Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
Nat Chem Biol ; 15(2): 206, 2019 02.
Article in English | MEDLINE | ID: mdl-30591731

ABSTRACT

In the version of the article originally published, the x axis of the graph in Fig. 4d was incorrectly labeled as "Retention time (min)". It should read "Reaction time (min)". The 'deceased' footnote was also formatted incorrectly when published. The footnote text itself should include the name of co-author Tara A. Gianoulis in addition to the previous link to her name in the author list through footnote number 10. The errors have been corrected in the HTML and PDF versions of the article.

2.
Nat Chem Biol ; 14(6): 556-564, 2018 06.
Article in English | MEDLINE | ID: mdl-29713061

ABSTRACT

The soil microbiome can produce, resist, or degrade antibiotics and even catabolize them. While resistance genes are widely distributed in the soil, there is a dearth of knowledge concerning antibiotic catabolism. Here we describe a pathway for penicillin catabolism in four isolates. Genomic and transcriptomic sequencing revealed ß-lactamase, amidase, and phenylacetic acid catabolon upregulation. Knocking out part of the phenylacetic acid catabolon or an apparent penicillin utilization operon (put) resulted in loss of penicillin catabolism in one isolate. A hydrolase from the put operon was found to degrade in vitro benzylpenicilloic acid, the ß-lactamase penicillin product. To test the generality of this strategy, an Escherichia coli strain was engineered to co-express a ß-lactamase and a penicillin amidase or the put operon, enabling it to grow using penicillin or benzylpenicilloic acid, respectively. Elucidation of additional pathways may allow bioremediation of antibiotic-contaminated soils and discovery of antibiotic-remodeling enzymes with industrial utility.


Subject(s)
Microbiota , Open Reading Frames , Soil Microbiology , beta-Lactams/metabolism , Amidohydrolases/metabolism , Burkholderia , Cloning, Molecular , Gene Expression Regulation, Bacterial , Genome , Hydrolases/metabolism , Microbial Sensitivity Tests , Operon , Penicillins/metabolism , Phenylacetates/metabolism , Phylogeny , Pseudomonas , Soil , Transcriptome , Up-Regulation , beta-Lactamases/metabolism
3.
Genome Announc ; 5(32)2017 Aug 10.
Article in English | MEDLINE | ID: mdl-28798166

ABSTRACT

Most antibiotics are derived from the soil, but their catabolism there, which is necessary to close the antibiotic carbon cycle, remains uncharacterized. We report the first draft genome sequences of soil Proteobacteria identified for subsisting solely on ß-lactams as their carbon sources. The genomes encode multiple ß-lactamases, although their antibiotic catabolic pathways remain enigmatic.

4.
Eur Respir J ; 47(4): 1082-92, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26917613

ABSTRACT

Increasing evidence suggests that the lung microbiome plays an important role in chronic obstructive pulmonary disease (COPD) severity. However, the dynamics of the lung microbiome during COPD exacerbations and its potential role in disease aetiology remain poorly understood.We completed a longitudinal 16S ribosomal RNA survey of the lung microbiome on 476 sputum samples collected from 87 subjects with COPD at four visits defined as stable state, exacerbation, 2 weeks post-therapy and 6 weeks recovery.Our analysis revealed a dynamic lung microbiota where changes appeared to be associated with exacerbation events and indicative of specific exacerbation phenotypes. Antibiotic and steroid treatments appear to have differential effects on the lung microbiome. We depict a microbial interaction network for the lung microbiome and suggest that perturbation of a few bacterial operational taxonomic units, in particular Haemophilus spp., could greatly impact the overall microbial community structure. Furthermore, several serum and sputum biomarkers, in particular sputum interleukin-8, appear to be highly correlated with the structure and diversity of the microbiome.Our study furthers the understanding of lung microbiome dynamics in COPD patients and highlights its potential as a biomarker, and possibly a target, for future respiratory therapeutics.


Subject(s)
Lung/microbiology , Microbiota , Pulmonary Disease, Chronic Obstructive/microbiology , Administration, Oral , Adrenal Cortex Hormones/therapeutic use , Adult , Aged , Aged, 80 and over , Biomarkers/metabolism , Chemokines/metabolism , Female , Humans , Interleukin-8/metabolism , Longitudinal Studies , Male , Middle Aged , Phenotype , Principal Component Analysis , Pulmonary Disease, Chronic Obstructive/metabolism , Quality Control , RNA, Ribosomal, 16S/genetics , RNA, Ribosomal, 16S/metabolism , Sequence Analysis, DNA , Sputum/microbiology
5.
G3 (Bethesda) ; 6(3): 653-67, 2016 Jan 15.
Article in English | MEDLINE | ID: mdl-26772747

ABSTRACT

Transcriptional control of gene expression requires interactions between the cis-regulatory elements (CREs) controlling gene promoters. We developed a sensitive computational method to identify CRE combinations with conserved spacing that does not require genome alignments. When applied to seven sensu stricto and sensu lato Saccharomyces species, 80% of the predicted interactions displayed some evidence of combinatorial transcriptional behavior in several existing datasets including: (1) chromatin immunoprecipitation data for colocalization of transcription factors, (2) gene expression data for coexpression of predicted regulatory targets, and (3) gene ontology databases for common pathway membership of predicted regulatory targets. We tested several predicted CRE interactions with chromatin immunoprecipitation experiments in a wild-type strain and strains in which a predicted cofactor was deleted. Our experiments confirmed that transcription factor (TF) occupancy at the promoters of the CRE combination target genes depends on the predicted cofactor while occupancy of other promoters is independent of the predicted cofactor. Our method has the additional advantage of identifying regulatory differences between species. By analyzing the S. cerevisiae and S. bayanus genomes, we identified differences in combinatorial cis-regulation between the species and showed that the predicted changes in gene regulation explain several of the species-specific differences seen in gene expression datasets. In some instances, the same CRE combinations appear to regulate genes involved in distinct biological processes in the two different species. The results of this research demonstrate that (1) combinatorial cis-regulation can be inferred by multi-genome analysis and (2) combinatorial cis-regulation can explain differences in gene expression between species.


Subject(s)
Gene Expression Regulation, Fungal , Regulatory Sequences, Nucleic Acid , Saccharomyces/genetics , Chromatin Immunoprecipitation , Cluster Analysis , Gene Expression Profiling , Gene Regulatory Networks , High-Throughput Nucleotide Sequencing , Saccharomyces/metabolism , Signal Transduction , Transcription Factors/metabolism , Transcription, Genetic , Transcriptome
6.
PLoS One ; 10(12): e0145499, 2015.
Article in English | MEDLINE | ID: mdl-26709835

ABSTRACT

The gastrointestinal tract microbiome has been suggested as a potential therapeutic target for metabolic diseases such as obesity and Type 2 diabetes mellitus (T2DM). However, the relationship between changes in microbial communities and metabolic disease-phenotypes are still poorly understood. In this study, we used antibiotics with markedly different antibacterial spectra to modulate the gut microbiome in a diet-induced obesity mouse model and then measured relevant biochemical, hormonal and phenotypic biomarkers of obesity and T2DM. Mice fed a high-fat diet were treated with either ceftazidime (a primarily anti-Gram negative bacteria antibiotic) or vancomycin (mainly anti-Gram positive bacteria activity) in an escalating three-dose regimen. We also dosed animals with a well-known prebiotic weight-loss supplement, 10% oligofructose saccharide (10% OFS). Vancomycin treated mice showed little weight change and no improvement in glycemic control while ceftazidime and 10% OFS treatments induced significant weight loss. However, only ceftazidime showed significant, dose dependent improvement in key metabolic variables including glucose, insulin, protein tyrosine tyrosine (PYY) and glucagon-like peptide-1 (GLP-1). Subsequently, we confirmed the positive hyperglycemic control effects of ceftazidime in the Zucker diabetic fatty (ZDF) rat model. Metagenomic DNA sequencing of bacterial 16S rRNA gene regions V1-V3 showed that the microbiomes of ceftazidime dosed mice and rats were enriched for the phylum Firmicutes while 10% OFS treated mice had a greater abundance of Bacteroidetes. We show that specific changes in microbial community composition are associated with obesity and glycemic control phenotypes. More broadly, our study suggests that in vivo modulation of the microbiome warrants further investigation as a potential therapeutic strategy for metabolic diseases.


Subject(s)
Anti-Bacterial Agents/pharmacology , Diabetes Mellitus, Type 2/microbiology , Gastrointestinal Microbiome/drug effects , Obesity/microbiology , Animals , Ceftazidime/pharmacology , Diet/adverse effects , Disease Models, Animal , Male , Mice , Obesity/etiology , Phenotype , Prebiotics , Rats
7.
Antimicrob Agents Chemother ; 59(1): 289-98, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25348524

ABSTRACT

GSK2251052, a novel leucyl-tRNA synthetase (LeuRS) inhibitor, was in development for the treatment of infections caused by multidrug-resistant Gram-negative pathogens. In a phase II study (study LRS114688) evaluating the efficacy of GSK2251052 in complicated urinary tract infections, resistance developed very rapidly in 3 of 14 subjects enrolled, with ≥32-fold increases in the GSK2251052 MIC of the infecting pathogen being detected. A fourth subject did not exhibit the development of resistance in the baseline pathogen but posttherapy did present with a different pathogen resistant to GSK2251052. Whole-genome DNA sequencing of Escherichia coli isolates collected longitudinally from two study LRS114688 subjects confirmed that GSK2251052 resistance was due to specific mutations, selected on the first day of therapy, in the LeuRS editing domain. Phylogenetic analysis strongly suggested that resistant Escherichia coli isolates resulted from clonal expansion of baseline susceptible strains. This resistance development likely resulted from the confluence of multiple factors, of which only some can be assessed preclinically. Our study shows the challenges of developing antibiotics and the importance of clinical studies to evaluate their effect on disease pathogenesis. (These studies have been registered at ClinicalTrials.gov under registration no. NCT01381549 for the study of complicated urinary tract infections and registration no. NCT01381562 for the study of complicated intra-abdominal infections.).


Subject(s)
Boron Compounds/pharmacology , Drug Resistance, Bacterial/drug effects , Enzyme Inhibitors/pharmacology , Escherichia coli/drug effects , Leucine-tRNA Ligase/antagonists & inhibitors , Urinary Tract Infections/drug therapy , Anti-Bacterial Agents/pharmacology , Anti-Infective Agents, Urinary/pharmacology , Boron Compounds/therapeutic use , Escherichia coli/genetics , Escherichia coli/isolation & purification , Escherichia coli/pathogenicity , Escherichia coli Infections/drug therapy , Escherichia coli Infections/microbiology , Genome, Bacterial , Humans , Mutation , Phylogeny , Urinary Tract Infections/microbiology
8.
Antimicrob Agents Chemother ; 59(2): 1182-92, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25487798

ABSTRACT

GSK1322322 is a novel antibacterial agent under development, and it has known antibacterial activities against multidrug-resistant respiratory and skin pathogens through its inhibition of the bacterial peptide deformylase. Here, we used next-generation sequencing (NGS) of the bacterial 16S rRNA genes from stool samples collected from 61 healthy volunteers at the predosing and end-of-study time points to determine the effects of GSK1322322 on the gastrointestinal (GI) microbiota in a phase I, randomized, double-blind, and placebo-controlled study. GSK1322322 was administered either intravenously (i.v.) only or in an oral-i.v. combination in single- and repeat-dose-escalation infusions. Analysis of the 16S rRNA sequence data found no significant changes in the relative abundances of GI operational taxonomic units (OTUs) between the prestudy and end-of-study samples for either the placebo- or i.v.-only-treated subjects. However, oral-i.v. treatment resulted in significant decreases in some bacterial taxa, the Firmicutes and Bacteroidales, and increases in others, the Betaproteobacteria, Gammaproteobacteria, and Bifidobacteriaceae. Microbiome diversity plots clearly differentiated the end-of-study oral-i.v.-dosed samples from all others collected. The changes in genome function as inferred from species composition suggest an increase in bacterial transporter and xenobiotic metabolism pathways in these samples. A phylogenetic analysis of the peptide deformylase protein sequences collected from the published genomes of clinical isolates previously tested for GSK1322322 in vitro susceptibility and GI bacterial reference genomes suggests that antibiotic target homology is one of several factors that influences the response of GI microbiota to this antibiotic. Our study shows that dosing regimen and target class are important factors when considering the impact of antibiotic usage on GI microbiota. (This clinical trial was registered at the GlaxoSmithKline Clinical Study Register under study identifier PDF 113376.).


Subject(s)
Amidohydrolases/antagonists & inhibitors , Anti-Bacterial Agents/pharmacology , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Hydroxamic Acids/pharmacology , Microbiota/drug effects , Microbiota/genetics , Betaproteobacteria/drug effects , Betaproteobacteria/genetics , Bifidobacterium/drug effects , Bifidobacterium/genetics , Double-Blind Method , Gammaproteobacteria/drug effects , Gammaproteobacteria/genetics , Healthy Volunteers , High-Throughput Nucleotide Sequencing , Humans , RNA, Ribosomal, 16S/genetics
9.
PLoS One ; 9(7): e100778, 2014.
Article in English | MEDLINE | ID: mdl-24988476

ABSTRACT

UNLABELLED: Metformin, a biguanide derivate, has pleiotropic effects beyond glucose reduction, including improvement of lipid profiles and lowering microvascular and macrovascular complications associated with type 2 diabetes mellitus (T2DM). These effects have been ascribed to adenosine monophosphate-activated protein kinase (AMPK) activation in the liver and skeletal muscle. However, metformin effects are not attenuated when AMPK is knocked out and intravenous metformin is less effective than oral medication, raising the possibility of important gut pharmacology. We hypothesized that the pharmacology of metformin includes alteration of bile acid recirculation and gut microbiota resulting in enhanced enteroendocrine hormone secretion. In this study we evaluated T2DM subjects on and off metformin monotherapy to characterize the gut-based mechanisms of metformin. Subjects were studied at 4 time points: (i) at baseline on metformin, (ii) 7 days after stopping metformin, (iii) when fasting blood glucose (FBG) had risen by 25% after stopping metformin, and (iv) when FBG returned to baseline levels after restarting the metformin. At these timepoints we profiled glucose, insulin, gut hormones (glucagon-like peptide-1 (GLP-1), peptide tyrosine-tyrosine (PYY) and glucose-dependent insulinotropic peptide (GIP) and bile acids in blood, as well as duodenal and faecal bile acids and gut microbiota. We found that metformin withdrawal was associated with a reduction of active and total GLP-1 and elevation of serum bile acids, especially cholic acid and its conjugates. These effects reversed when metformin was restarted. Effects on circulating PYY were more modest, while GIP changes were negligible. Microbiota abundance of the phylum Firmicutes was positively correlated with changes in cholic acid and conjugates, while Bacteroidetes abundance was negatively correlated. Firmicutes and Bacteroidetes representation were also correlated with levels of serum PYY. Our study suggests that metformin has complex effects due to gut-based pharmacology which might provide insights into novel therapeutic approaches to treat T2DM and associated metabolic diseases. TRIAL REGISTRATION: www.ClinicalTrials.gov NCT01357876.


Subject(s)
Diabetes Mellitus, Type 2/drug therapy , Hypoglycemic Agents , Intestinal Mucosa , Intestines , Metformin , Microbiota/drug effects , Adolescent , Adult , Aged , Bile Acids and Salts/metabolism , Blood Glucose/metabolism , Female , Glucagon-Like Peptide 1/blood , Humans , Hypoglycemic Agents/administration & dosage , Hypoglycemic Agents/pharmacokinetics , Intestinal Mucosa/metabolism , Intestines/microbiology , Male , Metformin/administration & dosage , Metformin/pharmacokinetics , Middle Aged , Peptide YY/blood
10.
Nucleic Acids Res ; 40(Database issue): D162-8, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22140105

ABSTRACT

Saccharomyces cerevisiae is a primary model for studies of transcriptional control, and the specificities of most yeast transcription factors (TFs) have been determined by multiple methods. However, it is unclear which position weight matrices (PWMs) are most useful; for the roughly 200 TFs in yeast, there are over 1200 PWMs in the literature. To address this issue, we created ScerTF, a comprehensive database of 1226 motifs from 11 different sources. We identified a single matrix for each TF that best predicts in vivo data by benchmarking matrices against chromatin immunoprecipitation and TF deletion experiments. We also used in vivo data to optimize thresholds for identifying regulatory sites with each matrix. To correct for biases from different methods, we developed a strategy to combine matrices. These aligned matrices outperform the best available matrix for several TFs. We used the matrices to predict co-occurring regulatory elements in the genome and identified many known TF combinations. In addition, we predict new combinations and provide evidence of combinatorial regulation from gene expression data. The database is available through a web interface at http://ural.wustl.edu/ScerTF. The site allows users to search the database with a regulatory site or matrix to identify the TFs most likely to bind the input sequence.


Subject(s)
Databases, Genetic , Saccharomyces cerevisiae/genetics , Saccharomyces/genetics , Transcription Factors/metabolism , Binding Sites , Internet , Position-Specific Scoring Matrices , Promoter Regions, Genetic , Sequence Analysis, DNA , User-Computer Interface
11.
Protein Expr Purif ; 62(2): 223-9, 2008 Dec.
Article in English | MEDLINE | ID: mdl-18765283

ABSTRACT

We have developed a simple method for isolating and purifying plasma membrane proteins from various cell types. This one-step affinity-chromatography method uses the property of the lectin concanavalin A (ConA) and the technique of magnetic bead separation to obtain highly purified plasma membrane proteins from crude membrane preparations or cell lines. ConA is immobilized onto magnetic beads by binding biotinylated ConA to streptavidin magnetic beads. When these ConA magnetic beads were used to enrich plasma membranes from a crude membrane preparation, this procedure resulted in 3.7-fold enrichment of plasma membrane marker 5'-nucleotidase activity with 70% recovery of the activity in the crude membrane fraction of rat liver. In agreement with the results of 5'-nucleotidase activity, immunoblotting with antibodies specific for a rat liver plasma membrane protein, CEACAM1, indicated that CEACAM1 was enriched about threefold relative to that of the original membranes. In similar experiments, this method produced 13-fold enrichment of 5'-nucleotidase activity with 45% recovery of the activity from a total cell lysate of PC-3 cells and 7.1-fold enrichment of 5'-nucleotidase activity with 33% recovery of the activity from a total cell lysate of HeLa cells. These results suggest that this one-step purification method can be used to isolate total plasma membrane proteins from tissue or cells for the identification of membrane biomarkers.


Subject(s)
Biochemistry/methods , Cell Membrane/metabolism , Concanavalin A/metabolism , Magnetics , Membrane Proteins/isolation & purification , Microspheres , Animals , Cell Line, Tumor , Humans , Liver Extracts/metabolism , Membrane Proteins/metabolism , Rats , Streptavidin/metabolism
12.
Can J Microbiol ; 49(11): 683-98, 2003 Nov.
Article in English | MEDLINE | ID: mdl-14735218

ABSTRACT

The objective of this study was to determine if antifungal actinomycetes isolated from rhizosphere and non-rhizosphere soils exhibit different chitinase-like production and (or) induction patterns. Selected isolates from both habitats were compared. Chitinase-like levels and isoform characteristic patterns were evaluated over time in culture fluids of isolates grown on media containing different combinations of colloidal chitin and fungal cell wall (FCW) preparation. Supernatants were also subjected to native and non-native polyacrylamide gel electrophoresis (PAGE), using glycol chitin amended gels. For non-native PAGE, protein samples were denatured by two different approaches. Multiple active bands, ranging from 20 to 53 kDa and present in varying amounts, were detected in gels for most strains. Different substrate preferences were observed among strains, and different chitinase-like enzymes were produced, depending upon the substrate combinations used. The presence of FCW in the medium induced specific chitinase-like enzymes not observed otherwise. Enzymatic activities and profiles of the isolates, however, were strain and substrate specific rather than habitat specific. However, a sagebrush rhizosphere soil had a larger actinomycete community with higher chitinolytic activities than the nearby bulk soil. The use of PAGE to compare chitinase-like proteins induced in media with and without FCW was useful for identifying chitinase-like enzymes potentially involved in antifungal activity.


Subject(s)
Actinobacteria/enzymology , Chitinases/metabolism , Plant Roots/microbiology , Soil Microbiology , Streptomyces/enzymology , Actinobacteria/classification , Actinobacteria/genetics , Actinobacteria/isolation & purification , Artemisia/microbiology , Cell Wall/metabolism , Chitin/metabolism , Chitinases/chemistry , DNA, Ribosomal/analysis , Electrophoresis, Polyacrylamide Gel , Fungi/chemistry , RNA, Ribosomal, 16S/genetics , Sequence Analysis, DNA , Streptomyces/classification , Streptomyces/genetics , Streptomyces/isolation & purification
SELECTION OF CITATIONS
SEARCH DETAIL
...