Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
Infect Immun ; 92(2): e0031823, 2024 Feb 13.
Artículo en Inglés | MEDLINE | ID: mdl-38189339

RESUMEN

Inflammation has a pronounced impact on the intestinal ecosystem by driving an expansion of facultative anaerobic bacteria at the cost of obligate anaerobic microbiota. This pathogen "blooming" is also a hallmark of enteric Salmonella enterica serovar Typhimurium (S. Tm) infection. Here, we analyzed the contribution of bacterial and host factors to S. Tm "blooming" in a gnotobiotic mouse model for S. Tm-induced enterocolitis. Mice colonized with the Oligo-Mouse-Microbiota (OMM12), a minimal bacterial community, develop fulminant colitis by day 4 after oral infection with wild-type S. Tm but not with an avirulent mutant. Inflammation leads to a pronounced reduction in overall intestinal bacterial loads, distinct microbial community shifts, and pathogen blooming (relative abundance >50%). S. Tm mutants attenuated in inducing gut inflammation generally elicit less pronounced microbiota shifts and reduction in total bacterial loads. In contrast, S. Tm mutants in nitrate respiration, salmochelin production, and ethanolamine utilization induced strong inflammation and S. Tm "blooming." Therefore, individual Salmonella-specific inflammation-fitness factors seem to be of minor importance for competition against this minimal microbiota in the inflamed gut. Finally, we show that antibody-mediated neutrophil depletion normalized gut microbiota loads but not intestinal inflammation or microbiota shifts. This suggests that neutrophils equally reduce pathogen and commensal bacterial loads in the inflamed gut.


Asunto(s)
Enterocolitis , Microbiota , Salmonelosis Animal , Ratones , Animales , Salmonella typhimurium , Serogrupo , Bacterias , Inflamación , Modelos Animales de Enfermedad , Vida Libre de Gérmenes , Salmonelosis Animal/microbiología
2.
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
3.
Nat Microbiol ; 4(12): 2164-2174, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31591555

RESUMEN

The microbiota confers colonization resistance, which blocks Salmonella gut colonization1. As diet affects microbiota composition, we studied whether food composition shifts enhance susceptibility to infection. Shifting mice to diets with reduced fibre or elevated fat content for 24 h boosted Salmonella Typhimurium or Escherichia coli gut colonization and plasmid transfer. Here, we studied the effect of dietary fat. Colonization resistance was restored within 48 h of return to maintenance diet. Salmonella gut colonization was also boosted by two oral doses of oleic acid or bile salts. These pathogen blooms required Salmonella's AcrAB/TolC-dependent bile resistance. Our data indicate that fat-elicited bile promoted Salmonella gut colonization. Both E. coli and Salmonella show much higher bile resistance than the microbiota. Correspondingly, competitive E. coli can be protective in the fat-challenged gut. Diet shifts and fat-elicited bile promote S. Typhimurium gut infections in mice lacking E. coli in their microbiota. This mouse model may be useful for studying pathogen-microbiota-host interactions, the protective effect of E. coli, to analyse the spread of resistance plasmids and assess the impact of food components on the infection process.


Asunto(s)
Grasas de la Dieta/administración & dosificación , Escherichia coli/fisiología , Microbioma Gastrointestinal , Interacciones Microbianas , Salmonella typhimurium/fisiología , Alimentación Animal , Animales , Ácidos y Sales Biliares/administración & dosificación , Femenino , Interacciones Huésped-Patógeno , Masculino , Ratones , Ratones Endogámicos C57BL , Ácidos Oléicos/administración & dosificación
4.
Cell Host Microbe ; 25(3): 389-403.e6, 2019 Mar 13.
Artículo en Inglés | MEDLINE | ID: mdl-30870621

RESUMEN

Commensal fungi of the mammalian skin, such as those of the genus Malassezia, are associated with atopic dermatitis and other common inflammatory skin disorders. Understanding of the causative relationship between fungal commensalism and disease manifestation remains incomplete. By developing a murine epicutaneous infection model, we found Malassezia spp. selectively induce IL-17 and related cytokines. This response is key in preventing fungal overgrowth on the skin, as disruption of the IL-23-IL-17 axis compromises Malassezia-specific cutaneous immunity. Under conditions of impaired skin integrity, mimicking a hallmark of atopic dermatitis, the presence of Malassezia dramatically aggravates cutaneous inflammation, which again was IL-23 and IL-17 dependent. Consistently, we found a CCR6+ Th17 subset of memory T cells to be Malassezia specific in both healthy individuals and atopic dermatitis patients, whereby the latter showed enhanced frequency of these cells. Thus, the Malassezia-induced type 17 response is pivotal in orchestrating antifungal immunity and in actively promoting skin inflammation.


Asunto(s)
Citocinas/metabolismo , Dermatitis Atópica/patología , Dermatomicosis/microbiología , Dermatomicosis/patología , Malassezia/inmunología , Células Th17/inmunología , Adulto , Animales , Modelos Animales de Enfermedad , Femenino , Humanos , Masculino , Ratones Endogámicos C57BL , Persona de Mediana Edad , Adulto Joven
5.
Front Immunol ; 9: 1344, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29951069

RESUMEN

CD4+Foxp3+ Treg cells are essential for maintaining self-tolerance and preventing excessive immune responses. In the context of Th1 immune responses, co-expression of the Th1 transcription factor T-bet with Foxp3 is essential for Treg cells to control Th1 responses. T-bet-dependent expression of CXCR3 directs Treg cells to the site of inflammation. However, the suppressive mediators enabling effective control of Th1 responses at this site are unknown. In this study, we determined the signature of CXCR3+ Treg cells arising in Th1 settings and defined universal features of Treg cells in this context using multiple Th1-dominated infection models. Our analysis defined a set of Th1-specific co-inhibitory receptors and cytotoxic molecules that are specifically expressed in Treg cells during Th1 immune responses in mice and humans. Among these, we identified the novel co-inhibitory receptor CD85k as a functional predictor for Treg-mediated suppression specifically of Th1 responses, which could be explored therapeutically for selective immune suppression in autoimmunity.

6.
PLoS Pathog ; 14(5): e1007069, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29782555

RESUMEN

The opportunistic fungal pathogen Candida albicans frequently causes diseases such as oropharyngeal candidiasis (OPC) in immunocompromised individuals. Although it is well appreciated that the cytokine IL-17 is crucial for protective immunity against OPC, the cellular source and the regulation of this cytokine during infection are still a matter of debate. Here, we directly visualized IL-17 production in the tongue of experimentally infected mice, thereby demonstrating that this key cytokine is expressed by three complementary subsets of CD90+ leukocytes: RAG-dependent αß and γδ T cells, as well as RAG-independent ILCs. To determine the regulation of IL-17 production at the onset of OPC, we investigated in detail the myeloid compartment of the tongue and found a heterogeneous and dynamic mononuclear phagocyte (MNP) network in the infected tongue that consists of Zbtb46-Langerin- macrophages, Zbtb46+Langerin+ dendritic cells (DCs) and Ly6C+ inflammatory monocytes. Of those, the Langerin+ DC population stands out by its unique capacity to co-produce the cytokines IL-1ß, IL-6 and IL-23, all of which promote IL-17 induction in response to C. albicans in the oral mucosa. The critical role of Langerin+ DCs for the innate IL-17 response was confirmed by depletion of this cellular subset in vivo, which compromised IL-17 induction during OPC. In conclusion, our work revealed key regulatory factors and their cellular sources of innate IL-17-dependent antifungal immunity in the oral mucosa.


Asunto(s)
Antígenos de Superficie/inmunología , Candida albicans/inmunología , Candidiasis Bucal/inmunología , Células Dendríticas/inmunología , Interleucina-17/biosíntesis , Lectinas Tipo C/inmunología , Lectinas de Unión a Manosa/inmunología , Mucosa Bucal/inmunología , Animales , Candidiasis Bucal/microbiología , Citocinas/inmunología , Femenino , Citometría de Flujo , Interleucina-1beta/biosíntesis , Interleucina-23/biosíntesis , Interleucina-23/inmunología , Interleucina-6/biosíntesis , Leucocitos/inmunología , Masculino , Ratones , Ratones Endogámicos C57BL , Sistema Mononuclear Fagocítico/inmunología , Mucosa Bucal/citología , Mucosa Bucal/microbiología , Neutrófilos/inmunología , Organismos Libres de Patógenos Específicos , Bazo/citología , Bazo/inmunología , Antígenos Thy-1/inmunología , Lengua/citología , Lengua/inmunología , Lengua/microbiología
7.
Front Microbiol ; 8: 2044, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-29104568

RESUMEN

To understand how bacteria evolve and adapt to their environment, it can be relevant to monitor phenotypic changes that occur in a population. Single cell level analyses and sorting of mutant cells according to a particular phenotypic readout can constitute efficient strategies. However, when the phenotype of interest is expressed heterogeneously in ancestral isogenic populations of cells, single cell level sorting approaches are not optimal. Phenotypic heterogeneity can for instance make no-expression mutant cells indistinguishable from a subpopulation of wild-type cells transiently not expressing the phenotype. The analysis of clonal populations (e.g., isolated colonies), in which the average phenotype is measured, can circumvent this issue. Indeed, no-expression mutants form negative populations while wild-type clones form populations in which average expression of the phenotype yields a positive signal. We present here an optimized colony immunoblot protocol and a semi-automated image analysis pipeline (ImageJ macro) allowing for rapid detection of clones harboring mutations that affect the heterogeneous (i.e., bimodal) expression of the Type Three Secretion System-1 (TTSS-1) in Salmonella enterica serovar Typhimurium. We show that this protocol can efficiently differentiate clones expressing TTSS-1 at various levels in mixed populations. We were able to detect the emergence of hilC mutants in which the proportion of cells expressing TTSS-1 was reduced compared to the ancestor. We could also follow changes in the frequency of different mutants during long-term infections. This demonstrates that our protocol constitutes a tractable approach to assess semi-quantitatively the evolutionary dynamics of heterogeneous phenotypes, such as the expression of virulence genes, in bacterial populations.

8.
Cell Host Microbe ; 20(2): 238-49, 2016 Aug 10.
Artículo en Inglés | MEDLINE | ID: mdl-27453483

RESUMEN

Salmonella Typhimurium (S.Tm) causes acute enteropathy resolving after 4-7 days. Strikingly, antibiotic therapy does not accelerate disease resolution. We screened for factors blocking remission using a S.Tm enterocolitis model. The antibiotic ciprofloxacin clears pathogen stool loads within 3-24 hr, while gut pathology resolves more slowly (ψ50: ∼48 hr, remission: 6-9 days). This delayed resolution is mediated by an interferon-γ (IFN-γ)-dependent response that is triggered during acute infection and continues throughout therapy. Specifically, IFN-γ production by mucosal T and NK cells retards disease resolution by maintaining signaling through the transcriptional regulator STAT1 and boosting expression of inflammatory mediators like IL-1ß, TNF, and iNOS. Additionally, sustained IFN-γ fosters phagocyte accumulation and hampers antimicrobial defense mediated by IL-22 and the lectin REGIIIß. These findings reveal a role for IFN-γ in delaying resolution of intestinal inflammation and may inform therapies for acute Salmonella enteropathy, chronic inflammatory bowel diseases, or disease resolution during antibiotic treatment.


Asunto(s)
Antibacterianos/administración & dosificación , Enterocolitis/patología , Tracto Gastrointestinal/patología , Interferón gamma/metabolismo , Infecciones por Salmonella/patología , Salmonella typhimurium/crecimiento & desarrollo , Animales , Modelos Animales de Enfermedad , Enterocolitis/tratamiento farmacológico , Enterocolitis/inmunología , Enterocolitis/microbiología , Tracto Gastrointestinal/inmunología , Tracto Gastrointestinal/microbiología , Factores Inmunológicos/metabolismo , Mucosa Intestinal/inmunología , Mucosa Intestinal/microbiología , Mucosa Intestinal/patología , Células Asesinas Naturales/inmunología , Ratones Endogámicos C57BL , Fagocitos/inmunología , Infecciones por Salmonella/tratamiento farmacológico , Infecciones por Salmonella/inmunología , Infecciones por Salmonella/microbiología , Salmonella typhimurium/efectos de los fármacos , Transducción de Señal , Linfocitos T/inmunología
9.
PLoS Pathog ; 12(6): e1005723, 2016 06.
Artículo en Inglés | MEDLINE | ID: mdl-27341123

RESUMEN

Salmonella Typhimurium (S.Tm) is a common cause of self-limiting diarrhea. The mucosal inflammation is thought to arise from a standoff between the pathogen's virulence factors and the host's mucosal innate immune defenses, particularly the mucosal NAIP/NLRC4 inflammasome. However, it had remained unclear how this switches the gut from homeostasis to inflammation. This was studied using the streptomycin mouse model. S.Tm infections in knockout mice, cytokine inhibition and -injection experiments revealed that caspase-1 (not -11) dependent IL-18 is pivotal for inducing acute inflammation. IL-18 boosted NK cell chemoattractants and enhanced the NK cells' migratory capacity, thus promoting mucosal accumulation of mature, activated NK cells. NK cell depletion and Prf-/- ablation (but not granulocyte-depletion or T-cell deficiency) delayed tissue inflammation. Our data suggest an NK cell perforin response as one limiting factor in mounting gut mucosal inflammation. Thus, IL-18-elicited NK cell perforin responses seem to be critical for coordinating mucosal inflammation during early infection, when S.Tm strongly relies on virulence factors detectable by the inflammasome. This may have broad relevance for mucosal defense against microbial pathogens.


Asunto(s)
Interleucina-18/biosíntesis , Mucosa Intestinal/inmunología , Células Asesinas Naturales/inmunología , Proteínas Citotóxicas Formadoras de Poros/inmunología , Infecciones por Salmonella/inmunología , Animales , Caspasa 1 , Quimiotaxis de Leucocito/inmunología , Modelos Animales de Enfermedad , Citometría de Flujo , Inmunidad Innata/inmunología , Inflamasomas/inmunología , Inflamación/inmunología , Interleucina-18/inmunología , Cinética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Microscopía Confocal , Reacción en Cadena de la Polimerasa , Salmonella typhimurium/inmunología
11.
PLoS Pathog ; 10(12): e1004557, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25522364

RESUMEN

Topological, chemical and immunological barriers are thought to limit infection by enteropathogenic bacteria. However, in many cases these barriers and their consequences for the infection process remain incompletely understood. Here, we employed a mouse model for Salmonella colitis and a mixed inoculum approach to identify barriers limiting the gut luminal pathogen population. Mice were infected via the oral route with wild type S. Typhimurium (S. Tm) and/or mixtures of phenotypically identical but differentially tagged S. Tm strains ("WITS", wild-type isogenic tagged strains), which can be individually tracked by quantitative real-time PCR. WITS dilution experiments identified a substantial loss in tag/genetic diversity within the gut luminal S. Tm population by days 2-4 post infection. The diversity-loss was not attributable to overgrowth by S. Tm mutants, but required inflammation, Gr-1+ cells (mainly neutrophilic granulocytes) and most likely NADPH-oxidase-mediated defense, but not iNOS. Mathematical modelling indicated that inflammation inflicts a bottleneck transiently restricting the gut luminal S. Tm population to approximately 6000 cells and plating experiments verified a transient, inflammation- and Gr-1+ cell-dependent dip in the gut luminal S. Tm population at day 2 post infection. We conclude that granulocytes, an important clinical hallmark of S. Tm-induced inflammation, impose a drastic bottleneck upon the pathogen population. This extends the current view of inflammation-fuelled gut-luminal Salmonella growth by establishing the host response in the intestinal lumen as a double-edged sword, fostering and diminishing colonization in a dynamic equilibrium. Our work identifies a potent immune defense against gut infection and reveals a potential Achilles' heel of the infection process which might be targeted for therapy.


Asunto(s)
Colitis/microbiología , Colitis/patología , Tracto Gastrointestinal/microbiología , Tracto Gastrointestinal/patología , Granulocitos/patología , Salmonelosis Animal/patología , Salmonella typhimurium/crecimiento & desarrollo , Animales , Antibacterianos/uso terapéutico , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Ciego/metabolismo , Ciego/microbiología , Ciego/patología , Colitis/tratamiento farmacológico , Modelos Animales de Enfermedad , Femenino , Tracto Gastrointestinal/metabolismo , Masculino , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Ratones , Ratones Endogámicos C57BL , Microbiota/fisiología , Modelos Teóricos , Mutación , Salmonelosis Animal/tratamiento farmacológico , Salmonelosis Animal/microbiología , Salmonella typhimurium/genética , Salmonella typhimurium/metabolismo , Estreptomicina/uso terapéutico
12.
J Immunol ; 193(8): 4223-34, 2014 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-25210121

RESUMEN

Recent results indicate a significant contribution of innate immune signaling to maintain mucosal homeostasis, but the precise underlying signal transduction pathways are ill-defined. By comparative analysis of intestinal epithelial cells isolated from conventionally raised and germ-free mice, as well as animals deficient in the adaptor molecules MyD88 and TRIF, the TLR3 and TLR4, as well as the type I and III IFN receptors, we demonstrate significant TLR-mediated signaling under homeostatic conditions. Surprisingly, homeostatic expression of Reg3γ and Paneth cell enteric antimicrobial peptides critically relied on TRIF and, in part, TLR3 but was independent of IFN receptor signaling. Reduced antimicrobial peptide expression was associated with significantly lower numbers of Paneth cells and a reduced Paneth cell maturation and differentiation factor expression in TRIF mutant compared with wild-type epithelium. This phenotype was not transferred to TRIF-sufficient germ-free animals during cohousing. Low antimicrobial peptide expression in TRIF-deficient mice caused reduced immediate killing of orally administered bacteria but was not associated with significant alterations in the overall composition of the enteric microbiota. The phenotype was rapidly restored in a TRIF-independent fashion after transient epithelial damage. Our results identify TRIF signaling as a truly homeostatic pathway to maintain intestinal epithelial barrier function revealing fundamental differences in the innate immune signaling between mucosal homeostasis and tissue repair.


Asunto(s)
Proteínas Adaptadoras del Transporte Vesicular/inmunología , Péptidos Catiónicos Antimicrobianos/inmunología , Inmunidad Innata/inmunología , Mucosa Intestinal/inmunología , Listeria monocytogenes/inmunología , Proteínas/metabolismo , Salmonella typhimurium/inmunología , Proteínas Adaptadoras del Transporte Vesicular/genética , Animales , Péptidos Catiónicos Antimicrobianos/biosíntesis , Ciclinas/metabolismo , Mucosa Intestinal/microbiología , Listeriosis/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Factor 88 de Diferenciación Mieloide/genética , Proteínas Asociadas a Pancreatitis , Células de Paneth/metabolismo , Receptores de Interferón/genética , Infecciones por Salmonella/inmunología , Transducción de Señal/inmunología , Receptor Toll-Like 3/genética , Receptor Toll-Like 4/genética
13.
Cell Host Microbe ; 16(2): 237-248, 2014 Aug 13.
Artículo en Inglés | MEDLINE | ID: mdl-25121751

RESUMEN

The gut mucosal epithelium separates the host from the microbiota, but enteropathogens such as Salmonella Typhimurium (S.Tm) can invade and breach this barrier. Defenses against such acute insults remain incompletely understood. Using a murine model of Salmonella enterocolitis, we analyzed mechanisms limiting pathogen loads in the epithelium during early infection. Although the epithelium-invading S.Tm replicate initially, this intraepithelial replicative niche is restricted by expulsion of infected enterocytes into the lumen. This mechanism is compromised if inflammasome components (NAIP1-6, NLRC4, caspase-1/-11) are deleted, or ablated specifically in the epithelium, resulting in ∼100-fold higher intraepithelial loads and accelerated lymph node colonization. Interestingly, the cytokines downstream of inflammasome activation, interleukin (IL)-1α/ß and IL-18, appear dispensable for epithelial restriction of early infection. These data establish the role of an epithelium-intrinsic inflammasome, which drives expulsion of infected cells to restrict the pathogen's intraepithelial proliferation. This may represent a general defense mechanism against mucosal infections.


Asunto(s)
Proteínas Reguladoras de la Apoptosis/fisiología , Proteínas de Unión al Calcio/fisiología , Enterocitos/microbiología , Proteína Inhibidora de la Apoptosis Neuronal/fisiología , Infecciones por Salmonella/inmunología , Salmonella typhimurium/inmunología , Animales , Ciego/microbiología , Ciego/patología , Enterocitos/inmunología , Interacciones Huésped-Patógeno , Inflamasomas , Mucosa Intestinal/inmunología , Mucosa Intestinal/microbiología , Mucosa Intestinal/patología , Ratones Endogámicos C57BL , Ratones Noqueados
14.
Curr Biol ; 24(17): 2000-5, 2014 Sep 08.
Artículo en Inglés | MEDLINE | ID: mdl-25131673

RESUMEN

Antibiotics are powerful therapeutics but are not equally effective against all cells in bacterial populations. Bacteria that express an antibiotic-tolerant phenotype ("persisters") can evade treatment [1]. Persisters can cause relapses of the infection after the end of the therapy [2]. It is still poorly understood whether persistence affects the evolution of bacterial virulence. During infections, persisters have been found preferentially at particular sites within the host [3, 4]. If bacterial virulence factors are required to reach such sites, treatment with antibiotics could impose selection on the expression of virulence genes, in addition to their well-established effects on bacterial resistance. Here, we report that treatment with antibiotics selects for virulence and fosters transmissibility of Salmonella Typhimurium. In a mouse model for Salmonella diarrhea, treatment with the broad-spectrum antibiotic ciprofloxacin reverses the outcome of competition between wild-type bacteria and avirulent mutants that can spontaneously arise during within-host evolution [5]. While avirulent mutants take over the gut lumen and abolish disease transmission in untreated mice, ciprofloxacin tilts the balance in favor of virulent, wild-type bacteria. This is explained by the need for virulence factors to invade gut tissues and form a persistent reservoir. Avirulent mutants remain in the gut lumen and are eradicated. Upon cessation of antibiotic treatment, tissue-lodged wild-type pathogens reseed the gut lumen and thereby facilitate disease transmissibility to new hosts. Our results suggest a general principle by which antibiotic treatment can promote cooperative virulence during within-host evolution, increase duration of transmissibility, and thereby enhance the spread of an infectious disease.


Asunto(s)
Salmonella typhimurium/genética , Salmonella typhimurium/patogenicidad , Selección Genética , Animales , Antibacterianos/farmacología , Diarrea/microbiología , Diarrea/prevención & control , Interacciones Huésped-Patógeno/efectos de los fármacos , Inflamación/microbiología , Inflamación/patología , Intestinos/microbiología , Ratones , Ratones Endogámicos C57BL , Interacciones Microbianas , Infecciones por Salmonella/microbiología , Infecciones por Salmonella/prevención & control , Infecciones por Salmonella/transmisión , Salmonella typhimurium/efectos de los fármacos , Salmonella typhimurium/crecimiento & desarrollo , Virulencia , Factores de Virulencia
15.
PLoS Biol ; 12(2): e1001793, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24558351

RESUMEN

In vivo, antibiotics are often much less efficient than ex vivo and relapses can occur. The reasons for poor in vivo activity are still not completely understood. We have studied the fluoroquinolone antibiotic ciprofloxacin in an animal model for complicated Salmonellosis. High-dose ciprofloxacin treatment efficiently reduced pathogen loads in feces and most organs. However, the cecum draining lymph node (cLN), the gut tissue, and the spleen retained surviving bacteria. In cLN, approximately 10%-20% of the bacteria remained viable. These phenotypically tolerant bacteria lodged mostly within CD103⁺CX3CR1⁻CD11c⁺ dendritic cells, remained genetically susceptible to ciprofloxacin, were sufficient to reinitiate infection after the end of the therapy, and displayed an extremely slow growth rate, as shown by mathematical analysis of infections with mixed inocula and segregative plasmid experiments. The slow growth was sufficient to explain recalcitrance to antibiotics treatment. Therefore, slow-growing antibiotic-tolerant bacteria lodged within dendritic cells can explain poor in vivo antibiotic activity and relapse. Administration of LPS or CpG, known elicitors of innate immune defense, reduced the loads of tolerant bacteria. Thus, manipulating innate immunity may augment the in vivo activity of antibiotics.


Asunto(s)
Antibacterianos/farmacología , Ciprofloxacina/farmacología , Células Dendríticas/microbiología , Ganglios Linfáticos/inmunología , Infecciones por Salmonella/inmunología , Salmonella typhimurium/inmunología , Animales , Carga Bacteriana/efectos de los fármacos , Ciego , Diarrea/tratamiento farmacológico , Diarrea/inmunología , Diarrea/microbiología , Farmacorresistencia Bacteriana , Lipopolisacáridos/farmacología , Ganglios Linfáticos/microbiología , Ratones , Ratones Endogámicos C57BL , Pruebas de Sensibilidad Microbiana , Fenotipo , Infecciones por Salmonella/tratamiento farmacológico , Infecciones por Salmonella/microbiología , Salmonella typhimurium/efectos de los fármacos , Salmonella typhimurium/crecimiento & desarrollo
16.
J Leukoc Biol ; 89(1): 105-11, 2011 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-20884647

RESUMEN

Macrophages represent the first defense line against bacterial infection and therefore, play a crucial role in early inflammatory response. In this study, we investigated the role of MAPKs and MKP-1 activation in regulation of an early inflammatory response in RAW 264.7 macrophage cells. We induced the inflammatory response by treating the macrophages with LPS and inhibited an early inflammatory response by using ferulaldehyde, a water-soluble end-product of dietary polyphenol degradation that we found previously to exert its beneficial anti-inflammatory effects during the early phase of in vivo inflammation. We found that LPS-induced ROS and nitrogen species formations were reduced by ferulaldehyde in a concentration-dependent manner, and ferulaldehyde protected mitochondria against LPS-induced rapid and massive membrane depolarization. LPS induced early suppression of MKP-1, which was accompanied by activation of JNK, ERK, and p38 MAPK. By reversing LPS-induced early suppression of MKP-1, ferulaldehyde diminished MAPK activation, thereby inhibiting NF-κB activation, mitochondrial depolarization, and ROS production. Taken together, our data suggest that ferulaldehyde exerts its early anti-inflammatory effect by preserving the mitochondrial membrane integrity and shifting the expression of MKP-1 forward in time in macrophages.


Asunto(s)
Aldehídos/farmacología , Fosfatasa 1 de Especificidad Dual/metabolismo , Flavonoides/farmacología , Lipopolisacáridos/farmacología , Macrófagos/efectos de los fármacos , Macrófagos/enzimología , Fenoles/farmacología , Transducción de Señal/efectos de los fármacos , Animales , Línea Celular , Fosfatasa 1 de Especificidad Dual/genética , Activación Enzimática/efectos de los fármacos , Citometría de Flujo , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Ratones , Proteínas Quinasas Activadas por Mitógenos/metabolismo , FN-kappa B/metabolismo , Dióxido de Nitrógeno/metabolismo , Fosforilación/efectos de los fármacos , Polifenoles , ARN Mensajero/genética , ARN Mensajero/metabolismo , Especies Reactivas de Oxígeno/metabolismo
17.
PLoS Pathog ; 6(11): e1001194, 2010 Nov 18.
Artículo en Inglés | MEDLINE | ID: mdl-21124989

RESUMEN

The epithelium efficiently attracts immune cells upon infection despite the low number of pathogenic microbes and moderate levels of secreted chemokines per cell. Here we examined whether horizontal intercellular communication between cells may contribute to a coordinated response of the epithelium. Listeria monocytogenes infection, transfection, and microinjection of individual cells within a polarized intestinal epithelial cell layer were performed and activation was determined at the single cell level by fluorescence microscopy and flow cytometry. Surprisingly, chemokine production after L. monocytogenes infection was primarily observed in non-infected epithelial cells despite invasion-dependent cell activation. Whereas horizontal communication was independent of gap junction formation, cytokine secretion, ion fluxes, or nitric oxide synthesis, NADPH oxidase (Nox) 4-dependent oxygen radical formation was required and sufficient to induce indirect epithelial cell activation. This is the first report to describe epithelial cell-cell communication in response to innate immune activation. Epithelial communication facilitates a coordinated infectious host defence at the very early stage of microbial infection.


Asunto(s)
Comunicación Celular , Células Epiteliales/microbiología , Intestino Delgado/inmunología , Listeria monocytogenes/patogenicidad , Listeriosis/inmunología , Animales , Toxinas Bacterianas/metabolismo , Western Blotting , Adhesión Celular , Células Cultivadas , Citocinas/metabolismo , Citometría de Flujo , Técnica del Anticuerpo Fluorescente , Proteínas de Choque Térmico/metabolismo , Proteínas Hemolisinas/metabolismo , Inmunidad Innata , Immunoblotting , Inmunoprecipitación , Intestino Delgado/metabolismo , Intestino Delgado/microbiología , Listeriosis/microbiología , Listeriosis/patología , Ratones , NADPH Oxidasa 4 , NADPH Oxidasas/genética , NADPH Oxidasas/metabolismo , Óxido Nítrico/metabolismo , ARN Mensajero/genética , Especies Reactivas de Oxígeno/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA