Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 325
Filtrar
1.
Int J Mol Sci ; 24(14)2023 Jul 14.
Artículo en Inglés | MEDLINE | ID: mdl-37511208

RESUMEN

The high-pathogenicity island (HPI) was initially identified in Yersinia and can be horizontally transferred to Escherichia coli to produce yersiniabactin (Ybt), which enhances the pathogenicity of E. coli by competing with the host for Fe3+. Pyroptosis is gasdermin-induced necrotic cell death. It involves the permeabilization of the cell membrane and is accompanied by an inflammatory response. It is still unclear whether Ybt HPI can cause intestinal epithelial cells to undergo pyroptosis and contribute to gut inflammation during E. coli infection. In this study, we infected intestinal epithelial cells of mice with E. coli ZB-1 and the Ybt-deficient strain ZB-1Δirp2. Our findings demonstrate that Ybt-producing E. coli is more toxic and exacerbates gut inflammation during systemic infection. Mechanistically, our results suggest the involvement of the NLRP3/caspase-1/GSDMD pathway in E. coli infection. Ybt promotes the assembly and activation of the NLRP3 inflammasome, leading to GSDMD cleavage into GSDMD-N and promoting the pyroptosis of intestinal epithelial cells, ultimately aggravating gut inflammation. Notably, NLRP3 knockdown alleviated these phenomena, and the binding of free Ybt to NLRP3 may be the trigger. Overall, our results show that Ybt HPI enhances the pathogenicity of E. coli and induces pyroptosis via the NLRP3 pathway, which is a new mechanism through which E. coli promotes gut inflammation. Furthermore, we screened drugs targeting NLRP3 from an existing drug library, providing a list of potential drug candidates for the treatment of gut injury caused by E. coli.


Asunto(s)
Células Epiteliales , Infecciones por Escherichia coli , Escherichia coli , Mucosa Intestinal , Proteína con Dominio Pirina 3 de la Familia NLR , Piroptosis , Animales , Ratones , Enterocitos/metabolismo , Enterocitos/microbiología , Células Epiteliales/metabolismo , Células Epiteliales/microbiología , Escherichia coli/metabolismo , Escherichia coli/patogenicidad , Infecciones por Escherichia coli/metabolismo , Inflamasomas/metabolismo , Inflamación/metabolismo , Mucosa Intestinal/citología , Mucosa Intestinal/metabolismo , Mucosa Intestinal/microbiología , Proteína con Dominio Pirina 3 de la Familia NLR/genética , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Piroptosis/fisiología
2.
Nat Commun ; 14(1): 456, 2023 01 28.
Artículo en Inglés | MEDLINE | ID: mdl-36709326

RESUMEN

Host bottlenecks prevent many infections before the onset of disease by eliminating invading pathogens. By monitoring the diversity of a barcoded population of the diarrhea causing bacterium Citrobacter rodentium during colonization of its natural host, mice, we determine the number of cells that found the infection by establishing a replicative niche. In female mice the size of the pathogen's founding population scales with dose and is controlled by a severe yet slow-acting bottleneck. Reducing stomach acid or changing host genotype modestly relaxes the bottleneck without breaking the fractional relationship between dose and founders. In contrast, disrupting the microbiota causes the founding population to no longer scale with the size of the inoculum and allows the pathogen to infect at almost any dose, indicating that the microbiota creates the dominant bottleneck. Further, in the absence of competition with the microbiota, the diversity of the pathogen population slowly contracts as the population is overtaken by bacteria having lost the critical virulence island, the locus of enterocyte effacement (LEE). Collectively, our findings reveal that the mechanisms of protection by colonization bottlenecks are reflected in and can be generally defined by the impact of dose on the pathogen's founding population.


Asunto(s)
Bacterias , Infecciones por Enterobacteriaceae , Femenino , Animales , Ratones , Virulencia/genética , Factores de Virulencia/genética , Enterocitos/microbiología , Diarrea , Citrobacter rodentium/genética , Infecciones por Enterobacteriaceae/microbiología
3.
Nat Commun ; 13(1): 17, 2022 01 10.
Artículo en Inglés | MEDLINE | ID: mdl-35013162

RESUMEN

Defense against intracellular infection has been extensively studied in vertebrate hosts, but less is known about invertebrate hosts; specifically, the transcription factors that induce defense against intracellular intestinal infection in the model nematode Caenorhabditis elegans remain understudied. Two different types of intracellular pathogens that naturally infect the C. elegans intestine are the Orsay virus, which is an RNA virus, and microsporidia, which comprise a phylum of fungal pathogens. Despite their molecular differences, these pathogens induce a common host transcriptional response called the intracellular pathogen response (IPR). Here we show that zip-1 is an IPR regulator that functions downstream of all known IPR-activating and regulatory pathways. zip-1 encodes a putative bZIP transcription factor, and we show that zip-1 controls induction of a subset of genes upon IPR activation. ZIP-1 protein is expressed in the nuclei of intestinal cells, and is at least partially required in the intestine to upregulate IPR gene expression. Importantly, zip-1 promotes resistance to infection by the Orsay virus and by microsporidia in intestinal cells. Altogether, our results indicate that zip-1 represents a central hub for triggers of the IPR, and that this transcription factor has a protective function against intracellular pathogen infection in C. elegans.


Asunto(s)
Factores de Transcripción con Cremalleras de Leucina de Carácter Básico , Caenorhabditis elegans , Enterocitos , Interacciones Huésped-Patógeno/fisiología , Animales , Factores de Transcripción con Cremalleras de Leucina de Carácter Básico/inmunología , Factores de Transcripción con Cremalleras de Leucina de Carácter Básico/metabolismo , Caenorhabditis elegans/inmunología , Caenorhabditis elegans/microbiología , Caenorhabditis elegans/virología , Proteínas de Caenorhabditis elegans/inmunología , Proteínas de Caenorhabditis elegans/metabolismo , Enterocitos/inmunología , Enterocitos/microbiología , Enterocitos/virología , Inmunidad Innata/fisiología , Intestinos/microbiología , Intestinos/virología , Invertebrados/inmunología , Microsporidios/patogenicidad , Virus ARN/patogenicidad
4.
Gut Microbes ; 13(1): 1973836, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34542008

RESUMEN

Salmonella pathogenicity island (SPI) 2 type three secretion system (T3SS)-mediated effector molecules facilitate bacterial survival in phagocytes but their role in the intestinal epithelium in vivo remains ill-defined. Using our neonatal murine infection model in combination with SPI2 reporter technology and RNA-Seq of sorted primary enterocytes, we demonstrate expression of SPI2 effector molecules by intraepithelial Salmonella Typhimurium (S. Typhimurium). Contrary to expectation, immunostaining revealed that infection with SPI2 T3SS-mutants resulted in significantly enlarged intraepithelial Salmonella-containing vacuoles (SCV) with altered cellular positioning, suggesting impaired apical to basolateral transmigration. Also, infection with isogenic tagged S. Typhimurium strains revealed a reduced spread of intraepithelial SPI2 T3SS mutant S. Typhimurium to systemic body sites. These results suggest that SPI2 T3SS effector molecules contribute to enterocyte apical to basolateral transmigration of the SCV during the early stage of the infection.


Asunto(s)
Proteínas Bacterianas/metabolismo , Mucosa Intestinal/microbiología , Proteínas de la Membrana/metabolismo , Salmonella typhimurium/metabolismo , Sistemas de Secreción Tipo III/metabolismo , Vacuolas/microbiología , Animales , Proteínas Bacterianas/genética , Células Cultivadas , Enterocitos/metabolismo , Enterocitos/microbiología , Mucosa Intestinal/citología , Macrófagos/inmunología , Proteínas de la Membrana/genética , Ratones , Ratones Endogámicos C57BL , RNA-Seq , Infecciones por Salmonella/patología , Uniones Estrechas/microbiología , Sistemas de Secreción Tipo III/genética , Vacuolas/metabolismo
5.
Oxid Med Cell Longev ; 2021: 8936907, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34373770

RESUMEN

Probiotics are widely used for protection against stress-induced intestinal dysfunction. Oxidative stress plays a critical role in gastrointestinal disorders. It is established that probiotics alleviate oxidative stress; however, the mechanism of action has not been elucidated. We developed an in vitro intestinal porcine epithelial cells (IPEC-J2) model of oxidative stress to explore the antioxidant effect and potential mode of action of Lactobacillus plantarum ZLP001. The IPEC-J2 cells were preincubated with and without L. plantarum ZLP001 for 3 h and then exposed to hydrogen peroxide (H2O2) for 4 h. Pretreatment with L. plantarum ZLP001 protected IPEC-J2 cells against H2O2-induced oxidative damage as indicated by cell viability assays and significantly alleviated apoptosis elicited by H2O2. L. plantarum ZLP001 pretreatment decreased reactive oxygen species production and the cellular malondialdehyde concentration and increased the mitochondrial membrane potential compared with H2O2 treatment alone, suggesting that L. plantarum ZLP001 promotes the maintenance of redox homeostasis in the cells. Furthermore, L. plantarum ZLP001 regulated the expression and generation of some antioxidant enzymes, thereby activating the antioxidant defense system. Treatment with L. plantarum ZLP001 led to nuclear erythroid 2-related factor 2 (Nrf2) enrichment in the nucleus compared with H2O2 treatment alone. Knockdown of Nrf2 significantly weakened the alleviating effect of L. plantarum ZLP001 on antioxidant stress in IPEC-J2 cells, suggesting that Nrf2 is involved in the antioxidative effect of L. plantarum ZLP001. Collectively, these results indicate that L. plantarum ZLP001 is a promising probiotic bacterium that can potentially alleviate oxidative stress.


Asunto(s)
Enterocitos/metabolismo , Microbioma Gastrointestinal , Lactobacillus plantarum/patogenicidad , Estrés Oxidativo , Animales , Células Cultivadas , Enterocitos/efectos de los fármacos , Enterocitos/microbiología , Peróxido de Hidrógeno/toxicidad , Íleon/citología , Íleon/metabolismo , Íleon/microbiología , Porcinos
6.
Vet Res ; 52(1): 90, 2021 Jun 19.
Artículo en Inglés | MEDLINE | ID: mdl-34147126

RESUMEN

Lawsonia intracellularis is endemic to swine herds worldwide, however much is still unknown regarding its impact on intestinal function. Thus, this study aimed to characterize the impact of L. intracellularis on digestive function, and how vaccination mitigates these impacts. Thirty-six L. intracellularis negative barrows were assigned to treatment groups (n = 12/trt): (1) nonvaccinated, L. intracellularis negative (NC); (2) nonvaccinated, L intracellularis challenged (PC); and (3) L. intracellularis challenged, vaccinated (Enterisol® Ileitis, Boehringer Ingelheim) 7 weeks pre-challenge (VAC). On days post-inoculation (dpi) 0 PC and VAC pigs were inoculated with L. intracellularis. From dpi 19-21 fecal samples were collected for apparent total tract digestibility (ATTD) and at dpi 21, pigs were euthanized for sample collection. Post-inoculation, ADG was reduced in PC pigs compared with NC (41%, P < 0.001) and VAC (25%, P < 0.001) pigs. Ileal gross lesion severity was greater in PC pigs compared with NC (P = 0.003) and VAC (P = 0.018) pigs. Dry matter, organic matter, nitrogen, and energy ATTD were reduced in PC pigs compared with NC pigs (P ≤ 0.001 for all). RNAscope in situ hybridization revealed abolition of sucrase-isomaltase transcript in the ileum of PC pigs compared with NC and VAC pigs (P < 0.01). Conversely, abundance of stem cell signaling markers Wnt3, Hes1, and p27Kip1 were increased in PC pigs compared with NC pigs (P ≤ 0.085). Taken together, these data demonstrate that reduced digestibility during L. intracellularis challenge is partially driven by abolition of digestive machinery in lesioned tissue. Further, vaccination mitigated several of these effects, likely from lower bacterial burden and reduced disease severity.


Asunto(s)
Infecciones por Desulfovibrionaceae/veterinaria , Enterocitos/microbiología , Lawsonia (Bacteria)/fisiología , Oligo-1,6-Glucosidasa/deficiencia , Sacarasa/deficiencia , Animales , Infecciones por Desulfovibrionaceae/enzimología , Infecciones por Desulfovibrionaceae/microbiología , Infecciones por Desulfovibrionaceae/fisiopatología , Enterocitos/enzimología , Sus scrofa , Porcinos , Enfermedades de los Porcinos/enzimología , Enfermedades de los Porcinos/microbiología , Enfermedades de los Porcinos/fisiopatología
7.
PLoS One ; 16(6): e0253242, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34129624

RESUMEN

Streptococcus agalactiae (group B streptococcus or GBS) is a commensal bacterium that can frequently behave as a pathogen, particularly in the neonatal period and in the elderly. The gut is a primary site of GBS colonization and a potential port of entry during neonatal infections caused by hypervirulent clonal complex 17 (CC17) strains. Here we studied the interactions between the prototypical CC17 BM110 strain and polarized enterocytes using the Caco-2 cell line. GBS could adhere to and invade these cells through their apical or basolateral surfaces. Basolateral invasion was considerably more efficient than apical invasion and predominated under conditions resulting in weakening of cell-to-cell junctions. Bacterial internalization occurred by a mechanism involving caveolae- and lipid raft-dependent endocytosis and actin re-organization, but not clathrin-dependent endocytosis. In the first steps of Caco-2 invasion, GBS colocalized with the early endocytic marker EEA-1, to later reside in acidic vacuoles. Taken together, these data suggest that CC17 GBS selectively adheres to the lateral surface of enterocytes from which it enters through caveolar lipid rafts using a classical, actin-dependent endocytic pathway. These data may be useful to develop alternative preventive strategies aimed at blocking GBS invasion of the intestinal barrier.


Asunto(s)
Enterocitos/microbiología , Infecciones Estreptocócicas/microbiología , Streptococcus agalactiae/patogenicidad , Adhesión Bacteriana , Células CACO-2/microbiología , Endocitosis , Humanos , Uniones Intercelulares/microbiología , Microscopía Fluorescente , Streptococcus agalactiae/fisiología , Virulencia
8.
Microbiol Spectr ; 9(1): e0000321, 2021 09 03.
Artículo en Inglés | MEDLINE | ID: mdl-34106568

RESUMEN

Gastrointestinal infections cause significant morbidity and mortality worldwide. The complexity of human biology and limited insights into host-specific infection mechanisms are key barriers to current therapeutic development. Here, we demonstrate that two-dimensional epithelial monolayers derived from human intestinal organoids, combined with in vivo-like bacterial culturing conditions, provide significant advancements for the study of enteropathogens. Monolayers from the terminal ileum, cecum, and ascending colon recapitulated the composition of the gastrointestinal epithelium, in which several techniques were used to detect the presence of enterocytes, mucus-producing goblet cells, and other cell types following differentiation. Importantly, the addition of receptor activator of nuclear factor kappa-B ligand (RANKL) increased the presence of M cells, critical antigen-sampling cells often exploited by enteric pathogens. For infections, bacteria were grown under in vivo-like conditions known to induce virulence. Overall, interesting patterns of tissue tropism and clinical manifestations were observed. Shigella flexneri adhered efficiently to the cecum and colon; however, invasion in the colon was best following RANKL treatment. Both Salmonella enterica serovars Typhi and Typhimurium displayed different infection patterns, with S. Typhimurium causing more destruction of the terminal ileum and S. Typhi infecting the cecum more efficiently than the ileum, particularly with regard to adherence. Finally, various pathovars of Escherichia coli validated the model by confirming only adherence was observed with these strains. This work demonstrates that the combination of human-derived tissue with targeted bacterial growth conditions enables powerful analyses of human-specific infections that could lead to important insights into pathogenesis and accelerate future vaccine development. IMPORTANCE While traditional laboratory techniques and animal models have provided valuable knowledge in discerning virulence mechanisms of enteric pathogens, the complexity of the human gastrointestinal tract has hindered our understanding of physiologically relevant, human-specific interactions; and thus, has significantly delayed successful vaccine development. The human intestinal organoid-derived epithelial monolayer (HIODEM) model closely recapitulates the diverse cell populations of the intestine, allowing for the study of human-specific infections. Differentiation conditions permit the expansion of various cell populations, including M cells that are vital to immune recognition and the establishment of infection by some bacteria. We provide details of reproducible culture methods and infection conditions for the analyses of Shigella, Salmonella, and pathogenic Escherichia coli in which tissue tropism and pathogen-specific infection patterns were detected. This system will be vital for future studies that explore infection conditions, health status, or epigenetic differences and will serve as a novel screening platform for therapeutic development.


Asunto(s)
Técnicas de Cultivo de Célula/métodos , Infecciones por Enterobacteriaceae/microbiología , Enterobacteriaceae/fisiología , Tracto Gastrointestinal/microbiología , Organoides/microbiología , Enterobacteriaceae/genética , Enterobacteriaceae/patogenicidad , Enterocitos/microbiología , Células Epiteliales/citología , Células Epiteliales/microbiología , Epitelio/microbiología , Tracto Gastrointestinal/citología , Humanos , Organoides/citología , Virulencia
9.
mBio ; 12(3)2021 05 18.
Artículo en Inglés | MEDLINE | ID: mdl-34006652

RESUMEN

The intestinal epithelium is a primary interface for engagement of the host response by foodborne pathogens, like Salmonella enterica Typhimurium. While the interaction of S Typhimurium with the mammalian host has been well studied in transformed epithelial cell lines or in the complex intestinal environment in vivo, few tractable models recapitulate key features of the intestine. Human intestinal organoids (HIOs) contain a polarized epithelium with functionally differentiated cell subtypes, including enterocytes and goblet cells and a supporting mesenchymal cell layer. HIOs contain luminal space that supports bacterial replication, are more amenable to experimental manipulation than animals and are more reflective of physiological host responses. Here, we use the HIO model to define host transcriptional responses to S Typhimurium infection, also determining host pathways dependent on Salmonella pathogenicity island-1 (SPI-1)- and -2 (SPI-2)-encoded type 3 secretion systems (T3SS). Consistent with prior findings, we find that S Typhimurium strongly stimulates proinflammatory gene expression. Infection-induced cytokine gene expression was rapid, transient, and largely independent of SPI-1 T3SS-mediated invasion, likely due to continued luminal stimulation. Notably, S Typhimurium infection led to significant downregulation of host genes associated with cell cycle and DNA repair, leading to a reduction in cellular proliferation, dependent on SPI-1 and SPI-2 T3SS. The transcriptional profile of cell cycle-associated target genes implicates multiple miRNAs as mediators of S Typhimurium-dependent cell cycle suppression. These findings from Salmonella-infected HIOs delineate common and distinct contributions of SPI-1 and SPI-2 T3SSs in inducing early host responses during enteric infection and reinforce host cell proliferation as a process targeted by SalmonellaIMPORTANCESalmonella enterica serovar Typhimurium (S Typhimurium) causes a significant health burden worldwide, yet host responses to initial stages of intestinal infection remain poorly understood. Due to differences in infection outcome between mice and humans, physiological human host responses driven by major virulence determinants of Salmonella have been more challenging to evaluate. Here, we use the three-dimensional human intestinal organoid model to define early responses to infection with wild-type S Typhimurium and mutants defective in the SPI-1 or SPI-2 type-3 secretion systems. While both secretion system mutants show defects in mouse models of oral Salmonella infection, the specific contributions of each secretion system are less well understood. We show that S Typhimurium upregulates proinflammatory pathways independently of either secretion system, while the downregulation of the host cell cycle pathways relies on both SPI-1 and SPI-2. These findings lay the groundwork for future studies investigating how SPI-1- and SPI-2-driven host responses affect infection outcome and show the potential of this model to study host-pathogen interactions with other serovars to understand how initial interactions with the intestinal epithelium may affect pathogenesis.


Asunto(s)
Proteínas Bacterianas/genética , Enterocitos/microbiología , Perfilación de la Expresión Génica , Interacciones Huésped-Patógeno/genética , Proteínas de la Membrana/genética , Organoides/microbiología , Salmonella typhimurium/genética , Línea Celular , Regulación Bacteriana de la Expresión Génica , Humanos , Mucosa Intestinal/microbiología , Intestinos/citología , Intestinos/microbiología , Salmonella typhimurium/patogenicidad , Serogrupo , Factores de Virulencia
10.
BMC Microbiol ; 20(1): 205, 2020 07 11.
Artículo en Inglés | MEDLINE | ID: mdl-32652929

RESUMEN

BACKGROUND: The intestinal tract undergoes a period of cellular maturation during early life, primarily characterized by the organization of epithelial cells into specialized crypt and villus structures. These processes are in part mediated by the acquisition of microbes. Infants delivered at term typically harbor a stable, low diversity microbiota characterized by an overrepresentation of various Bacilli spp., while pre-term infants are colonized by an assortment of bacteria during the first several weeks after delivery. However, the functional effects of these changes on intestinal epithelium homeostasis and maturation remain unclear. To study these effects, human neonate feces were obtained from term and pre-term infants. Fecal 16S rDNA sequencing and global untargeted LC-MS were performed to characterize microbial composition and metabolites from each population. Murine enteral organoids (enteroids) were cultured with 0.22 µm filtered stool supernatant pooled from term or pre-term infants. RESULTS: Term and pre-term microbial communities differed significantly from each other by principle components analysis (PCoA, PERMANOVA p < 0.001), with the pre-term microbiome characterized by increased OTU diversity (Wilcox test p < 0.01). Term communities were less diverse and dominated by Bacilli (81.54%). Pre-term stools had an increased abundance of vitamins, amino acid derivatives and unconjugated bile acids. Pathway analysis revealed a significant increase in multiple metabolic pathways in pre-term samples mapped to E. coli using the KEGG database related to the fermentation of various amino acids and vitamin biosynthesis. Enteroids cultured with supernatant from pre-term stools proliferated at a higher rate than those cultured with supernatant from term stools (cell viability: 207% vs. 147.7%, p < 0.01), grew larger (area: 81,189µm2 vs. 41,777µm2, p < 0.001), and bud at a higher rate (6.5 vs. 4, p < 0.01). Additionally, genes involved in stem cell proliferation were upregulated in pre-term stool treated enteroid cultures (Lgr5, Ephb2, Ascl2 Sox9) but not term stool treated enteroids. CONCLUSIONS: Our findings indicate that microbial metabolites from the more diverse gut microbiome associated with pre-term infants facilitate stem cell proliferation. Therefore, perturbations of the pre-term microbiota may impair intestinal homeostasis.


Asunto(s)
Bacterias/clasificación , Enterocitos/citología , Metabolómica/métodos , Nacimiento Prematuro/microbiología , ARN Ribosómico 16S/genética , Animales , Animales Recién Nacidos , Bacterias/química , Bacterias/genética , Bacterias/aislamiento & purificación , Biomarcadores/metabolismo , Proliferación Celular , ADN Bacteriano/genética , ADN Ribosómico/genética , Enterocitos/microbiología , Heces/microbiología , Microbioma Gastrointestinal , Regulación de la Expresión Génica , Humanos , Recién Nacido , Ratones , Técnicas de Cultivo de Órganos , Organoides/química , Organoides/citología , Organoides/microbiología , Filogenia , Nacimiento a Término
11.
Infect Immun ; 88(9)2020 08 19.
Artículo en Inglés | MEDLINE | ID: mdl-32631916

RESUMEN

Enteric pathogens exploit chemical and nutrient signaling to gauge their location within a host and control expression of traits important for infection. Ethanolamine-containing molecules are essential in host physiology and play important roles in intestinal processes. The transcription factor EutR is conserved in the Enterobacteriaceae and is required for ethanolamine sensing and metabolism. In enterohemorrhagic Escherichia coli (EHEC) O157:H7, EutR responds to ethanolamine to activate expression of traits required for host colonization and disease; however, the importance of EutR to EHEC intestinal infection has not been examined. Because EHEC does not naturally colonize or cause disease in mice, we employed the natural murine pathogen Citrobacter rodentium as a model of EHEC virulence to investigate the importance of EutR in vivo EHEC and C. rodentium possess the locus of enterocyte effacement (LEE), which is the canonical virulence trait of attaching and effacing pathogens. Our findings demonstrate that ethanolamine sensing and EutR-dependent regulation of the LEE are conserved in C. rodentium Moreover, during infection, EutR is required for maximal LEE expression, colonization, and transmission efficiency. These findings reveal that EutR not only is important for persistence during the primary host infection cycle but also is required for maintenance in a host population.


Asunto(s)
Citrobacter rodentium/genética , Infecciones por Enterobacteriaceae/microbiología , Escherichia coli Enterohemorrágica/genética , Proteínas de Escherichia coli/genética , Etanolamina/metabolismo , Regulación Bacteriana de la Expresión Génica , Fosfoproteínas/genética , Factores de Transcripción/genética , Animales , Citrobacter rodentium/patogenicidad , Recuento de Colonia Microbiana , Secuencia Conservada , Infecciones por Enterobacteriaceae/genética , Infecciones por Enterobacteriaceae/patología , Infecciones por Enterobacteriaceae/transmisión , Enterocitos/microbiología , Enterocitos/patología , Escherichia coli Enterohemorrágica/patogenicidad , Proteínas de Escherichia coli/metabolismo , Femenino , Interacciones Microbiota-Huesped/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Fosfoproteínas/metabolismo , Isoformas de Proteínas/deficiencia , Isoformas de Proteínas/genética , Transducción de Señal , Factores de Transcripción/deficiencia , Virulencia
12.
Vet Pathol ; 57(4): 565-576, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32527210

RESUMEN

Enterospora nucleophila is a microsporidian responsible for an emaciative disease in gilthead sea bream (Sparus aurata). Its intranuclear development and the lack of in vitro and in vivo models hinder its research. This study investigated the associated lesions, its detection by quantitative polymerase chain reaction, and the cellular immune response of naturally infected fish. The intensity of infection in the intestine was correlated with stunted growth and reduced body condition. At the beginning of the outbreaks, infection prevalence was highest in intestine and stomach, and in subsequent months, the prevalence decreased in the intestine and increased in hematopoietic organs and stomach. In heavy infections, the intestine had histologic lesions of enterocyte hypercellularity and proliferation of rodlet cells. Infected enterocytes had E. nucleophila spores in the cytoplasm, and a pyknotic nucleus, karyorhexis or karyolysis. Lymphocytes were present at the base of the mucosa, and eosinophilic granule cells were located between the enterocytes. In intestinal submucosa, macrophage aggregates containing spores were surrounded by lymphocytes and granulocytes, with submucosal infiltration of granulocytes. Macrophage aggregates appeared to develop into granulomata with necrotic areas containing parasite remnants. Immunohistochemistry revealed mast cells as the main type of granulocyte involved. Abundant IgM+ and IgT+ cells were identified by in situ hybridization in the submucosa when intracytoplasmic stages were present. This study describes the lesions of E. nucleophila in gilthead sea bream, an important aquaculture species.


Asunto(s)
Enfermedades de los Peces/microbiología , Microsporidios/aislamiento & purificación , Microsporidiosis/veterinaria , Dorada/microbiología , Animales , Acuicultura , Núcleo Celular/microbiología , Núcleo Celular/patología , Citoplasma/microbiología , Citoplasma/patología , Enterocitos/microbiología , Enterocitos/patología , Enfermedades de los Peces/patología , Granulocitos/microbiología , Granulocitos/patología , Granuloma/microbiología , Granuloma/patología , Histocitoquímica/veterinaria , Inmunidad Celular , Hibridación in Situ/veterinaria , Intestinos/microbiología , Intestinos/patología , Microsporidios/clasificación , Microsporidios/ultraestructura , Microsporidiosis/patología , Reacción en Cadena en Tiempo Real de la Polimerasa/veterinaria , Dorada/crecimiento & desarrollo
13.
PLoS Pathog ; 16(4): e1008446, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-32282860

RESUMEN

Microfold (M) cell host-pathogen interaction studies would benefit from the visual analysis of dynamic cellular and microbial interplays. We adapted a human in vitro M cell model to physiological bacterial infections, expression of fluorescent localization reporters and long-term three-dimensional time-lapse microscopy. This approach allows following key steps of M cell infection dynamics at subcellular resolution, from the apical onset to basolateral epithelial dissemination. We focused on the intracellular pathogen Shigella flexneri, classically reported to transcytose through M cells to initiate bacillary dysentery in humans, while eliciting poorly protective immune responses. Our workflow was critical to reveal that S. flexneri develops a bimodal lifestyle within M cells leading to rapid transcytosis or delayed vacuolar rupture, followed by direct actin motility-based propagation to neighboring enterocytes. Moreover, we show that Listeria monocytogenes, another intracellular pathogen sharing a tropism for M cells, disseminates in a similar manner and evades M cell transcytosis completely. We established that actin-based M cell-to-enterocyte spread is the major dissemination pathway for both pathogens and avoids their exposure to basolateral compartments in our system. Our results challenge the notion that intracellular pathogens are readily transcytosed by M cells to inductive immune compartments in vivo, providing a potential mechanism for their ability to evade adaptive immunity.


Asunto(s)
Disentería Bacilar/microbiología , Enterocitos/microbiología , Células Epiteliales/microbiología , Listeria monocytogenes/fisiología , Listeriosis/microbiología , Shigella flexneri/fisiología , Células CACO-2 , Humanos , Listeria monocytogenes/genética , Shigella flexneri/genética
14.
Vet Microbiol ; 242: 108596, 2020 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-32122600

RESUMEN

Gut is often subject to infection by different pathogens like Y. enterocolitica. To date, biotypes (BTs) 1A have been considered as non-pathogenic, because they do not express plasmid of virulence pYV; however, BTs 1A strains present other chromosomic virulence genes and recent studies suggest an implication of this microorganism in reactive arthritis. Although many studies highlighted the molecular basis of pathogenesis of Ye infection, scanty data are available about several environmental BTs 1A strains, often isolated in cases of foodborne disease but not included in pathogenicity studies. The aim of our work was to verify the ability of different Ye 1A strains to adhere and penetrate IPEC-J2 cells and to modulate intestinal innate immunity. Our results showed that all strains under study were able to adhere and penetrate enterocytes, causing inflammatory responses. Indeed, adhesion and invasion of enterocytes is an essential step in Ye pathogenesis (Fàbrega and Vila, 2012). Moreover, our data suggest the possible involvement of strains Ye2/O:9 in reactive arthritis, due to their ability (i) to penetrate enterocytes as pathogenic Ye1/O:8 strains do, and (ii) to increase IL-6, IL-8, IL-12 and IL-18 release. Lastly, our results confirm that IPEC-J2 cells are a very good model to evaluate host-pathogen interaction, and indicate IL-8, TNF-α, TLRs1 and 4 as possible markers of the ability of Ye strains to penetrate enterocytes. Moreover, we showed that Ye strains differently affect the host's innate immune responses.


Asunto(s)
Enterocitos/inmunología , Enterocitos/microbiología , Interacciones Microbiota-Huesped/inmunología , Inmunidad Innata , Yeyuno/citología , Yersiniosis/inmunología , Animales , Adhesión Bacteriana , Línea Celular , Citocinas/inmunología , Yeyuno/inmunología , Yeyuno/microbiología , Porcinos , Virulencia , Yersinia enterocolitica/clasificación
15.
Cell Host Microbe ; 27(3): 358-375.e7, 2020 Mar 11.
Artículo en Inglés | MEDLINE | ID: mdl-32101704

RESUMEN

Despite the recognized capacity of the gut microbiota to regulate intestinal lipid metabolism, the role of specific commensal species remains undefined. Here, we aimed to understand the bacterial effectors and molecular mechanisms by which Lactobacillus paracasei and Escherichia coli regulate lipid metabolism in enterocytes. We show that L-lactate produced by L. paracasei inhibits chylomicron secretion from enterocytes and promotes lipid storage by a mechanism involving L-lactate absorption by enterocytes, its conversion to malonyl-CoA, and the subsequent inhibition of lipid beta-oxidation. In contrast, acetate produced by E. coli also inhibits chylomicron secretion by enterocytes but promotes lipid oxidation by a mechanism involving acetate absorption by enterocytes, its metabolism to acetyl-CoA and AMP, and the subsequent upregulation of the AMPK/PGC-1α/PPARα pathway. Our study opens perspectives for developing specific bacteria- and metabolite-based therapeutic interventions against obesity, atherosclerosis, and malnutrition by targeting lipid metabolism in enterocytes.


Asunto(s)
Enterocitos/metabolismo , Escherichia coli/metabolismo , Fermentación , Lacticaseibacillus paracasei/metabolismo , Metabolismo de los Lípidos , Simbiosis , Animales , Línea Celular , Quilomicrones , Enterocitos/microbiología , Femenino , Intestinos/microbiología , Ratones Endogámicos C57BL
16.
Int J Mol Sci ; 20(22)2019 Nov 12.
Artículo en Inglés | MEDLINE | ID: mdl-31726759

RESUMEN

LF82, a prototype of adherent-invasive E. coli (AIEC), is able to adhere to, invade, survive and replicate into intestinal epithelial cells. LF82 is able to enhance either its adhesion and invasion by up-regulating carcinoembryonic antigen-related cell adhesion molecule 6 (CEACAM-6), the main cell surface molecule for bacterial adhesion, and its intracellular survival by inducing host DNA damage, thus blocking the cellular cycle. Lactoferrin (Lf) is a multifunctional cationic glycoprotein of natural immunity, exerting an anti-invasive activity against LF82 when added to Caco-2 cells at the moment of infection. Here, the infection of 12 h Lf pre-treated Caco-2 cells was carried out at a time of 0 or 3 or 10 h after Lf removal from culture medium. The effect of Lf pre-treatment on LF82 invasiveness, survival, cell DNA damage, CEACAM-6 expression, apoptosis induction, as well as on Lf subcellular localization, has been evaluated. Lf, even if removed from culture medium, reduced LF82 invasion and survival as well as bacteria-induced DNA damage in Caco-2 cells independently from induction of apoptosis, modulation of CEACAM-6 expression and Lf sub-cellular localization. At our knowledge, this is the first study showing that the sole Lf pre-treatment can activate protective intracellular pathways, reducing LF82 invasiveness, intracellular survival and cell-DNA damages.


Asunto(s)
Diferenciación Celular , Daño del ADN , Enterocitos , Escherichia coli Enteropatógena/crecimiento & desarrollo , Infecciones por Escherichia coli , Lactoferrina/farmacología , Animales , Células CACO-2 , Bovinos , Enterocitos/metabolismo , Enterocitos/microbiología , Enterocitos/patología , Infecciones por Escherichia coli/tratamiento farmacológico , Infecciones por Escherichia coli/metabolismo , Infecciones por Escherichia coli/patología , Humanos
17.
PLoS One ; 14(11): e0216762, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31675374

RESUMEN

Enterococcus faecalis is a ubiquitous intestinal symbiont and common early colonizer of the neonatal gut. Although colonization with E. faecalis has been previously associated with decreased pathology of necrotizing enterocolitis (NEC), these bacteria have been also implicated as opportunistic pathogens. Here we characterized 21 strains of E. faecalis, naturally occurring in 4-day-old rats, for potentially pathogenic properties and ability to colonize the neonatal gut. The strains differed in hemolysis, gelatin liquefaction, antibiotic resistance, biofilm formation, and ability to activate the pro-inflammatory transcription factor NF-κB in cultured enterocytes. Only 3 strains, BB70, 224, and BB24 appreciably colonized the neonatal intestine on day 4 after artificial introduction with the first feeding. The best colonizer, strain BB70, effectively displaced E. faecalis of maternal origin. Whereas BB70 and BB24 significantly increased NEC pathology, strain 224 significantly protected from NEC. Our results show that different strains of E. faecalis may be pathogenic or protective in experimental NEC.


Asunto(s)
Enterococcus faecalis/patogenicidad , Enterocolitis Necrotizante/microbiología , Animales , Animales Recién Nacidos , Modelos Animales de Enfermedad , Enterococcus faecalis/clasificación , Enterococcus faecalis/genética , Enterocolitis Necrotizante/patología , Enterocolitis Necrotizante/prevención & control , Enterocitos/microbiología , Enterocitos/patología , Femenino , Variación Genética , Humanos , Recién Nacido , Intestinos/microbiología , Intestinos/patología , Fenotipo , Embarazo , Probióticos/uso terapéutico , Ratas , Ratas Sprague-Dawley , Especificidad de la Especie , Virulencia
18.
Nat Commun ; 10(1): 4365, 2019 09 25.
Artículo en Inglés | MEDLINE | ID: mdl-31554796

RESUMEN

Epithelia are exposed to diverse types of stress and damage from pathogens and the environment, and respond by regenerating. Yet, the proximal mechanisms that sense epithelial damage remain poorly understood. Here we report that p38 signaling is activated in adult Drosophila midgut enterocytes in response to diverse stresses including pathogenic bacterial infection and chemical and mechanical insult. Two upstream kinases, Ask1 and Licorne (MKK3), are required for p38 activation following infection, oxidative stress, detergent exposure and wounding. Ask1-p38 signaling in enterocytes is required upon infection to promote full intestinal stem cell (ISC) activation and regeneration, partly through Upd3/Jak-Stat signaling. Furthermore, reactive oxygen species (ROS) produced by the NADPH oxidase Nox in enterocytes, are required for p38 activation in enterocytes following infection or wounding, and for ISC activation upon infection or detergent exposure. We propose that Nox-ROS-Ask1-MKK3-p38 signaling in enterocytes integrates multiple different stresses to induce regeneration.


Asunto(s)
Proteínas de Drosophila/metabolismo , Intestinos/fisiopatología , MAP Quinasa Quinasa 3/metabolismo , Quinasas Quinasa Quinasa PAM/metabolismo , NADPH Oxidasas/metabolismo , Regeneración/fisiología , Transducción de Señal , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Animales , Animales Modificados Genéticamente , Infecciones Bacterianas/microbiología , Proteínas de Drosophila/genética , Drosophila melanogaster/genética , Drosophila melanogaster/metabolismo , Enterocitos/metabolismo , Enterocitos/microbiología , Mucosa Intestinal/metabolismo , Mucosa Intestinal/microbiología , Mucosa Intestinal/fisiopatología , Intestinos/microbiología , Intestinos/patología , MAP Quinasa Quinasa 3/genética , Quinasas Quinasa Quinasa PAM/genética , NADPH Oxidasas/genética , Estrés Oxidativo , Regeneración/genética , Células Madre/metabolismo , Células Madre/microbiología , Estrés Mecánico , Proteínas Quinasas p38 Activadas por Mitógenos/genética
19.
Proc Natl Acad Sci U S A ; 116(34): 16961-16970, 2019 08 20.
Artículo en Inglés | MEDLINE | ID: mdl-31391308

RESUMEN

Intestinal epithelial cell (IEC) shedding is a fundamental response to intestinal damage, yet underlying mechanisms and functions have been difficult to define. Here we model chronic intestinal damage in zebrafish larvae using the nonsteroidal antiinflammatory drug (NSAID) Glafenine. Glafenine induced the unfolded protein response (UPR) and inflammatory pathways in IECs, leading to delamination. Glafenine-induced inflammation was augmented by microbial colonization and associated with changes in intestinal and environmental microbiotas. IEC shedding was a UPR-dependent protective response to Glafenine that restricts inflammation and promotes animal survival. Other NSAIDs did not induce IEC delamination; however, Glafenine also displays off-target inhibition of multidrug resistance (MDR) efflux pumps. We found a subset of MDR inhibitors also induced IEC delamination, implicating MDR efflux pumps as cellular targets underlying Glafenine-induced enteropathy. These results implicate IEC delamination as a protective UPR-mediated response to chemical injury, and uncover an essential role for MDR efflux pumps in intestinal homeostasis.


Asunto(s)
Antiinflamatorios no Esteroideos , Enterocitos/metabolismo , Microbioma Gastrointestinal , Glafenina/efectos adversos , Enfermedades Intestinales , Pez Cebra , Transportadoras de Casetes de Unión a ATP/antagonistas & inhibidores , Transportadoras de Casetes de Unión a ATP/metabolismo , Animales , Antiinflamatorios no Esteroideos/efectos adversos , Antiinflamatorios no Esteroideos/farmacología , Enterocitos/microbiología , Enterocitos/patología , Glafenina/farmacología , Inflamación/inducido químicamente , Inflamación/metabolismo , Inflamación/microbiología , Inflamación/patología , Enfermedades Intestinales/inducido químicamente , Enfermedades Intestinales/metabolismo , Enfermedades Intestinales/microbiología , Enfermedades Intestinales/patología , Pez Cebra/metabolismo , Pez Cebra/microbiología
20.
Dis Model Mech ; 12(9)2019 09 12.
Artículo en Inglés | MEDLINE | ID: mdl-31413153

RESUMEN

The intestine is the primary reservoir of Candida albicans that can cause systemic infections in immunocompromised patients. In this reservoir, the fungus exists as a harmless commensal. However, antibiotic treatment can disturb the bacterial microbiota, facilitating fungal overgrowth and favoring pathogenicity. The current in vitro gut models that are used to study the pathogenesis of C. albicans investigate the state in which C. albicans behaves as a pathogen rather than as a commensal. We present a novel in vitro gut model in which the fungal pathogenicity is reduced to a minimum by increasing the biological complexity. In this model, enterocytes represent the epithelial barrier and goblet cells limit C. albicans adhesion and invasion. Significant protection against C. albicans-induced necrotic damage was achieved by the introduction of a microbiota of antagonistic lactobacilli. We demonstrated a time-, dose- and species-dependent protective effect against C. albicans-induced cytotoxicity. This required bacterial growth, which relied on the presence of host cells, but was not dependent on the competition for adhesion sites. Lactobacillus rhamnosus reduced hyphal elongation, a key virulence attribute. Furthermore, bacterial-driven shedding of hyphae from the epithelial surface, associated with apoptotic epithelial cells, was identified as a main and novel mechanism of damage protection. However, host cell apoptosis was not the driving mechanism behind shedding. Collectively, we established an in vitro gut model that can be used to experimentally dissect commensal-like interactions of C. albicans with a bacterial microbiota and the host epithelial barrier. We also discovered fungal shedding as a novel mechanism by which bacteria contribute to the protection of epithelial surfaces.This article has an associated First Person interview with the joint first authors of the paper.


Asunto(s)
Candida albicans/fisiología , Tracto Gastrointestinal/microbiología , Lactobacillus/fisiología , Modelos Biológicos , Adhesividad , Apoptosis , Células CACO-2 , Agregación Celular , Enterocitos/microbiología , Enterocitos/patología , Enterocitos/ultraestructura , Epitelio/microbiología , Epitelio/patología , Regulación de la Expresión Génica , Glucosa/metabolismo , Interacciones Huésped-Patógeno , Humanos , Hifa/metabolismo , Lactatos/metabolismo , Lactobacillus/crecimiento & desarrollo , Viabilidad Microbiana , Oxígeno/metabolismo , Polisacáridos Bacterianos/metabolismo , Especificidad de la Especie , Estrés Fisiológico , Factores de Tiempo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...