Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 34
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
PLoS Pathog ; 16(1): e1008251, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31961914

RESUMEN

Patients with cystic fibrosis (CF) have altered fecal microbiomes compared to those of healthy controls. The magnitude of this dysbiosis correlates with measures of CF gastrointestinal (GI) disease, including GI inflammation and nutrient malabsorption. However, whether this dysbiosis is caused by mutations in the CFTR gene, the underlying defect in CF, or whether CF-associated dysbiosis augments GI disease was not clear. To test the relationships between CFTR dysfunction, microbes, and intestinal health, we established a germ-free (GF) CF mouse model and demonstrated that CFTR gene mutations are sufficient to alter the GI microbiome. Furthermore, flow cytometric analysis demonstrated that colonized CF mice have increased mesenteric lymph node and spleen TH17+ cells compared with non-CF mice, suggesting that CFTR defects alter adaptive immune responses. Our findings demonstrate that CFTR mutations modulate both the host adaptive immune response and the intestinal microbiome.


Asunto(s)
Regulador de Conductancia de Transmembrana de Fibrosis Quística/genética , Fibrosis Quística/microbiología , Disbiosis/microbiología , Microbioma Gastrointestinal , Animales , Bacterias/clasificación , Bacterias/genética , Bacterias/aislamiento & purificación , Fibrosis Quística/genética , Fibrosis Quística/inmunología , Regulador de Conductancia de Transmembrana de Fibrosis Quística/inmunología , Modelos Animales de Enfermedad , Disbiosis/genética , Disbiosis/inmunología , Femenino , Humanos , Intestinos/inmunología , Intestinos/microbiología , Masculino , Ratones , Ratones Endogámicos C57BL , Mutación
2.
BMC Microbiol ; 21(1): 247, 2021 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-34525965

RESUMEN

BACKGROUND: Infants with cystic fibrosis (CF) suffer from gastrointestinal (GI) complications, including pancreatic insufficiency and intestinal inflammation, which have been associated with impaired nutrition and growth. Recent evidence identified altered fecal microbiota taxonomic compositions in infants with CF relative to healthy infants that were characterized by differences in the abundances of taxa associated with GI health and nutrition. Furthermore, these taxonomic differences were more pronounced in low length infants with CF, suggesting a potential link to linear growth failure. We hypothesized that these differences would entail shifts in the microbiome's functional capacities that could contribute to inflammation and nutritional failure in infants with CF. RESULTS: To test this hypothesis, we compared fecal microbial metagenomic content between healthy infants and infants with CF, supplemented with an analysis of fecal metabolomes in infants with CF. We identified notable differences in CF fecal microbial functional capacities, including metabolic and environmental response functions, compared to healthy infants that intensified during the first year of life. A machine learning-based longitudinal metagenomic age analysis of healthy and CF fecal metagenomic functional profiles further demonstrated that these differences are characterized by a CF-associated delay in the development of these functional capacities. Moreover, we found metagenomic differences in functions related to metabolism among infants with CF that were associated with diet and antibiotic exposure, and identified several taxa as potential drivers of these functional differences. An integrated metagenomic and metabolomic analysis further revealed that abundances of several fecal GI metabolites important for nutrient absorption, including three bile acids, correlated with specific microbes in infants with CF. CONCLUSIONS: Our results highlight several metagenomic and metabolomic factors, including bile acids and other microbial metabolites, that may impact nutrition, growth, and GI health in infants with CF. These factors could serve as promising avenues for novel microbiome-based therapeutics to improve health outcomes in these infants.


Asunto(s)
Fibrosis Quística/complicaciones , Fibrosis Quística/microbiología , Disbiosis/complicaciones , Heces/microbiología , Enfermedades Gastrointestinales/etiología , Metaboloma , Metagenoma , Enfermedades Gastrointestinales/microbiología , Enfermedades Gastrointestinales/fisiopatología , Humanos , Lactante , Estudios Longitudinales , Metabolómica/métodos , Estudios Prospectivos
3.
Proc Natl Acad Sci U S A ; 115(7): 1605-1610, 2018 02 13.
Artículo en Inglés | MEDLINE | ID: mdl-29378945

RESUMEN

The mature human gut microbiota is established during the first years of life, and altered intestinal microbiomes have been associated with several human health disorders. Escherichia coli usually represents less than 1% of the human intestinal microbiome, whereas in cystic fibrosis (CF), greater than 50% relative abundance is common and correlates with intestinal inflammation and fecal fat malabsorption. Despite the proliferation of E. coli and other Proteobacteria in conditions involving chronic gastrointestinal tract inflammation, little is known about adaptation of specific characteristics associated with microbiota clonal expansion. We show that E. coli isolated from fecal samples of young children with CF has adapted to growth on glycerol, a major component of fecal fat. E. coli isolates from different CF patients demonstrate an increased growth rate in the presence of glycerol compared with E. coli from healthy controls, and unrelated CF E. coli strains have independently acquired this growth trait. Furthermore, CF and control E. coli isolates have differential gene expression when grown in minimal media with glycerol as the sole carbon source. While CF isolates display a growth-promoting transcriptional profile, control isolates engage stress and stationary-phase programs, which likely results in slower growth rates. Our results indicate that there is selection of unique characteristics within the microbiome of individuals with CF, which could contribute to individual disease outcomes.


Asunto(s)
Fibrosis Quística/microbiología , Infecciones por Escherichia coli/microbiología , Escherichia coli/patogenicidad , Heces/microbiología , Microbioma Gastrointestinal/genética , Intestinos/microbiología , Estudios de Casos y Controles , Preescolar , Fibrosis Quística/genética , Fibrosis Quística/patología , Grasas de la Dieta/metabolismo , Infecciones por Escherichia coli/genética , Infecciones por Escherichia coli/patología , Redes Reguladoras de Genes , Glicerol/metabolismo , Humanos , Lactante , Fosfolípidos/metabolismo , Filogenia , Estados Unidos
4.
Thorax ; 75(9): 780-790, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32631930

RESUMEN

RATIONALE: The most common antibiotic used to treat people with cystic fibrosis (PWCF) is inhaled tobramycin, administered as maintenance therapy for chronic Pseudomonas aeruginosa lung infections. While the effects of inhaled tobramycin on P. aeruginosa abundance and lung function diminish with continued therapy, this maintenance treatment is known to improve long-term outcomes, underscoring how little is known about why antibiotics work in CF infections, what their effects are on complex CF sputum microbiomes and how to improve these treatments. OBJECTIVES: To rigorously define the effect of maintenance tobramycin on CF sputum microbiome characteristics. METHODS AND MEASUREMENTS: We collected sputum from 30 PWCF at standardised times before, during and after a single month-long course of maintenance inhaled tobramycin. We used traditional culture, quantitative PCR and metagenomic sequencing to define the dynamic effects of this treatment on sputum microbiomes, including abundance changes in both clinically targeted and untargeted bacteria, as well as functional gene categories. MAIN RESULTS: CF sputum microbiota changed most markedly by 1 week of antibiotic therapy and plateaued thereafter, and this shift was largely driven by changes in non-dominant taxa. The genetically conferred functional capacities (ie, metagenomes) of subjects' sputum communities changed little with antibiotic perturbation, despite taxonomic shifts, suggesting functional redundancy within the CF sputum microbiome. CONCLUSIONS: Maintenance treatment with inhaled tobramycin, an antibiotic with demonstrated long-term mortality benefit, primarily impacted clinically untargeted bacteria in CF sputum, highlighting the importance of monitoring the non-canonical effects of antibiotics and other treatments to accurately define and improve their clinical impact.


Asunto(s)
Antibacterianos/farmacología , Bacterias , Fibrosis Quística/microbiología , Microbiota/efectos de los fármacos , Esputo/microbiología , Tobramicina/farmacología , Administración por Inhalación , Adolescente , Adulto , Anciano , Antibacterianos/uso terapéutico , Bacterias/genética , Bacterias/aislamiento & purificación , Infecciones Bacterianas/prevención & control , Niño , Fibrosis Quística/fisiopatología , Volumen Espiratorio Forzado , Humanos , Quimioterapia de Mantención , Metagenoma/efectos de los fármacos , Persona de Mediana Edad , Índice de Severidad de la Enfermedad , Factores de Tiempo , Tobramicina/uso terapéutico , Adulto Joven
5.
J Clin Gastroenterol ; 52(2): 155-163, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-28030510

RESUMEN

GOAL: To determine the effect of the specific carbohydrate diet (SCD) on active inflammatory bowel disease (IBD). BACKGROUND: IBD is a chronic idiopathic inflammatory intestinal disorder associated with fecal dysbiosis. Diet is a potential therapeutic option for IBD based on the hypothesis that changing the fecal dysbiosis could decrease intestinal inflammation. STUDY: Pediatric patients with mild to moderate IBD defined by pediatric Crohn's disease activity index (PCDAI 10-45) or pediatric ulcerative colitis activity index (PUCAI 10-65) were enrolled into a prospective study of the SCD. Patients started SCD with follow-up evaluations at 2, 4, 8, and 12 weeks. PCDAI/PUCAI, laboratory studies were assessed. RESULTS: Twelve patients, ages 10 to 17 years, were enrolled. Mean PCDAI decreased from 28.1±8.8 to 4.6±10.3 at 12 weeks. Mean PUCAI decreased from 28.3±23.1 to 6.7±11.6 at 12 weeks. Dietary therapy was ineffective for 2 patients while 2 individuals were unable to maintain the diet. Mean C-reactive protein decreased from 24.1±22.3 to 7.1±0.4 mg/L at 12 weeks in Seattle Cohort (nL<8.0 mg/L) and decreased from 20.7±10.9 to 4.8±4.5 mg/L at 12 weeks in Atlanta Cohort (nL<4.9 mg/L). Stool microbiome analysis showed a distinctive dysbiosis for each individual in most prediet microbiomes with significant changes in microbial composition after dietary change. CONCLUSIONS: SCD therapy in IBD is associated with clinical and laboratory improvements as well as concomitant changes in the fecal microbiome. Further prospective studies are required to fully assess the safety and efficacy of dietary therapy in patients with IBD.


Asunto(s)
Colitis Ulcerosa/dietoterapia , Enfermedad de Crohn/dietoterapia , Disbiosis/dietoterapia , Heces/microbiología , Adolescente , Proteína C-Reactiva/metabolismo , Niño , Colitis Ulcerosa/fisiopatología , Enfermedad de Crohn/fisiopatología , Carbohidratos de la Dieta/administración & dosificación , Femenino , Estudios de Seguimiento , Humanos , Masculino , Estudios Prospectivos , Índice de Severidad de la Enfermedad , Factores de Tiempo
6.
Proc Natl Acad Sci U S A ; 112(10): E1096-105, 2015 Mar 10.
Artículo en Inglés | MEDLINE | ID: mdl-25713353

RESUMEN

We previously reported that lagging-strand genes accumulate mutations faster than those encoded on the leading strand in Bacillus subtilis. Although we proposed that orientation-specific encounters between replication and transcription underlie this phenomenon, the mechanism leading to the increased mutagenesis of lagging-strand genes remained unknown. Here, we report that the transcription-dependent and orientation-specific differences in mutation rates of genes require the B. subtilis Y-family polymerase, PolY1 (yqjH). We find that without PolY1, association of the replicative helicase, DnaC, and the recombination protein, RecA, with lagging-strand genes increases in a transcription-dependent manner. These data suggest that PolY1 promotes efficient replisome progression through lagging-strand genes, thereby reducing potentially detrimental breaks and single-stranded DNA at these loci. Y-family polymerases can alleviate potential obstacles to replisome progression by facilitating DNA lesion bypass, extension of D-loops, or excision repair. We find that the nucleotide excision repair (NER) proteins UvrA, UvrB, and UvrC, but not RecA, are required for transcription-dependent asymmetry in mutation rates of genes in the two orientations. Furthermore, we find that the transcription-coupling repair factor Mfd functions in the same pathway as PolY1 and is also required for increased mutagenesis of lagging-strand genes. Experimental and SNP analyses of B. subtilis genomes show mutational footprints consistent with these findings. We propose that the interplay between replication and transcription increases lesion susceptibility of, specifically, lagging-strand genes, activating an Mfd-dependent error-prone NER mechanism. We propose that this process, at least partially, underlies the accelerated evolution of lagging-strand genes.


Asunto(s)
Bacillus subtilis/genética , Genes Bacterianos , Daño del ADN , Replicación del ADN , Mutagénesis , Transcripción Genética
7.
J Bacteriol ; 197(12): 2027-35, 2015 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-25845845

RESUMEN

UNLABELLED: Acinetobacter baumannii is a Gram-negative bacterial pathogen notorious for causing serious nosocomial infections that resist antibiotic therapy. Research to identify factors responsible for the pathogen's success has been limited by the resources available for genome-scale experimental studies. This report describes the development of several such resources for A. baumannii strain AB5075, a recently characterized wound isolate that is multidrug resistant and displays robust virulence in animal models. We report the completion and annotation of the genome sequence, the construction of a comprehensive ordered transposon mutant library, the extension of high-coverage transposon mutant pool sequencing (Tn-seq) to the strain, and the identification of the genes essential for growth on nutrient-rich agar. These resources should facilitate large-scale genetic analysis of virulence, resistance, and other clinically relevant traits that make A. baumannii a formidable public health threat. IMPORTANCE: Acinetobacter baumannii is one of six bacterial pathogens primarily responsible for antibiotic-resistant infections that have become the scourge of health care facilities worldwide. Eliminating such infections requires a deeper understanding of the factors that enable the pathogen to persist in hospital environments, establish infections, and resist antibiotics. We present a set of resources that should accelerate genome-scale genetic characterization of these traits for a reference isolate of A. baumannii that is highly virulent and representative of current outbreak strains.


Asunto(s)
Infecciones por Acinetobacter/microbiología , Acinetobacter baumannii/genética , Enfermedades Transmisibles Emergentes/microbiología , Genoma Bacteriano , Infecciones por Acinetobacter/epidemiología , Acinetobacter baumannii/clasificación , Enfermedades Transmisibles Emergentes/epidemiología , Elementos Transponibles de ADN/genética , ADN Bacteriano/genética , Biblioteca de Genes , Humanos , Mutación , Plásmidos
8.
Am J Pathol ; 184(5): 1309-22, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24637292

RESUMEN

Cystic fibrosis (CF) is a multiorgan disease caused by loss of a functional cystic fibrosis transmembrane conductance regulator (CFTR) chloride channel in many epithelia of the body. Here we report the pathology observed in the gastrointestinal organs of juvenile to adult CFTR-knockout ferrets. CF gastrointestinal manifestations included gastric ulceration, intestinal bacterial overgrowth with villous atrophy, and rectal prolapse. Metagenomic phylogenetic analysis of fecal microbiota by deep sequencing revealed considerable genotype-independent microbial diversity between animals, with the majority of taxa overlapping between CF and non-CF pairs. CF hepatic manifestations were variable, but included steatosis, necrosis, biliary hyperplasia, and biliary fibrosis. Gallbladder cystic mucosal hyperplasia was commonly found in 67% of CF animals. The majority of CF animals (85%) had pancreatic abnormalities, including extensive fibrosis, loss of exocrine pancreas, and islet disorganization. Interestingly, 2 of 13 CF animals retained predominantly normal pancreatic histology (84% to 94%) at time of death. Fecal elastase-1 levels from these CF animals were similar to non-CF controls, whereas all other CF animals evaluated were pancreatic insufficient (<2 µg elastase-1 per gram of feces). These findings suggest that genetic factors likely influence the extent of exocrine pancreas disease in CF ferrets and have implications for the etiology of pancreatic sufficiency in CF patients. In summary, these studies demonstrate that the CF ferret model develops gastrointestinal pathology similar to CF patients.


Asunto(s)
Envejecimiento/patología , Regulador de Conductancia de Transmembrana de Fibrosis Quística/deficiencia , Tracto Gastrointestinal/patología , Técnicas de Inactivación de Genes , Animales , Atrofia , Bacterias/crecimiento & desarrollo , Fibrosis Quística/microbiología , Fibrosis Quística/patología , Regulador de Conductancia de Transmembrana de Fibrosis Quística/metabolismo , Hurones , Tracto Gastrointestinal/anomalías , Humanos , Moco/metabolismo , Especificidad de Órganos
9.
Proc Natl Acad Sci U S A ; 109(41): E2823-31, 2012 Oct 09.
Artículo en Inglés | MEDLINE | ID: mdl-22988113

RESUMEN

Quorum sensing allows bacteria to sense and respond to changes in population density. Acyl-homoserine lactones serve as quorum-sensing signals for many Proteobacteria, and acyl-homoserine lactone signaling is known to control cooperative activities. Quorum-controlled activities vary from one species to another. Quorum-sensing controls a constellation of genes in the opportunistic pathogen Pseudomonas aeruginosa, which thrives in a number of habitats ranging from soil and water to animal hosts. We hypothesized that there would be significant variation in quorum-sensing regulons among strains of P. aeruginosa isolated from different habitats and that differences in the quorum-sensing regulons might reveal insights about the ecology of P. aeruginosa. As a test of our hypothesis we used RNA-seq to identify quorum-controlled genes in seven P. aeruginosa isolates of diverse origins. Although our approach certainly overlooks some quorum-sensing-regulated genes we found a shared set of genes, i.e., a core quorum-controlled gene set, and we identified distinct, strain-variable sets of quorum-controlled genes, i.e., accessory genes. Some quorum-controlled genes in some strains were not present in the genomes of other strains. We detected a correlation between traits encoded by some genes in the strain-variable subsets of the quorum regulons and the ecology of the isolates. These findings indicate a role for quorum sensing in extension of the range of habitats in which a species can thrive. This study also provides a framework for understanding the molecular mechanisms by which quorum-sensing systems operate, the evolutionary pressures by which they are maintained, and their importance in disparate ecological contexts.


Asunto(s)
Variación Genética , Pseudomonas aeruginosa/genética , Percepción de Quorum/genética , Regulón , Análisis por Conglomerados , Fibrosis Quística/microbiología , Microbiología Ambiental , Perfilación de la Expresión Génica , Regulación Bacteriana de la Expresión Génica , Genes Bacterianos/genética , Genoma Bacteriano/genética , Humanos , Solanum lycopersicum/microbiología , Datos de Secuencia Molecular , Mutación , Análisis de Secuencia por Matrices de Oligonucleótidos , Infecciones por Pseudomonas/microbiología , Pseudomonas aeruginosa/clasificación , Pseudomonas aeruginosa/aislamiento & purificación , Análisis de Secuencia de ADN , Especificidad de la Especie
10.
Proc Natl Acad Sci U S A ; 109(35): E2343-52, 2012 Aug 28.
Artículo en Inglés | MEDLINE | ID: mdl-22837397

RESUMEN

Genome-wide association studies can identify common differences that contribute to human phenotypic diversity and disease. When genome-wide association studies are combined with approaches that test how variants alter physiology, biological insights can emerge. Here, we used such an approach to reveal regulation of cell death by the methionine salvage pathway. A common SNP associated with reduced expression of a putative methionine salvage pathway dehydratase, apoptotic protease activating factor 1 (APAF1)-interacting protein (APIP), was associated with increased caspase-1-mediated cell death in response to Salmonella. The role of APIP in methionine salvage was confirmed by growth assays with methionine-deficient media and quantitation of the methionine salvage substrate, 5'-methylthioadenosine. Reducing expression of APIP or exogenous addition of 5'-methylthioadenosine increased Salmonellae-induced cell death. Consistent with APIP originally being identified as an inhibitor of caspase-9-dependent apoptosis, the same allele was also associated with increased sensitivity to the chemotherapeutic agent carboplatin. Our results show that common human variation affecting expression of a single gene can alter susceptibility to two distinct cell death programs. Furthermore, the same allele that promotes cell death is associated with improved survival of individuals with systemic inflammatory response syndrome, suggesting a possible evolutionary pressure that may explain the geographic pattern observed for the frequency of this SNP. Our study shows that in vitro association screens of disease-related traits can not only reveal human genetic differences that contribute to disease but also provide unexpected insights into cell biology.


Asunto(s)
Proteínas Reguladoras de la Apoptosis/genética , Apoptosis/fisiología , Caspasa 1/genética , Metionina/metabolismo , Infecciones por Salmonella , Salmonella typhimurium/inmunología , Adulto , Anciano , Anciano de 80 o más Años , Animales , Apoptosis/genética , Proteínas Reguladoras de la Apoptosis/metabolismo , Células de la Médula Ósea/citología , Caspasa 1/metabolismo , Caspasa 9/metabolismo , Desoxiadenosinas/metabolismo , Predisposición Genética a la Enfermedad/genética , Variación Genética , Células HEK293 , Proyecto Mapa de Haplotipos , Humanos , Ratones , Ratones Endogámicos C57BL , Persona de Mediana Edad , Polimorfismo de Nucleótido Simple/genética , Sitios de Carácter Cuantitativo , Infecciones por Salmonella/genética , Infecciones por Salmonella/metabolismo , Infecciones por Salmonella/patología , Tionucleósidos/metabolismo , Adulto Joven
11.
J Bacteriol ; 196(22): 3862-71, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25182491

RESUMEN

Burkholderia pseudomallei, Burkholderia thailandensis, and Burkholderia mallei (the Bptm group) are close relatives with very different lifestyles: B. pseudomallei is an opportunistic pathogen, B. thailandensis is a nonpathogenic saprophyte, and B. mallei is a host-restricted pathogen. The acyl-homoserine lactone quorum-sensing (QS) systems of these three species show a high level of conservation. We used transcriptome sequencing (RNA-seq) to define the quorum-sensing regulon in each species, and we performed a cross-species analysis of the QS-controlled orthologs. Our analysis revealed a core set of QS-regulated genes in all three species, as well as QS-controlled factors shared by only two species or unique to a given species. This global survey of the QS regulons of B. pseudomallei, B. thailandensis, and B. mallei serves as a platform for predicting which QS-controlled processes might be important in different bacterial niches and contribute to the pathogenesis of B. pseudomallei and B. mallei.


Asunto(s)
Burkholderia/genética , Burkholderia/fisiología , Percepción de Quorum/fisiología , Regulón/fisiología , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Burkholderia/clasificación , Burkholderia mallei/clasificación , Burkholderia mallei/genética , Burkholderia mallei/fisiología , Burkholderia pseudomallei/clasificación , Burkholderia pseudomallei/genética , Burkholderia pseudomallei/fisiología , Regulación Bacteriana de la Expresión Génica/fisiología , Especificidad de la Especie
12.
Clin Infect Dis ; 58(3): 396-9, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24178246

RESUMEN

Cystic fibrosis gastrointestinal disease includes nutrient malabsorption and intestinal inflammation. We show that the abundances of Escherichia coli in fecal microbiota were significantly higher in young children with cystic fibrosis than in controls and correlated with fecal measures of nutrient malabsorption and inflammation, suggesting that E. coli could contribute to cystic fibrosis gastrointestinal dysfunction.


Asunto(s)
Fibrosis Quística/complicaciones , Disbiosis/complicaciones , Disbiosis/microbiología , Infecciones por Escherichia coli/complicaciones , Infecciones por Escherichia coli/microbiología , Enfermedades Gastrointestinales/microbiología , Enfermedades Gastrointestinales/patología , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Niño , Preescolar , Heces/microbiología , Femenino , Enfermedades Gastrointestinales/etiología , Humanos , Lactante , Recién Nacido , Masculino , Persona de Mediana Edad , Adulto Joven
13.
BMC Genomics ; 15: 355, 2014 May 10.
Artículo en Inglés | MEDLINE | ID: mdl-24886041

RESUMEN

BACKGROUND: Shigella dysenteriae type 1 (Sd1) causes recurrent epidemics of dysentery associated with high mortality in many regions of the world. Sd1 infects humans at very low infectious doses (10 CFU), and treatment is complicated by the rapid emergence of antibiotic resistant Sd1 strains. Sd1 is only detected in the context of human infections, and the circumstances under which epidemics emerge and regress remain unknown. RESULTS: Phylogenomic analyses of 56 isolates collected worldwide over the past 60 years indicate that the Sd1 clone responsible for the recent pandemics emerged at the turn of the 20th century, and that the two world wars likely played a pivotal role for its dissemination. Several lineages remain ubiquitous and their phylogeny indicates several recent intercontinental transfers. Our comparative genomics analysis reveals that isolates responsible for separate outbreaks, though closely related to one another, have independently accumulated antibiotic resistance genes, suggesting that there is little or no selection to retain these genes in-between outbreaks. The genomes appear to be subjected to genetic drift that affects a number of functions currently used by diagnostic tools to identify Sd1, which could lead to the potential failure of such tools. CONCLUSIONS: Taken together, the Sd1 population structure and pattern of evolution suggest a recent emergence and a possible human carrier state that could play an important role in the epidemic pattern of infections of this human-specific pathogen. This analysis highlights the important role of whole-genome sequencing in studying pathogens for which epidemiological or laboratory investigations are particularly challenging.


Asunto(s)
Disentería Bacilar/epidemiología , Shigella dysenteriae/genética , Antibacterianos/farmacología , Brotes de Enfermedades , Farmacorresistencia Bacteriana/efectos de los fármacos , Disentería Bacilar/historia , Evolución Molecular , Variación Genética , Genoma Bacteriano , Genómica , Secuenciación de Nucleótidos de Alto Rendimiento , Historia del Siglo XX , Humanos , Filogenia , Análisis de Secuencia de ADN , Shigella dysenteriae/clasificación , Shigella dysenteriae/aislamiento & purificación
14.
J Bacteriol ; 194(24): 6965-6, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-23209222

RESUMEN

Here we report the complete, accurate 1.89-Mb genome sequence of Francisella tularensis subsp. holarctica strain FSC200, isolated in 1998 in the Swedish municipality Ljusdal, which is in an area where tularemia is highly endemic. This genome is important because strain FSC200 has been extensively used for functional and genetic studies of Francisella and is well-characterized.


Asunto(s)
Francisella tularensis/genética , Genoma Bacteriano , Tularemia/microbiología , Técnicas de Tipificación Bacteriana , Secuencia de Bases , Preescolar , ADN Bacteriano/genética , Femenino , Francisella tularensis/aislamiento & purificación , Humanos , Datos de Secuencia Molecular , Análisis de Secuencia de ADN , Suecia
15.
Am J Hum Genet ; 85(2): 214-27, 2009 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-19664744

RESUMEN

Recent progress in cataloguing common genetic variation has made possible genome-wide studies that are beginning to elucidate the causes and consequences of our genetic differences. Approaches that provide a mechanistic understanding of how genetic variants function to alter disease susceptibility and why they were substrates of natural selection would complement other approaches to human-genome analysis. Here we use a novel cell-based screen of bacterial infection to identify human variation in Salmonella-induced cell death. A loss-of-function allele of CARD8, a reported inhibitor of the proinflammatory protease caspase-1, was associated with increased cell death in vitro (p = 0.013). The validity of this association was demonstrated through overexpression of alternative alleles and RNA interference in cells of varying genotype. Comparison of mammalian CARD8 orthologs and examination of variation among different human populations suggest that the increase in infectious-disease burden associated with larger animal groups (i.e., herds and colonies), and possibly human population expansion, may have naturally selected for loss of CARD8. We also find that the loss-of-function CARD8 allele shows a modest association with an increased risk of systemic inflammatory response syndrome in a small study (p = 0.05). Therefore, a by-product of the selected benefit of loss of CARD8 could be increased inflammatory diseases. These results demonstrate the utility of genome-wide cell-based association screens with microbes in the identification of naturally selected variants that can impact human health.


Asunto(s)
Infecciones Bacterianas/genética , Variación Genética , Genoma Humano , Estudio de Asociación del Genoma Completo , Fenómenos del Sistema Inmunológico , Alelos , Proteínas Adaptadoras de Señalización CARD/genética , Genética de Población , Genotipo , Humanos , Proteínas de Neoplasias/genética , Polimorfismo de Nucleótido Simple , Salmonella typhimurium/genética , Salmonella typhimurium/metabolismo
16.
Appl Environ Microbiol ; 78(19): 6812-8, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22798355

RESUMEN

High-throughput sequencing of cDNA prepared from RNA, an approach known as RNA-seq, is coming into increasing use as a method for transcriptome analysis. Despite its many advantages, widespread adoption of the technique has been hampered by a lack of easy-to-use, integrated, open-source tools for analyzing the nucleotide sequence data that are generated. Here we describe Xpression, an integrated tool for processing prokaryotic RNA-seq data. The tool is easy to use and is fully automated. It performs all essential processing tasks, including nucleotide sequence extraction, alignment, quantification, normalization, and visualization. Importantly, Xpression processes multiplexed and strand-specific nucleotide sequence data. It extracts and trims specific sequences from files and separately quantifies sense and antisense reads in the final results. Outputs from the tool can also be conveniently used in downstream analysis. In this paper, we show the utility of Xpression to process strand-specific RNA-seq data to identify genes regulated by CouR, a transcription factor that controls p-coumarate degradation by the bacterium Rhodopseudomonas palustris.


Asunto(s)
Biología Computacional/métodos , Ácidos Cumáricos/metabolismo , Perfilación de la Expresión Génica/métodos , Redes y Vías Metabólicas/genética , Regulón , Rhodopseudomonas/genética , Rhodopseudomonas/metabolismo , Secuenciación de Nucleótidos de Alto Rendimiento , Propionatos , ARN Bacteriano/biosíntesis , ARN Bacteriano/genética , Análisis de Secuencia de ADN/métodos
17.
PLoS Pathog ; 4(2): e24, 2008 Feb 08.
Artículo en Inglés | MEDLINE | ID: mdl-18266468

RESUMEN

Francisella tularensis (Ft) is a highly infectious gram-negative bacterium and the causative agent of the human disease tularemia. Ft is designated a class A select agent by the Centers for Disease Control and Prevention. Human clinical isolates of Ft produce lipid A of similar structure to Ft subspecies novicida (Fn), a pathogen of mice. We identified three enzymes required for Fn lipid A carbohydrate modifications, specifically the presence of mannose (flmF1), galactosamine (flmF2), or both carbohydrates (flmK). Mutants lacking either galactosamine (flmF2) or galactosamine/mannose (flmK) addition to their lipid A were attenuated in mice by both pulmonary and subcutaneous routes of infection. In addition, aerosolization of the mutants (flmF2 and flmK) provided protection against challenge with wild-type (WT) Fn, whereas subcutaneous administration of only the flmK mutant provided protection from challenge with WT Fn. Furthermore, infection of an alveolar macrophage cell line by the flmK mutant induced higher levels of tumor necrosis factor-alpha (TNF-alpha) and macrophage inhibitory protein-2 (MIP-2) when compared to infection with WT Fn. Bone marrow-derived macrophages (BMMø) from Toll-like receptor 4 (TLR4) and TLR2/4 knockout mice infected with the flmK mutant also produced significantly higher amounts of interleukin-6 (IL-6) and MIP-2 than BMMø infected with WT Fn. However, production of IL-6 and MIP-2 was undetectable in BMMø from MyD88(-/-) mice infected with either strain. MyD88(-/-) mice were also susceptible to flmK mutant infection. We hypothesize that the ability of the flmK mutant to activate pro-inflammatory cytokine/chemokine production and innate immune responses mediated by the MyD88 signaling pathway may be responsible for its attenuation, leading to the induction of protective immunity by this mutant.


Asunto(s)
Francisella tularensis/fisiología , Genes Bacterianos/genética , Lípido A/metabolismo , Tularemia/microbiología , Animales , Biomarcadores/metabolismo , Células de la Médula Ósea/metabolismo , Células de la Médula Ósea/microbiología , Línea Celular , Modelos Animales de Enfermedad , Femenino , Silenciador del Gen , Inmunidad Innata/fisiología , Macrófagos Alveolares/metabolismo , Macrófagos Alveolares/microbiología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Mutación , Factor 88 de Diferenciación Mieloide/deficiencia , Factor 88 de Diferenciación Mieloide/genética , Factor 88 de Diferenciación Mieloide/metabolismo , Transducción de Señal , Organismos Libres de Patógenos Específicos , Tularemia/genética , Tularemia/inmunología
18.
Nat Med ; 26(2): 215-221, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31959989

RESUMEN

Most infants with cystic fibrosis (CF) have pancreatic exocrine insufficiency that results in nutrient malabsorption and requires oral pancreatic enzyme replacement. Newborn screening for CF has enabled earlier diagnosis, nutritional intervention and enzyme replacement for these infants, allowing most infants with CF to achieve their weight goals by 12 months of age1. Nevertheless, most infants with CF continue to have poor linear growth during their first year of life1. Although this early linear growth failure is associated with worse long-term respiratory function and survival2,3, the determinants of body length in infants with CF have not been defined. Several characteristics of the CF gastrointestinal (GI) tract, including inflammation, maldigestion and malabsorption, may promote intestinal dysbiosis4,5. As GI microbiome activities are known to affect endocrine functions6,7, the intestinal microbiome of infants with CF may also impact growth. We identified an early, progressive fecal dysbiosis that distinguished infants with CF and low length from infants with CF and normal length. This dysbiosis included altered abundances of taxa that perform functions that are important for GI health, nutrient harvest and growth hormone signaling, including decreased abundance of Bacteroidetes and increased abundance of Proteobacteria. Thus, the GI microbiota represent a potential therapeutic target for the correction of low linear growth in infants with CF.


Asunto(s)
Fibrosis Quística/microbiología , Disbiosis/microbiología , Heces/microbiología , Trastornos del Crecimiento/etiología , Tamaño Corporal , Estudios de Casos y Controles , Femenino , Microbioma Gastrointestinal , Tracto Gastrointestinal/microbiología , Humanos , Lactante , Recién Nacido , Inflamación , Estudios Longitudinales , Masculino , Análisis Multivariante , Mutación , Tamizaje Neonatal , Estudios Prospectivos , Análisis de Secuencia de ADN
19.
Infect Immun ; 77(1): 232-44, 2009 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-18955478

RESUMEN

Francisella tularensis is a gram-negative, highly infectious, aerosolizable facultative intracellular pathogen that causes the potentially life-threatening disease tularemia. To date there is no approved vaccine available, and little is known about the molecular mechanisms important for infection, survival, and dissemination at different times of infection. We report the first whole-genome screen using an inhalation mouse model to monitor infection in the lung and dissemination to the liver and spleen. We queried a comprehensive library of 2,998 sequence-defined transposon insertion mutants in Francisella novicida strain U112 using a microarray-based negative-selection screen. We were able to track the behavior of 1,029 annotated genes, equivalent to a detection rate of 75% and corresponding to approximately 57% of the entire F. novicida genome. As expected, most transposon mutants retained the ability to colonize, but 125 candidate virulence genes (12%) could not be detected in at least one of the three organs. They fell into a variety of functional categories, with one-third having no annotated function and a statistically significant enrichment of genes involved in transcription. Based on the observation that behavior during complex pool infections correlated with the degree of attenuation during single-strain infection we identified nine genes expected to strongly contribute to infection. These included two genes, those for ATP synthase C (FTN_1645) and thioredoxin (FTN_1415), that when mutated allowed increased host survival and conferred protection in vaccination experiments.


Asunto(s)
Proteínas Bacterianas/genética , Proteínas Bacterianas/fisiología , Francisella/genética , Francisella/patogenicidad , Tularemia/microbiología , Factores de Virulencia/genética , Factores de Virulencia/fisiología , Animales , Recuento de Colonia Microbiana , Elementos Transponibles de ADN , Genes Bacterianos , Hígado/microbiología , Pulmón/microbiología , Ratones , Análisis por Micromatrices , Mutagénesis Insercional , Bazo/microbiología , Análisis de Supervivencia , Virulencia
20.
Cell Rep ; 26(8): 2227-2240.e5, 2019 02 19.
Artículo en Inglés | MEDLINE | ID: mdl-30784601

RESUMEN

Metagenomic sequencing is a promising approach for identifying and characterizing organisms and their functional characteristics in complex, polymicrobial infections, such as airway infections in people with cystic fibrosis. These analyses are often hampered, however, by overwhelming quantities of human DNA, yielding only a small proportion of microbial reads for analysis. In addition, many abundant microbes in respiratory samples can produce large quantities of extracellular bacterial DNA originating either from biofilms or dead cells. We describe a method for simultaneously depleting DNA from intact human cells and extracellular DNA (human and bacterial) in sputum, using selective lysis of eukaryotic cells and endonuclease digestion. We show that this method increases microbial sequencing depth and, consequently, both the number of taxa detected and coverage of individual genes such as those involved in antibiotic resistance. This finding underscores the substantial impact of DNA from sources other than live bacteria in microbiological analyses of complex, chronic infection specimens.


Asunto(s)
Infecciones Bacterianas/microbiología , Código de Barras del ADN Taxonómico/métodos , Metagenoma , Metagenómica/métodos , Microbiota , Esputo/microbiología , Infecciones Bacterianas/diagnóstico , Humanos , Técnicas de Diagnóstico Molecular/métodos , Mucosa Respiratoria/metabolismo , Mucosa Respiratoria/microbiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA