Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Proc Natl Acad Sci U S A ; 120(48): e2315503120, 2023 Nov 28.
Artículo en Inglés | MEDLINE | ID: mdl-37988464

RESUMEN

Gasdermins (GSDMs) share a common functional domain structure and are best known for their capacity to form membrane pores. These pores are hallmarks of a specific form of cell death called pyroptosis and mediate the secretion of pro-inflammatory cytokines such as interleukin 1ß (IL1ß) and interleukin 18 (IL18). Thereby, Gasdermins have been implicated in various immune responses against cancer and infectious diseases such as acute Salmonella Typhimurium (S.Tm) gut infection. However, to date, we lack a comprehensive functional assessment of the different Gasdermins (GSDMA-E) during S.Tm infection in vivo. Here, we used epithelium-specific ablation, bone marrow chimeras, and mouse lines lacking individual Gasdermins, combinations of Gasdermins or even all Gasdermins (GSDMA1-3C1-4DE) at once and performed littermate-controlled oral S.Tm infections in streptomycin-pretreated mice to investigate the impact of all murine Gasdermins. While GSDMA, C, and E appear dispensable, we show that GSDMD i) restricts S.Tm loads in the gut tissue and systemic organs, ii) controls gut inflammation kinetics, and iii) prevents epithelium disruption by 72 h of the infection. Full protection requires GSDMD expression by both bone-marrow-derived lamina propria cells and intestinal epithelial cells (IECs). In vivo experiments as well as 3D-, 2D-, and chimeric enteroid infections further show that infected IEC extrusion proceeds also without GSDMD, but that GSDMD controls the permeabilization and morphology of the extruding IECs, affects extrusion kinetics, and promotes overall mucosal barrier capacity. As such, this work identifies a unique multipronged role of GSDMD among the Gasdermins for mucosal tissue defense against a common enteric pathogen.


Asunto(s)
Gasderminas , Infecciones por Salmonella , Animales , Ratones , Infecciones por Salmonella/prevención & control , Salmonella typhimurium , Inflamación , Células Epiteliales , Inflamasomas
2.
PLoS Pathog ; 19(6): e1011235, 2023 06.
Artículo en Inglés | MEDLINE | ID: mdl-37384776

RESUMEN

Recruitment of neutrophils into and across the gut mucosa is a cardinal feature of intestinal inflammation in response to enteric infections. Previous work using the model pathogen Salmonella enterica serovar Typhimurium (S.Tm) established that invasion of intestinal epithelial cells by S.Tm leads to recruitment of neutrophils into the gut lumen, where they can reduce pathogen loads transiently. Notably, a fraction of the pathogen population can survive this defense, re-grow to high density, and continue triggering enteropathy. However, the functions of intraluminal neutrophils in the defense against enteric pathogens and their effects on preventing or aggravating epithelial damage are still not fully understood. Here, we address this question via neutrophil depletion in different mouse models of Salmonella colitis, which differ in their degree of enteropathy. In an antibiotic pretreated mouse model, neutrophil depletion by an anti-Ly6G antibody exacerbated epithelial damage. This could be linked to compromised neutrophil-mediated elimination and reduced physical blocking of the gut-luminal S.Tm population, such that the pathogen density remained high near the epithelial surface throughout the infection. Control infections with a ssaV mutant and gentamicin-mediated elimination of gut-luminal pathogens further supported that neutrophils are protecting the luminal surface of the gut epithelium. Neutrophil depletion in germ-free and gnotobiotic mice hinted that the microbiota can modulate the infection kinetics and ameliorate epithelium-disruptive enteropathy even in the absence of neutrophil-protection. Together, our data indicate that the well-known protective effect of the microbiota is augmented by intraluminal neutrophils. After antibiotic-mediated microbiota disruption, neutrophils are central for maintaining epithelial barrier integrity during acute Salmonella-induced gut inflammation, by limiting the sustained pathogen assault on the epithelium in a critical window of the infection.


Asunto(s)
Neutrófilos , Infecciones por Salmonella , Animales , Ratones , Salmonella typhimurium , Células Epiteliales , Antibacterianos , Inflamación , Epitelio , Mucosa Intestinal
3.
J Exp Med ; 218(11)2021 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-34529751

RESUMEN

Intestinal epithelial cell (IEC) NF-κB signaling regulates the balance between mucosal homeostasis and inflammation. It is not fully understood which signals tune this balance and how bacterial exposure elicits the process. Pure LPS induces epithelial NF-κB activation in vivo. However, we found that in mice, IECs do not respond directly to LPS. Instead, tissue-resident lamina propria intercrypt macrophages sense LPS via TLR4 and rapidly secrete TNF to elicit epithelial NF-κB signaling in their immediate neighborhood. This response pattern is relevant also during oral enteropathogen infection. The macrophage-TNF-IEC axis avoids responses to luminal microbiota LPS but enables crypt- or tissue-scale epithelial NF-κB responses in proportion to the microbial threat. Thereby, intercrypt macrophages fulfill important sentinel functions as first responders to Gram-negative microbes breaching the epithelial barrier. The tunability of this crypt response allows the induction of defense mechanisms at an appropriate scale according to the localization and intensity of microbial triggers.


Asunto(s)
Antibacterianos/metabolismo , Células Epiteliales/metabolismo , Mucosa Intestinal/metabolismo , Intestinos/metabolismo , FN-kappa B/metabolismo , Factores de Necrosis Tumoral/metabolismo , Animales , Regulación de la Expresión Génica/fisiología , Inflamación/metabolismo , Macrófagos/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Transducción de Señal/fisiología
4.
PLoS One ; 9(10): e110187, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25303479

RESUMEN

Salmonella enterica is a common cause of diarrhea. For eliciting disease, the pathogen has to colonize the gut lumen, a site colonized by the microbiota. This process/initial stage is incompletely understood. Recent work established that one particular strain, Salmonella enterica subspecies 1 serovar Typhimurium strain SL1344, employs the hyb H2-hydrogenase for consuming microbiota-derived H2 to support gut luminal pathogen growth: Protons from the H2-splitting reaction contribute to the proton gradient across the outer bacterial membrane which can be harvested for ATP production or for import of carbon sources. However, it remained unclear, if other Salmonella strains would use the same strategy. In particular, earlier work had left unanswered if strain ATCC14028 might use H2 for growth at systemic sites. To clarify the role of the hydrogenases, it seems important to establish if H2 is used at systemic sites or in the gut and if Salmonella strains may differ with respect to the host sites where they require H2 in vivo. In order to resolve this, we constructed a strain lacking all three H2-hydrogenases of ATCC14028 (14028hyd3) and performed competitive infection experiments. Upon intragastric inoculation, 14028hyd3 was present at 100-fold lower numbers than 14028WT in the stool and at systemic sites. In contrast, i.v. inoculation led to equivalent systemic loads of 14028hyd3 and the wild type strain. However, the pathogen population spreading to the gut lumen featured again up to 100-fold attenuation of 14028hyd3. Therefore, ATCC14028 requires H2-hydrogenases for growth in the gut lumen and not at systemic sites. This extends previous work on ATCC14028 and supports the notion that H2-utilization might be a general feature of S. Typhimurium gut colonization.


Asunto(s)
Tracto Gastrointestinal/microbiología , Hidrogenasas/metabolismo , Salmonella typhimurium/enzimología , Salmonella typhimurium/crecimiento & desarrollo , Animales , Modelos Animales de Enfermedad , Ratones , Infecciones por Salmonella/microbiología , Salmonella typhimurium/patogenicidad , Factores de Virulencia
5.
Cell Microbiol ; 16(12): 1723-35, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25052734

RESUMEN

Salmonella enterica serovar Typhimurium (S. Tm) is a facultative intracellular pathogen that induces entry into non-phagocytic cells by a Type III secretion system (TTSS) and cognate effector proteins. Upon host cell entry, S. Tm expresses a second TTSS and subverts intracellular trafficking to create a replicative niche - the Salmonella-containing vacuole (SCV). SopE, a guanidyl exchange factor (GEF) for Rac1 and Cdc42, is translocated by the TTSS-1 upon host cell contact and promotes entry through triggering of actin-dependent ruffles. After host cell entry, the bulk of SopE undergoes proteasomal degradation. Here we show that a subfraction is however detectable on the nascent SCV membrane up to ∼ 6 h post infection. Membrane localization of SopE and the closely related SopE2 differentially depend on the Rho-GTPase-binding GEF domain, and to some extent involves also the unstructured N-terminus. SopE localizes transiently to the early SCV, dependent on continuous synthesis and secretion by the TTSS-1 during the intracellular state. Mutant strains lacking SopE or SopE2 are attenuated in early intracellular replication, while complementation restores this defect. Hence, the present study reveals an unanticipated role for SopE and SopE2 in establishing the Salmonella replicative niche, and further emphasizes the importance of entry effectors in later stages of host-cell manipulation.


Asunto(s)
Proteínas Bacterianas/análisis , Interacciones Huésped-Patógeno , Membranas Intracelulares/química , Salmonella typhimurium/crecimiento & desarrollo , Vacuolas/química , Vacuolas/microbiología , Animales , Análisis Mutacional de ADN , Células HeLa , Humanos , Ratones , Células 3T3 NIH , Transporte de Proteínas , Infecciones por Salmonella/microbiología
6.
Antimicrob Agents Chemother ; 56(5): 2295-304, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-22354292

RESUMEN

Nontyphoidal Salmonella (NTS) species cause self-limiting diarrhea and sometimes severe disease. Antibiotic treatment is considered only in severe cases and immune-compromised patients. The beneficial effects of antibiotic therapy and the consequences for adaptive immune responses are not well understood. We used a mouse model for Salmonella diarrhea to assess the effects of per os treatment with ciprofloxacin (15 mg/kg of body weight intragastrically 2 times/day, 5 days) or parenteral ceftriaxone (50 mg/kg intraperitoneally, 5 days), two common drugs used in human patients. The therapeutic and adverse effects were assessed with respect to generation of a protective adaptive immune response, fecal pathogen excretion, and the emergence of nonsymptomatic excreters. In the mouse model, both therapies reduced disease severity and reduced the level of fecal shedding. In line with clinical data, in most animals, a rebound of pathogen gut colonization/fecal shedding was observed 2 to 12 days after the end of the treatment. Yet, levels of pathogen shedding and frequency of appearance of nonsymptomatic excreters did not differ from those for untreated controls. Moreover, mice treated intraperitoneally with ceftriaxone developed an adaptive immunity protecting the mice from enteropathy in wild-type Salmonella enterica serovar Typhimurium challenge infections. In contrast, the mice treated intragastrically with ciprofloxacin were not protected. Thus, antibiotic treatment regimens can disrupt the adaptive immune response, but treatment regimens may be optimized in order to preserve the generation of protective immunity. It might be of interest to determine whether this also pertains to human patients. In this case, the mouse model might be a tool for further mechanistic studies.


Asunto(s)
Inmunidad Adaptativa/efectos de los fármacos , Ceftriaxona/administración & dosificación , Ciprofloxacina/administración & dosificación , Diarrea/tratamiento farmacológico , Infecciones por Salmonella/tratamiento farmacológico , Salmonella typhimurium/efectos de los fármacos , Administración Oral , Animales , Antibacterianos/administración & dosificación , Antibacterianos/uso terapéutico , Ceftriaxona/uso terapéutico , Ciprofloxacina/uso terapéutico , Recuento de Colonia Microbiana , Diarrea/inmunología , Diarrea/microbiología , Modelos Animales de Enfermedad , Esquema de Medicación , Heces/microbiología , Infusiones Parenterales , Ratones , Ratones Endogámicos C57BL , Infecciones por Salmonella/inmunología , Infecciones por Salmonella/microbiología , Salmonella typhimurium/inmunología , Índice de Severidad de la Enfermedad
7.
Proc Natl Acad Sci U S A ; 109(4): 1269-74, 2012 Jan 24.
Artículo en Inglés | MEDLINE | ID: mdl-22232693

RESUMEN

The mammalian gut harbors a dense microbial community interacting in multiple ways, including horizontal gene transfer (HGT). Pangenome analyses established particularly high levels of genetic flux between Gram-negative Enterobacteriaceae. However, the mechanisms fostering intraenterobacterial HGT are incompletely understood. Using a mouse colitis model, we found that Salmonella-inflicted enteropathy elicits parallel blooms of the pathogen and of resident commensal Escherichia coli. These blooms boosted conjugative HGT of the colicin-plasmid p2 from Salmonella enterica serovar Typhimurium to E. coli. Transconjugation efficiencies of ~100% in vivo were attributable to high intrinsic p2-transfer rates. Plasmid-encoded fitness benefits contributed little. Under normal conditions, HGT was blocked by the commensal microbiota inhibiting contact-dependent conjugation between Enterobacteriaceae. Our data show that pathogen-driven inflammatory responses in the gut can generate transient enterobacterial blooms in which conjugative transfer occurs at unprecedented rates. These blooms may favor reassortment of plasmid-encoded genes between pathogens and commensals fostering the spread of fitness-, virulence-, and antibiotic-resistance determinants.


Asunto(s)
Evolución Biológica , Colitis/microbiología , Enterobacteriaceae/genética , Transferencia de Gen Horizontal/genética , Animales , Plásmidos de Bacteriocinas/genética , Secuencia de Bases , Biología Computacional , Cartilla de ADN/genética , Enterobacteriaceae/crecimiento & desarrollo , Escherichia coli/genética , Ratones , Datos de Secuencia Molecular , Análisis de Secuencia por Matrices de Oligonucleótidos , Filogenia , ARN Ribosómico 16S/genética , Salmonella typhimurium/genética , Alineación de Secuencia , Análisis de Secuencia de ADN
8.
PLoS One ; 6(7): e22459, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21829463

RESUMEN

Enteropathogenic bacteria are a frequent cause of diarrhea worldwide. The mucosal defenses against infection are not completely understood. We have used the streptomycin mouse model for Salmonella Typhimurium diarrhea to analyze the role of interferon gamma receptor (IFN-γR)-signaling in mucosal defense. IFN-γ is known to contribute to acute S. Typhimurium diarrhea. We have compared the acute mucosal inflammation in IFN-γR(-/-) mice and wild type animals. IFN-γR(-/-) mice harbored increased pathogen loads in the mucosal epithelium and the lamina propria. Surprisingly, the epithelium of the IFN-γR(-/-) mice did not show the dramatic "loss" of mucus-filled goblet cell vacuoles, a hallmark of the wild type mucosal infection. Using bone marrow chimeric mice we established that IFN-γR-signaling in stromal cells (e.g. goblet cells, enterocytes) controlled mucus excretion/vacuole loss by goblet cells. In contrast, IFN-γR-signaling in bone marrow-derived cells (e.g. macrophages, DCs, PMNs) was required for restricting pathogen growth in the gut tissue. Thus IFN-γR-signaling influences different mucosal responses to infection, including not only pathogen restriction in the lamina propria, but, as shown here, also goblet cell function.


Asunto(s)
Células Caliciformes/fisiología , Receptores de Interferón/fisiología , Infecciones por Salmonella/inmunología , Salmonella typhimurium/inmunología , Células del Estroma/inmunología , Animales , Médula Ósea/inmunología , Médula Ósea/metabolismo , Médula Ósea/microbiología , Células Cultivadas , Técnica del Anticuerpo Fluorescente , Interferón gamma/metabolismo , Mucosa Intestinal/inmunología , Mucosa Intestinal/metabolismo , Mucosa Intestinal/microbiología , Macrófagos/inmunología , Macrófagos/metabolismo , Macrófagos/microbiología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Infecciones por Salmonella/microbiología , Salmonella typhimurium/genética , Transducción de Señal , Receptor de Interferón gamma
9.
PLoS One ; 5(11): e13804, 2010 Nov 23.
Artículo en Inglés | MEDLINE | ID: mdl-21124903

RESUMEN

Salmonella enterica subspecies 1 serovar Typhimurium (S. Typhimurium) causes diarrhea and acute inflammation of the intestinal mucosa. The pro-inflammatory cytokines IL-17A and IL-17F are strongly induced in the infected mucosa but their contribution in driving the tissue inflammation is not understood. We have used the streptomycin mouse model to analyze the role of IL-17A and IL-17F and their cognate receptor IL-17RA in S. Typhimurium enterocolitis. Neutralization of IL-17A and IL-17F did not affect mucosal inflammation triggered by infection or spread of S. Typhimurium to systemic sites by 48 h p.i. Similarly, Il17ra(-/-) mice did not display any reduction in infection or inflammation by 12 h p.i. The same results were obtained using S. Typhimurium variants infecting via the TTSS1 type III secretion system, the TTSS1 effector SipA or the TTSS1 effector SopE. Moreover, the expression pattern of 45 genes encoding chemokines/cytokines (including CXCL1, CXCL2, IL-17A, IL-17F, IL-1α, IL-1ß, IFNγ, CXCL-10, CXCL-9, IL-6, CCL3, CCL4) and antibacterial molecules was not affected by Il17ra deficiency by 12 h p.i. Thus, in spite of the strong increase in Il17a/Il17f mRNA in the infected mucosa, IL-17RA signaling seems to be dispensable for eliciting the acute disease. Future work will have to address whether this is attributable to redundancy in the cytokine signaling network.


Asunto(s)
Enterocolitis/inmunología , Interleucina-17/inmunología , Salmonelosis Animal/inmunología , Salmonella typhimurium/inmunología , Animales , Ciego/inmunología , Ciego/metabolismo , Quimiocinas/genética , Citocinas/genética , Enterocolitis/genética , Femenino , Perfilación de la Expresión Génica , Interleucina-17/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Mutación , Receptores de Interleucina-17/deficiencia , Receptores de Interleucina-17/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Salmonelosis Animal/genética , Salmonella typhimurium/genética , Transducción de Señal/inmunología , Factores de Tiempo , Vacunación/métodos
10.
Infect Immun ; 78(8): 3420-31, 2010 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-20515923

RESUMEN

The immune system comprises an innate and an adaptive immune response to combat pathogenic agents. The human enteropathogen Salmonella enterica serovar Typhimurium invades the intestinal mucosa and triggers an early innate proinflammatory host gene response, which results in diarrheal disease. Several host factors, including transcription factors and transcription coregulators, are involved in the acute early response to Salmonella infection. We found in a mouse model of enterocolitis induced by S. Typhimurium that the absence of the nuclear protein poly(ADP-ribose) polymerase 1 (PARP1), a previously described cofactor for NF-kappaB-mediated proinflammatory gene expression, is associated with a delayed proinflammatory immune response after Salmonella infection. Our data reveal that PARP1 is expressed in the proliferative zone of cecum crypts, where it is required for the efficient expression of proinflammatory genes, many of which are related to interferon signaling. Consequently, animals lacking PARP1 show impaired infiltration of immune cells into the gut, with severely delayed inflammation.


Asunto(s)
Poli(ADP-Ribosa) Polimerasas/inmunología , Salmonelosis Animal/inmunología , Salmonelosis Animal/patología , Salmonella typhimurium/inmunología , Salmonella typhimurium/patogenicidad , Animales , Ciego/inmunología , Ciego/patología , Modelos Animales de Enfermedad , Histocitoquímica , Humanos , Inmunohistoquímica , Inflamación/inmunología , Inflamación/patología , Ratones , Ratones Endogámicos C57BL , Microscopía , Microscopía Fluorescente , Poli(ADP-Ribosa) Polimerasa-1 , Poli(ADP-Ribosa) Polimerasas/deficiencia
11.
J Infect Dis ; 199(2): 243-52, 2009 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-19072552

RESUMEN

Dietary exposure to prion-contaminated materials has caused kuru and variant Creutzfeldt-Jakob disease in humans and transmissible spongiform encephalopathies (TSEs) in cattle, mink, and felines. The epidemiology of dietary prion infections suggests that host genetic modifiers and possibly exogenous cofactors may play a decisive role in determining disease susceptibility. However, few cofactors influencing susceptibility to prion infection have been identified. In the present study, we investigated whether colitis might represent one such cofactor. We report that moderate colitis caused by an attenuated Salmonella strain more than doubles the susceptibility of mice to oral prion infection and modestly accelerates the development of disease after prion challenge. The prion protein was up-regulated in intestines and mesenteric lymph nodes of mice with colitis, providing a possible mechanism for the effect of colitis on the pathogenesis of prion disease. Therefore, moderate intestinal inflammation at the time of prion exposure may constitute one of the elusive risk factors underlying the development of TSE.


Asunto(s)
Enterocolitis/complicaciones , Enfermedades de la Boca/complicaciones , Enfermedades por Prión/complicaciones , Priones/patogenicidad , Infecciones por Salmonella/complicaciones , Salmonella typhimurium/patogenicidad , Animales , Ciego/metabolismo , Susceptibilidad a Enfermedades , Enterocolitis/microbiología , Ratones , Ratones Endogámicos C57BL , Enfermedades de la Boca/metabolismo , Proteínas PrPC/metabolismo , Proteínas PrPSc/metabolismo , Enfermedades por Prión/metabolismo , Priones/metabolismo , Factores de Riesgo , Infecciones por Salmonella/microbiología , Salmonella typhimurium/genética , Scrapie/complicaciones , Scrapie/metabolismo , Organismos Libres de Patógenos Específicos
12.
J Exp Med ; 205(2): 437-50, 2008 Feb 18.
Artículo en Inglés | MEDLINE | ID: mdl-18268033

RESUMEN

Intestinal dendritic cells (DCs) are believed to sample and present commensal bacteria to the gut-associated immune system to maintain immune homeostasis. How antigen sampling pathways handle intestinal pathogens remains elusive. We present a murine colitogenic Salmonella infection model that is highly dependent on DCs. Conditional DC depletion experiments revealed that intestinal virulence of S. Typhimurium SL1344 DeltainvG mutant lacking a functional type 3 secretion system-1 (DeltainvG)critically required DCs for invasion across the epithelium. The DC-dependency was limited to the early phase of infection when bacteria colocalized with CD11c(+)CX3CR1(+) mucosal DCs. At later stages, the bacteria became associated with other (CD11c(-)CX3CR1(-)) lamina propria cells, DC depletion no longer attenuated the pathology, and a MyD88-dependent mucosal inflammation was initiated. Using bone marrow chimeric mice, we showed that the MyD88 signaling within hematopoietic cells, which are distinct from DCs, was required and sufficient for induction of the colitis. Moreover, MyD88-deficient DCs supported transepithelial uptake of the bacteria and the induction of MyD88-dependent colitis. These results establish that pathogen sampling by DCs is a discrete, and MyD88-independent, step during the initiation of a mucosal innate immune response to bacterial infection in vivo.


Asunto(s)
Colitis/inmunología , Células Dendríticas/inmunología , Mucosa Intestinal/inmunología , Infecciones por Salmonella/inmunología , Salmonella typhimurium , Animales , Antígenos CD11/inmunología , Receptor 1 de Quimiocinas CX3C , Ciego/inmunología , Ciego/patología , Colitis/patología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Factor 88 de Diferenciación Mieloide/deficiencia , Factor 88 de Diferenciación Mieloide/genética , Factor 88 de Diferenciación Mieloide/inmunología , Receptores de Quimiocina/inmunología , Infecciones por Salmonella/patología
13.
Cell Microbiol ; 10(5): 1166-80, 2008 May.
Artículo en Inglés | MEDLINE | ID: mdl-18241212

RESUMEN

The mammalian intestine is colonized by a dense bacterial community, called microbiota. The microbiota shields from intestinal infection (colonization resistance). Recently, we have shown that enteropathogenic Salmonella spp. can exploit inflammation to compete with the intestinal microbiota. The mechanisms explaining the enhanced pathogen growth in the inflamed intestine are elusive. Here, we analysed the function of bacterial flagella in the inflamed intestine using a mouse model for acute Salmonella Typhimurium enterocolitis. Mutations affecting flagellar assembly (Fla(-)) and chemotaxis (Che(-)) impaired the pathogen's fitness in the inflamed intestine, but not in the normal gut. This was attributable to a localized source of high-energy nutrients (e.g. galactose-containing glyco-conjugates, mucin) released as an element of the mucosal defence. Motility allows Salmonella Typhimurium to benefit from these nutrients and utilize them for enhanced growth. Thus, nutrient availability contributes to enhanced pathogen growth in the inflamed intestine. Strategies interfering with bacterial motility or nutrient availability might offer starting points for therapeutic approaches.


Asunto(s)
Mucosa Intestinal/microbiología , Salmonella typhimurium/fisiología , Animales , Ciego/microbiología , Flagelos/fisiología , Galactosa/metabolismo , Ratones , Ratones Endogámicos C57BL , Factor 88 de Diferenciación Mieloide/metabolismo , Operón , Salmonelosis Animal/microbiología , Salmonella typhimurium/genética , Organismos Libres de Patógenos Específicos
14.
PLoS Biol ; 5(10): 2177-89, 2007 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-17760501

RESUMEN

Most mucosal surfaces of the mammalian body are colonized by microbial communities ("microbiota"). A high density of commensal microbiota inhabits the intestine and shields from infection ("colonization resistance"). The virulence strategies allowing enteropathogenic bacteria to successfully compete with the microbiota and overcome colonization resistance are poorly understood. Here, we investigated manipulation of the intestinal microbiota by the enteropathogenic bacterium Salmonella enterica subspecies 1 serovar Typhimurium (S. Tm) in a mouse colitis model: we found that inflammatory host responses induced by S. Tm changed microbiota composition and suppressed its growth. In contrast to wild-type S. Tm, an avirulent invGsseD mutant failing to trigger colitis was outcompeted by the microbiota. This competitive defect was reverted if inflammation was provided concomitantly by mixed infection with wild-type S. Tm or in mice (IL10(-/-), VILLIN-HA(CL4-CD8)) with inflammatory bowel disease. Thus, inflammation is necessary and sufficient for overcoming colonization resistance. This reveals a new concept in infectious disease: in contrast to current thinking, inflammation is not always detrimental for the pathogen. Triggering the host's immune defence can shift the balance between the protective microbiota and the pathogen in favour of the pathogen.


Asunto(s)
Colitis/microbiología , Intestinos/microbiología , Infecciones por Salmonella/microbiología , Salmonella typhimurium/aislamiento & purificación , Animales , Bacterias/clasificación , Bacterias/genética , Bacterias/aislamiento & purificación , Colitis/patología , Femenino , Genotipo , Hibridación Fluorescente in Situ , Intestinos/patología , Masculino , Ratones , Ratones Endogámicos C3H , Ratones Endogámicos C57BL , Ratones Endogámicos , Microscopía Fluorescente , Modelos Animales , Datos de Secuencia Molecular , Mutación , Filogenia , ARN Ribosómico 16S/genética , Infecciones por Salmonella/patología , Salmonella typhimurium/clasificación , Salmonella typhimurium/genética , Análisis de Secuencia de ADN
15.
Infect Immun ; 74(9): 5047-57, 2006 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-16926396

RESUMEN

Salmonella enterica subspecies 1 serovar Typhimurium is an enteric bacterial pathogen infecting a broad range of hosts. In susceptible Nramp1(-/-) (Slc11alpha1(-/-)) mice, serovar Typhimurium cannot efficiently colonize the intestine but causes a systemic typhoid-like infection. However, after pretreatment with streptomycin, these susceptible (C57BL/6 and BALB/c) mice develop acute serovar Typhimurium-induced colitis (M. Barthel et al., Infect. Immun. 71:2839-2858, 2003). It was not clear whether resistant Nramp1(+/+) (Slc11alpha1(+/+)) mouse strains would similarly develop colitis. Here we compared serovar Typhimurium infection in streptomycin-pretreated susceptible (C57BL/6) and resistant (DBA/2 and 129Sv/Ev) mouse strains: We found that acute colitis (days 1 and 3 postinfection) is strikingly similar in susceptible and resistant mice. In 129Sv/Ev mice we followed the serovar Typhimurium infection for as long as 6 weeks. After the initial phase of acute colitis, these animals developed chronic crypt-destructive colitis, including ulceration, crypt abscesses, pronounced mucosal and submucosal infiltrates, overshooting regeneration of the epithelium, and crypt branching. Moreover, we observed inflammation of the gall duct epithelium (cholangitis) in the 129Sv/Ev mice between days 14 and 43 of infection. Cholangitis was not attributable to side effects of the streptomycin treatment. Furthermore, chronic infection of 129Sv/Ev mice in a typhoid fever model did not lead to cholangitis. We propose that streptomycin-pretreated 129Sv/Ev mice provide a robust murine model for chronic enteric salmonellosis including complications such as cholangitis.


Asunto(s)
Colangitis/microbiología , Colitis/microbiología , Modelos Animales de Enfermedad , Ratones/microbiología , Infecciones por Salmonella/microbiología , Salmonella typhimurium , Animales , Proteínas de Transporte de Catión/genética , Colangitis/genética , Colangitis/patología , Enfermedad Crónica , Colitis/genética , Colitis/patología , Susceptibilidad a Enfermedades , Femenino , Masculino , Ratones Endogámicos DBA , Ratones Mutantes , Infecciones por Salmonella/genética , Infecciones por Salmonella/patología , Estreptomicina/administración & dosificación
16.
J Immunol ; 174(3): 1675-85, 2005 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-15661931

RESUMEN

Salmonella typhimurium can colonize the gut, invade intestinal tissues, and cause enterocolitis. In vitro studies suggest different mechanisms leading to mucosal inflammation, including 1) direct modulation of proinflammatory signaling by bacterial type III effector proteins and 2) disruption or penetration of the intestinal epithelium so that penetrating bacteria or bacterial products can trigger innate immunity (i.e., TLR signaling). We studied these mechanisms in vivo using streptomycin-pretreated wild-type and knockout mice including MyD88(-/-) animals lacking an adaptor molecule required for signaling via most TLRs. The Salmonella SPI-1 and the SPI-2 type III secretion systems (TTSS) contributed to inflammation. Mutants that retain only a functional SPI-1 (M556; sseD::aphT) or a SPI-2 TTSS (SB161; DeltainvG) caused attenuated colitis, which reflected distinct aspects of the colitis caused by wild-type S. typhimurium: M556 caused diffuse cecal inflammation that did not require MyD88 signaling. In contrast, SB161 induced focal mucosal inflammation requiring MyD88. M556 but not SB161 was found in intestinal epithelial cells. In the lamina propria, M556 and SB161 appeared to reside in different leukocyte cell populations as indicated by differential CD11c staining. Only the SPI-2-dependent inflammatory pathway required aroA-dependent intracellular growth. Thus, S. typhimurium can use two independent mechanisms to elicit colitis in vivo: SPI-1-dependent and MyD88-independent signaling to epithelial cells and SPI-2-dependent intracellular proliferation in the lamina propria triggering MyD88-dependent innate immune responses.


Asunto(s)
Antígenos de Diferenciación/fisiología , Proteínas Bacterianas/fisiología , Colitis/inmunología , Colitis/microbiología , Proteínas de la Membrana/fisiología , Receptores Inmunológicos/fisiología , Salmonella typhimurium/patogenicidad , Transducción de Señal/inmunología , 3-Fosfoshikimato 1-Carboxiviniltransferasa , Proteínas Adaptadoras Transductoras de Señales , Transferasas Alquil y Aril/genética , Animales , Antígenos de Diferenciación/genética , Proteínas Bacterianas/genética , Colitis/patología , Eliminación de Gen , Mucosa Intestinal/inmunología , Mucosa Intestinal/microbiología , Mucosa Intestinal/patología , Leucocitos/inmunología , Leucocitos/microbiología , Leucocitos/patología , Tejido Linfoide/inmunología , Tejido Linfoide/microbiología , Tejido Linfoide/patología , Proteínas de la Membrana/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Factor 88 de Diferenciación Mieloide , Receptores Inmunológicos/deficiencia , Receptores Inmunológicos/genética , Salmonella typhimurium/enzimología , Salmonella typhimurium/genética , Salmonella typhimurium/crecimiento & desarrollo , Transducción de Señal/genética
17.
Infect Immun ; 72(2): 795-809, 2004 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-14742523

RESUMEN

Salmonella enterica subspecies 1 serovar Typhimurium (serovar Typhimurium) induces enterocolitis in humans and cattle. The mechanisms of enteric salmonellosis have been studied most extensively in calf infection models. The previous studies established that effector protein translocation into host cells via the Salmonella pathogenicity island 1 (SPI-1) type III secretion system (TTSS) is of central importance in serovar Typhimurium enterocolitis. We recently found that orally streptomycin-pretreated mice provide an alternative model for serovar Typhimurium colitis. In this model the SPI-1 TTSS also plays a key role in the elicitation of intestinal inflammation. However, whether intestinal inflammation in calves and intestinal inflammation in streptomycin-pretreated mice are induced by the same SPI-1 effector proteins is still unclear. Therefore, we analyzed the role of the SPI-1 effector proteins SopB/SigD, SopE, SopE2, and SipA/SspA in elicitation of intestinal inflammation in the murine model. We found that sipA, sopE, and, to a lesser degree, sopE2 contribute to murine colitis, but we could not assign an inflammation phenotype to sopB. These findings are in line with previous studies performed with orally infected calves. Extending these observations, we demonstrated that in addition to SipA, SopE and SopE2 can induce intestinal inflammation independent of each other and in the absence of SopB. In conclusion, our data corroborate the finding that streptomycin-pretreated mice provide a useful model for studying the molecular mechanisms of serovar Typhimurium colitis and are an important starting point for analysis of the molecular events triggered by SopE, SopE2, and SipA in vivo.


Asunto(s)
Proteínas Bacterianas/fisiología , Colitis/etiología , Factores de Intercambio de Guanina Nucleótido/fisiología , Proteínas de Microfilamentos/fisiología , Salmonelosis Animal/etiología , Salmonella typhimurium/patogenicidad , Estreptomicina/farmacología , Animales , Células COS , Femenino , Ratones , Ratones Endogámicos C57BL
18.
Infect Immun ; 71(5): 2839-58, 2003 May.
Artículo en Inglés | MEDLINE | ID: mdl-12704158

RESUMEN

Salmonella enterica subspecies 1 serovar Typhimurium is a principal cause of human enterocolitis. For unknown reasons, in mice serovar Typhimurium does not provoke intestinal inflammation but rather targets the gut-associated lymphatic tissues and causes a systemic typhoid-like infection. The lack of a suitable murine model has limited the analysis of the pathogenetic mechanisms of intestinal salmonellosis. We describe here how streptomycin-pretreated mice provide a mouse model for serovar Typhimurium colitis. Serovar Typhimurium colitis in streptomycin-pretreated mice resembles many aspects of the human infection, including epithelial ulceration, edema, induction of intercellular adhesion molecule 1, and massive infiltration of PMN/CD18(+) cells. This pathology is strongly dependent on protein translocation via the serovar Typhimurium SPI1 type III secretion system. Using a lymphotoxin beta-receptor knockout mouse strain that lacks all lymph nodes and organized gut-associated lymphatic tissues, we demonstrate that Peyer's patches and mesenteric lymph nodes are dispensable for the initiation of murine serovar Typhimurium colitis. Our results demonstrate that streptomycin-pretreated mice offer a unique infection model that allows for the first time to use mutants of both the pathogen and the host to study the molecular mechanisms of enteric salmonellosis.


Asunto(s)
Colitis/etiología , Modelos Animales de Enfermedad , Salmonelosis Animal/etiología , Salmonella typhimurium/patogenicidad , Estreptomicina/farmacología , Animales , Proteínas Bacterianas/metabolismo , Colitis/patología , Femenino , Intestinos/inmunología , Intestinos/microbiología , Intestinos/patología , Lactobacillus/patogenicidad , Tejido Linfoide/inmunología , Receptor beta de Linfotoxina , Ratones , Ratones Endogámicos C57BL , Ganglios Linfáticos Agregados/fisiología , Receptores del Factor de Necrosis Tumoral/fisiología , Salmonelosis Animal/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...