Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 92
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Nat Immunol ; 24(12): 2108-2120, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-37932457

RESUMEN

Regulatory T cells (Treg cells) are instrumental in establishing immunological tolerance. However, the precise effector mechanisms by which Treg cells control a specific type of immune response in a given tissue remains unresolved. By simultaneously studying Treg cells from different tissue origins under systemic autoimmunity, in the present study we show that interleukin (IL)-27 is specifically produced by intestinal Treg cells to regulate helper T17 cell (TH17 cell) immunity. Selectively increased intestinal TH17 cell responses in mice with Treg cell-specific IL-27 ablation led to exacerbated intestinal inflammation and colitis-associated cancer, but also helped protect against enteric bacterial infection. Furthermore, single-cell transcriptomic analysis has identified a CD83+CD62Llo Treg cell subset that is distinct from previously characterized intestinal Treg cell populations as the main IL-27 producers. Collectively, our study uncovers a new Treg cell suppression mechanism crucial for controlling a specific type of immune response in a particular tissue and provides further mechanistic insights into tissue-specific Treg cell-mediated immune regulation.


Asunto(s)
Interleucina-27 , Linfocitos T Reguladores , Ratones , Animales , Linfocitos T Colaboradores-Inductores , Tolerancia Inmunológica , Inmunidad Celular , Células Th17
2.
Immunity ; 48(5): 839-841, 2018 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-29768168

RESUMEN

Molecular mechanisms connecting the gut-brain axis to immunity remain elusive. In this issue of Immunity, Labed et al. (2018) demonstrate that two evolutionarily conserved signaling mechanisms, the neuronal muscarinic and the epithelial Wnt pathways, together induce antimicrobial peptide expression that protects Caenorhabditis elegans against intestinal infection.


Asunto(s)
Proteínas de Caenorhabditis elegans , Vía de Señalización Wnt , Acetilcolina , Animales , Antiinfecciosos , Caenorhabditis elegans , Colinérgicos , Emociones
3.
Nature ; 579(7797): 123-129, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-32103176

RESUMEN

A mosaic of cross-phylum chemical interactions occurs between all metazoans and their microbiomes. A number of molecular families that are known to be produced by the microbiome have a marked effect on the balance between health and disease1-9. Considering the diversity of the human microbiome (which numbers over 40,000 operational taxonomic units10), the effect of the microbiome on the chemistry of an entire animal remains underexplored. Here we use mass spectrometry informatics and data visualization approaches11-13 to provide an assessment of the effects of the microbiome on the chemistry of an entire mammal by comparing metabolomics data from germ-free and specific-pathogen-free mice. We found that the microbiota affects the chemistry of all organs. This included the amino acid conjugations of host bile acids that were used to produce phenylalanocholic acid, tyrosocholic acid and leucocholic acid, which have not previously been characterized despite extensive research on bile-acid chemistry14. These bile-acid conjugates were also found in humans, and were enriched in patients with inflammatory bowel disease or cystic fibrosis. These compounds agonized the farnesoid X receptor in vitro, and mice gavaged with the compounds showed reduced expression of bile-acid synthesis genes in vivo. Further studies are required to confirm whether these compounds have a physiological role in the host, and whether they contribute to gut diseases that are associated with microbiome dysbiosis.


Asunto(s)
Ácidos y Sales Biliares/biosíntesis , Ácidos y Sales Biliares/química , Metabolómica , Microbiota/fisiología , Animales , Ácidos y Sales Biliares/metabolismo , Ácido Cólico/biosíntesis , Ácido Cólico/química , Ácido Cólico/metabolismo , Fibrosis Quística/genética , Fibrosis Quística/metabolismo , Fibrosis Quística/microbiología , Vida Libre de Gérmenes , Humanos , Enfermedades Inflamatorias del Intestino/genética , Enfermedades Inflamatorias del Intestino/metabolismo , Enfermedades Inflamatorias del Intestino/microbiología , Ratones , Receptores Citoplasmáticos y Nucleares/genética , Receptores Citoplasmáticos y Nucleares/metabolismo
4.
Infect Immun ; 92(5): e0009924, 2024 May 07.
Artículo en Inglés | MEDLINE | ID: mdl-38557196

RESUMEN

The mouse pathogen Citrobacter rodentium is utilized as a model organism for studying infections caused by the human pathogens enteropathogenic Escherichia coli (EPEC) and enterohemorrhagic E. coli (EHEC) and to elucidate mechanisms of mucosal immunity. In response to C. rodentium infection, innate lymphoid cells and T cells secrete interleukin (IL)-22, a cytokine that promotes mucosal barrier function. IL-22 plays a pivotal role in enabling mice to survive and recover from C. rodentium infection, although the exact mechanisms involved remain incompletely understood. Here, we investigated whether particular components of the host response downstream of IL-22 contribute to the cytokine's protective effects during C. rodentium infection. In line with previous research, mice lacking the IL-22 gene (Il22-/- mice) were highly susceptible to C. rodentium infection. To elucidate the role of specific antimicrobial proteins modulated by IL-22, we infected the following knockout mice: S100A9-/- (calprotectin), Lcn2-/- (lipocalin-2), Reg3b-/- (Reg3ß), Reg3g-/- (Reg3γ), and C3-/- (C3). All knockout mice tested displayed a considerable level of resistance to C. rodentium infection, and none phenocopied the lethality observed in Il22-/- mice. By investigating another arm of the IL-22 response, we observed that C. rodentium-infected Il22-/- mice exhibited an overall decrease in gene expression related to intestinal barrier integrity as well as significantly elevated colonic inflammation, gut permeability, and pathogen levels in the spleen. Taken together, these results indicate that host resistance to lethal C. rodentium infection may depend on multiple antimicrobial responses acting in concert, or that other IL-22-regulated processes, such as tissue repair and maintenance of epithelial integrity, play crucial roles in host defense to attaching and effacing pathogens.


Asunto(s)
Citrobacter rodentium , Infecciones por Enterobacteriaceae , Interleucina-22 , Animales , Ratones , Citrobacter rodentium/inmunología , Modelos Animales de Enfermedad , Infecciones por Enterobacteriaceae/inmunología , Infecciones por Enterobacteriaceae/microbiología , Interleucina-22/genética , Interleucina-22/metabolismo , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas Asociadas a Pancreatitis/genética , Proteínas Asociadas a Pancreatitis/metabolismo , Proteínas Asociadas a Pancreatitis/inmunología
5.
J Am Chem Soc ; 146(11): 7708-7722, 2024 Mar 20.
Artículo en Inglés | MEDLINE | ID: mdl-38457782

RESUMEN

Developing new antibiotics and delivery strategies is of critical importance for treating infections caused by Gram-negative bacterial pathogens. Hijacking bacterial iron uptake machinery, such as that of the siderophore enterobactin (Ent), represents one promising approach toward these goals. Here, we report a novel Ent-inspired siderophore-antibiotic conjugate (SAC) employing an alternative siderophore moiety as the delivery vector and demonstrate the potency of our SACs harboring the ß-lactam antibiotic ampicillin (Amp) against multiple pathogenic Gram-negative bacterial strains. We establish the ability of N,N',N''-(nitrilotris(ethane-2,1-diyl))tris(2,3-dihydroxybenzamide) (TRENCAM, hereafter TC), a synthetic mimic of Ent, to facilitate drug delivery across the outer membrane (OM) of Gram-negative pathogens. Conjugation of Amp to a new monofunctionalized TC scaffold affords TC-Amp, which displays markedly enhanced antibacterial activity against the gastrointestinal pathogen Salmonella enterica serovar Typhimurium (STm) compared with unmodified Amp. Bacterial uptake, antibiotic susceptibility, and microscopy studies with STm show that the TC moiety facilitates TC-Amp uptake by the OM receptors FepA and IroN and that the Amp warhead inhibits penicillin-binding proteins. Moreover, TC-Amp achieves targeted activity, selectively killing STm in the presence of a commensal lactobacillus. Remarkably, we uncover that TC-Amp and its Ent-based predecessor Ent-Amp achieve enhanced antibacterial activity against diverse Gram-negative ESKAPE pathogens that express Ent uptake machinery, including strains that possess intrinsic ß-lactam resistance. TC-Amp and Ent-Amp exhibit potency comparable to that of the FDA-approved SAC cefiderocol against Gram-negative pathogens. These results demonstrate the effective application of native and appropriately designed nonnative siderophores as vectors for drug delivery across the OM of multiple Gram-negative bacterial pathogens.


Asunto(s)
Sideróforos , beta-Lactamas , Sideróforos/farmacología , beta-Lactamas/farmacología , Lactamas , Antibacterianos/farmacología , Enterobactina/farmacología , Enterobactina/metabolismo , Bacterias Gramnegativas , Hierro
6.
Immunity ; 40(2): 262-73, 2014 Feb 20.
Artículo en Inglés | MEDLINE | ID: mdl-24508234

RESUMEN

Interleukin-22 (IL-22) is highly induced in response to infections with a variety of pathogens, and its main functions are considered to be tissue repair and host defense at mucosal surfaces. Here we showed that IL-22 has a unique role during infection in that its expression suppressed the intestinal microbiota and enhanced the colonization of a pathogen. IL-22 induced the expression of antimicrobial proteins, including lipocalin-2 and calprotectin, which sequester essential metal ions from microbes. Because Salmonella enterica ser. Typhimurium can overcome metal ion starvation mediated by lipocalin-2 and calprotectin via alternative pathways, IL-22 boosted its colonization of the inflamed intestine by suppressing commensal Enterobacteriaceae, which are susceptible to the antimicrobial proteins. Thus, IL-22 tipped the balance between pathogenic and commensal bacteria in favor of a pathogen. Taken together, IL-22 induction can be exploited by pathogens to suppress the growth of their closest competitors, thereby enhancing pathogen colonization of mucosal surfaces.


Asunto(s)
Interacciones Huésped-Patógeno , Interleucinas/inmunología , Intestinos/microbiología , Infecciones por Salmonella/inmunología , Infecciones por Salmonella/microbiología , Simbiosis/inmunología , Animales , Citocinas/metabolismo , Interleucinas/genética , Ratones , Ratones Endogámicos C57BL , Modelos Biológicos , Reacción en Cadena en Tiempo Real de la Polimerasa , Regulación hacia Arriba , Interleucina-22
7.
Proc Natl Acad Sci U S A ; 117(40): 24998-25007, 2020 10 06.
Artículo en Inglés | MEDLINE | ID: mdl-32958643

RESUMEN

Infections elicit immune adaptations to enable pathogen resistance and/or tolerance and are associated with compositional shifts of the intestinal microbiome. However, a comprehensive understanding of how infections with pathogens that exhibit distinct capability to spread and/or persist differentially change the microbiome, the underlying mechanisms, and the relative contribution of individual commensal species to immune cell adaptations is still lacking. Here, we discovered that mouse infection with a fast-spreading and persistent (but not a slow-spreading acute) isolate of lymphocytic choriomeningitis virus induced large-scale microbiome shifts characterized by increased Verrucomicrobia and reduced Firmicute/Bacteroidetes ratio. Remarkably, the most profound microbiome changes occurred transiently after infection with the fast-spreading persistent isolate, were uncoupled from sustained viral loads, and were instead largely caused by CD8 T cell responses and/or CD8 T cell-induced anorexia. Among the taxa enriched by infection with the fast-spreading virus, Akkermansia muciniphila, broadly regarded as a beneficial commensal, bloomed upon starvation and in a CD8 T cell-dependent manner. Strikingly, oral administration of A. muciniphila suppressed selected effector features of CD8 T cells in the context of both infections. Our findings define unique microbiome differences after chronic versus acute viral infections and identify CD8 T cell responses and downstream anorexia as driver mechanisms of microbial dysbiosis after infection with a fast-spreading virus. Our data also highlight potential context-dependent effects of probiotics and suggest a model in which changes in host behavior and downstream microbiome dysbiosis may constitute a previously unrecognized negative feedback loop that contributes to CD8 T cell adaptations after infections with fast-spreading and/or persistent pathogens.


Asunto(s)
Anorexia/inmunología , Antígenos CD8/inmunología , Memoria Inmunológica/inmunología , Coriomeningitis Linfocítica/inmunología , Virosis/inmunología , Akkermansia , Animales , Anorexia/microbiología , Anorexia/virología , Antígenos CD8/metabolismo , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/microbiología , Disbiosis/inmunología , Disbiosis/microbiología , Disbiosis/virología , Firmicutes/inmunología , Firmicutes/metabolismo , Microbioma Gastrointestinal/inmunología , Humanos , Coriomeningitis Linfocítica/microbiología , Coriomeningitis Linfocítica/patología , Virus de la Coriomeningitis Linfocítica/patogenicidad , Ratones , Linfocitos T/inmunología , Linfocitos T/microbiología , Verrucomicrobia/inmunología , Verrucomicrobia/patogenicidad , Virosis/microbiología , Virosis/patología
8.
Immunity ; 39(2): 206-7, 2013 Aug 22.
Artículo en Inglés | MEDLINE | ID: mdl-23973218

RESUMEN

In this issue of Immunity, Zelante et al. (2013) and Qiu et al. (2013) provide mechanistic insights into functional interactions between commensal microbes and innate lymphoid cells via the aryl hydrocarbon receptor.


Asunto(s)
Candida albicans/inmunología , Colitis/inmunología , Interleucinas/metabolismo , Limosilactobacillus reuteri/metabolismo , Receptores de Hidrocarburo de Aril/metabolismo , Células Th17/inmunología , Triptófano/metabolismo , Animales , Femenino , Interleucina-22
9.
Nature ; 540(7632): 280-283, 2016 12 08.
Artículo en Inglés | MEDLINE | ID: mdl-27798599

RESUMEN

The Enterobacteriaceae are a family of Gram-negative bacteria that include commensal organisms as well as primary and opportunistic pathogens that are among the leading causes of morbidity and mortality worldwide. Although Enterobacteriaceae often comprise less than 1% of a healthy intestine's microbiota, some of these organisms can bloom in the inflamed gut; expansion of enterobacteria is a hallmark of microbial imbalance known as dysbiosis. Microcins are small secreted proteins that possess antimicrobial activity in vitro, but whose role in vivo has been unclear. Here we demonstrate that microcins enable the probiotic bacterium Escherichia coli Nissle 1917 (EcN) to limit the expansion of competing Enterobacteriaceae (including pathogens and pathobionts) during intestinal inflammation. Microcin-producing EcN limits the growth of competitors in the inflamed intestine, including commensal E. coli, adherent-invasive E. coli and the related pathogen Salmonella enterica. Moreover, only therapeutic administration of the wild-type, microcin-producing EcN to mice previously infected with S. enterica substantially reduced intestinal colonization by the pathogen. Our work provides the first evidence that microcins mediate inter- and intraspecies competition among the Enterobacteriaceae in the inflamed gut. Moreover, we show that microcins can act as narrow-spectrum therapeutics to inhibit enteric pathogens and reduce enterobacterial blooms.


Asunto(s)
Bacteriocinas/metabolismo , Enterobacteriaceae/crecimiento & desarrollo , Escherichia coli/metabolismo , Inflamación/microbiología , Inflamación/patología , Intestinos/microbiología , Intestinos/patología , Animales , Bacteriocinas/genética , Bacteriocinas/uso terapéutico , Disbiosis/microbiología , Enterobacteriaceae/patogenicidad , Escherichia coli/clasificación , Escherichia coli/crecimiento & desarrollo , Femenino , Inflamación/metabolismo , Mucosa Intestinal/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Probióticos/metabolismo , Salmonella enterica/crecimiento & desarrollo , Salmonella enterica/patogenicidad , Simbiosis
10.
J Infect Dis ; 223(12 Suppl 2): S307-S313, 2021 06 16.
Artículo en Inglés | MEDLINE | ID: mdl-33330928

RESUMEN

Infections caused by Gram-negative bacteria can be challenging to treat due to the outer membrane permeability barrier and the increasing emergence of antibiotic resistance. During infection, Gram-negative pathogens must acquire iron, an essential nutrient, in the host. Many Gram-negative bacteria utilize sophisticated iron acquisition machineries based on siderophores, small molecules that bind iron with high affinity. In this review, we provide an overview of siderophore-mediated iron acquisition in Enterobacteriaceae and show how these systems provide a foundation for the conceptualization and development of approaches to prevent and/or treat bacterial infections. Differences between the siderophore-based iron uptake machineries of pathogenic Enterobacteriaceae and commensal microbes may lead to the development of selective "Trojan-horse" antimicrobials and immunization strategies that will not harm the host microbiota.


Asunto(s)
Antibacterianos/metabolismo , Enterobacteriaceae/efectos de los fármacos , Hierro/metabolismo , Sideróforos/metabolismo , Animales , Bacteriocinas/metabolismo , Bacteriocinas/farmacología , Enterobacteriaceae/crecimiento & desarrollo , Enterobacteriaceae/metabolismo , Bacterias Gramnegativas/efectos de los fármacos , Bacterias Gramnegativas/crecimiento & desarrollo , Bacterias Gramnegativas/metabolismo , Humanos , Inmunización , Microbiota/efectos de los fármacos , Sideróforos/inmunología
11.
Semin Cell Dev Biol ; 88: 129-137, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-29432952

RESUMEN

The gastrointestinal tract is a complex environment in which the host immune system interacts with a diverse array of microorganisms, both symbiotic and pathogenic. As such, mobilizing a rapid and appropriate antimicrobial response depending on the nature of each stimulus is crucial for maintaining the balance between homeostasis and inflammation in the gut. Here we focus on the mechanisms by which intestinal antimicrobial peptides regulate microbial communities during dysbiosis and infection. We also discuss classes of bacterial peptides that contribute to reducing enteric pathogen outgrowth. This review aims to provide a comprehensive overview on the interplay of diverse antimicrobial responses with enteric pathogens and the gut microbiota.


Asunto(s)
Bacteriocinas/inmunología , Defensinas/inmunología , Disbiosis/prevención & control , Tracto Gastrointestinal/inmunología , Mucosa Intestinal/inmunología , Animales , Bacteriocinas/biosíntesis , Bacteriocinas/farmacología , Catelicidinas/biosíntesis , Catelicidinas/inmunología , Catelicidinas/farmacología , Defensinas/biosíntesis , Defensinas/farmacología , Disbiosis/inmunología , Disbiosis/microbiología , Microbioma Gastrointestinal/inmunología , Tracto Gastrointestinal/efectos de los fármacos , Tracto Gastrointestinal/microbiología , Expresión Génica/inmunología , Humanos , Inmunidad Mucosa/efectos de los fármacos , Inflamación , Mucosa Intestinal/efectos de los fármacos , Mucosa Intestinal/microbiología , Lipocalina 2/biosíntesis , Lipocalina 2/inmunología , Lipocalina 2/farmacología , Muramidasa/biosíntesis , Muramidasa/inmunología , Muramidasa/farmacología , Simbiosis/inmunología
12.
J Immunol ; 203(2): 532-543, 2019 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-31142601

RESUMEN

Gut lymphocytes and the microbiota establish a reciprocal relationship that impacts the host immune response. Class I-restricted T cell-associated molecule (CRTAM) is a cell adhesion molecule expressed by intraepithelial T cells and is required for their retention in the gut. In this study, we show that CRTAM expression affects gut microbiota composition under homeostatic conditions. Moreover, Crtam-/- mice infected with the intestinal pathogen Salmonella exhibit reduced Th17 responses, lower levels of inflammation, and reduced Salmonella burden, which is accompanied by expansion of other microbial taxa. Thus, CRTAM enhances susceptibility to Salmonella, likely by promoting the inflammatory response that promotes the pathogen's growth. We also found that the gut microbiota from wild-type mice, but not from Crtam-/- mice, induces CRTAM expression and Th17 responses in ex-germ-free mice during Salmonella infection. Our study demonstrates a reciprocal relationship between CRTAM expression and the gut microbiota, which ultimately impacts the host response to enteric pathogens.


Asunto(s)
Microbioma Gastrointestinal/inmunología , Inmunoglobulinas/inmunología , Linfocitos T/inmunología , Animales , Femenino , Inflamación/inmunología , Intestinos/inmunología , Masculino , Ratones , Salmonella/inmunología , Infecciones por Salmonella/inmunología , Células Th17/inmunología
13.
Nature ; 580(7805): 594-595, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-32313162
14.
J Immunol ; 199(9): 3326-3335, 2017 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-28978694

RESUMEN

We describe a novel B cell-associated cytokine, encoded by an uncharacterized gene (C17orf99; chromosome 17 open reading frame 99), that is expressed in bone marrow and fetal liver and whose expression is also induced in peripheral B cells upon activation. C17orf99 is only present in mammalian genomes, and it encodes a small (∼27-kDa) secreted protein unrelated to other cytokine families, suggesting a function in mammalian immune responses. Accordingly, C17orf99 expression is induced in the mammary gland upon the onset of lactation, and a C17orf99-/- mouse exhibits reduced levels of IgA in the serum, gut, feces, and lactating mammary gland. C17orf99-/- mice have smaller and fewer Peyer's patches and lower numbers of IgA-secreting cells. The microbiome of C17orf99-/- mice exhibits altered composition, likely a consequence of the reduced levels of IgA in the gut. Although naive B cells can express C17orf99 upon activation, their production increases following culture with various cytokines, including IL-4 and TGF-ß1, suggesting that differentiation can result in the expansion of C17orf99-producing B cells during some immune responses. Taken together, these observations indicate that C17orf99 encodes a novel B cell-associated cytokine, which we have called IL-40, that plays an important role in humoral immune responses and may also play a role in B cell development. Importantly, IL-40 is also expressed by human activated B cells and by several human B cell lymphomas. The latter observations suggest that it may play a role in the pathogenesis of certain human diseases.


Asunto(s)
Linfocitos B/inmunología , Regulación de la Expresión Génica/inmunología , Interleucinas/inmunología , Ganglios Linfáticos Agregados/inmunología , Animales , Humanos , Inmunoglobulina A/inmunología , Interleucinas/genética , Células Jurkat , Linfoma de Células B/genética , Linfoma de Células B/inmunología , Ratones , Ratones Noqueados
15.
Proc Natl Acad Sci U S A ; 113(47): 13462-13467, 2016 11 22.
Artículo en Inglés | MEDLINE | ID: mdl-27821741

RESUMEN

Infections with Gram-negative pathogens pose a serious threat to public health. This scenario is exacerbated by increases in antibiotic resistance and the limited availability of vaccines and therapeutic tools to combat these infections. Here, we report an immunization approach that targets siderophores, which are small molecules exported by enteric Gram-negative pathogens to acquire iron, an essential nutrient, in the host. Because siderophores are nonimmunogenic, we designed and synthesized conjugates of a native siderophore and the immunogenic carrier protein cholera toxin subunit B (CTB). Mice immunized with the CTB-siderophore conjugate developed anti-siderophore antibodies in the gut mucosa, and when mice were infected with the enteric pathogen Salmonella, they exhibited reduced intestinal colonization and reduced systemic dissemination of the pathogen. Moreover, analysis of the gut microbiota revealed that reduction of Salmonella colonization in the inflamed gut was accompanied by expansion of Lactobacillus spp., which are beneficial commensal organisms that thrive in similar locales as Enterobacteriaceae. Collectively, our results demonstrate that anti-siderophore antibodies inhibit Salmonella colonization. Because siderophore-mediated iron acquisition is a virulence trait shared by many bacterial and fungal pathogens, blocking microbial iron acquisition by siderophore-based immunization or other siderophore-targeted approaches may represent a novel strategy to prevent and ameliorate a broad range of infections.


Asunto(s)
Enterobacteriaceae/crecimiento & desarrollo , Enterobacteriaceae/inmunología , Inmunización , Sideróforos/inmunología , Animales , Formación de Anticuerpos , Recuento de Colonia Microbiana , Femenino , Microbioma Gastrointestinal , Inmunidad Mucosa/inmunología , Inflamación/patología , Ratones Endogámicos C57BL , Sideróforos/química
16.
J Am Chem Soc ; 140(15): 5193-5201, 2018 04 18.
Artículo en Inglés | MEDLINE | ID: mdl-29578687

RESUMEN

Enteric Gram-negative bacteria, including Escherichia coli, biosynthesize and deploy the triscatecholate siderophore enterobactin (Ent) in the vertebrate host to acquire iron, an essential nutrient. We report that Ent-Cipro, a synthetic siderophore-antibiotic conjugate based on the native Ent platform that harbors an alkyl linker at one of the catechols with a ciprofloxacin cargo attached, affords targeted antibacterial activity against E. coli strains that express the pathogen-associated iroA gene cluster. Attachment of the siderophore to ciprofloxacin, a DNA gyrase inhibitor and broad-spectrum antibiotic that is used to treat infections caused by E. coli, generates an inactive prodrug and guides the antibiotic into the cytoplasm of bacteria that express the Ent uptake machinery (FepABCDG). Intracellular hydrolysis of the siderophore restores the activity of the antibiotic. Remarkably, Fes, the cytoplasmic Ent hydrolase expressed by all E. coli, does not contribute to Ent-Cipro activation. Instead, this processing step requires IroD, a cytoplasmic hydrolase that is expressed only by E. coli that harbor the iroA gene cluster and are predominantly pathogenic. In the uropathogenic E. coli UTI89 and CFT073, Ent-Cipro provides antibacterial activity comparable to unmodified ciprofloxacin. This work highlights the potential of leveraging and targeting pathogen-associated microbial enzymes in narrow-spectrum antibacterial approaches. Moreover, because E. coli include harmless gut commensals as well as resident microbes that can contribute to disease, Ent-Cipro may provide a valuable chemical tool for strain-selective modulation of the microbiota.


Asunto(s)
Antibacterianos/farmacología , Ciprofloxacina/farmacología , Enterobactina/farmacología , Escherichia coli/efectos de los fármacos , Esterasas/metabolismo , Antibacterianos/química , Antibacterianos/metabolismo , Biocatálisis , Ciprofloxacina/química , Ciprofloxacina/metabolismo , Relación Dosis-Respuesta a Droga , Enterobactina/química , Enterobactina/metabolismo , Hidrólisis , Pruebas de Sensibilidad Microbiana , Conformación Molecular , Relación Estructura-Actividad
17.
Trends Immunol ; 36(2): 112-20, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25582038

RESUMEN

Pathogens have evolved clever strategies to evade and in some cases exploit the attacks of an activated immune system. Salmonella enterica is one such pathogen, exploiting multiple aspects of host defense to promote its replication in the host. Here we review recent findings on the mechanisms by which Salmonella establishes systemic and chronic infection, including strategies involving manipulation of innate immune signaling and inflammatory forms of cell death, as well as immune evasion by establishing residency in M2 macrophages. We also examine recent evidence showing that the oxidative environment and the high levels of antimicrobial proteins produced in response to localized Salmonella gastrointestinal infection enable the pathogen to successfully outcompete the resident gut microbiota.


Asunto(s)
Interacciones Huésped-Patógeno/inmunología , Inmunidad , Infecciones por Salmonella/inmunología , Salmonella/inmunología , Animales , Humanos , Mucosa Intestinal/metabolismo , Intestinos/inmunología , Intestinos/microbiología , Estrés Oxidativo , Especies de Nitrógeno Reactivo/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Infecciones por Salmonella/metabolismo , Receptores Toll-Like/metabolismo
18.
J Immunol ; 194(9): 4081-7, 2015 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-25888704

RESUMEN

The mammalian intestine harbors a community of trillions of microbes, collectively known as the gut microbiota, which coevolved with the host in a mutually beneficial relationship. Among the numerous gut microbial species, certain commensal bacteria are known to provide health benefits to the host when administered in adequate amounts and, as such, are labeled "probiotics." We review some of the mechanisms by which probiotics and other beneficial commensals provide colonization resistance to pathogens. The battle for similar nutrients and the bacterial secretion of antimicrobials provide a direct means of competition between beneficial and harmful microbes. Beneficial microbes can also indirectly diminish pathogen colonization by stimulating the development of innate and adaptive immunity, as well as the function of the mucosal barrier. Altogether, we gather and present evidence that beneficial microbes cooperate with host immunity in an effort to shut out pathogens.


Asunto(s)
Bacterias/crecimiento & desarrollo , Bacterias/inmunología , Interacciones Huésped-Patógeno/inmunología , Inmunidad/inmunología , Microbiota/inmunología , Simbiosis/inmunología , Animales , Bacterias/patogenicidad , Humanos
19.
Proc Natl Acad Sci U S A ; 111(47): E5076-85, 2014 Nov 25.
Artículo en Inglés | MEDLINE | ID: mdl-25385646

RESUMEN

The mammalian target of rapamycin (mTOR) is a kinase that functions in two distinct complexes, mTORC1 and mTORC2. In peripheral B cells, complete deletion of mTOR suppresses germinal center B-cell responses, including class switching and somatic hypermutation. The allosteric mTORC1 inhibitor rapamycin blocks proliferation and differentiation, but lower doses can promote protective IgM responses. To elucidate the complexity of mTOR signaling in B cells further, we used ATP-competitive mTOR kinase inhibitors (TOR-KIs), which inhibit both mTORC1 and mTORC2. Although TOR-KIs are in clinical development for cancer, their effects on mature lymphocytes are largely unknown. We show that high concentrations of TOR-KIs suppress B-cell proliferation and differentiation, yet lower concentrations that preserve proliferation increase the fraction of B cells undergoing class switching in vitro. Transient treatment of mice with the TOR-KI compound AZD8055 increased titers of class-switched high-affinity antibodies to a hapten-protein conjugate. Mechanistic investigation identified opposing roles for mTORC1 and mTORC2 in B-cell differentiation and showed that TOR-KIs enhance class switching in a manner dependent on forkhead box, subgroup O (FoxO) transcription factors. These observations emphasize the distinct actions of TOR-KIs compared with rapamycin and suggest that TOR-KIs might be useful to enhance production of class-switched antibodies following vaccination.


Asunto(s)
Cambio de Clase de Inmunoglobulina/efectos de los fármacos , Complejos Multiproteicos/antagonistas & inhibidores , Inhibidores de Proteínas Quinasas/farmacología , Serina-Treonina Quinasas TOR/antagonistas & inhibidores , Regulación Alostérica , Animales , Inmunoglobulina G/biosíntesis , Diana Mecanicista del Complejo 2 de la Rapamicina , Ratones , Ratones Endogámicos C57BL , Sirolimus/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA