Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 34
Filtrar
1.
Trends Immunol ; 45(2): 94-102, 2024 02.
Artículo en Inglés | MEDLINE | ID: mdl-38216387

RESUMEN

The intestinal microbiota has a pervasive influence on mammalian innate immunity fortifying defenses to infection in tissues throughout the host. How intestinal microbes control innate defenses in systemic tissues is, however, poorly defined. In our opinion, there are three core challenges that need addressing to advance our understanding of how the intestinal microbiota controls innate immunity systemically: first, deciphering how signals from intestinal microbes are transmitted to distal tissues; second, unraveling how intestinal microbes prime systemic innate immunity without inducing widespread immunopathology; and third, identifying which intestinal microbes control systemic immunity. Here, we propose answers to these problems which provide a framework for understanding how microbes in the intestine can regulate innate immunity systemically.


Asunto(s)
Microbioma Gastrointestinal , Microbiota , Animales , Humanos , Inmunidad Innata , Intestinos , Mucosa Intestinal , Mamíferos
2.
PLoS Pathog ; 17(1): e1009191, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33465156

RESUMEN

The immunological impact of individual commensal species within the microbiota is poorly understood limiting the use of commensals to treat disease. Here, we systematically profile the immunological fingerprint of commensals from the major phyla in the human intestine (Actinobacteria, Bacteroidetes, Firmicutes and Proteobacteria) to reveal taxonomic patterns in immune activation and use this information to rationally design commensal communities to enhance antibacterial defenses and combat intestinal inflammation. We reveal that Bacteroidetes and Firmicutes have distinct effects on intestinal immunity by differentially inducing primary and secondary response genes. Within these phyla, the immunostimulatory capacity of commensals from the Bacteroidia class (Bacteroidetes phyla) reflects their robustness of TLR4 activation and Bacteroidia communities rely solely on this receptor for their effects on intestinal immunity. By contrast, within the Clostridia class (Firmicutes phyla) it reflects the degree of TLR2 and TLR4 activation, and communities of Clostridia signal via both of these receptors to exert their effects on intestinal immunity. By analyzing the receptors, intracellular signaling components and transcription factors that are engaged by different commensal species, we identify canonical NF-κB signaling as a critical rheostat which grades the degree of immune stimulation commensals elicit. Guided by this immunological analysis, we constructed a cross-phylum consortium of commensals (Bacteroides uniformis, Bacteroides ovatus, Peptostreptococcus anaerobius and Clostridium histolyticum) which enhances innate TLR, IL6 and macrophages-dependent defenses against intestinal colonization by vancomycin resistant Enterococci, and fortifies mucosal barrier function during pathological intestinal inflammation through the same pathway. Critically, the setpoint of intestinal immunity established by this consortium is calibrated by canonical NF-κB signaling. Thus, by profiling the immunological impact of major human commensal species our work paves the way for rational microbiota reengineering to protect against antibiotic resistant infections and to treat intestinal inflammation.


Asunto(s)
Bacterias/inmunología , Inflamación/prevención & control , Enfermedades Intestinales/prevención & control , Mucosa Intestinal/inmunología , Animales , Bacterias/clasificación , Bacterias/metabolismo , Femenino , Humanos , Inflamación/inmunología , Inflamación/microbiología , Enfermedades Intestinales/inmunología , Enfermedades Intestinales/microbiología , Mucosa Intestinal/metabolismo , Mucosa Intestinal/microbiología , Masculino , Ratones Endogámicos C57BL , FN-kappa B/genética , FN-kappa B/metabolismo , Filogenia , Receptores Toll-Like/genética , Receptores Toll-Like/metabolismo
4.
Genes Immun ; 22(5-6): 255-267, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-33947987

RESUMEN

Colonization by the microbiota provides one of our most effective barriers against infection by pathogenic microbes. The microbiota protects against infection by priming immune defenses, by metabolic exclusion of pathogens from their preferred niches, and through direct antimicrobial antagonism. Disruption of the microbiota, especially by antibiotics, is a major risk factor for bacterial pathogen colonization. Restoration of the microbiota through microbiota transplantation has been shown to be an effective way to reduce pathogen burden in the intestine but comes with a number of drawbacks, including the possibility of transferring other pathogens into the host, lack of standardization, and potential disruption to host metabolism. More refined methods to exploit the power of the microbiota would allow us to utilize its protective power without the drawbacks of fecal microbiota transplantation. To achieve this requires detailed understanding of which members of the microbiota protect against specific pathogens and the mechanistic basis for their effects. In this review, we will discuss the clinical and experimental evidence that has begun to reveal which members of the microbiota protect against some of the most troublesome antibiotic-resistant pathogens: Klebsiella pneumoniae, vancomycin-resistant enterococci, and Clostridioides difficile.


Asunto(s)
Clostridioides difficile , Microbiota , Antibacterianos/farmacología , Antibacterianos/uso terapéutico , Klebsiella pneumoniae
5.
PLoS Pathog ; 15(12): e1008006, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31830135

RESUMEN

Shigella flexneri is historically regarded as the primary agent of bacillary dysentery, yet the closely-related Shigella sonnei is replacing S. flexneri, especially in developing countries. The underlying reasons for this dramatic shift are mostly unknown. Using a zebrafish (Danio rerio) model of Shigella infection, we discover that S. sonnei is more virulent than S. flexneri in vivo. Whole animal dual-RNAseq and testing of bacterial mutants suggest that S. sonnei virulence depends on its O-antigen oligosaccharide (which is unique among Shigella species). We show in vivo using zebrafish and ex vivo using human neutrophils that S. sonnei O-antigen can mediate neutrophil tolerance. Consistent with this, we demonstrate that O-antigen enables S. sonnei to resist phagolysosome acidification and promotes neutrophil cell death. Chemical inhibition or promotion of phagolysosome maturation respectively decreases and increases neutrophil control of S. sonnei and zebrafish survival. Strikingly, larvae primed with a sublethal dose of S. sonnei are protected against a secondary lethal dose of S. sonnei in an O-antigen-dependent manner, indicating that exposure to O-antigen can train the innate immune system against S. sonnei. Collectively, these findings reveal O-antigen as an important therapeutic target against bacillary dysentery, and may explain the rapidly increasing S. sonnei burden in developing countries.


Asunto(s)
Neutrófilos/inmunología , Antígenos O/inmunología , Shigella sonnei/inmunología , Shigella sonnei/patogenicidad , Virulencia/inmunología , Animales , Disentería Bacilar , Humanos , Pez Cebra
6.
PLoS Pathog ; 14(5): e1007052, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29750817

RESUMEN

To survive diverse host environments, the human pathogen Streptococcus pneumoniae must prevent its self-produced, extremely high levels of peroxide from reacting with intracellular iron. However, the regulatory mechanism(s) by which the pneumococcus accomplishes this balance remains largely enigmatic, as this pathogen and other related streptococci lack all known redox-sensing transcription factors. Here we describe a two-component-derived response regulator, RitR, as the archetype for a novel family of redox sensors in a subset of streptococcal species. We show that RitR works to both repress iron transport and enable nasopharyngeal colonization through a mechanism that exploits a single cysteine (Cys128) redox switch located within its linker domain. Biochemical experiments and phylogenetics reveal that RitR has diverged from the canonical two-component virulence regulator CovR to instead dimerize and bind DNA only upon Cys128 oxidation in air-rich environments. Atomic structures show that Cys128 oxidation initiates a "helical unravelling" of the RitR linker region, suggesting a mechanism by which the DNA-binding domain is then released to interact with its cognate regulatory DNA. Expanded computational studies indicate this mechanism could be shared by many microbial species outside the streptococcus genus.


Asunto(s)
Proteínas Represoras/metabolismo , Streptococcus pneumoniae/metabolismo , Proteínas Bacterianas/metabolismo , Cisteína/metabolismo , Regulación Bacteriana de la Expresión Génica/genética , Peróxido de Hidrógeno/metabolismo , Transporte Iónico/fisiología , Hierro/metabolismo , Oxidación-Reducción , Elementos de Respuesta/fisiología , Transducción de Señal , Streptococcus pneumoniae/genética , Streptococcus pneumoniae/patogenicidad , Streptococcus pyogenes/genética , Streptococcus pyogenes/metabolismo , Factores de Transcripción/metabolismo , Factores de Transcripción/fisiología , Virulencia/genética
7.
Gut ; 68(10): 1791-1800, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-30816855

RESUMEN

OBJECTIVE: Faecal microbiota transplant (FMT) effectively treats recurrent Clostridioides difficile infection (rCDI), but its mechanisms of action remain poorly defined. Certain bile acids affect C. difficile germination or vegetative growth. We hypothesised that loss of gut microbiota-derived bile salt hydrolases (BSHs) predisposes to CDI by perturbing gut bile metabolism, and that BSH restitution is a key mediator of FMT's efficacy in treating the condition. DESIGN: Using stool collected from patients and donors pre-FMT/post-FMT for rCDI, we performed 16S rRNA gene sequencing, ultra performance liquid chromatography mass spectrometry (UPLC-MS) bile acid profiling, BSH activity measurement, and qPCR of bsh/baiCD genes involved in bile metabolism. Human data were validated in C. difficile batch cultures and a C57BL/6 mouse model of rCDI. RESULTS: From metataxonomics, pre-FMT stool demonstrated a reduced proportion of BSH-producing bacterial species compared with donors/post-FMT. Pre-FMT stool was enriched in taurocholic acid (TCA, a potent C. difficile germinant); TCA levels negatively correlated with key bacterial genera containing BSH-producing organisms. Post-FMT samples demonstrated recovered BSH activity and bsh/baiCD gene copy number compared with pretreatment (p<0.05). In batch cultures, supernatant from engineered bsh-expressing E. coli and naturally BSH-producing organisms (Bacteroides ovatus, Collinsella aerofaciens, Bacteroides vulgatus and Blautia obeum) reduced TCA-mediated C. difficile germination relative to culture supernatant of wild-type (BSH-negative) E. coli. C. difficile total viable counts were ~70% reduced in an rCDI mouse model after administration of E. coli expressing highly active BSH relative to mice administered BSH-negative E. coli (p<0.05). CONCLUSION: Restoration of gut BSH functionality contributes to the efficacy of FMT in treating rCDI.


Asunto(s)
Amidohidrolasas/farmacología , Clostridioides difficile/genética , Infecciones por Clostridium/terapia , ADN Bacteriano/genética , Trasplante de Microbiota Fecal/métodos , Microbioma Gastrointestinal/fisiología , Animales , Infecciones por Clostridium/microbiología , Modelos Animales de Enfermedad , Femenino , Ácido Glicocólico , Humanos , Ratones , Ratones Endogámicos C57BL , Recurrencia , Espectrometría de Masas en Tándem
8.
Gastroenterology ; 155(5): 1495-1507.e15, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-30025704

RESUMEN

BACKGROUND & AIMS: Fecal microbiota transplantation (FMT) is effective for treating recurrent Clostridioides difficile infection (CDI), but there are concerns about its long-term safety. Understanding the mechanisms of the effects of FMT could help us design safer, targeted therapies. We aimed to identify microbial metabolites that are important for C difficile growth. METHODS: We used a CDI chemostat model as a tool to study the effects of FMT in vitro. The following analyses were performed: C difficile plate counts, 16S rRNA gene sequencing, proton nuclear magnetic resonance spectroscopy, and ultra-performance liquid chromatography and mass spectrometry bile acid profiling. FMT mixtures were prepared using fresh fecal samples provided by donors enrolled in an FMT program in the United Kingdom. Results from chemostat experiments were validated using human stool samples, C difficile batch cultures, and C57BL/6 mice with CDI. Human stool samples were collected from 16 patients with recurrent CDI and healthy donors (n = 5) participating in an FMT trial in Canada. RESULTS: In the CDI chemostat model, clindamycin decreased valerate and deoxycholic acid concentrations and increased C difficile total viable counts and valerate precursors, taurocholic acid, and succinate concentrations. After we stopped adding clindamycin, levels of bile acids and succinate recovered, whereas levels of valerate and valerate precursors did not. In the CDI chemostat model, FMT increased valerate concentrations and decreased C difficile total viable counts (94% decrease), spore counts (86% decrease), and valerate precursor concentrations; concentrations of bile acids were unchanged. In stool samples from patients with CDI, valerate was depleted before FMT but restored after FMT. Clostridioides difficile batch cultures confirmed that valerate decreased vegetative growth, and that taurocholic acid was required for germination but had no effect on vegetative growth. Clostridioides difficile total viable counts were decreased by 95% in mice with CDI given glycerol trivalerate compared with phosphate buffered saline. CONCLUSIONS: We identified valerate as a metabolite that is depleted with clindamycin and only recovered with FMT. Valerate is a target for a rationally designed recurrent CDI therapy.


Asunto(s)
Clostridioides difficile/efectos de los fármacos , Infecciones por Clostridium/terapia , Microbioma Gastrointestinal , Valeratos/farmacología , Animales , Ácidos y Sales Biliares/análisis , Cromatografía Líquida de Alta Presión , Clindamicina/farmacología , Clostridioides difficile/crecimiento & desarrollo , Heces/química , Femenino , Cromatografía de Gases y Espectrometría de Masas , Espectroscopía de Resonancia Magnética , Ratones Endogámicos C57BL , Esporas Bacterianas , Triglicéridos/uso terapéutico , Valeratos/metabolismo
9.
Blood ; 127(20): 2460-71, 2016 05 19.
Artículo en Inglés | MEDLINE | ID: mdl-26989200

RESUMEN

Maintenance of myeloid cell homeostasis requires continuous turnover of phagocytes from the bloodstream, yet whether environmental signals influence phagocyte longevity in the absence of inflammation remains unknown. Here, we show that the gut microbiota regulates the steady-state cellular lifespan of neutrophils and inflammatory monocytes, the 2 most abundant circulating myeloid cells and key contributors to inflammatory responses. Treatment of mice with broad-spectrum antibiotics, or with the gut-restricted aminoglycoside neomycin alone, accelerated phagocyte turnover and increased the rates of their spontaneous apoptosis. Metagenomic analyses revealed that neomycin altered the abundance of intestinal bacteria bearing γ-d-glutamyl-meso-diaminopimelic acid, a ligand for the intracellular peptidoglycan sensor Nod1. Accordingly, signaling through Nod1 was both necessary and sufficient to mediate the stimulatory influence of the flora on myeloid cell longevity. Stimulation of Nod1 signaling increased the frequency of lymphocytes in the murine intestine producing the proinflammatory cytokine interleukin 17A (IL-17A), and liberation of IL-17A was required for transmission of Nod1-dependent signals to circulating phagocytes. Together, these results define a mechanism through which intestinal microbes govern a central component of myeloid homeostasis and suggest perturbations of commensal communities can influence steady-state regulation of cell fate.


Asunto(s)
Microbioma Gastrointestinal/fisiología , Homeostasis , Peptidoglicano/farmacología , Fagocitos/citología , Traslado Adoptivo , Animales , Animales Congénicos , Antibacterianos/farmacología , Apoptosis/efectos de los fármacos , Supervivencia Celular/fisiología , Ácido Diaminopimélico/análogos & derivados , Ácido Diaminopimélico/farmacología , Femenino , Microbioma Gastrointestinal/efectos de los fármacos , Vida Libre de Gérmenes , Interleucina-17/fisiología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Monocitos/citología , Neutrófilos/citología , Proteína Adaptadora de Señalización NOD1/deficiencia , Proteína Adaptadora de Señalización NOD1/fisiología , Proteína Adaptadora de Señalización NOD2/deficiencia , Proteína Adaptadora de Señalización NOD2/fisiología , Fagocitos/efectos de los fármacos , Receptor Toll-Like 2/deficiencia , Receptor Toll-Like 2/fisiología , Receptor Toll-Like 4/deficiencia , Receptor Toll-Like 4/fisiología
10.
Immunology ; 150(1): 1-6, 2017 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-27311879

RESUMEN

The skin and mucosal epithelia of humans and other mammals are permanently colonized by large microbial communities (the microbiota). Due to this life-long association with the microbiota, these microbes have an extensive influence over the physiology of their host organism. It is now becoming apparent that nearly all tissues and organ systems, whether in direct contact with the microbiota or in deeper host sites, are under microbial influence. The immune system is perhaps the most profoundly affected, with the microbiota programming both its innate and adaptive arms. The regulation of immunity by the microbiota helps to protect the host against intestinal and extra-intestinal infection by many classes of pathogen. In this review, we will discuss the experimental evidence supporting a role for the microbiota in regulating host defences to extra-intestinal infection, draw together common mechanistic themes, including the central role of pattern recognition receptors, and outline outstanding questions that need to be answered.


Asunto(s)
Inmunidad Adaptativa , Bacterias/inmunología , Inmunidad Mucosa , Infecciones/inmunología , Microbiota/inmunología , Animales , Homeostasis , Humanos , Sistema Inmunológico , Infecciones/microbiología , Receptores de Reconocimiento de Patrones/metabolismo
11.
Immunol Rev ; 243(1): 9-25, 2011 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-21884164

RESUMEN

Initial recognition of bacteria by the innate immune system is thought to occur primarily by germline-encoded pattern recognition receptors (PRRs). These receptors are present in multiple compartments of host cells and are thus capable of surveying both the intracellular and extracellular milieu for bacteria. It has generally been presumed that the cellular location of these receptors dictates what type of bacteria they respond to: extracellular bacteria being recognized by cell surface receptors, such as certain Toll-like receptors, and bacteria that are capable of breaching the plasma membrane and entering the cytoplasm, being sensed by cytoplasmic receptors, including the Nod-like receptors (NLRs). Increasingly, it is becoming apparent that this is a false dichotomy and that extracellular bacteria can be sensed by cytoplasmic PRRs and this is crucial for controlling the levels of these bacteria. In this review, we discuss the role of two NLRs, Nod1 and Nod2, in the recognition of and response to extracellular bacteria.


Asunto(s)
Infecciones Bacterianas/inmunología , Espacio Extracelular , Espacio Intracelular , Proteína Adaptadora de Señalización NOD1/inmunología , Proteína Adaptadora de Señalización NOD2/inmunología , Animales , Espacio Extracelular/inmunología , Espacio Extracelular/microbiología , Interacciones Huésped-Patógeno , Humanos , Inmunidad Innata , Espacio Intracelular/inmunología , Espacio Intracelular/microbiología , Proteína Adaptadora de Señalización NOD1/metabolismo , Proteína Adaptadora de Señalización NOD2/metabolismo , Transducción de Señal/inmunología
12.
Infect Immun ; 82(11): 4596-606, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25135683

RESUMEN

The commensal microbiota is a major regulator of the immune system. The majority of commensal bacteria inhabit the gastrointestinal tract and are known to regulate local mucosal defenses against intestinal pathogens. There is growing appreciation that the commensal microbiota also regulates immune responses at extraintestinal sites. Currently, however, it is unclear how this influences host defenses against bacterial infection outside the intestine. Microbiota depletion caused significant defects in the early innate response to lung infection by the major human pathogen Klebsiella pneumoniae. After microbiota depletion, early clearance of K. pneumoniae was impaired, and this could be rescued by administration of bacterial Nod-like receptor (NLR) ligands (the NOD1 ligand MurNAcTri(DAP) and NOD2 ligand muramyl dipeptide [MDP]) but not bacterial Toll-like receptor (TLR) ligands. Importantly, NLR ligands from the gastrointestinal, but not upper respiratory, tract rescued host defenses in the lung. Defects in early innate immunity were found to be due to reduced reactive oxygen species-mediated killing of bacteria by alveolar macrophages. These data show that bacterial signals from the intestine have a profound influence on establishing the levels of antibacterial defenses in distal tissues.


Asunto(s)
Acetilmuramil-Alanil-Isoglutamina/farmacología , Inmunidad Innata , Infecciones por Klebsiella/inmunología , Enfermedades Pulmonares/inmunología , Proteínas Adaptadoras de Señalización NOD/metabolismo , Animales , Regulación de la Expresión Génica/inmunología , Intestinos/microbiología , Infecciones por Klebsiella/microbiología , Klebsiella pneumoniae/inmunología , Ligandos , Enfermedades Pulmonares/microbiología , Macrófagos Alveolares/fisiología , Ratones , Ratones Endogámicos C57BL , Especies Reactivas de Oxígeno , Simbiosis
13.
Chem Sci ; 14(9): 2336-2341, 2023 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-36873831

RESUMEN

Many studies have shown chemistry proceeds differently in small volumes compared to bulk phases. However, few studies exist elucidating spontaneous means by which small volumes can form in Nature. Such studies are critical in understanding the formation of life in microcompartments. In this study, we track in real-time the coalescence of two or more water microdroplets adsorbed on an electrified surface in a 1,2-dichloroethane continuous phase by electrogenerated chemiluminescence (ECL) imaging, uncovering the spontaneous generation of multiple emulsions inside the resulting water droplets. During the fusion of adsorbed water droplets with each other on the electrode surface, volumes of organic and water phases are entrapped in between and detected respectively as ECL not-emitting and emitting regions. The diameter of those confined environments inside the water droplets can be less than a micrometer, as described by scanning electron microscopy data. This study adds a new mechanism for the generation of micro- and nano-emulsions and provides insight into confinement techniques under abiotic conditions as well as new potential strategies in microfluidic devices.

14.
Chem Sci ; 14(27): 7595, 2023 Jul 12.
Artículo en Inglés | MEDLINE | ID: mdl-37449081

RESUMEN

[This corrects the article DOI: 10.1039/D2SC06553C.].

15.
Nat Commun ; 14(1): 705, 2023 Feb 09.
Artículo en Inglés | MEDLINE | ID: mdl-36759528

RESUMEN

Emulsions are critical across a broad spectrum of industries. Unfortunately, emulsification requires a significant driving force for droplet dispersion. Here, we demonstrate a mechanism of spontaneous droplet formation (emulsification), where the interfacial solute flux promotes droplet formation at the liquid-liquid interface when a phase transfer agent is present. We have termed this phenomenon fluxification. For example, when HAuCl4 is dissolved in an aqueous phase and [NBu4][ClO4] is dissolved in an oil phase, emulsion droplets (both water-in-oil and oil-in-water) can be observed at the interface for various oil phases (1,2-dichloroethane, dichloromethane, chloroform, and nitrobenzene). Emulsification occurs when AuCl4- interacts with NBu4+, a well-known phase-transfer agent, and transfers into the oil phase while ClO4- transfers into the aqueous phase to maintain electroneutrality. The phase transfer of SCN- and Fe(CN)63- also produce droplets. We propose a microscopic mechanism of droplet formation and discuss design principles by tuning experimental parameters.

16.
Cell Host Microbe ; 31(9): 1433-1449.e9, 2023 09 13.
Artículo en Inglés | MEDLINE | ID: mdl-37582375

RESUMEN

The intestinal microbiota regulates immunity across organ systems. Which symbionts control systemic immunity, the mechanisms they use, and how they avoid widespread inflammatory damage are unclear. We uncover host tolerance and resistance mechanisms that allow Firmicutes from the human microbiota to control systemic immunity without inducing immunopathology. Intestinal processing releases Firmicute glycoconjugates that disseminate, resulting in release of cytokine IL-34 that stimulates macrophages and enhances defenses against pneumonia, sepsis, and meningitis. Despite systemic penetration of Firmicutes, immune homeostasis is maintained through feedback control whereby IL-34-mediated mTORC1 activation in macrophages clears polymeric glycoconjugates from peripheral tissues. Smaller glycoconjugates evading this clearance mechanism are tolerated through sequestration by albumin, which acts as an inflammatory buffer constraining their immunological impact. Without these resistance and tolerance mechanisms, Firmicutes drive catastrophic organ damage and cachexia via IL-1ß. This reveals how Firmicutes are safely assimilated into systemic immunity to protect against infection without threatening host viability.


Asunto(s)
Firmicutes , Microbiota , Humanos , Simbiosis , Tolerancia Inmunológica , Citocinas , Interleucinas , Inmunidad Innata
17.
Nat Commun ; 14(1): 5094, 2023 08 22.
Artículo en Inglés | MEDLINE | ID: mdl-37607936

RESUMEN

The intestine is the primary colonisation site for carbapenem-resistant Enterobacteriaceae (CRE) and serves as a reservoir of CRE that cause invasive infections (e.g. bloodstream infections). Broad-spectrum antibiotics disrupt colonisation resistance mediated by the gut microbiota, promoting the expansion of CRE within the intestine. Here, we show that antibiotic-induced reduction of gut microbial populations leads to an enrichment of nutrients and depletion of inhibitory metabolites, which enhances CRE growth. Antibiotics decrease the abundance of gut commensals (including Bifidobacteriaceae and Bacteroidales) in ex vivo cultures of human faecal microbiota; this is accompanied by depletion of microbial metabolites and enrichment of nutrients. We measure the nutrient utilisation abilities, nutrient preferences, and metabolite inhibition susceptibilities of several CRE strains. We find that CRE can use the nutrients (enriched after antibiotic treatment) as carbon and nitrogen sources for growth. These nutrients also increase in faeces from antibiotic-treated mice and decrease following intestinal colonisation with carbapenem-resistant Escherichia coli. Furthermore, certain microbial metabolites (depleted upon antibiotic treatment) inhibit CRE growth. Our results show that killing gut commensals with antibiotics facilitates CRE colonisation by enriching nutrients and depleting inhibitory microbial metabolites.


Asunto(s)
Actinobacteria , Enterobacteriaceae Resistentes a los Carbapenémicos , Neoplasias Intestinales , Humanos , Animales , Ratones , Antibacterianos/farmacología , Bacteroidetes , Escherichia coli , Nutrientes
18.
J Phys Chem Lett ; 13(15): 3338-3341, 2022 Apr 21.
Artículo en Inglés | MEDLINE | ID: mdl-35394768

RESUMEN

Chemistry in confined volumes, such as aqueous droplets, is different from bulk, continuous water. However, few techniques are available to probe interfacial reactivity in complex, multiphase environments. Here, we demonstrate preferential electroreduction at the oil|water|conductor (three-phase) interface. Electrodeposition of cobalt and nickel results in ringlike structures that can be characterized with tens of nanometers precision in scanning electron microscopy and energy dispersive X-ray spectroscopy. To demonstrate the generalizability of these observations, we show that electroreduction of resazurin to fluorescent resorufin occurs preferentially at the three-phase boundary. The preferential electroreduction does not depend on droplet geometry. These results, grounded in three-phase boundary reactivity, are highly important across all fields of chemistry and biology because they highlight how the interface can change chemistry in unexpected ways.


Asunto(s)
Agua , Agua/química
19.
J Clin Invest ; 132(8)2022 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-35239513

RESUMEN

The respiratory tract surface is protected from inhaled pathogens by a secreted layer of mucus rich in mucin glycoproteins. Abnormal mucus accumulation is a cardinal feature of chronic respiratory diseases, but the relationship between mucus and pathogens during exacerbations is poorly understood. We identified elevations in airway mucin 5AC (MUC5AC) and MUC5B concentrations during spontaneous and experimentally induced chronic obstructive pulmonary disease (COPD) exacerbations. MUC5AC was more sensitive to changes in expression during exacerbation and was therefore more predictably associated with viral load, inflammation, symptom severity, decrements in lung function, and secondary bacterial infections. MUC5AC was functionally related to inflammation, as Muc5ac-deficient (Muc5ac-/-) mice had attenuated RV-induced (RV-induced) airway inflammation, and exogenous MUC5AC glycoprotein administration augmented inflammatory responses and increased the release of extracellular adenosine triphosphate (ATP) in mice and human airway epithelial cell cultures. Hydrolysis of ATP suppressed MUC5AC augmentation of RV-induced inflammation in mice. Therapeutic suppression of mucin production using an EGFR antagonist ameliorated immunopathology in a mouse COPD exacerbation model. The coordinated virus induction of MUC5AC and MUC5B expression suggests that non-Th2 mechanisms trigger mucin hypersecretion during exacerbations. Our data identified a proinflammatory role for MUC5AC during viral infection and suggest that MUC5AC inhibition may ameliorate COPD exacerbations.


Asunto(s)
Mucina 5AC , Enfermedad Pulmonar Obstructiva Crónica , Adenosina Trifosfato/metabolismo , Animales , Modelos Animales de Enfermedad , Humanos , Inflamación/metabolismo , Ratones , Mucina 5AC/genética , Mucina 5AC/metabolismo , Mucina 5B/genética , Mucina 5B/metabolismo , Moco/metabolismo , Enfermedad Pulmonar Obstructiva Crónica/metabolismo , Enfermedad Pulmonar Obstructiva Crónica/virología , Mucosa Respiratoria/metabolismo , Mucosa Respiratoria/patología
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda