence or absence of FHL2, a eukaryotic partner of ElrA, does not affect the ElrA-dependent mechanism preventing macrophage migration. However, we highlight a partial contribution of FHL2 in ElrA-mediated virulence in vivo. Our results indicate that ElrA plays at least a dual role of which anti-phagocytic activity may contribute to dissemination of extracellular E. faecalis during infection.


Asunto(s)
Enterococcus faecalis/metabolismo , Infecciones por Bacterias Grampositivas/microbiología , Proteína Estafilocócica A/metabolismo , Factores de Virulencia/metabolismo , Virulencia/fisiología , Animales , Proteínas Bacterianas/metabolismo , Células CACO-2 , Línea Celular , Línea Celular Tumoral , Células HeLa , Células Hep G2 , Humanos , Leucina/metabolismo , Macrófagos/metabolismo , Macrófagos/microbiología , Ratones , Células RAW 264.7
8.
J Proteome Res ; 13(12): 6046-57, 2014 Dec 05.
Artículo en Inglés | MEDLINE | ID: mdl-25383790

RESUMEN

Protein phosphorylation is a major mechanism of signal transduction in bacteria. Here, we analyzed the proteome and phosphoproteome of a wild-type strain of the food-borne pathogen Listeria monocytogenes that was grown in either chemically defined medium or rich medium containing glucose. We then compared these results with those obtained from an isogenic prfA* mutant that produced a constitutively active form of PrfA, the main transcriptional activator of virulence genes. In the prfA* mutant grown in rich medium, we identified 256 peptides that were phosphorylated on serine (S), threonine (T), or tyrosine (Y) residues, with a S/T/Y ratio of 155:75:12. Strikingly, we detected five novel phosphosites on the virulence protein ActA. This protein was known to be phosphorylated by a cellular kinase in the infected host, but phosphorylation by a listerial kinase had not previously been reported. Unexpectedly, SILAC experiments with the prfA* mutant grown in chemically defined medium revealed that, in addition to previously described PrfA-regulated proteins, several other proteins were significantly overproduced, among them were several proteins involved in purine biosynthesis. This work provides new information for our understanding of the correlation among protein phosphorylation, virulence mechanisms, and carbon metabolism.


Asunto(s)
Proteínas Bacterianas/metabolismo , Listeria monocytogenes/metabolismo , Factores de Terminación de Péptidos/metabolismo , Fosfoproteínas/metabolismo , Proteoma/metabolismo , Proteómica/métodos , Proteínas Bacterianas/análisis , Proteínas Bacterianas/genética , Sitios de Unión/genética , Cromatografía Liquida , Medios de Cultivo/química , Medios de Cultivo/farmacología , Glucosa/farmacología , Listeria monocytogenes/genética , Listeria monocytogenes/patogenicidad , Mutación , Factores de Terminación de Péptidos/análisis , Factores de Terminación de Péptidos/genética , Péptidos/análisis , Péptidos/genética , Péptidos/metabolismo , Fosfoproteínas/análisis , Fosfoproteínas/genética , Fosforilación/efectos de los fármacos , Proteoma/análisis , Proteoma/genética , Purinas/biosíntesis , Serina/genética , Serina/metabolismo , Espectrometría de Masas en Tándem , Treonina/genética , Treonina/metabolismo , Tirosina/genética , Tirosina/metabolismo , Virulencia/genética
9.
mBio ; 5(2): e00969-14, 2014 Mar 25.
Artículo en Inglés | MEDLINE | ID: mdl-24667708

RESUMEN

For nearly 3 decades, listeriologists and immunologists have used mainly three strains of the same serovar (1/2a) to analyze the virulence of the bacterial pathogen Listeria monocytogenes. The genomes of two of these strains, EGD-e and 10403S, were released in 2001 and 2008, respectively. Here we report the genome sequence of the third reference strain, EGD, and extensive genomic and phenotypic comparisons of the three strains. Strikingly, EGD-e is genetically highly distinct from EGD (29,016 single nucleotide polymorphisms [SNPs]) and 10403S (30,296 SNPs), and is more related to serovar 1/2c than 1/2a strains. We also found that while EGD and 10403S strains are genetically very close (317 SNPs), EGD has a point mutation in the transcriptional regulator PrfA (PrfA*), leading to constitutive expression of several major virulence genes. We generated an EGD-e PrfA* mutant and showed that EGD behaves like this strain in vitro, with slower growth in broth and higher invasiveness in human cells than those of EGD-e and 10403S. In contrast, bacterial counts in blood, liver, and spleen during infection in mice revealed that EGD and 10403S are less virulent than EGD-e, which is itself less virulent than EGD-e PrfA*. Thus, constitutive expression of PrfA-regulated virulence genes does not appear to provide a significant advantage to the EGD strain during infection in vivo, highlighting the fact that in vitro invasion assays are not sufficient for evaluating the pathogenic potential of L. monocytogenes strains. Together, our results pave the way for deciphering unexplained differences or discrepancies in experiments using different L. monocytogenes strains. IMPORTANCE Over the past 3 decades, Listeria has become a model organism for host-pathogen interactions, leading to critical discoveries in a broad range of fields, including bacterial gene regulation, cell biology, and bacterial pathophysiology. Scientists studying Listeria use primarily three pathogenic strains: EGD, EGD-e, and 10403S. Despite many studies on EGD, it is the only one of the three strains whose genome has not been sequenced. Here we report the sequence of its genome and a series of important genomic and phenotypic differences between the three strains, in particular, a critical mutation in EGD's PrfA, the main regulator of Listeria virulence. Our results show that the three strains display differences which may play an important role in the virulence differences observed between the strains. Our findings will be of critical relevance to listeriologists and immunologists who have used or may use Listeria as a tool to study the pathophysiology of listeriosis and immune responses.


Asunto(s)
ADN Bacteriano/química , ADN Bacteriano/genética , Variación Genética , Genoma Bacteriano , Listeria monocytogenes/genética , Listeria monocytogenes/patogenicidad , Animales , Sangre/microbiología , Modelos Animales de Enfermedad , Humanos , Listeriosis/microbiología , Listeriosis/patología , Hígado/microbiología , Ratones , Ratones Endogámicos BALB C , Datos de Secuencia Molecular , Análisis de Secuencia de ADN , Bazo/microbiología , Virulencia
10.
PLoS Genet ; 10(1): e1004065, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24415952

RESUMEN

The human bacterial pathogen Listeria monocytogenes is emerging as a model organism to study RNA-mediated regulation in pathogenic bacteria. A class of non-coding RNAs called CRISPRs (clustered regularly interspaced short palindromic repeats) has been described to confer bacterial resistance against invading bacteriophages and conjugative plasmids. CRISPR function relies on the activity of CRISPR associated (cas) genes that encode a large family of proteins with nuclease or helicase activities and DNA and RNA binding domains. Here, we characterized a CRISPR element (RliB) that is expressed and processed in the L. monocytogenes strain EGD-e, which is completely devoid of cas genes. Structural probing revealed that RliB has an unexpected secondary structure comprising basepair interactions between the repeats and the adjacent spacers in place of canonical hairpins formed by the palindromic repeats. Moreover, in contrast to other CRISPR-Cas systems identified in Listeria, RliB-CRISPR is ubiquitously present among Listeria genomes at the same genomic locus and is never associated with the cas genes. We showed that RliB-CRISPR is a substrate for the endogenously encoded polynucleotide phosphorylase (PNPase) enzyme. The spacers of the different Listeria RliB-CRISPRs share many sequences with temperate and virulent phages. Furthermore, we show that a cas-less RliB-CRISPR lowers the acquisition frequency of a plasmid carrying the matching protospacer, provided that trans encoded cas genes of a second CRISPR-Cas system are present in the genome. Importantly, we show that PNPase is required for RliB-CRISPR mediated DNA interference. Altogether, our data reveal a yet undescribed CRISPR system whose both processing and activity depend on PNPase, highlighting a new and unexpected function for PNPase in "CRISPRology".


Asunto(s)
Sistemas CRISPR-Cas/genética , Repeticiones Palindrómicas Cortas Agrupadas y Regularmente Espaciadas/genética , Listeria monocytogenes/genética , Polirribonucleótido Nucleotidiltransferasa/genética , ARN no Traducido/genética , Proteínas Bacterianas , Proteínas de Unión al ADN/genética , Genoma Bacteriano , Humanos , Polirribonucleótido Nucleotidiltransferasa/metabolismo , Proteínas de Unión al ARN/genética
11.
mBio ; 4(6): e00707-13, 2013 Dec 10.
Artículo en Inglés | MEDLINE | ID: mdl-24327339

RESUMEN

UNLABELLED: The intestinal tract is the largest reservoir of microbes in the human body. The intestinal microbiota is thought to be able to modulate alterations of the gut induced by enteropathogens, thereby maintaining homeostasis. Listeria monocytogenes is the agent of listeriosis, an infection transmitted to humans upon ingestion of contaminated food. Crossing of the intestinal barrier is a critical step of the infection before dissemination into deeper organs. Here, we investigated the role of the intestinal microbiota in the regulation of host protein-coding genes and microRNA (miRNA or miR) expression during Listeria infection. We first established the intestinal miRNA signatures corresponding to the 10 most highly expressed miRNAs in the murine ileum of conventional and germfree mice, noninfected and infected with Listeria. Next, we identified 6 miRNAs whose expression decreased upon Listeria infection in conventional mice. Strikingly, five of these miRNA expression variations (in miR-143, miR-148a, miR-200b, miR-200c, and miR-378) were dependent on the presence of the microbiota. In addition, as is already known, protein-coding genes were highly affected by infection in both conventional and germfree mice. By crossing bioinformatically the predicted targets of the miRNAs to our whole-genome transcriptomic data, we revealed an miRNA-mRNA network that suggested miRNA-mediated global regulation during intestinal infection. Other recent studies have revealed an miRNA response to either bacterial pathogens or commensal bacteria. In contrast, our work provides an unprecedented insight into the impact of the intestinal microbiota on host transcriptional reprogramming during infection by a human pathogen. IMPORTANCE: While the crucial role of miRNAs in regulating the host response to bacterial infection is increasingly recognized, the involvement of the intestinal microbiota in the regulation of miRNA expression has not been explored in detail. Here, we investigated the impact of the intestinal microbiota on the regulation of protein-coding genes and miRNA expression in a host infected by L. monocytogenes, a food-borne pathogen. We show that the microbiota interferes with the microRNA response upon oral Listeria infection and identify several protein-coding target genes whose expression correlates inversely with that of the miRNA. Further investigations of the regulatory networks involving miR-143, miR-148a, miR-200b, miR-200c, and miR-378 will provide new insights into the impact of the intestinal microbiota on the host upon bacterial infection.


Asunto(s)
Interacciones Huésped-Patógeno , Íleon/inmunología , Íleon/microbiología , Listeria monocytogenes/inmunología , Listeriosis/inmunología , MicroARNs/metabolismo , Microbiota/inmunología , Animales , Femenino , Perfilación de la Expresión Génica , Ratones , Ratones Endogámicos C57BL , Transcripción Genética
13.
Nat Rev Microbiol ; 11(2): 75-82, 2013 02.
Artículo en Inglés | MEDLINE | ID: mdl-23268228

RESUMEN

In recent years, non-coding RNAs have emerged as key regulators of gene expression. Among these RNAs, the antisense RNAs (asRNAs) are particularly abundant, but in most cases the function and mechanism of action for a particular asRNA remains elusive. Here, we highlight a recently discovered paradigm termed the excludon, which defines a genomic locus encoding an unusually long asRNA that spans divergent genes or operons with related or opposing functions. Because these asRNAs can inhibit the expression of one operon while functioning as an mRNA for the adjacent operon, they act as fine-tuning regulatory switches in bacteria.


Asunto(s)
Bacterias/genética , Regulación Bacteriana de la Expresión Génica/fisiología , ARN sin Sentido/genética , ARN Bacteriano/genética , Bacterias/metabolismo , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Genoma Bacteriano , Operón , Biosíntesis de Proteínas , ARN sin Sentido/metabolismo , ARN Bacteriano/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Transactivadores/genética , Transcriptoma
14.
Proc Natl Acad Sci U S A ; 109(41): 16684-9, 2012 Oct 09.
Artículo en Inglés | MEDLINE | ID: mdl-23012479

RESUMEN

Listeria monocytogenes is a foodborne pathogen that crosses the intestinal barrier and disseminates within the host. Here, we report a unique comprehensive analysis of the impact of two Lactobacillus species, Lactobacillus paracasei CNCM I-3689 and Lactobacillus casei BL23, on L. monocytogenes and orally acquired listeriosis in a gnotobiotic humanized mouse model. We first assessed the effect of treatment with each Lactobacillus on L. monocytogenes counts in host tissues and showed that each decreases L. monocytogenes systemic dissemination in orally inoculated mice. A whole genome intestinal transcriptomic analysis revealed that each Lactobacillus changes expression of a specific subset of genes during infection, with IFN-stimulated genes (ISGs) being the most affected by both lactobacilli. We also examined microRNA (miR) expression and showed that three miRs (miR-192, miR-200b, and miR-215) are repressed during L. monocytogenes infection. Treatment with each Lactobacillus increased miR-192 expression, whereas only L. casei association increased miR-200b and miR-215 expression. Finally, we showed that treatment with each Lactobacillus significantly reshaped the L. monocytogenes transcriptome and up-regulated transcription of L. monocytogenes genes encoding enzymes allowing utilization of intestinal carbon and nitrogen sources in particular genes involved in propanediol and ethanolamine catabolism and cobalamin biosynthesis. Altogether, these data reveal that the modulation of L. monocytogenes infection by treatment with lactobacilli correlates with a decrease in host gene expression, in particular ISGs, miR regulation, and a dramatic reshaping of L. monocytogenes transcriptome.


Asunto(s)
Antibiosis/fisiología , Lactobacillus/fisiología , Listeria monocytogenes/genética , Listeriosis/genética , Animales , Expresión Génica/efectos de los fármacos , Genes Bacterianos/genética , Interacciones Huésped-Patógeno/genética , Interferones/farmacología , Mucosa Intestinal/metabolismo , Intestinos/microbiología , Lactobacillus/clasificación , Lacticaseibacillus casei/fisiología , Listeria monocytogenes/fisiología , Listeriosis/microbiología , Ratones , Ratones Transgénicos , MicroARNs/genética , Boca/microbiología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transcriptoma/genética
15.
Mol Syst Biol ; 8: 583, 2012 May 22.
Artículo en Inglés | MEDLINE | ID: mdl-22617957

RESUMEN

Listeria monocytogenes is a human, food-borne pathogen. Genomic comparisons between L. monocytogenes and Listeria innocua, a closely related non-pathogenic species, were pivotal in the identification of protein-coding genes essential for virulence. However, no comprehensive comparison has focused on the non-coding genome. We used strand-specific cDNA sequencing to produce genome-wide transcription start site maps for both organisms, and developed a publicly available integrative browser to visualize and analyze both transcriptomes in different growth conditions and genetic backgrounds. Our data revealed conservation across most transcripts, but significant divergence between the species in a subset of non-coding RNAs. In L. monocytogenes, we identified 113 small RNAs (33 novel) and 70 antisense RNAs (53 novel), significantly increasing the repertoire of ncRNAs in this species. Remarkably, we identified a class of long antisense transcripts (lasRNAs) that overlap one gene while also serving as the 5' UTR of the adjacent divergent gene. Experimental evidence suggests that lasRNAs transcription inhibits expression of one operon while activating the expression of another. Such a lasRNA/operon structure, that we named 'excludon', might represent a novel form of regulation in bacteria.


Asunto(s)
Listeria/genética , Listeria/patogenicidad , ARN sin Sentido/genética , Transcriptoma , Regiones no Traducidas 5' , Secuencia de Bases , Evolución Biológica , Secuencia Conservada , Regulación Bacteriana de la Expresión Génica , Genoma Bacteriano , Listeria monocytogenes/genética , Listeria monocytogenes/patogenicidad , Datos de Secuencia Molecular , Operón , ARN sin Sentido/metabolismo , ARN Bacteriano , ARN no Traducido/genética , ARN no Traducido/metabolismo , Sitio de Iniciación de la Transcripción
17.
Infect Immun ; 79(6): 2489-98, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21444667

RESUMEN

Intracellular bacterial pathogens manipulate host cell functions by producing enzymes that stimulate or antagonize signal transduction. The Listeria monocytogenes genome contains a gene, lmo1800, encoding a protein with a conserved motif of conventional tyrosine phosphatases. Here, we report that the lmo1800-encoded protein LipA is secreted by Listeria and displays tyrosine as well as lipid phosphatase activity in vitro. Bacteria lacking LipA are severely attenuated in virulence in vivo, thus revealing a so-far-undescribed enzymatic activity involved in Listeria infection.


Asunto(s)
Proteínas Bacterianas/fisiología , Listeria monocytogenes/patogenicidad , Listeriosis/microbiología , Factores de Virulencia/fisiología , Animales , Western Blotting , Línea Celular , Ensayo de Inmunoadsorción Enzimática , Técnica de Placa Hemolítica , Listeria monocytogenes/enzimología , Macrófagos/microbiología , Ratones , Ratones Endogámicos C57BL , Proteínas Tirosina Fosfatasas/fisiología
18.
Immunol Rev ; 240(1): 160-84, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21349093

RESUMEN

Listeria monocytogenes is an intracellular bacterium responsible for a disease characterized by several clinical features, such as septicemia, brain infection, abortion, and perinatal infection. These clinical features are attributed to its amazing capacity to cross several barriers in susceptible hosts. Intracellular infection is a consequence of the bacterium's capacity to enter a wide variety of mammalian cells, to not only survive but also replicate therein, and to its faculty to spread from one cell to the next, thereby escaping the humoral immune response. Here, we review both the well-established and the newly discovered strategies used by this bacterium to achieve this intracellular lifestyle while escaping from the host innate immune response. More than ever, Listeria appears as a model system and a reference in infection biology.


Asunto(s)
Listeria monocytogenes/inmunología , Listeriosis/inmunología , Listeriosis/fisiopatología , Humanos , Evasión Inmune , Listeria monocytogenes/crecimiento & desarrollo , Interacciones Microbianas , Modelos Biológicos
19.
Eur J Immunol ; 40(12): 3458-71, 2010 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21108467

RESUMEN

Control of pulmonary pathogens constitutes a challenging task as successful immune responses need to be mounted without damaging the lung parenchyma. Using immunofluorescence microscopy and flow cytometry, we analyzed in the mouse the initial innate immune response that follows intranasal inoculation of Brucella abortus. Bacteria were absent from parenchymal dendritic cells (DC) but present in alveolar macrophages in which they replicated. When the number of alveolar macrophages was reduced prior to Brucella infection, small numbers of pulmonary DC were infected and a massive recruitment of TNF-α- and iNOS-producing DC ensued. Coincidentally, Brucella disseminated to the lung-draining mediastinal lymph nodes (LN) where they replicated in both migratory DC and migratory alveolar macrophages. Together, these results demonstrate that alveolar macrophages are critical regulators of the initial innate immune response against Brucella within the lungs and show that pulmonary DC and alveolar macrophages play rather distinct roles in the control of microbial burden.


Asunto(s)
Brucella abortus/inmunología , Brucelosis/inmunología , Células Dendríticas/metabolismo , Ganglios Linfáticos/inmunología , Macrófagos Alveolares/metabolismo , Animales , Carga Bacteriana , Brucella abortus/crecimiento & desarrollo , Brucella abortus/patogenicidad , Brucelosis/microbiología , Brucelosis/transmisión , Recuento de Células , Movimiento Celular/inmunología , Células Dendríticas/inmunología , Células Dendríticas/microbiología , Células Dendríticas/patología , Inmunidad Innata , Pulmón/inmunología , Pulmón/microbiología , Pulmón/patología , Ganglios Linfáticos/microbiología , Macrófagos Alveolares/inmunología , Macrófagos Alveolares/microbiología , Macrófagos Alveolares/patología , Ratones , Ratones Endogámicos C57BL , Óxido Nítrico Sintasa de Tipo II/genética , Óxido Nítrico Sintasa de Tipo II/metabolismo , Factor de Necrosis Tumoral alfa/genética , Factor de Necrosis Tumoral alfa/metabolismo
20.
Immunity ; 31(2): 197-208, 2009 Aug 21.
Artículo en Inglés | MEDLINE | ID: mdl-19682930

RESUMEN

Despite compromised T cell antigen receptor (TCR) signaling, mice in which tyrosine 136 of the adaptor linker for activation of T cells (LAT) was constitutively mutated (Lat(Y136F) mice) accumulate CD4(+) T cells that trigger autoimmunity and inflammation. Here we show that equipping postthymic CD4(+) T cells with LATY136F molecules or rendering them deficient in LAT molecules triggers a lymphoproliferative disorder dependent on prior TCR engagement. Therefore, such disorders required neither faulty thymic T cell maturation nor LATY136F molecules. Unexpectedly, in CD4(+) T cells recently deprived of LAT, the proximal triggering module of the TCR induced a spectrum of protein tyrosine phosphorylation that largely overlapped the one observed in the presence of LAT. The fact that such LAT-independent signals result in lymphoproliferative disorders with excessive cytokine production demonstrates that LAT constitutes a key negative regulator of the triggering module and of the LAT-independent branches of the TCR signaling cassette.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/inmunología , Antígenos CD28/inmunología , Linfocitos T CD4-Positivos/inmunología , Antígenos de Histocompatibilidad Clase II/inmunología , Trastornos Linfoproliferativos/inmunología , Proteínas de la Membrana/inmunología , Fosfoproteínas/inmunología , Receptores de Antígenos de Linfocitos T/inmunología , Proteínas Adaptadoras Transductoras de Señales/genética , Traslado Adoptivo , Animales , Antígenos CD28/metabolismo , Linfocitos T CD4-Positivos/metabolismo , Citocinas/inmunología , Citocinas/metabolismo , Antígenos de Histocompatibilidad Clase II/metabolismo , Trastornos Linfoproliferativos/genética , Proteínas de la Membrana/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Mutantes , Mutación , Fosfoproteínas/genética , Fosforilación/inmunología , Receptores de Antígenos de Linfocitos T/metabolismo , Transducción de Señal/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...