Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
1.
Blood ; 141(14): 1755-1767, 2023 04 06.
Artículo en Inglés | MEDLINE | ID: mdl-36574344

RESUMEN

CD4+FOXP3+ regulatory T cells (Tregs) have demonstrated efficacy in the prevention and treatment of graft-versus-host disease (GVHD). Preclinical and clinical studies indicate that Tregs are able to protect from GVHD without interfering with the graft-versus-tumor (GVT) effect of hematopoietic cell transplantation (HCT), although the underlying molecular mechanisms are largely unknown. To elucidate Treg suppressive function during in vivo suppression of acute GVHD, we performed paired T-cell receptor (TCRα and ΤCRß genes) repertoire sequencing and RNA sequencing analysis on conventional T cells (Tcons) and Tregs before and after transplantation in a major histocompatibility complex -mismatched mouse model of HCT. We show that both Tregs and Tcons underwent clonal restriction, and Tregs did not interfere with the activation of alloreactive Tcon clones and the breadth of their TCR repertoire but markedly suppressed their expansion. Transcriptomic analysis revealed that Tregs predominantly affected the transcriptome of CD4 Tcons and, to a lesser extent, that of CD8 Tcons, thus modulating the transcription of genes encoding pro- and anti-inflammatory molecules as well as enzymes involved in metabolic processes, inducing a switch from glycolysis to oxidative phosphorylation. Finally, Tregs did not interfere with the induction of gene sets involved in the GVT effect. Our results shed light onto the mechanisms of acute GVHD suppression by Tregs and will support the clinical translation of this immunoregulatory approach.


Asunto(s)
Enfermedad Injerto contra Huésped , Trasplante de Células Madre Hematopoyéticas , Animales , Ratones , Linfocitos T Reguladores/patología , Transcriptoma , Enfermedad Injerto contra Huésped/genética , Enfermedad Injerto contra Huésped/prevención & control , Enfermedad Injerto contra Huésped/patología , Proteínas/genética
2.
Nature ; 572(7771): 665-669, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-31435014

RESUMEN

Intestinal commensal bacteria can inhibit dense colonization of the gut by vancomycin-resistant Enterococcus faecium (VRE), a leading cause of hospital-acquired infections1,2. A four-strained consortium of commensal bacteria that contains Blautia producta BPSCSK can reverse antibiotic-induced susceptibility to VRE infection3. Here we show that BPSCSK reduces growth of VRE by secreting a lantibiotic that is similar to the nisin-A produced by Lactococcus lactis. Although the growth of VRE is inhibited by BPSCSK and L. lactis in vitro, only BPSCSK colonizes the colon and reduces VRE density in vivo. In comparison to nisin-A, the BPSCSK lantibiotic has reduced activity against intestinal commensal bacteria. In patients at high risk of VRE infection, high abundance of the lantibiotic gene is associated with reduced density of E. faecium. In germ-free mice transplanted with patient-derived faeces, resistance to VRE colonization correlates with abundance of the lantibiotic gene. Lantibiotic-producing commensal strains of the gastrointestinal tract reduce colonization by VRE and represent potential probiotic agents to re-establish resistance to VRE.


Asunto(s)
Bacteriocinas/metabolismo , Bacteriocinas/farmacología , Enterococcus faecium/efectos de los fármacos , Lactococcus lactis/metabolismo , Probióticos , Resistencia a la Vancomicina/efectos de los fármacos , Enterococos Resistentes a la Vancomicina/efectos de los fármacos , Animales , Antibacterianos/biosíntesis , Antibacterianos/aislamiento & purificación , Antibacterianos/metabolismo , Antibacterianos/farmacología , Bacteriocinas/genética , Bacteriocinas/aislamiento & purificación , Enterococcus faecium/crecimiento & desarrollo , Enterococcus faecium/aislamiento & purificación , Heces/microbiología , Femenino , Tracto Gastrointestinal/efectos de los fármacos , Tracto Gastrointestinal/microbiología , Vida Libre de Gérmenes , Bacterias Grampositivas/efectos de los fármacos , Bacterias Grampositivas/crecimiento & desarrollo , Humanos , Lactococcus lactis/química , Lactococcus lactis/crecimiento & desarrollo , Lactococcus lactis/fisiología , Ratones , Pruebas de Sensibilidad Microbiana , Microbiota/genética , Nisina/química , Nisina/farmacología , Simbiosis/efectos de los fármacos , Vancomicina/farmacología , Enterococos Resistentes a la Vancomicina/crecimiento & desarrollo , Enterococos Resistentes a la Vancomicina/aislamiento & purificación
3.
Eur J Immunol ; 52(10): 1547-1560, 2022 10.
Artículo en Inglés | MEDLINE | ID: mdl-35985020

RESUMEN

Tissue-resident memory T cells (Trm), and particularly the CD8+ subset, have been shown to play a pivotal role in protection against infections and tumors. Studies in animal models and human tissues have highlighted that, while a core functional program is shared by Trm at all anatomical sites, distinct tissues imprint unique features through specific molecular cues. The intestinal tissue is often the target of pathogens for local proliferation and penetration into the host systemic circulation, as well as a prominent site of tumorigenesis. Therefore, promoting the formation of Trm at this location is an appealing therapeutic option. The various segments composing the gastrointestinal tract present distinctive histological and functional characteristics, which may reflect on the imprinting of unique functional features in the respective Trm populations. What these features are, and whether they can effectively be harnessed to promote local and systemic immunity, is still under investigation. Here, we review how Trm are generated and maintained in distinct intestinal niches, analyzing the required molecular signals and the models utilized to uncover them. We also discuss evidence for a protective role of Trm against infectious agents and tumors. Finally, we integrate the knowledge obtained from animal models with that gathered from human studies.


Asunto(s)
Memoria Inmunológica , Neoplasias , Animales , Linfocitos T CD8-positivos , Humanos , Intestinos/patología , Células T de Memoria
4.
Infect Immun ; 88(4)2020 03 23.
Artículo en Inglés | MEDLINE | ID: mdl-31964746

RESUMEN

The complex bacterial populations that constitute the gut microbiota can harbor antibiotic resistance genes (ARGs), including those encoding ß-lactamase enzymes (BLA), which degrade commonly prescribed antibiotics such as ampicillin. The prevalence of such genes in commensal bacteria has been increased in recent years by the wide use of antibiotics in human populations and in livestock. While transfer of ARGs between bacterial species has well-established dramatic public health implications, these genes can also function in trans within bacterial consortia, where antibiotic-resistant bacteria can provide antibiotic-sensitive neighbors with leaky protection from drugs, as shown both in vitro and in vivo, in models of lung and subcutaneous coinfection. However, whether the expression of ARGs by harmless commensal bacterial species can destroy antibiotics in the intestinal lumen and shield antibiotic-sensitive pathogens is unknown. To address this question, we colonized germfree or wild-type mice with a model intestinal commensal strain of Escherichia coli that produces either functional or defective BLA. Mice were subsequently infected with Listeria monocytogenes or Clostridioides difficile, followed by treatment with oral ampicillin. The production of functional BLA by commensal E. coli markedly reduced clearance of these pathogens and enhanced systemic dissemination during ampicillin treatment. Pathogen resistance was independent of ARG acquisition via horizontal gene transfer but instead relied on antibiotic degradation in the intestinal lumen by BLA. We conclude that commensal bacteria that have acquired ARGs can mediate shielding of pathogens from the bactericidal effects of antibiotics.


Asunto(s)
Ampicilina/metabolismo , Antibacterianos/metabolismo , Clostridioides difficile/efectos de los fármacos , Escherichia coli/metabolismo , Intestinos/microbiología , Listeria monocytogenes/efectos de los fármacos , beta-Lactamasas/metabolismo , Ampicilina/administración & dosificación , Ampicilina/farmacología , Animales , Antibacterianos/administración & dosificación , Antibacterianos/farmacología , Clostridioides difficile/crecimiento & desarrollo , Farmacorresistencia Bacteriana , Escherichia coli/enzimología , Escherichia coli/crecimiento & desarrollo , Hidrólisis , Ratones , Interacciones Microbianas , Viabilidad Microbiana/efectos de los fármacos
5.
Infect Immun ; 88(4)2020 03 23.
Artículo en Inglés | MEDLINE | ID: mdl-31907198

RESUMEN

Antibiotic treatment of patients undergoing complex medical treatments can deplete commensal bacterial strains from the intestinal microbiota, thereby reducing colonization resistance against a wide range of antibiotic-resistant pathogens. Loss of colonization resistance can lead to marked expansion of vancomycin-resistant Enterococcus faecium (VRE), Klebsiella pneumoniae, and Escherichia coli in the intestinal lumen, predisposing patients to bloodstream invasion and sepsis. The impact of intestinal domination by these antibiotic-resistant pathogens on mucosal immune defenses and epithelial and mucin-mediated barrier integrity is unclear. We used a mouse model to study the impact of intestinal domination by antibiotic-resistant bacterial species and strains on the colonic mucosa. Intestinal colonization with K. pneumoniae, Proteus mirabilis, or Enterobacter cloacae promoted greater recruitment of neutrophils to the colonic mucosa. To test the hypothesis that the residual microbiota influences the severity of colitis caused by infection with Clostridioides difficile, we coinfected mice that were colonized with ampicillin-resistant bacteria with a virulent strain of C. difficile and monitored colonization and pathogenesis. Despite the compositional differences in the gut microbiota, the severity of C. difficile infection (CDI) and mortality did not differ significantly between mice colonized with different ampicillin-resistant bacterial species. Our results suggest that the virulence mechanisms enabling CDI and epithelial destruction outweigh the relatively minor impact of less-virulent antibiotic-resistant intestinal bacteria on the outcome of CDI.


Asunto(s)
Antibacterianos/administración & dosificación , Infecciones por Clostridium/fisiopatología , Farmacorresistencia Bacteriana , Enterobacter cloacae/crecimiento & desarrollo , Infecciones por Enterobacteriaceae/complicaciones , Klebsiella pneumoniae/crecimiento & desarrollo , Proteus mirabilis/crecimiento & desarrollo , Animales , Infecciones por Clostridium/microbiología , Colitis/microbiología , Colitis/fisiopatología , Modelos Animales de Enfermedad , Enterobacter cloacae/efectos de los fármacos , Infecciones por Enterobacteriaceae/tratamiento farmacológico , Klebsiella pneumoniae/efectos de los fármacos , Ratones , Interacciones Microbianas , Proteus mirabilis/efectos de los fármacos , Análisis de Supervivencia
6.
PLoS Pathog ; 11(10): e1005164, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26431538

RESUMEN

Candida spp. can cause severe and chronic mucocutaneous and systemic infections in immunocompromised individuals. Protection from mucocutaneous candidiasis depends on T helper cells, in particular those secreting IL-17. The events regulating T cell activation and differentiation toward effector fates in response to fungal invasion in different tissues are poorly understood. Here we generated a Candida-specific TCR transgenic mouse reactive to a novel endogenous antigen that is conserved in multiple distant species of Candida, including the clinically highly relevant C. albicans and C. glabrata. Using TCR transgenic T cells in combination with an experimental model of oropharyngeal candidiasis (OPC) we investigated antigen presentation and Th17 priming by different subsets of dendritic cells (DCs) present in the infected oral mucosa. Candida-derived endogenous antigen accesses the draining lymph nodes and is directly presented by migratory DCs. Tissue-resident Flt3L-dependent DCs and CCR2-dependent monocyte-derived DCs collaborate in antigen presentation and T cell priming during OPC. In contrast, Langerhans cells, which are also present in the oral mucosa and have been shown to prime Th17 cells in the skin, are not required for induction of the Candida-specific T cell response upon oral challenge. This highlights the functional compartmentalization of specific DC subsets in different tissues. These data provide important new insights to our understanding of tissue-specific antifungal immunity.


Asunto(s)
Candidiasis Bucal/inmunología , Células Dendríticas/inmunología , Activación de Linfocitos/inmunología , Células Th17/inmunología , Animales , Presentación de Antígeno/inmunología , Antígenos Fúngicos/inmunología , Modelos Animales de Enfermedad , Epítopos de Linfocito T/inmunología , Citometría de Flujo , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Receptores de Antígenos de Linfocitos T/genética
7.
Proc Natl Acad Sci U S A ; 111(50): 17965-70, 2014 Dec 16.
Artículo en Inglés | MEDLINE | ID: mdl-25453106

RESUMEN

The use of neutralizing antibodies to identify the most effective antigen has been proposed as a strategy to design vaccines capable of eliciting protective B-cell immunity. In this study, we analyzed the human antibody response to cytomegalovirus (human cytomegalovirus, HCMV) infection and found that antibodies to glycoprotein (g)B, a surface glycoprotein that has been developed as a HCMV vaccine, were primarily nonneutralizing. In contrast, most of the antibodies to the complex formed by gH, gL, protein (p)UL128, pUL130, and pUL131 (the gHgLpUL128L pentamer) neutralized HCMV infection with high potency. Based on this analysis, we developed a single polycistronic vector encoding the five pentamer genes separated by "self-cleaving" 2A peptides to generate a stably transfected CHO cell line constitutively secreting high levels of recombinant pentamer that displayed the functional antigenic sites targeted by human neutralizing antibodies. Immunization of mice with the pentamer formulated with different adjuvants elicited HCMV neutralizing antibody titers that persisted to high levels over time and that were a hundred- to thousand-fold higher than those found in individuals that recovered from primary HCMV infection. Sera from mice immunized with the pentamer vaccine neutralized infection of both epithelial cells and fibroblasts and prevented cell-to-cell spread and viral dissemination from endothelial cells to leukocytes. Neutralizing monoclonal antibodies from immunized mice showed the same potency as human antibodies and targeted the same as well as additional sites on the pentamer. These results illustrate with a relevant example a general and practical approach of analytic vaccinology for the development of subunit vaccines against complex pathogens.


Asunto(s)
Anticuerpos Neutralizantes/inmunología , Antígenos Virales/inmunología , Vacunas contra Citomegalovirus/inmunología , Diseño de Fármacos , Animales , Anticuerpos Monoclonales/inmunología , Antígenos Virales/aislamiento & purificación , Células CHO , Cricetinae , Cricetulus , Ensayo de Inmunoadsorción Enzimática , Vectores Genéticos/genética , Células HEK293 , Humanos , Ratones , Estadísticas no Paramétricas , Vacunas de Subunidad/inmunología
8.
Mucosal Immunol ; 2024 May 21.
Artículo en Inglés | MEDLINE | ID: mdl-38782240

RESUMEN

Upon infection, CD8+ T cells that have been primed in the draining lymph nodes migrate to the invaded tissue, where they receive cues prompting their differentiation into tissue-resident memory cells (Trm), which display niche-specific transcriptional features. Despite the importance of these cells, our understanding of their molecular landscape and the signals that dictate their development remains limited, particularly in specific anatomical niches such as the large intestine (LI). Here, we report that LI Trm-generated following oral infection exhibits a distinct transcriptional profile compared to Trm in other tissues. Notably, we observe that local cues play a crucial role in the preferential establishment of LI Trm, favoring precursors that migrate to the tissue early during infection. Our investigations identify cognate antigen recognition as a major driver of Trm differentiation at this anatomical site. Local antigen presentation not only promotes the proliferation of effector cells and memory precursors but also facilitates the acquisition of transcriptional features characteristic of gut Trm. Thus, antigen recognition in the LI favors the establishment of Trm by impacting T cell expansion and gene expression.

9.
Gut Microbes ; 16(1): 2340486, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38659243

RESUMEN

Carbapenem-resistant Klebsiella pneumoniae (CR-Kp) is a significant threat to public health worldwide. The primary reservoir for CR-Kp is the intestinal tract. There, the bacterium is usually present at low density but can bloom following antibiotic treatment, mostly in hospital settings. The impact of disturbances in the intestinal environment on the fitness, survival, expansion, and drug susceptibility of this pathogen is not well-understood, yet it may be relevant to devise strategies to tackle CR-Kp colonization and infection. Here, we adopted an in vivo model to examine the transcriptional adaptation of a CR-Kp clinical isolate to immune activation in the intestine. We report that as early as 6 hours following host treatment with anti-CD3 antibody, CR-Kp underwent rapid transcriptional changes including downregulation of genes involved in sugar utilization and amino acid biosynthesis and upregulation of genes involved in amino acid uptake and catabolism, antibiotic resistance, and stress response. In agreement with these findings, treatment increased the concentration of oxidative species and amino acids in the mouse intestine. Genes encoding for proteins containing the domain of unknown function (DUF) 1471 were strongly upregulated, however their deletion did not impair CR-Kp fitness in vivo upon immune activation. Transcription factor enrichment analysis identified the global regulator cAMP-Receptor Protein, CRP, as a potential orchestrator of the observed transcriptional signature. In keeping with the recognized role of CRP in regulating utilization of alternative carbon sources, crp deletion in CR-Kp resulted in strongly impaired gut colonization, although this effect was not amplified by immune activation. Thus, following intestinal colonization, which occurs in a CRP-dependent manner, CR-Kp can rapidly respond to immune cues by implementing a well-defined and complex transcriptional program whose direct relevance toward bacterial fitness warrants further investigation. Additional analyses utilizing this model may identify key factors to tackle CR-Kp colonization of the intestine.


Asunto(s)
Antibacterianos , Intestinos , Infecciones por Klebsiella , Klebsiella pneumoniae , Klebsiella pneumoniae/genética , Klebsiella pneumoniae/efectos de los fármacos , Klebsiella pneumoniae/inmunología , Animales , Ratones , Infecciones por Klebsiella/microbiología , Infecciones por Klebsiella/inmunología , Intestinos/microbiología , Intestinos/inmunología , Antibacterianos/farmacología , Enterobacteriaceae Resistentes a los Carbapenémicos/genética , Regulación Bacteriana de la Expresión Génica , Carbapenémicos/farmacología , Ratones Endogámicos C57BL , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Femenino , Humanos
10.
Cell Host Microbe ; 29(3): 378-393.e5, 2021 03 10.
Artículo en Inglés | MEDLINE | ID: mdl-33539766

RESUMEN

The gut microbiota produces metabolites that regulate host immunity, thereby impacting disease resistance and susceptibility. The extent to which commensal bacteria reciprocally respond to immune activation, however, remains largely unexplored. Herein, we colonized mice with four anaerobic symbionts and show that acute immune responses result in dramatic transcriptional reprogramming of these commensals with minimal changes in their relative abundance. Transcriptomic changes include induction of stress-response mediators and downregulation of carbohydrate-degrading factors such as polysaccharide utilization loci (PULs). Flagellin and anti-CD3 antibody, two distinct immune stimuli, induced similar transcriptional profiles, suggesting that commensal bacteria detect common effectors or activate shared pathways when facing different host responses. Immune activation altered the intestinal metabolome within 6 hours, decreasing luminal short-chain fatty acid and increasing aromatic metabolite concentrations. Thus, intestinal bacteria, prior to detectable shifts in community composition, respond to acute host immune activation by rapidly changing gene transcription and immunomodulatory metabolite production.


Asunto(s)
Microbioma Gastrointestinal/inmunología , Microbioma Gastrointestinal/fisiología , Intestinos/inmunología , Intestinos/microbiología , Animales , Bacterias/genética , Bacterias/metabolismo , Estudios Transversales , Regulación hacia Abajo , Ácidos Grasos Volátiles , Femenino , Flagelina , Microbioma Gastrointestinal/genética , Inflamación/inmunología , Metaboloma , Ratones , Ratones Endogámicos C57BL , ARN Ribosómico 16S , Simbiosis , Transcriptoma
11.
Nat Commun ; 11(1): 4475, 2020 09 08.
Artículo en Inglés | MEDLINE | ID: mdl-32901029

RESUMEN

Tissue resident memory CD8+ T cells (Trm) are poised for immediate reactivation at sites of pathogen entry and provide optimal protection of mucosal surfaces. The intestinal tract represents a portal of entry for many infectious agents; however, to date specific strategies to enhance Trm responses at this site are lacking. Here, we present TMDI (Transient Microbiota Depletion-boosted Immunization), an approach that leverages antibiotic treatment to temporarily restrain microbiota-mediated colonization resistance, and favor intestinal expansion to high densities of an orally-delivered Listeria monocytogenes strain carrying an antigen of choice. By augmenting the local chemotactic gradient as well as the antigenic load, this procedure generates a highly expanded pool of functional, antigen-specific intestinal Trm, ultimately enhancing protection against infectious re-challenge in mice. We propose that TMDI is a useful model to dissect the requirements for optimal Trm responses in the intestine, and also a potential platform to devise novel mucosal vaccination approaches.


Asunto(s)
Microbioma Gastrointestinal/inmunología , Inmunidad Mucosa , Administración Oral , Animales , Antígenos/administración & dosificación , Linfocitos T CD8-positivos/inmunología , Quimiotaxis/inmunología , Femenino , Microbioma Gastrointestinal/efectos de los fármacos , Interacciones Microbiota-Huesped/inmunología , Inmunidad Mucosa/efectos de los fármacos , Memoria Inmunológica , Listeria monocytogenes/crecimiento & desarrollo , Listeria monocytogenes/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ovalbúmina/administración & dosificación , Estreptomicina/administración & dosificación
12.
Clin Hematol Int ; 2(4): 156-164, 2020 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-34595456

RESUMEN

BACKGROUND: Monocytes are an essential cellular component of the innate immune system that support the host's effectiveness to combat a range of infectious pathogens. Hemopoietic cell transplantation (HCT) results in transient monocyte depletion, but the factors that regulate recovery of monocyte populations are not fully understood. In this study, we investigated whether the composition of the gastrointestinal microbiota is associated with the recovery of monocyte homeostasis after HCT. METHODS: We performed a single-center, prospective, pilot study of 18 recipients of either autologous or allogeneic HCT. Serial blood and stool samples were collected from each patient during their HCT hospitalization. Analysis of the gut microbiota was done using 16S rRNA gene sequencing, and flow cytometric analysis was used to characterize the phenotypic composition of monocyte populations. RESULTS: Dynamic fluctuations in monocyte reconstitution occurred after HCT, and large differences were observed in monocyte frequency among patients over time. Recovery of absolute monocyte counts and subsets showed significant variability across the heterogeneous transplant types and conditioning intensities; no relationship to the microbiota composition was observed in this small cohort. CONCLUSION: In this pilot study, a relationship between the microbiota composition and monocyte homeostasis could not be firmly established. However, we identify multivariate associations between clinical factors and monocyte reconstitution post-HCT. Our findings encourage further longitudinal surveillance of the intestinal microbiome and its link to immune reconstitution.

13.
mBio ; 10(6)2019 12 17.
Artículo en Inglés | MEDLINE | ID: mdl-31848289

RESUMEN

Listeria monocytogenes can cause a life-threatening illness when the foodborne pathogen spreads beyond the intestinal tract to distant organs. Many aspects of the intestinal phase of L. monocytogenes pathogenesis remain unknown. Here, we present a foodborne infection model using C57BL/6 mice that have been pretreated with streptomycin. In this model, as few as 100 L. monocytogenes CFU were required to cause self-limiting enterocolitis, and systemic dissemination followed previously reported routes. Using this model, we report that listeriolysin O (LLO) and actin assembly-inducing protein (ActA), two critical virulence determinants, were necessary for intestinal pathology and systemic spread but were dispensable for intestinal growth. Sequence tag-based analysis of microbial populations (STAMP) was used to investigate the within-host population dynamics of wild-type and LLO-deficient strains. The wild-type bacterial population experienced severe bottlenecks over the course of infection, and by 5 days, the intestinal population was highly enriched for bacteria originating from the gallbladder. In contrast, LLO-deficient strains did not efficiently disseminate and gain access to the gallbladder, and the intestinal population remained diverse. These findings suggest that systemic spread and establishment of a bacterial reservoir in the gallbladder imparts an intraspecies advantage in intestinal occupancy. Since intestinal L. monocytogenes is ultimately released into the environment, within-host population bottlenecks may provide purifying selection of virulence genes.IMPORTANCEListeria monocytogenes maintains capabilities for free-living growth in the environment and for intracellular replication in a wide range of hosts, including livestock and humans. Here, we characterized an enterocolitis model of foodborne L. monocytogenes infection. This work highlights a multiorgan trafficking circuit and reveals a fitness advantage for bacteria that successfully complete this cycle. Because virulence factors play critical roles in systemic dissemination and multiple bottlenecks occur as the bacterial population colonizes different tissue sites, this multiorgan trafficking circuit likely provides purifying selection of virulence genes. This study also serves as a foundation for future work using the L. monocytogenes-induced enterocolitis model to investigate the biology of L. monocytogenes in the intestinal environment.


Asunto(s)
Listeria monocytogenes/fisiología , Listeriosis/microbiología , Factores de Virulencia/genética , Animales , Modelos Animales de Enfermedad , Humanos , Mucosa Intestinal/metabolismo , Mucosa Intestinal/microbiología , Mucosa Intestinal/patología , Listeria monocytogenes/efectos de los fármacos , Listeria monocytogenes/patogenicidad , Listeriosis/tratamiento farmacológico , Listeriosis/transmisión , Ratones , Pruebas de Sensibilidad Microbiana , Especificidad de Órganos , Estreptomicina/farmacología , Estreptomicina/uso terapéutico , Virulencia/genética
14.
Cell Host Microbe ; 25(5): 695-705.e5, 2019 05 08.
Artículo en Inglés | MEDLINE | ID: mdl-31031170

RESUMEN

Vancomycin-resistant Enterococcus (VRE) are highly antibiotic-resistant and readily transmissible pathogens that cause severe infections in hospitalized patients. We discovered that lithocholic acid (LCA), a secondary bile acid prevalent in the cecum and colon of mice and humans, impairs separation of growing VRE diplococci, causing the formation of long chains and increased biofilm formation. Divalent cations reversed this LCA-induced switch to chaining and biofilm formation. Experimental evolution in the presence of LCA yielded mutations in the essential two-component kinase yycG/walK and three-component response regulator liaR that locked VRE in diplococcal mode, impaired biofilm formation, and increased susceptibility to the antibiotic daptomycin. These mutant VRE strains were deficient in host colonization because of their inability to compete with intestinal microbiota. This morphotype switch presents a potential non-bactericidal therapeutic target that may help clear VRE from the intestines of dominated patients, as occurs frequently during hematopoietic stem cell transplantation.


Asunto(s)
Ácidos y Sales Biliares/metabolismo , Colon/microbiología , Enterococcus faecium/efectos de los fármacos , Enterococcus faecium/crecimiento & desarrollo , Infecciones por Bacterias Grampositivas/microbiología , Enterococos Resistentes a la Vancomicina/efectos de los fármacos , Enterococos Resistentes a la Vancomicina/crecimiento & desarrollo , Animales , Portador Sano/microbiología , Ratones , Virulencia/efectos de los fármacos
15.
Pathogens ; 7(1)2017 Dec 22.
Artículo en Inglés | MEDLINE | ID: mdl-29271903

RESUMEN

Listeria monocytogenes is a foodborne pathogen that can cause febrile gastroenteritis in healthy subjects and systemic infections in immunocompromised individuals. Despite the high prevalence of L. monocytogenes in the environment and frequent contamination of uncooked meat and poultry products, infections with this pathogen are relatively uncommon, suggesting that protective defenses in the general population are effective. In the mammalian gastrointestinal tract, a variety of defense mechanisms prevent L. monocytogenes growth, epithelial penetration and systemic dissemination. Among these defenses, colonization resistance mediated by the gut microbiota is crucial in protection against a range of intestinal pathogens, including L. monocytogenes. Here we review defined mechanisms of defense against L. monocytogenes in the lumen of the gastro-intestinal tract, with particular emphasis on protection conferred by the autochthonous microbiota. We suggest that selected probiotic species derived from the microbiota may be developed for eventual clinical use to enhance resistance against L. monocytogenes infections.

16.
J Exp Med ; 214(7): 1973-1989, 2017 Jul 03.
Artículo en Inglés | MEDLINE | ID: mdl-28588016

RESUMEN

Listeria monocytogenes is a foodborne pathogen that causes septicemia, meningitis and chorioamnionitis and is associated with high mortality. Immunocompetent humans and animals, however, can tolerate high doses of L. monocytogenes without developing systemic disease. The intestinal microbiota provides colonization resistance against many orally acquired pathogens, and antibiotic-mediated depletion of the microbiota reduces host resistance to infection. Here we show that a diverse microbiota markedly reduces Listeria monocytogenes colonization of the gut lumen and prevents systemic dissemination. Antibiotic administration to mice before low dose oral inoculation increases L. monocytogenes growth in the intestine. In immunodeficient or chemotherapy-treated mice, the intestinal microbiota provides nonredundant defense against lethal, disseminated infection. We have assembled a consortium of commensal bacteria belonging to the Clostridiales order, which exerts in vitro antilisterial activity and confers in vivo resistance upon transfer into germ free mice. Thus, we demonstrate a defensive role of the gut microbiota against Listeria monocytogenes infection and identify intestinal commensal species that, by enhancing resistance against this pathogen, represent potential probiotics.


Asunto(s)
Microbioma Gastrointestinal/fisiología , Intestinos/microbiología , Listeria monocytogenes/fisiología , Listeriosis/microbiología , Animales , Antibacterianos/administración & dosificación , Antibacterianos/farmacología , Antibiosis/efectos de los fármacos , Heces/microbiología , Interacciones Huésped-Patógeno/genética , Humanos , Huésped Inmunocomprometido , Intestinos/efectos de los fármacos , Listeria monocytogenes/efectos de los fármacos , Listeriosis/genética , Listeriosis/mortalidad , Ratones Endogámicos C57BL , Ratones Noqueados , Análisis de Supervivencia , Tasa de Supervivencia , Factores de Tiempo
17.
Trends Mol Med ; 22(6): 458-478, 2016 06.
Artículo en Inglés | MEDLINE | ID: mdl-27178527

RESUMEN

The gut microbiota is a key player in many physiological and pathological processes occurring in humans. Recent investigations suggest that the efficacy of some clinical approaches depends on the action of commensal bacteria. Antibiotics are invaluable weapons to fight infectious diseases. However, by altering the composition and functions of the microbiota, they can also produce long-lasting deleterious effects for the host. The emergence of multidrug-resistant pathogens raises concerns about the common, and at times inappropriate, use of antimicrobial agents. Here we review the most recently discovered connections between host pathophysiology, microbiota, and antibiotics highlighting technological platforms, mechanistic insights, and clinical strategies to enhance resistance to diseases by preserving the beneficial functions of the microbiota.


Asunto(s)
Antibacterianos/metabolismo , Antibacterianos/farmacología , Bacterias/efectos de los fármacos , Microbioma Gastrointestinal/efectos de los fármacos , Animales , Bacterias/genética , Línea Celular , Farmacorresistencia Bacteriana , Microbioma Gastrointestinal/genética , Humanos , Sistema Inmunológico , Ratones
18.
Sci Transl Med ; 8(327): 327ra25, 2016 Feb 24.
Artículo en Inglés | MEDLINE | ID: mdl-26912904

RESUMEN

Antibiotic administration can disrupt the intestinal microbiota and down-regulate innate immune defenses, compromising colonization resistance against orally acquired bacterial pathogens. Vancomycin-resistant Enterococcus faecium (VRE), a major cause of antibiotic-resistant infections in hospitalized patients, thrives in the intestine when colonization resistance is compromised, achieving extremely high densities that can lead to bloodstream invasion and sepsis. Viral infections, by mechanisms that remain incompletely defined, can stimulate resistance against invading bacterial pathogens. We report that murine norovirus infection correlates with reduced density of VRE in the intestinal tract of mice with antibiotic-induced loss of colonization resistance. Resiquimod (R848), a synthetic ligand for Toll-like receptor 7 (TLR-7) that stimulates antiviral innate immune defenses, restores expression of the antimicrobial peptide Reg3γ and reestablishes colonization resistance against VRE in antibiotic-treated mice. Orally administered R848 triggers TLR-7 on CD11c(+) dendritic cells, inducing interleukin-23 (IL-23) expression followed by a burst of IL-22 secretion by innate lymphoid cells, leading to Reg3γ expression and restoration of colonization resistance against VRE. Our findings reveal that an orally bioavailable TLR-7 ligand that stimulates innate antiviral immune pathways in the intestine restores colonization resistance against a highly antibiotic-resistant bacterial pathogen.


Asunto(s)
Farmacorresistencia Bacteriana/efectos de los fármacos , Enterococcus/efectos de los fármacos , Enterococcus/crecimiento & desarrollo , Interleucinas/metabolismo , Receptor Toll-Like 7/metabolismo , Vancomicina/farmacología , Ampicilina/farmacología , Animales , Antígeno CD11c/metabolismo , Infecciones por Caliciviridae/complicaciones , Infecciones por Caliciviridae/patología , Infecciones por Caliciviridae/virología , Recuento de Colonia Microbiana , Células Dendríticas/efectos de los fármacos , Células Dendríticas/metabolismo , Gastroenteritis/complicaciones , Gastroenteritis/patología , Gastroenteritis/virología , Imidazoles/farmacología , Interferón Tipo I/metabolismo , Interleucina-1/metabolismo , Interleucina-23/metabolismo , Ligandos , Ratones Endogámicos C57BL , Norovirus/efectos de los fármacos , Norovirus/fisiología , Proteínas Asociadas a Pancreatitis , Proteínas/metabolismo , Transducción de Señal/efectos de los fármacos , Interleucina-22
19.
Science ; 347(6220): 400-6, 2015 Jan 23.
Artículo en Inglés | MEDLINE | ID: mdl-25477212

RESUMEN

Distinct types of CD4(+) T cells protect the host against different classes of pathogens. However, it is unclear whether a given pathogen induces a single type of polarized T cell. By combining antigenic stimulation and T cell receptor deep sequencing, we found that human pathogen- and vaccine-specific T helper 1 (T(H)1), T(H)2, and T(H)17 memory cells have different frequencies but comparable diversity and comprise not only clones polarized toward a single fate, but also clones whose progeny have acquired multiple fates. Single naïve T cells primed by a pathogen in vitro could also give rise to multiple fates. Our results unravel an unexpected degree of interclonal and intraclonal functional heterogeneity of the human T cell response and suggest that polarized responses result from preferential expansion rather than priming.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Candida albicans/inmunología , Interacciones Huésped-Patógeno/inmunología , Memoria Inmunológica , Mycobacterium tuberculosis/inmunología , Subgrupos de Linfocitos T/inmunología , Vacunas/inmunología , Secuencia de Aminoácidos , Células Cultivadas , Células Clonales , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Activación de Linfocitos , Datos de Secuencia Molecular , Receptores de Antígenos de Linfocitos T/genética , Células TH1/inmunología , Células Th17/inmunología , Células Th2/inmunología
20.
Nat Commun ; 6: 6431, 2015 Mar 16.
Artículo en Inglés | MEDLINE | ID: mdl-25775432

RESUMEN

T helper (TH) cell polarization during priming is modulated by a number of signals, but whether polarization to a given phenotype also influences recall responses of memory TH cells is relatively unknown. Here we show that miR-181a is selectively induced in both human and mouse naive T cells differentiating into the TH17, but not TH1 or TH2 subset. In human memory TH17 cells, miR-181a regulates responses to cognate antigens through modulation of ERK phosphorylation. By enhancing the signalling cascade from the T-cell receptor, such molecular network reduces the threshold of TH17 cell activation. Moreover, at a late time point, the same network induces a self-regulatory mechanism dependent on ID3, a negative regulator of transcription factors that control RORC expression, thus modulating TH17 activity. Our results demonstrate that the phenotype acquired by TH cells during priming contributes to their threshold of activation to secondary antigenic stimulations, thus influencing memory responses.


Asunto(s)
Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Memoria Inmunológica , MicroARNs/metabolismo , Células Th17/citología , Animales , Antígenos/química , Candida albicans/metabolismo , Diferenciación Celular , Femenino , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , MicroARNs/genética , Fenotipo , Fosforilación , Interferencia de ARN , Transducción de Señal
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA