Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 73
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Cell ; 169(3): 547-558.e15, 2017 04 20.
Artículo en Inglés | MEDLINE | ID: mdl-28431252

RESUMEN

The gut microbiota is implicated in numerous aspects of health and disease, but dissecting these connections is challenging because genetic tools for gut anaerobes are limited. Inducible promoters are particularly valuable tools because these platforms allow real-time analysis of the contribution of microbiome gene products to community assembly, host physiology, and disease. We developed a panel of tunable expression platforms for the prominent genus Bacteroides in which gene expression is controlled by a synthetic inducer. In the absence of inducer, promoter activity is fully repressed; addition of inducer rapidly increases gene expression by four to five orders of magnitude. Because the inducer is absent in mice and their diets, Bacteroides gene expression inside the gut can be modulated by providing the inducer in drinking water. We use this system to measure the dynamic relationship between commensal sialidase activity and liberation of mucosal sialic acid, a receptor and nutrient for pathogens. VIDEO ABSTRACT.


Asunto(s)
Bacteroides/genética , Microbioma Gastrointestinal , Ingeniería Genética/métodos , Animales , Bacteroides/clasificación , Expresión Génica , Humanos , Ratones , Neuraminidasa/metabolismo , Regiones Promotoras Genéticas
2.
Cell ; 158(5): 1000-1010, 2014 Aug 28.
Artículo en Inglés | MEDLINE | ID: mdl-25171403

RESUMEN

Specific members of the intestinal microbiota dramatically affect inflammatory bowel disease (IBD) in mice. In humans, however, identifying bacteria that preferentially affect disease susceptibility and severity remains a major challenge. Here, we used flow-cytometry-based bacterial cell sorting and 16S sequencing to characterize taxa-specific coating of the intestinal microbiota with immunoglobulin A (IgA-SEQ) and show that high IgA coating uniquely identifies colitogenic intestinal bacteria in a mouse model of microbiota-driven colitis. We then used IgA-SEQ and extensive anaerobic culturing of fecal bacteria from IBD patients to create personalized disease-associated gut microbiota culture collections with predefined levels of IgA coating. Using these collections, we found that intestinal bacteria selected on the basis of high coating with IgA conferred dramatic susceptibility to colitis in germ-free mice. Thus, our studies suggest that IgA coating identifies inflammatory commensals that preferentially drive intestinal disease. Targeted elimination of such bacteria may reduce, reverse, or even prevent disease development.


Asunto(s)
Colitis Ulcerosa/inmunología , Enfermedad de Crohn/inmunología , Inmunoglobulina A/inmunología , Microbiota , Animales , Colitis Ulcerosa/microbiología , Colitis Ulcerosa/patología , Enfermedad de Crohn/microbiología , Enfermedad de Crohn/patología , ADN Bacteriano/análisis , Disbiosis/inmunología , Disbiosis/microbiología , Humanos , Inflamasomas/inmunología , Inflamación/inmunología , Inflamación/microbiología , Intestinos/inmunología , Intestinos/microbiología , Ratones , Ratones Endogámicos C57BL , ARN Ribosómico 16S/análisis , Organismos Libres de Patógenos Específicos
3.
EMBO J ; 42(2): e112372, 2023 01 16.
Artículo en Inglés | MEDLINE | ID: mdl-36472247

RESUMEN

Protein synthesis is crucial for cell growth and survival yet one of the most energy-consuming cellular processes. How, then, do cells sustain protein synthesis under starvation conditions when energy is limited? To accelerate the translocation of mRNA-tRNAs through the ribosome, bacterial elongation factor G (EF-G) hydrolyzes energy-rich guanosine triphosphate (GTP) for every amino acid incorporated into a protein. Here, we identify an EF-G paralog-EF-G2-that supports translocation without hydrolyzing GTP in the gut commensal bacterium Bacteroides thetaiotaomicron. EF-G2's singular ability to sustain protein synthesis, albeit at slow rates, is crucial for bacterial gut colonization. EF-G2 is ~10-fold more abundant than canonical EF-G1 in bacteria harvested from murine ceca and, unlike EF-G1, specifically accumulates during carbon starvation. Moreover, we uncover a 26-residue region unique to EF-G2 that is essential for protein synthesis, EF-G2 dissociation from the ribosome, and responsible for the absence of GTPase activity. Our findings reveal how cells curb energy consumption while maintaining protein synthesis to advance fitness in nutrient-fluctuating environments.


Asunto(s)
Bacteroides , Factor G de Elongación Peptídica , Animales , Ratones , Bacteroides/genética , Bacteroides/metabolismo , Guanosina Trifosfato/metabolismo , Hidrólisis , Factor G de Elongación Peptídica/genética , Factor G de Elongación Peptídica/química , Ribosomas/metabolismo , ARN de Transferencia/metabolismo
4.
Nat Rev Genet ; 21(9): 526-540, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32533119

RESUMEN

It has been 10 years since the introduction of modern transposon-insertion sequencing (TIS) methods, which combine genome-wide transposon mutagenesis with high-throughput sequencing to estimate the fitness contribution or essentiality of each genetic component in a bacterial genome. Four TIS variations were published in 2009: transposon sequencing (Tn-Seq), transposon-directed insertion site sequencing (TraDIS), insertion sequencing (INSeq) and high-throughput insertion tracking by deep sequencing (HITS). TIS has since become an important tool for molecular microbiologists, being one of the few genome-wide techniques that directly links phenotype to genotype and ultimately can assign gene function. In this Review, we discuss the recent applications of TIS to answer overarching biological questions. We explore emerging and multidisciplinary methods that build on TIS, with an eye towards future applications.


Asunto(s)
Elementos Transponibles de ADN/genética , Secuenciación de Nucleótidos de Alto Rendimiento , Análisis de Secuencia de ADN , Animales , Humanos
5.
Nature ; 570(7762): 462-467, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-31158845

RESUMEN

Individuals vary widely in their responses to medicinal drugs, which can be dangerous and expensive owing to treatment delays and adverse effects. Although increasing evidence implicates the gut microbiome in this variability, the molecular mechanisms involved remain largely unknown. Here we show, by measuring the ability of 76 human gut bacteria from diverse clades to metabolize 271 orally administered drugs, that many drugs are chemically modified by microorganisms. We combined high-throughput genetic analyses with mass spectrometry to systematically identify microbial gene products that metabolize drugs. These microbiome-encoded enzymes can directly and substantially affect intestinal and systemic drug metabolism in mice, and can explain the drug-metabolizing activities of human gut bacteria and communities on the basis of their genomic contents. These causal links between the gene content and metabolic activities of the microbiota connect interpersonal variability in microbiomes to interpersonal differences in drug metabolism, which has implications for medical therapy and drug development across multiple disease indications.


Asunto(s)
Bacterias/genética , Bacterias/metabolismo , Microbioma Gastrointestinal/genética , Preparaciones Farmacéuticas/metabolismo , Animales , Bacterias/clasificación , Bacterias/enzimología , Bacteroides thetaiotaomicron/enzimología , Bacteroides thetaiotaomicron/genética , Bacteroides thetaiotaomicron/metabolismo , Diltiazem/metabolismo , Femenino , Microbioma Gastrointestinal/fisiología , Genoma Bacteriano/genética , Vida Libre de Gérmenes , Humanos , Masculino , Ratones , Preparaciones Farmacéuticas/administración & dosificación , Especificidad por Sustrato
6.
J Biol Chem ; 299(12): 105363, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-37863262

RESUMEN

Metformin is among the most prescribed medications worldwide and the first-line therapy for type 2 diabetes. However, gastrointestinal side effects are common and can be dose limiting. The total daily metformin dose frequently reaches several grams, and poor absorption results in high intestinal drug concentrations. Here, we report that metformin inhibits the activity of enteropeptidase and other digestive enzymes at drug concentrations predicted to occur in the human duodenum. Treatment of mouse gastrointestinal tissue with metformin reduces enteropeptidase activity; further, metformin-treated mice exhibit reduced enteropeptidase activity, reduced trypsin activity, and impaired protein digestion within the intestinal lumen. These results indicate that metformin-induced protein maldigestion could contribute to the gastrointestinal side effects and other impacts of this widely used drug.


Asunto(s)
Enteropeptidasa , Metformina , Proteolisis , Animales , Humanos , Ratones , Diabetes Mellitus Tipo 2/tratamiento farmacológico , Enteropeptidasa/metabolismo , Metformina/efectos adversos , Metformina/farmacología , Metformina/uso terapéutico , Proteolisis/efectos de los fármacos , Activación Enzimática/efectos de los fármacos , Tracto Gastrointestinal/enzimología , Tripsina/metabolismo , Hipoglucemiantes/farmacología , Hipoglucemiantes/uso terapéutico
7.
Immunity ; 43(5): 909-22, 2015 Nov 17.
Artículo en Inglés | MEDLINE | ID: mdl-26546281

RESUMEN

Microbe-induced receptor trafficking has emerged as an essential means to promote innate immune signal transduction. Upon detection of bacterial lipopolysaccharides (LPS), CD14 induces an inflammatory endocytosis pathway that delivers Toll-like receptor 4 (TLR4) to endosomes. Although several regulators of CD14-dependent TLR4 endocytosis have been identified, the cargo-selection mechanism during this process remains unknown. We reveal that, in contrast to classic cytosolic interactions that promoted the endocytosis of transmembrane receptors, TLR4 was selected as cargo for inflammatory endocytosis entirely through extracellular interactions. Mechanistically, the extracellular protein MD-2 bound to and dimerized TLR4 in order to promote this endocytic event. Our analysis of LPS variants from human pathogens and gut commensals revealed a common mechanism by which bacteria prevent inflammatory endocytosis. We suggest that evasion of CD14-dependent endocytosis is an attribute that transcends the concept of pathogenesis and might be a fundamental feature of bacteria that inhabit eukaryotic hosts.


Asunto(s)
Bacterias/inmunología , Endocitosis/inmunología , Evasión Inmune/inmunología , Receptores de Lipopolisacáridos/metabolismo , Receptor Toll-Like 4/metabolismo , Células Cultivadas , Humanos , Inflamación/inmunología , Lipopolisacáridos/inmunología , Antígeno 96 de los Linfocitos/inmunología , Transporte de Proteínas/inmunología , Transducción de Señal/inmunología , Receptor Toll-Like 4/inmunología
8.
Nature ; 534(7606): 213-7, 2016 06 09.
Artículo en Inglés | MEDLINE | ID: mdl-27279214

RESUMEN

Obesity, insulin resistance and the metabolic syndrome are associated with changes to the gut microbiota; however, the mechanism by which modifications to the gut microbiota might lead to these conditions is unknown. Here we show that increased production of acetate by an altered gut microbiota in rodents leads to activation of the parasympathetic nervous system, which, in turn, promotes increased glucose-stimulated insulin secretion, increased ghrelin secretion, hyperphagia, obesity and related sequelae. Together, these findings identify increased acetate production resulting from a nutrient-gut microbiota interaction and subsequent parasympathetic activation as possible therapeutic targets for obesity.


Asunto(s)
Acetatos/metabolismo , Encéfalo/fisiología , Microbioma Gastrointestinal/fisiología , Células Secretoras de Insulina/metabolismo , Síndrome Metabólico/metabolismo , Animales , Dieta Alta en Grasa , Ghrelina/metabolismo , Glucosa/metabolismo , Hiperfagia/metabolismo , Insulina/metabolismo , Secreción de Insulina , Obesidad/metabolismo , Sistema Nervioso Parasimpático/fisiología , Ratas
9.
Proc Natl Acad Sci U S A ; 116(1): 233-238, 2019 01 02.
Artículo en Inglés | MEDLINE | ID: mdl-30559205

RESUMEN

The composition of the gut microbiota is largely determined by environmental factors including the host diet. Dietary components are believed to influence the composition of the gut microbiota by serving as nutrients to a subset of microbes, thereby favoring their expansion. However, we now report that dietary fructose and glucose, which are prevalent in the Western diet, specifically silence a protein that is necessary for gut colonization, but not for utilization of these sugars, by the human gut commensal Bacteroides thetaiotaomicron Silencing by fructose and glucose requires the 5' leader region of the mRNA specifying the protein, designated Roc for regulator of colonization. Incorporation of the roc leader mRNA in front of a heterologous gene was sufficient for fructose and glucose to turn off expression of the corresponding protein. An engineered strain refractory to Roc silencing by these sugars outcompeted wild-type B. thetaiotaomicron in mice fed a diet rich in glucose and sucrose (a disaccharide composed of glucose and fructose), but not in mice fed a complex polysaccharide-rich diet. Our findings underscore a role for dietary sugars that escape absorption by the host intestine and reach the microbiota: regulation of gut colonization by beneficial microbes independently of supplying nutrients to the microbiota.


Asunto(s)
Proteínas Bacterianas/antagonistas & inhibidores , Bacteroides thetaiotaomicron/efectos de los fármacos , Carbohidratos de la Dieta/farmacología , Azúcares de la Dieta/farmacología , Microbioma Gastrointestinal/efectos de los fármacos , Animales , Proteínas Bacterianas/metabolismo , Fructosa/administración & dosificación , Fructosa/farmacología , Regulación Bacteriana de la Expresión Génica/efectos de los fármacos , Silenciador del Gen/efectos de los fármacos , Glucosa/administración & dosificación , Glucosa/farmacología , Ratones , Polisacáridos/administración & dosificación , Polisacáridos/farmacología , Simbiosis/efectos de los fármacos
10.
J Bacteriol ; 202(3)2020 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-31712278

RESUMEN

Bacteroides is one of the most prominent genera in the human gut microbiome, and study of this bacterial group provides insights into gut microbial ecology and pathogenesis. In this report, we introduce a negative selection system for rapid and efficient allelic exchange in wild Bacteroides species that does not require any alterations to the genetic background or a nutritionally defined culture medium. In this approach, dual antibacterial effectors normally delivered via type VI secretion are targeted to the bacterial periplasm under the control of tightly regulated anhydrotetracycline (aTC)-inducible promoters. Introduction of aTC selects for recombination events producing the desired genetic modification, and the dual effector design allows for broad applicability across strains that may have immunity to one counterselection effector. We demonstrate the utility of this approach across 21 human gut Bacteroides isolates representing diverse species, including strains isolated directly from human donors. We use this system to establish that antimicrobial peptide resistance in Bacteroides vulgatus is determined by the product of a gene that is not included in the genomes of previously genetically tractable members of the human gut microbiome.IMPORTANCE Human gut Bacteroides species exhibit strain-level differences in their physiology, ecology, and impact on human health and disease. However, existing approaches for genetic manipulation generally require construction of genetically modified parental strains for each microbe of interest or defined medium formulations. In this report, we introduce a robust and efficient strategy for targeted genetic manipulation of diverse wild-type Bacteroides species from the human gut. This system enables genetic investigation of members of human and animal microbiomes beyond existing model organisms.


Asunto(s)
Bacteroides/genética , Antibacterianos/farmacología , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Bacteroides/efectos de los fármacos , Bacteroides fragilis/efectos de los fármacos , Bacteroides fragilis/genética , Microbioma Gastrointestinal/efectos de los fármacos , Microbioma Gastrointestinal/genética , Tracto Gastrointestinal/microbiología , Humanos , Microbiota/efectos de los fármacos , Microbiota/genética , Polimixina B/farmacología
11.
PLoS Biol ; 15(5): e2001390, 2017 05.
Artículo en Inglés | MEDLINE | ID: mdl-28542173

RESUMEN

Campylobacter jejuni is one of the leading infectious causes of food-borne illness around the world. Its ability to persistently colonize the intestinal tract of a broad range of hosts, including food-producing animals, is central to its epidemiology since most infections are due to the consumption of contaminated food products. Using a highly saturated transposon insertion library combined with next-generation sequencing and a mouse model of infection, we have carried out a comprehensive genome-wide analysis of the fitness determinants for growth in vitro and in vivo of a highly pathogenic strain of C. jejuni. A comparison of the C. jejuni requirements to colonize the mouse intestine with those necessary to grow in different culture media in vitro, combined with isotopologue profiling and metabolic flow analysis, allowed us to identify its metabolic requirements to establish infection, including the ability to acquire certain nutrients, metabolize specific substrates, or maintain intracellular ion homeostasis. This comprehensive analysis has identified metabolic pathways that could provide the basis for the development of novel strategies to prevent C. jejuni colonization of food-producing animals or to treat human infections.


Asunto(s)
Proteínas Bacterianas/metabolismo , Infecciones por Campylobacter/microbiología , Campylobacter jejuni/fisiología , Proteínas de Transporte de Catión/metabolismo , Gastroenteritis/microbiología , Modelos Biológicos , Absorción Fisiológica , Aminoácidos/metabolismo , Animales , Antibacterianos/efectos adversos , Proteínas Bacterianas/genética , Campylobacter jejuni/crecimiento & desarrollo , Campylobacter jejuni/aislamiento & purificación , Proteínas de Transporte de Catión/genética , Elementos Transponibles de ADN , Disbiosis/inducido químicamente , Disbiosis/microbiología , Eliminación de Gen , Estudios de Asociación Genética , Genoma Bacteriano , Biblioteca Genómica , Ratones Endogámicos C57BL , Viabilidad Microbiana , Mutagénesis Insercional , Mutación
12.
Proc Natl Acad Sci U S A ; 114(48): E10446-E10454, 2017 11 28.
Artículo en Inglés | MEDLINE | ID: mdl-29133401

RESUMEN

Legionella pneumophila is the causative agent of a severe pneumonia called Legionnaires' disease. A single strain of L. pneumophila encodes a repertoire of over 300 different effector proteins that are delivered into host cells by the Dot/Icm type IV secretion system during infection. The large number of L. pneumophila effectors has been a limiting factor in assessing the importance of individual effectors for virulence. Here, a transposon insertion sequencing technology called INSeq was used to analyze replication of a pool of effector mutants in parallel both in a mouse model of infection and in cultured host cells. Loss-of-function mutations in genes encoding effector proteins resulted in host-specific or broad virulence phenotypes. Screen results were validated for several effector mutants displaying different virulence phenotypes using genetic complementation studies and infection assays. Specifically, loss-of-function mutations in the gene encoding LegC4 resulted in enhanced L. pneumophila in the lungs of infected mice but not within cultured host cells, which indicates LegC4 augments bacterial clearance by the host immune system. The effector proteins RavY and Lpg2505 were important for efficient replication within both mammalian and protozoan hosts. Further analysis of Lpg2505 revealed that this protein functions as a metaeffector that counteracts host cytotoxicity displayed by the effector protein SidI. Thus, this study identified a large cohort of effectors that contribute to L. pneumophila virulence positively or negatively and has demonstrated regulation of effector protein activities by cognate metaeffectors as being critical for host pathogenesis.


Asunto(s)
Proteínas Bacterianas/genética , Regulación Bacteriana de la Expresión Génica , Interacciones Huésped-Patógeno/inmunología , Legionella pneumophila/patogenicidad , Enfermedad de los Legionarios/inmunología , Animales , Proteínas Bacterianas/inmunología , Citotoxicidad Inmunológica , Modelos Animales de Enfermedad , Femenino , Secuenciación de Nucleótidos de Alto Rendimiento/métodos , Humanos , Legionella pneumophila/genética , Legionella pneumophila/inmunología , Enfermedad de los Legionarios/microbiología , Mutación con Pérdida de Función , Ratones , Ratones Endogámicos C57BL , Mutagénesis Insercional , Fenotipo , Virulencia/genética
13.
Proc Natl Acad Sci U S A ; 113(13): 3639-44, 2016 Mar 29.
Artículo en Inglés | MEDLINE | ID: mdl-26957597

RESUMEN

The human gut microbiome is a dynamic and densely populated microbial community that can provide important benefits to its host. Cooperation and competition for nutrients among its constituents only partially explain community composition and interpersonal variation. Notably, certain human-associated Bacteroidetes--one of two major phyla in the gut--also encode machinery for contact-dependent interbacterial antagonism, but its impact within gut microbial communities remains unknown. Here we report that prominent human gut symbionts persist in the gut through continuous attack on their immediate neighbors. Our analysis of just one of the hundreds of species in these communities reveals 12 candidate antibacterial effector loci that can exist in 32 combinations. Through the use of secretome studies, in vitro bacterial interaction assays and multiple mouse models, we uncover strain-specific effector/immunity repertoires that can predict interbacterial interactions in vitro and in vivo, and find that some of these strains avoid contact-dependent killing by accumulating immunity genes to effectors that they do not encode. Effector transmission rates in live animals can exceed 1 billion events per minute per gram of colonic contents, and multiphylum communities of human gut commensals can partially protect sensitive strains from these attacks. Together, these results suggest that gut microbes can determine their interactions through direct contact. An understanding of the strategies human gut symbionts have evolved to target other members of this community may provide new approaches for microbiome manipulation.


Asunto(s)
Microbioma Gastrointestinal/fisiología , Animales , Bacteroides fragilis/genética , Bacteroides fragilis/inmunología , Bacteroides fragilis/fisiología , Femenino , Microbioma Gastrointestinal/genética , Microbioma Gastrointestinal/inmunología , Genoma Bacteriano , Vida Libre de Gérmenes , Humanos , Masculino , Ratones , Modelos Animales , Filogenia , Simbiosis/genética , Simbiosis/inmunología , Simbiosis/fisiología , Sistemas de Secreción Tipo VI/genética , Sistemas de Secreción Tipo VI/inmunología , Sistemas de Secreción Tipo VI/fisiología
14.
Infect Immun ; 86(4)2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29339460

RESUMEN

Coxiella burnetii is an intracellular pathogen that replicates in a lysosome-derived vacuole. A determinant necessary for C. burnetii virulence is the Dot/Icm type IVB secretion system (T4SS). The Dot/Icm system delivers more than 100 proteins, called type IV effectors (T4Es), across the vacuolar membrane into the host cell cytosol. Several T4Es have been shown to be important for vacuolar biogenesis. Here, transposon (Tn) insertion sequencing technology (INSeq) was used to identify C. burnetii Nine Mile phase II mutants in an arrayed library, which facilitated the identification and clonal isolation of mutants deficient in 70 different T4E proteins. These effector mutants were screened in HeLa cells for deficiencies in Coxiella-containing vacuole (CCV) biogenesis. This screen identified and validated seven new T4Es that were important for vacuole biogenesis. Loss-of-function mutations in cbu0414 (coxH1), cbu0513, cbu0978 (cem3), cbu1387 (cem6), cbu1524 (caeA), cbu1752, or cbu2028 resulted in a small-vacuole phenotype. These seven mutant strains produced small CCVs in all cells tested, which included macrophage-like cells. The cbu2028::Tn mutant, though unable to develop large CCVs, had intracellular replication rates similar to the rate of the parental strain of C. burnetii, whereas the other six effector mutants defective in CCV biogenesis displayed significant reductions in intracellular replication. Vacuoles created by the cbu0513::Tn mutant did not accumulate lipidated microtubule-associated protein 1A/1B light chain 3 (LC3-II), suggesting a failure in fusion of the CCV with autophagosomes. These seven T4E proteins add to the growing repertoire of C. burnetii factors that contribute to CCV biogenesis.


Asunto(s)
Proteínas Bacterianas/metabolismo , Coxiella burnetii/fisiología , Fiebre Q/metabolismo , Fiebre Q/microbiología , Autofagosomas/metabolismo , Sistemas de Secreción Bacterianos , Coxiella burnetii/genética , Coxiella burnetii/patogenicidad , Elementos Transponibles de ADN , Células Epiteliales/metabolismo , Células Epiteliales/microbiología , Humanos , Lisosomas/metabolismo , Macrófagos/metabolismo , Macrófagos/microbiología , Mutación , Transporte de Proteínas , Vacuolas/metabolismo
15.
Annu Rev Microbiol ; 67: 459-75, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24024637

RESUMEN

The complex and intimate relationship between humans and their gut microbial communities is becoming less obscure, due in part to large-scale gut microbial genome-sequencing projects and culture-independent surveys of the composition and gene content of these communities. These studies build upon, and are complemented by, experimental efforts to define underlying mechanisms of host-microbe interactions in simplified model systems. This review highlights the intersection of these approaches. Experimental studies now leverage the advances in high-throughput DNA sequencing that have driven the explosion of microbial genome and community profiling projects, and the loss-of-function and gain-of-function strategies long employed in model organisms are now being extended to microbial genes, species, and communities from the human gut. These developments promise to deepen our understanding of human gut host-microbiota relationships and are readily applicable to other host-associated and free-living microbial communities.


Asunto(s)
Bacterias/genética , Tracto Gastrointestinal/microbiología , Metagenómica , Microbiota , Bacterias/aislamiento & purificación , Bacterias/metabolismo , Humanos , Metagenoma , Metagenómica/métodos
16.
J Surg Res ; 231: 331-337, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-30278949

RESUMEN

BACKGROUND: The microbiome of the gastrointestinal tract is a vast collection of microorganisms implicated in numerous aspects of normal physiology and disease pathogenesis. The use of gnotobiotic mouse models, with single or specific communities of microbes comprising the microbiome, can enhance our understanding of the microbiome-host relationship. We hypothesized that gnotobiotic mice would exhibit differences in mucosal homeostasis when compared with mice with conventional flora (CF). MATERIALS AND METHODS: Single-organism gnotobiotic mice were generated containing Escherichia coli MG1655, Akkermansia muciniphila, Bacteroides eggerthii, and Clostridium symbiosum, representing four of the major phyla present in the gastrointestinal tract. Distal ileal segments were harvested from adult mice, and histologic sections were H&E stained and used to measure villus height and crypt depth. Immunohistochemistry was performed with Ki67 and TUNEL as markers of proliferation and apoptosis, respectively. RESULTS: When compared to the ileum from CF mice, the ileum from all groups of gnotobiotic mice had significant increases in nearly all measured parameters. In addition, significant differences were seen among certain gnotobiotic groups for villus height, crypt depth, and apoptosis. CONCLUSIONS: Single-organism gnotobiotic mice demonstrate enhanced morphometric parameters compared with mice with CF and show differences in growth patterns among bacterial species. These findings suggest unique interactions between individual bacteria and the host animal which hold potential for future therapeutic strategies aimed at mucosal restoration. The mechanisms involved in this process therefore warrant further study.


Asunto(s)
Microbioma Gastrointestinal/fisiología , Homeostasis , Íleon/fisiología , Mucosa Intestinal/fisiología , Animales , Apoptosis , Biomarcadores/metabolismo , Proliferación Celular , Vida Libre de Gérmenes , Íleon/anatomía & histología , Íleon/microbiología , Inmunohistoquímica , Etiquetado Corte-Fin in Situ , Mucosa Intestinal/anatomía & histología , Mucosa Intestinal/microbiología , Masculino , Ratones , Ratones Endogámicos C57BL
18.
Nature ; 474(7351): 327-36, 2011 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-21677749

RESUMEN

Marked changes in socio-economic status, cultural traditions, population growth and agriculture are affecting diets worldwide. Understanding how our diet and nutritional status influence the composition and dynamic operations of our gut microbial communities, and the innate and adaptive arms of our immune system, represents an area of scientific need, opportunity and challenge. The insights gleaned should help to address several pressing global health problems.


Asunto(s)
Tracto Gastrointestinal/inmunología , Tracto Gastrointestinal/microbiología , Sistema Inmunológico/inmunología , Metagenoma/inmunología , Fenómenos Fisiológicos de la Nutrición/fisiología , Animales , Dieta/tendencias , Humanos , Metagenómica , Leche Humana/inmunología , Obesidad/inmunología , Obesidad/microbiología
19.
Proc Natl Acad Sci U S A ; 111(48): 17284-9, 2014 Dec 02.
Artículo en Inglés | MEDLINE | ID: mdl-25404340

RESUMEN

Animal epithelial tissue becomes reproducibly colonized by specific environmental bacteria. The bacteria (microbiota) perform critical functions for the host's tissue development, immune system development, and nutrition; yet the processes by which bacterial diversity in the environment is selected to assemble the correct communities in the host are unclear. To understand the molecular determinants of microbiota selection, we examined colonization of a simplified model in which the light organ of Euprymna scolopes squid is colonized exclusively by Vibrio fischeri bacteria. We applied high-throughput insertion sequencing to identify which bacterial genes are required during host colonization. A library of over 41,000 unique transposon insertions was analyzed before and after colonization of 1,500 squid hatchlings. Mutants that were reproducibly depleted following squid colonization represented 380 genes, including 37 that encode known colonization factors. Validation of select mutants in defined competitions against the wild-type strain identified nine mutants that exhibited a reproducible colonization defect. Some of the colonization factors identified included genes predicted to influence copper regulation and secretion. Other mutants exhibited defects in biofilm development, which is required for aggregation in host mucus and initiation of colonization. Biofilm formation in culture and in vivo was abolished in a strain lacking the cytoplasmic chaperone DnaJ, suggesting an important role for protein quality control during the elaboration of bacterial biofilm in the context of an intact host immune system. Overall these data suggest that cellular stress responses and biofilm regulation are critical processes underlying the reproducible colonization of animal hosts by specific microbial symbionts.


Asunto(s)
Aliivibrio fischeri/genética , Aliivibrio fischeri/fisiología , Decapodiformes/microbiología , Microbiota/genética , Animales , Proteínas Bacterianas/genética , Biopelículas , Elementos Transponibles de ADN/genética , Regulación Bacteriana de la Expresión Génica , Genes Bacterianos/genética , Secuenciación de Nucleótidos de Alto Rendimiento , Interacciones Huésped-Patógeno , Mutagénesis Insercional , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Simbiosis
20.
Genes Dev ; 23(2): 249-59, 2009 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-19171785

RESUMEN

The genome of the opportunistic pathogen Pseudomonas aeruginosa encodes over 60 two-component sensor kinases and uses several (including RetS and GacS) to reciprocally regulate the production of virulence factors involved in the development of acute or chronic infections. We demonstrate that RetS modulates the phosphorylation state of GacS by a direct and specific interaction between these two membrane-bound sensors. The RetS-GacS interaction can be observed in vitro, in heterologous systems in vivo, and in P. aeruginosa. This function does not require the predicted RetS phosphorelay residues and provides a mechanism for integrating multiple signals without cross-phosphorylation from sensors to noncognate response regulators. These results suggest that multiple two-component systems found in a single bacterium can form multisensor signaling networks while maintaining specific phosphorelay pathways that remain insulated from detrimental cross-talk.


Asunto(s)
Fenotipo , Proteínas Quinasas/metabolismo , Infecciones por Pseudomonas/microbiología , Pseudomonas aeruginosa/enzimología , Pseudomonas aeruginosa/patogenicidad , Enfermedad Aguda , Proteínas Bacterianas/metabolismo , Enfermedad Crónica , Citoplasma , Fosforilación , Estructura Terciaria de Proteína , Factores de Transcripción/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA