Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 77
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Cell Host Microbe ; 32(6): 863-874, 2024 Jun 12.
Artículo en Inglés | MEDLINE | ID: mdl-38870902

RESUMEN

The composition and function of the gut microbiota are intimately tied to nutrient acquisition strategies and metabolism, with significant implications for host health. Both dietary and host-intrinsic factors influence community structure and the basic modes of bacterial energy metabolism. The intestinal tract is rich in carbon and nitrogen sources; however, limited access to oxygen restricts energy-generating reactions to fermentation. By contrast, increased availability of electron acceptors during episodes of intestinal inflammation results in phylum-level changes in gut microbiota composition, suggesting that bacterial energy metabolism is a key driver of gut microbiota function. In this review article, we will illustrate diverse examples of microbial nutrient acquisition strategies in the context of habitat filters and anatomical location and the central role of energy metabolism in shaping metabolic strategies to support bacterial growth in the mammalian gut.


Asunto(s)
Bacterias , Metabolismo Energético , Microbioma Gastrointestinal , Nutrientes , Microbioma Gastrointestinal/fisiología , Humanos , Nutrientes/metabolismo , Animales , Bacterias/metabolismo , Bacterias/crecimiento & desarrollo , Tracto Gastrointestinal/microbiología , Tracto Gastrointestinal/metabolismo , Nitrógeno/metabolismo , Carbono/metabolismo , Fermentación
2.
mBio ; 15(6): e0035024, 2024 Jun 12.
Artículo en Inglés | MEDLINE | ID: mdl-38682906

RESUMEN

Enteric pathogens such as Salmonella enterica serovar Typhimurium experience spatial and temporal changes to the metabolic landscape throughout infection. Host reactive oxygen and nitrogen species non-enzymatically convert monosaccharides to alpha hydroxy acids, including L-tartrate. Salmonella utilizes L-tartrate early during infection to support fumarate respiration, while L-tartrate utilization ceases at later time points due to the increased availability of exogenous electron acceptors such as tetrathionate, nitrate, and oxygen. It remains unknown how Salmonella regulates its gene expression to metabolically adapt to changing nutritional environments. Here, we investigated how the transcriptional regulation for L-tartrate metabolism in Salmonella is influenced by infection-relevant cues. L-tartrate induces the transcription of ttdBAU, genes involved in L-tartrate utilization. L-tartrate metabolism is negatively regulated by two previously uncharacterized transcriptional regulators TtdV (STM3357) and TtdW (STM3358), and both TtdV and TtdW are required for the sensing of L-tartrate. The electron acceptors nitrate, tetrathionate, and oxygen repress ttdBAU transcription via the two-component system ArcAB. Furthermore, the regulation of L-tartrate metabolism is required for optimal fitness in a mouse model of Salmonella-induced colitis. TtdV, TtdW, and ArcAB allow for the integration of two cues, i.e., substrate availability and availability of exogenous electron acceptors, to control L-tartrate metabolism. Our findings provide novel insights into how Salmonella prioritizes the utilization of different electron acceptors for respiration as it experiences transitional nutrient availability throughout infection. IMPORTANCE: Bacterial pathogens must adapt their gene expression profiles to cope with diverse environments encountered during infection. This coordinated process is carried out by the integration of cues that the pathogen senses to fine-tune gene expression in a spatiotemporal manner. Many studies have elucidated the regulatory mechanisms of how Salmonella sense metabolites in the gut to activate or repress its virulence program; however, our understanding of how Salmonella coordinates its gene expression to maximize the utilization of carbon and energy sources found in transitional nutrient niches is not well understood. In this study, we discovered how Salmonella integrates two infection-relevant cues, substrate availability and exogenous electron acceptors, to control L-tartrate metabolism. From our experiments, we propose a model for how L-tartrate metabolism is regulated in response to different metabolic cues in addition to characterizing two previously unknown transcriptional regulators. This study expands our understanding of how microbes combine metabolic cues to enhance fitness during infection.


Asunto(s)
Proteínas Bacterianas , Regulación Bacteriana de la Expresión Génica , Salmonella typhimurium , Tartratos , Salmonella typhimurium/genética , Salmonella typhimurium/metabolismo , Ratones , Animales , Tartratos/metabolismo , Proteínas Bacterianas/metabolismo , Proteínas Bacterianas/genética , Infecciones por Salmonella/microbiología , Femenino
3.
bioRxiv ; 2024 Feb 06.
Artículo en Inglés | MEDLINE | ID: mdl-38370731

RESUMEN

Enteric pathogens such as Salmonella enterica serovar Typhimurium experience spatial and temporal changes to the metabolic landscape throughout infection. Host reactive oxygen and nitrogen species non-enzymatically convert monosaccharides to alpha hydroxy acids, including L-tartrate. Salmonella utilizes L-tartrate early during infection to support fumarate respiration, while L-tartrate utilization ceases at later time points due to the increased availability of exogenous electron acceptors such as tetrathionate, nitrate, and oxygen. It remains unknown how Salmonella regulates its gene expression to metabolically adapt to changing nutritional environments. Here, we investigated how the transcriptional regulation for L-tartrate metabolism in Salmonella is influenced by infection-relevant cues. L-tartrate induces the transcription of ttdBAU, genes involved in L-tartrate utilization. L-tartrate metabolism is negatively regulated by two previously uncharacterized transcriptional regulators TtdV (STM3357) and TtdW (STM3358), and both TtdV and TtdW are required for sensing of L-tartrate. The electron acceptors nitrate, tetrathionate, and oxygen repress ttdBAU transcription via the two-component system ArcAB. Furthermore, regulation of L-tartrate metabolism is required for optimal fitness in a mouse model of Salmonella-induced colitis. TtdV, TtdW, and ArcAB allow for the integration of two cues, substrate availability and availability of exogenous electron acceptors, to control L-tartrate metabolism. Our findings provide novel insights into how Salmonella prioritizes utilization of different electron acceptors for respiration as it experiences transitional nutrient availability throughout infection.

4.
PLoS Biol ; 22(1): e3002486, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-38236896

RESUMEN

Acute gastrointestinal infection with intracellular pathogens like Salmonella Typhimurium triggers the release of the proinflammatory cytokine interleukin 1ß (IL-1ß). However, the role of IL-1ß in intestinal defense against Salmonella remains unclear. Here, we show that IL-1ß production is detrimental during Salmonella infection. Mice lacking IL-1ß (IL-1ß -/-) failed to recruit neutrophils to the gut during infection, which reduced tissue damage and prevented depletion of short-chain fatty acid (SCFA)-producing commensals. Changes in epithelial cell metabolism that typically support pathogen expansion, such as switching energy production from fatty acid oxidation to fermentation, were absent in infected IL-1ß -/- mice which inhibited Salmonella expansion. Additionally, we found that IL-1ß induces expression of complement anaphylatoxins and suppresses the complement-inactivator carboxypeptidase N (CPN1). Disrupting this process via IL-1ß loss prevented mortality in Salmonella-infected IL-1ß -/- mice. Finally, we found that IL-1ß expression correlates with expression of the complement receptor in patients suffering from sepsis, but not uninfected patients and healthy individuals. Thus, Salmonella exploits IL-1ß signaling to outcompete commensal microbes and establish gut colonization. Moreover, our findings identify the intersection of IL-1ß signaling and the complement system as key host factors involved in controlling mortality during invasive Salmonellosis.


Asunto(s)
Interleucina-1beta , Infecciones por Salmonella , Animales , Humanos , Ratones , Interleucina-1beta/genética , Interleucina-1beta/metabolismo , Neutrófilos/metabolismo , Infecciones por Salmonella/metabolismo , Salmonella typhimurium/metabolismo , Virulencia
5.
Proc Natl Acad Sci U S A ; 120(50): e2316579120, 2023 Dec 12.
Artículo en Inglés | MEDLINE | ID: mdl-38048456

RESUMEN

The gut microbiota plays a role in many human diseases, but high-throughput sequence analysis does not provide a straightforward path for defining healthy microbial communities. Therefore, understanding mechanisms that drive compositional changes during disease (gut dysbiosis) continues to be a central goal in microbiome research. Insights from the microbial pathogenesis field show that an ecological cause for gut dysbiosis is an increased availability of host-derived respiratory electron acceptors, which are dominant drivers of microbial community composition. Similar changes in the host environment also drive gut dysbiosis in several chronic human illnesses, and a better understanding of the underlying mechanisms informs approaches to causatively link compositional changes in the gut microbiota to an exacerbation of symptoms. The emerging picture suggests that homeostasis is maintained by host functions that control the availability of resources governing microbial growth. Defining dysbiosis as a weakening of these host functions directs attention to the underlying cause and identifies potential targets for therapeutic intervention.


Asunto(s)
Microbioma Gastrointestinal , Microbiota , Humanos , Disbiosis
6.
bioRxiv ; 2023 Dec 08.
Artículo en Inglés | MEDLINE | ID: mdl-38106073

RESUMEN

Louis Pasteur's experiments on tartaric acid laid the foundation for our understanding of molecular chirality, but major questions remain. By comparing the optical activity of naturally-occurring tartaric acid with chemically-synthesized paratartaric acid, Pasteur realized that naturally-occurring tartaric acid contained only L-tartaric acid while paratartaric acid consisted of a racemic mixture of D- and L-tartaric acid. Curiously, D-tartaric acid has no known natural source, yet several gut bacteria specifically degrade D-tartaric acid. Here, we investigated the oxidation of monosaccharides by inflammatory reactive oxygen and nitrogen species. We found that this reaction yields an array of alpha hydroxy carboxylic acids, including tartaric acid isomers. Utilization of inflammation- derived D- and L-tartaric acid enhanced colonization by Salmonella Typhimurium and E. coli in murine models of gut inflammation. Our findings suggest that byproducts of inflammatory radical metabolism, such as tartrate and other alpha hydroxy carboxylic acids, create transient nutrient niches for enteric pathogens and other potentially harmful bacteria. Furthermore, this work illustrates that inflammatory radicals generate a zoo of molecules, some of which may erroneously presumed to be xenobiotics.

7.
mBio ; 14(4): e0092123, 2023 08 31.
Artículo en Inglés | MEDLINE | ID: mdl-37498116

RESUMEN

Salmonella enterica serovar Typhimurium induces intestinal inflammation to create a niche that fosters the outgrowth of the pathogen over the gut microbiota. Under inflammatory conditions, Salmonella utilizes terminal electron acceptors generated as byproducts of intestinal inflammation to generate cellular energy through respiration. However, the electron donating reactions in these electron transport chains are poorly understood. Here, we investigated how formate utilization through the respiratory formate dehydrogenase-N (FdnGHI) and formate dehydrogenase-O (FdoGHI) contribute to gut colonization of Salmonella. Both enzymes fulfilled redundant roles in enhancing fitness in a mouse model of Salmonella-induced colitis, and coupled to tetrathionate, nitrate, and oxygen respiration. The formic acid utilized by Salmonella during infection was generated by its own pyruvate-formate lyase as well as the gut microbiota. Transcription of formate dehydrogenases and pyruvate-formate lyase was significantly higher in bacteria residing in the mucus layer compared to the lumen. Furthermore, formate utilization conferred a more pronounced fitness advantage in the mucus, indicating that formate production and degradation occurred predominantly in the mucus layer. Our results provide new insights into how Salmonella adapts its energy metabolism to the local microenvironment in the gut. IMPORTANCE Bacterial pathogens must not only evade immune responses but also adapt their metabolism to successfully colonize their host. The microenvironments encountered by enteric pathogens differ based on anatomical location, such as small versus large intestine, spatial stratification by host factors, such as mucus layer and antimicrobial peptides, and distinct commensal microbial communities that inhabit these microenvironments. Our understanding of how Salmonella populations adapt its metabolism to different environments in the gut is incomplete. In the current study, we discovered that Salmonella utilizes formate as an electron donor to support respiration, and that formate oxidation predominantly occurs in the mucus layer. Our experiments suggest that spatially distinct Salmonella populations in the mucus layer and the lumen differ in their energy metabolism. Our findings enhance our understanding of the spatial nature of microbial metabolism and may have implications for other enteric pathogens as well as commensal host-associated microbial communities.


Asunto(s)
Liasas , Salmonelosis Animal , Animales , Ratones , Salmonella typhimurium/metabolismo , Serogrupo , Salmonelosis Animal/microbiología , Bacterias , Inflamación , Formiatos/metabolismo , Moco , Piruvatos/metabolismo , Liasas/metabolismo
8.
Microbiome ; 10(1): 200, 2022 11 26.
Artículo en Inglés | MEDLINE | ID: mdl-36434690

RESUMEN

BACKGROUND: Intestinal inflammation disrupts the microbiota composition leading to an expansion of Enterobacteriaceae family members (dysbiosis). Associated with this shift in microbiota composition is a profound change in the metabolic landscape of the intestine. It is unclear how changes in metabolite availability during gut inflammation impact microbial and host physiology. RESULTS: We investigated microbial and host lactate metabolism in murine models of infectious and non-infectious colitis. During inflammation-associated dysbiosis, lactate levels in the gut lumen increased. The disease-associated spike in lactate availability was significantly reduced in mice lacking the lactate dehydrogenase A subunit in intestinal epithelial cells. Commensal E. coli and pathogenic Salmonella, representative Enterobacteriaceae family members, utilized lactate via the respiratory L-lactate dehydrogenase LldD to increase fitness. Furthermore, mice lacking the lactate dehydrogenase A subunit in intestinal epithelial cells exhibited lower levels of inflammation in a model of non-infectious colitis. CONCLUSIONS: The release of lactate by intestinal epithelial cells during gut inflammation impacts the metabolism of gut-associated microbial communities. These findings suggest that during intestinal inflammation and dysbiosis, changes in metabolite availability can perpetuate colitis-associated disturbances of microbiota composition. Video Abstract.


Asunto(s)
Colitis , Microbioma Gastrointestinal , Ratones , Animales , Disbiosis , Escherichia coli/metabolismo , Ácido Láctico/metabolismo , Lactato Deshidrogenasa 5 , Ratones Endogámicos C57BL , Inflamación/patología , Colitis/patología , Enterobacteriaceae/metabolismo
9.
Cell Host Microbe ; 29(10): 1531-1544.e9, 2021 10 13.
Artículo en Inglés | MEDLINE | ID: mdl-34536347

RESUMEN

The minimal genetic requirements for microbes to survive within multiorganism communities, including host-pathogen interactions, remain poorly understood. Here, we combined targeted gene mutagenesis with phenotype-guided genetic reassembly to identify a cooperative network of SPI-2 T3SS effector genes that are sufficient for Salmonella Typhimurium (STm) to cause disease in a natural host organism. Five SPI-2 effector genes support pathogen survival within the host cell cytoplasm by coordinating bacterial replication with Salmonella-containing vacuole (SCV) division. Unexpectedly, this minimal genetic repertoire does not support STm systemic infection of mice. In vivo screening revealed a second effector-gene network, encoded by the spv operon, that expands the life cycle of STm from growth in cells to deep-tissue colonization in a murine model of typhoid fever. Comparison between Salmonella infection models suggests how cooperation between effector genes drives tissue tropism in a pathogen group.


Asunto(s)
Proteínas Bacterianas/genética , Redes Reguladoras de Genes , Infecciones por Salmonella/microbiología , Salmonella typhimurium/genética , Animales , Proteínas Bacterianas/metabolismo , Citoplasma/microbiología , Femenino , Islas Genómicas , Interacciones Huésped-Patógeno , Humanos , Ratones , Ratones Endogámicos C57BL , Viabilidad Microbiana , Operón , Salmonella typhimurium/crecimiento & desarrollo , Salmonella typhimurium/patogenicidad , Salmonella typhimurium/fisiología , Tropismo , Sistemas de Secreción Tipo III/genética , Sistemas de Secreción Tipo III/metabolismo , Virulencia
10.
Braz J Microbiol ; 52(4): 1745-1753, 2021 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-34251610

RESUMEN

Historically, clinical microbiological laboratories have often relied on isolation of pure cultures and phenotypic testing to identify microorganisms. These clinical tests are often based on specific biochemical reactions, growth characteristics, colony morphology, and other physiological aspects. The features used for identification in clinical laboratories are highly conserved and specific for a given group of microbes. We speculate that these features might be the result of evolutionary selection and thus may reflect aspects of the life cycle of the organism and pathogenesis. Indeed, several of the metabolic pathways targeted by diagnostic tests in some cases may represent mechanisms for host colonization or pathogenesis. Examples include, but are not restricted to, Staphylococcus aureus, Pseudomonas aeruginosa, Klebsiella pneumoniae, Salmonella enterica, Shigella spp., and enteroinvasive Escherichia coli (EIEC). Here, we provide an overview of how some common tests reflect molecular mechanisms of bacterial pathogenesis.


Asunto(s)
Infecciones Bacterianas , Disentería Bacilar , Adaptación al Huésped , Bacterias/inmunología , Infecciones Bacterianas/diagnóstico , Infecciones Bacterianas/microbiología , Infecciones Bacterianas/patología , Disentería Bacilar/diagnóstico , Disentería Bacilar/microbiología , Disentería Bacilar/fisiopatología , Humanos , Laboratorios Clínicos
11.
Elife ; 102021 06 04.
Artículo en Inglés | MEDLINE | ID: mdl-34085924

RESUMEN

The composition of gut-associated microbial communities changes during intestinal inflammation, including an expansion of Enterobacteriaceae populations. The mechanisms underlying microbiota changes during inflammation are incompletely understood. Here, we analyzed previously published metagenomic datasets with a focus on microbial hydrogen metabolism. The bacterial genomes in the inflamed murine gut and in patients with inflammatory bowel disease contained more genes encoding predicted hydrogen-utilizing hydrogenases compared to communities found under non-inflamed conditions. To validate these findings, we investigated hydrogen metabolism of Escherichia coli, a representative Enterobacteriaceae, in mouse models of colitis. E. coli mutants lacking hydrogenase-1 and hydrogenase-2 displayed decreased fitness during colonization of the inflamed cecum and colon. Utilization of molecular hydrogen was in part dependent on respiration of inflammation-derived electron acceptors. This work highlights the contribution of hydrogenases to alterations of the gut microbiota in the context of non-infectious colitis.


Asunto(s)
Ciego/microbiología , Colitis/inducido químicamente , Colitis/microbiología , Colon/microbiología , Infecciones por Escherichia coli/microbiología , Escherichia coli/metabolismo , Microbioma Gastrointestinal , Hidrógeno/metabolismo , Animales , Ciego/metabolismo , Ciego/patología , Colitis/metabolismo , Colitis/patología , Colon/metabolismo , Colon/patología , Bases de Datos Genéticas , Sulfato de Dextran , Modelos Animales de Enfermedad , Disbiosis , Escherichia coli/genética , Escherichia coli/crecimiento & desarrollo , Infecciones por Escherichia coli/metabolismo , Infecciones por Escherichia coli/patología , Proteínas de Escherichia coli/genética , Proteínas de Escherichia coli/metabolismo , Femenino , Humanos , Hidrogenasas/genética , Hidrogenasas/metabolismo , Interleucina-10/genética , Interleucina-10/metabolismo , Masculino , Metagenoma , Metagenómica , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Piroxicam
12.
Cell ; 183(3): 650-665.e15, 2020 10 29.
Artículo en Inglés | MEDLINE | ID: mdl-33031742

RESUMEN

Endocannabinoids are host-derived lipid hormones that fundamentally impact gastrointestinal (GI) biology. The use of cannabis and other exocannabinoids as anecdotal treatments for various GI disorders inspired the search for mechanisms by which these compounds mediate their effects, which led to the discovery of the mammalian endocannabinoid system. Dysregulated endocannabinoid signaling was linked to inflammation and the gut microbiota. However, the effects of endocannabinoids on host susceptibility to infection has not been explored. Here, we show that mice with elevated levels of the endocannabinoid 2-arachidonoyl glycerol (2-AG) are protected from enteric infection by Enterobacteriaceae pathogens. 2-AG directly modulates pathogen function by inhibiting virulence programs essential for successful infection. Furthermore, 2-AG antagonizes the bacterial receptor QseC, a histidine kinase encoded within the core Enterobacteriaceae genome that promotes the activation of pathogen-associated type three secretion systems. Taken together, our findings establish that endocannabinoids are directly sensed by bacteria and can modulate bacterial function.


Asunto(s)
Endocannabinoides/metabolismo , Enterobacteriaceae/patogenicidad , Animales , Ácidos Araquidónicos/química , Ácidos Araquidónicos/metabolismo , Adhesión Bacteriana , Proteínas Bacterianas/metabolismo , Sistemas de Secreción Bacterianos/metabolismo , Citrobacter rodentium/patogenicidad , Colon/microbiología , Colon/patología , Endocannabinoides/química , Infecciones por Enterobacteriaceae/microbiología , Femenino , Microbioma Gastrointestinal , Glicéridos/química , Glicéridos/metabolismo , Células HeLa , Interacciones Huésped-Patógeno , Humanos , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Monoacilglicerol Lipasas/metabolismo , Salmonella/patogenicidad , Virulencia
13.
Cell Host Microbe ; 28(6): 780-788.e5, 2020 12 09.
Artículo en Inglés | MEDLINE | ID: mdl-33053375

RESUMEN

The intestinal epithelium separates host tissue and gut-associated microbial communities. During inflammation, the host releases reactive oxygen and nitrogen species as an antimicrobial response. The impact of these radicals on gut microbes is incompletely understood. We discovered that the cryptic appBCX genes, predicted to encode a cytochrome bd-II oxidase, conferred a fitness advantage for E. coli in chemical and genetic models of non-infectious colitis. This fitness advantage was absent in mice that lacked epithelial NADPH oxidase 1 (NOX1) activity. In laboratory growth experiments, supplementation with exogenous hydrogen peroxide enhanced E. coli growth through AppBCX-mediated respiration in a catalase-dependent manner. We conclude that epithelial-derived reactive oxygen species are degraded in the gut lumen, which gives rise to molecular oxygen that supports the aerobic respiration of E. coli. This work illustrates how epithelial host responses intersect with gut microbial metabolism in the context of gut inflammation.


Asunto(s)
Complejo IV de Transporte de Electrones/fisiología , Escherichia coli/fisiología , Inflamación/metabolismo , Mucosa Intestinal/metabolismo , NADPH Oxidasa 1/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Aerobiosis , Animales , Colitis/inducido químicamente , ADN Bacteriano , Modelos Animales de Enfermedad , Proteínas de Escherichia coli/fisiología , Femenino , Microbioma Gastrointestinal , Interacciones Microbiota-Huesped , Peróxido de Hidrógeno/metabolismo , Inflamación/microbiología , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Microbiota , NADPH Oxidasa 1/genética , Oxígeno/metabolismo
15.
Proc Natl Acad Sci U S A ; 117(22): 12387-12393, 2020 06 02.
Artículo en Inglés | MEDLINE | ID: mdl-32409599

RESUMEN

Microbiota, host and dietary metabolites/signals compose the rich gut chemical environment, which profoundly impacts virulence of enteric pathogens. Enterohemorrhagic Escherichia coli (EHEC) engages a syringe-like machinery named type-III secretion system (T3SS) to inject effectors within host cells that lead to intestinal colonization and disease. We previously conducted a high-throughput screen to identify metabolic pathways that affect T3SS expression. Here we show that in the presence of arginine, the arginine sensor ArgR, identified through this screen, directly activates expression of the genes encoding the T3SS. Exogenously added arginine induces EHEC virulence gene expression in vitro. Congruently, a mutant deficient in arginine transport (ΔartP) had decreased virulence gene expression. ArgR also augments murine disease caused by Citrobacter rodentium, which is a murine pathogen extensively employed as a surrogate animal model for EHEC. The source of arginine sensed by C. rodentium is not dietary. At the peak of C. rodentium infection, increased arginine concentration in the colon correlated with down-regulation of the host SLC7A2 transporter. This increase in the concentration of colonic arginine promotes virulence gene expression in C. rodentium Arginine is an important modulator of the host immune response to pathogens. Here we add that arginine also directly impacts bacterial virulence. These findings suggest that a delicate balance between host and pathogen responses to arginine occur during disease progression.


Asunto(s)
Citrobacter rodentium/metabolismo , Infecciones por Enterobacteriaceae/microbiología , Escherichia coli Enterohemorrágica/metabolismo , Infecciones por Escherichia coli/microbiología , Regulación Bacteriana de la Expresión Génica , Animales , Arginina/metabolismo , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Citrobacter rodentium/genética , Citrobacter rodentium/patogenicidad , Escherichia coli Enterohemorrágica/genética , Escherichia coli Enterohemorrágica/patogenicidad , Humanos , Ratones , Ratones Endogámicos C3H , Virulencia , Factores de Virulencia/genética , Factores de Virulencia/metabolismo
16.
Cell Host Microbe ; 27(3): 376-388.e8, 2020 03 11.
Artículo en Inglés | MEDLINE | ID: mdl-32075741

RESUMEN

During short-lived perturbations, such as inflammation, the gut microbiota exhibits resilience and reverts to its original configuration. Although microbial access to the micronutrient iron is decreased during colitis, pathogens can scavenge iron by using siderophores. How commensal bacteria acquire iron during gut inflammation is incompletely understood. Curiously, the human commensal Bacteroides thetaiotaomicron does not produce siderophores but grows under iron-limiting conditions using enterobacterial siderophores. Using RNA-seq, we identify B. thetaiotaomicron genes that were upregulated during Salmonella-induced gut inflammation and were predicted to be involved in iron uptake. Mutants in the xusABC locus (BT2063-2065) were defective for xenosiderophore-mediated iron uptake in vitro. In the normal mouse gut, the XusABC system was dispensable, while a xusA mutant colonized poorly during colitis. This work identifies xenosiderophore utilization as a critical mechanism for B. thetaiotaomicron to sustain colonization during inflammation and suggests a mechanism of how interphylum iron metabolism contributes to gut microbiota resilience.


Asunto(s)
Bacteroides thetaiotaomicron/metabolismo , Colitis/microbiología , Enterobacteriaceae/genética , Microbioma Gastrointestinal , Hierro/metabolismo , Sideróforos/genética , Animales , Bacteroides thetaiotaomicron/genética , Femenino , Genes Bacterianos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , RNA-Seq , Simbiosis
17.
J Exp Med ; 216(10): 2378-2393, 2019 10 07.
Artículo en Inglés | MEDLINE | ID: mdl-31358565

RESUMEN

Chronic inflammation and gut microbiota dysbiosis, in particular the bloom of genotoxin-producing E. coli strains, are risk factors for the development of colorectal cancer. Here, we sought to determine whether precision editing of gut microbiota metabolism and composition could decrease the risk for tumor development in mouse models of colitis-associated colorectal cancer (CAC). Expansion of experimentally introduced E. coli strains in the azoxymethane/dextran sulfate sodium colitis model was driven by molybdoenzyme-dependent metabolic pathways. Oral administration of sodium tungstate inhibited E. coli molybdoenzymes and selectively decreased gut colonization with genotoxin-producing E. coli and other Enterobacteriaceae. Restricting the bloom of Enterobacteriaceae decreased intestinal inflammation and reduced the incidence of colonic tumors in two models of CAC, the azoxymethane/dextran sulfate sodium colitis model and azoxymethane-treated, Il10-deficient mice. We conclude that metabolic targeting of protumoral Enterobacteriaceae during chronic inflammation is a suitable strategy to prevent the development of malignancies arising from gut microbiota dysbiosis.


Asunto(s)
Colitis/microbiología , Neoplasias Colorrectales/microbiología , Disbiosis/microbiología , Microbioma Gastrointestinal , Neoplasias Experimentales/microbiología , Animales , Colitis/inducido químicamente , Colitis/genética , Colitis/patología , Neoplasias Colorrectales/inducido químicamente , Neoplasias Colorrectales/genética , Sulfato de Dextran/toxicidad , Disbiosis/inducido químicamente , Disbiosis/genética , Escherichia coli/crecimiento & desarrollo , Interleucina-10/deficiencia , Ratones , Neoplasias Experimentales/inducido químicamente , Neoplasias Experimentales/genética
18.
mBio ; 10(3)2019 05 28.
Artículo en Inglés | MEDLINE | ID: mdl-31138751

RESUMEN

Oral infection of C57BL/6J mice with Toxoplasma gondii results in a marked bacterial dysbiosis and the development of severe pathology in the distal small intestine that is dependent on CD4+ T cells and interferon gamma (IFN-γ). This dysbiosis and bacterial translocation contribute to the development of ileal pathology, but the factors that support the bloom of bacterial pathobionts are unclear. The use of microbial community profiling and shotgun metagenomics revealed that Toxoplasma infection induces a dysbiosis dominated by Enterobacteriaceae and an increased potential for nitrate respiration. In vivo experiments using bacterial metabolic mutants revealed that during this infection, host-derived nitrate supports the expansion of Enterobacteriaceae in the ileum via nitrate respiration. Additional experiments with infected mice indicate that the IFN-γ/STAT1/iNOS axis, while essential for parasite control, also supplies a pool of nitrate that serves as a source for anaerobic respiration and supports overgrowth of Enterobacteriaceae Together, these data reveal a trade-off in intestinal immunity after oral infection of C57BL/6J mice with T. gondii, in which inducible nitric oxide synthase (iNOS) is required for parasite control, while this host enzyme is responsible for specific modification of the composition of the microbiome that contributes to pathology.IMPORTANCEToxoplasma gondii is a protozoan parasite and a leading cause of foodborne illness. Infection is initiated when the parasite invades the intestinal epithelium, and in many host species, this leads to intense inflammation and a dramatic disruption of the normal microbial ecosystem that resides in the healthy gut (the so-called microbiome). One characteristic change in the microbiome during infection with Toxoplasma-as well as numerous other pathogens-is the overgrowth of Escherichia coli or similar bacteria and a breakdown of commensal containment leading to seeding of peripheral organs with gut bacteria and subsequent sepsis. Our findings provide one clear explanation for how this process is regulated, thereby improving our understanding of the relationship between parasite infection, inflammation, and disease. Furthermore, our results could serve as the basis for the development of novel therapeutics to reduce the potential for harmful bacteria to bloom in the gut during infection.


Asunto(s)
Disbiosis/inmunología , Microbioma Gastrointestinal , Intestino Delgado/inmunología , Intestino Delgado/patología , Activación de Macrófagos , Nitratos/metabolismo , Toxoplasmosis Animal/inmunología , Animales , Citocinas/análisis , Enterobacteriaceae/clasificación , Femenino , Inflamación , Interferón gamma/inmunología , Intestino Delgado/microbiología , Intestino Delgado/parasitología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Óxido Nítrico Sintasa de Tipo II/inmunología , Toxoplasma/patogenicidad , Toxoplasmosis Animal/microbiología
19.
PLoS Pathog ; 15(4): e1007745, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-31009517

RESUMEN

The mechanisms by which the gut luminal environment is disturbed by the immune system to foster pathogenic bacterial growth and survival remain incompletely understood. Here, we show that STAT2 dependent type I IFN signaling contributes to the inflammatory environment by disrupting hypoxia enabling the pathogenic S. Typhimurium to outgrow the microbiota. Stat2-/- mice infected with S. Typhimurium exhibited impaired type I IFN induced transcriptional responses in cecal tissue and reduced bacterial burden in the intestinal lumen compared to infected wild-type mice. Although inflammatory pathology was similar between wild-type and Stat2-/- mice, we observed decreased hypoxia in the gut tissue of Stat2-/- mice. Neutrophil numbers were similar in wild-type and Stat2-/- mice, yet Stat2-/- mice showed reduced levels of myeloperoxidase activity. In vitro, the neutrophils from Stat2-/- mice produced lower levels of superoxide anion upon stimulation with the bacterial ligand N-formylmethionyl-leucyl-phenylalanine (fMLP) in the presence of IFNα compared to neutrophils from wild-type mice, indicating that the neutrophils were less functional in Stat2-/- mice. Cytochrome bd-II oxidase-mediated respiration enhances S. Typhimurium fitness in wild-type mice, while in Stat2-/- deficiency, this respiratory pathway did not provide a fitness advantage. Furthermore, luminal expansion of S. Typhimurium in wild-type mice was blunted in Stat2-/- mice. Compared to wild-type mice which exhibited a significant perturbation in Bacteroidetes abundance, Stat2-/- mice exhibited significantly less perturbation and higher levels of Bacteroidetes upon S. Typhimurium infection. Our results highlight STAT2 dependent type I IFN mediated inflammation in the gut as a novel mechanism promoting luminal expansion of S. Typhimurium.


Asunto(s)
Disbiosis/inmunología , Gastroenteritis/inmunología , Inflamación/inmunología , Interferón Tipo I/inmunología , Factor de Transcripción STAT2/fisiología , Infecciones por Salmonella/inmunología , Salmonella typhimurium/inmunología , Animales , Células Cultivadas , Disbiosis/metabolismo , Disbiosis/patología , Femenino , Gastroenteritis/metabolismo , Gastroenteritis/microbiología , Gastroenteritis/patología , Inflamación/metabolismo , Inflamación/microbiología , Inflamación/patología , Interferón Tipo I/metabolismo , Intestinos/inmunología , Intestinos/microbiología , Intestinos/patología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Neutrófilos/inmunología , Neutrófilos/metabolismo , Neutrófilos/microbiología , Neutrófilos/patología , Factor de Transcripción STAT1/fisiología , Infecciones por Salmonella/metabolismo , Infecciones por Salmonella/microbiología , Infecciones por Salmonella/patología
20.
Infect Immun ; 87(4)2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30617205

RESUMEN

During Salmonella enterica serovar Typhimurium infection, host inflammation alters the metabolic environment of the gut lumen to favor the outgrowth of the pathogen at the expense of the microbiota. Inflammation-driven changes in host cell metabolism lead to the release of l-lactate and molecular oxygen from the tissue into the gut lumen. Salmonella utilizes lactate as an electron donor in conjunction with oxygen as the terminal electron acceptor to support gut colonization. Here, we investigated transcriptional regulation of the respiratory l-lactate dehydrogenase LldD in vitro and in mouse models of Salmonella infection. The two-component system ArcAB repressed transcription of l-lactate utilization genes under anaerobic conditions in vitro The ArcAB-mediated repression of lldD transcription was relieved under microaerobic conditions. Transcription of lldD was induced by l-lactate but not d-lactate. A mutant lacking the regulatory protein LldR failed to induce lldD transcription in response to l-lactate. Furthermore, the lldR mutant exhibited reduced transcription of l-lactate utilization genes and impaired fitness in murine models of infection. These data provide evidence that the host-derived metabolites oxygen and l-lactate serve as cues for Salmonella to regulate lactate oxidation metabolism on a transcriptional level.


Asunto(s)
Mucosa Intestinal/microbiología , Ácido Láctico/metabolismo , Infecciones por Salmonella/metabolismo , Salmonella typhimurium/crecimiento & desarrollo , Salmonella typhimurium/metabolismo , Animales , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Femenino , Regulación Bacteriana de la Expresión Génica , Humanos , Mucosa Intestinal/metabolismo , L-Lactato Deshidrogenasa/genética , L-Lactato Deshidrogenasa/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Oxígeno/metabolismo , Infecciones por Salmonella/microbiología , Salmonella typhimurium/enzimología , Salmonella typhimurium/genética , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...