Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
mBio ; 14(5): e0157323, 2023 Oct 31.
Article in English | MEDLINE | ID: mdl-37642463

ABSTRACT

IMPORTANCE: This work has broad relevance due to the ubiquity of dyes containing azo bonds in food and drugs. We report that azo dyes can be degraded by human gut bacteria through both enzymatic and nonenzymatic mechanisms, even from a single gut bacterial species. Furthermore, we revealed that environmental factors, oxygen, and L-Cysteine control the ability of E. coli to degrade azo dyes due to their impacts on bacterial transcription and metabolism. These results open up new opportunities to manipulate the azoreductase activity of the gut microbiome through the manipulation of host diet, suggest that azoreductase potential may be altered in patients suffering from gastrointestinal disease, and highlight the importance of studying bacterial enzymes for drug metabolism in their natural cellular and ecological context.


Subject(s)
Escherichia coli Proteins , Iron-Sulfur Proteins , Humans , Coloring Agents/metabolism , Anaerobiosis , Escherichia coli/metabolism , Bacteria/metabolism , Azo Compounds/chemistry , Azo Compounds/metabolism , Escherichia coli Proteins/genetics , Escherichia coli Proteins/metabolism , Iron-Sulfur Proteins/metabolism , Bacterial Proteins/metabolism
2.
Nat Microbiol ; 7(10): 1605-1620, 2022 10.
Article in English | MEDLINE | ID: mdl-36138165

ABSTRACT

Pharmaceuticals have extensive reciprocal interactions with the microbiome, but whether bacterial drug sensitivity and metabolism is driven by pathways conserved in host cells remains unclear. Here we show that anti-cancer fluoropyrimidine drugs inhibit the growth of gut bacterial strains from 6 phyla. In both Escherichia coli and mammalian cells, fluoropyrimidines disrupt pyrimidine metabolism. Proteobacteria and Firmicutes metabolized 5-fluorouracil to its inactive metabolite dihydrofluorouracil, mimicking the major host mechanism for drug clearance. The preTA operon was necessary and sufficient for 5-fluorouracil inactivation by E. coli, exhibited high catalytic efficiency for the reductive reaction, decreased the bioavailability and efficacy of oral fluoropyrimidine treatment in mice and was prevalent in the gut microbiomes of colorectal cancer patients. The conservation of both the targets and enzymes for metabolism of therapeutics across domains highlights the need to distinguish the relative contributions of human and microbial cells to drug efficacy and side-effect profiles.


Subject(s)
Antineoplastic Agents , Escherichia coli , Animals , Antineoplastic Agents/metabolism , Antineoplastic Agents/pharmacology , Bacteria/genetics , Escherichia coli/genetics , Escherichia coli/metabolism , Fluorouracil/metabolism , Fluorouracil/pharmacology , Humans , Mammals , Metabolic Networks and Pathways , Mice
3.
Cell Rep ; 37(5): 109930, 2021 11 02.
Article in English | MEDLINE | ID: mdl-34731631

ABSTRACT

Mechanistic insights into the role of the human microbiome in the predisposition to and treatment of disease are limited by the lack of methods to precisely add or remove microbial strains or genes from complex communities. Here, we demonstrate that engineered bacteriophage M13 can be used to deliver DNA to Escherichia coli within the mouse gastrointestinal (GI) tract. Delivery of a programmable exogenous CRISPR-Cas9 system enables the strain-specific depletion of fluorescently marked isogenic strains during competitive colonization and genomic deletions that encompass the target gene in mice colonized with a single strain. Multiple mechanisms allow E. coli to escape targeting, including loss of the CRISPR array or even the entire CRISPR-Cas9 system. These results provide a robust and experimentally tractable platform for microbiome editing, a foundation for the refinement of this approach to increase targeting efficiency, and a proof of concept for the extension to other phage-bacterial pairs of interest.


Subject(s)
Bacteriophage M13/genetics , CRISPR-Associated Protein 9/genetics , CRISPR-Cas Systems , Chromosome Deletion , Chromosomes, Bacterial , Clustered Regularly Interspaced Short Palindromic Repeats , Escherichia coli/genetics , Gastrointestinal Microbiome , Gene Editing , Animals , CRISPR-Associated Protein 9/metabolism , Escherichia coli/growth & development , Feces/microbiology , Female , Gene Expression Regulation, Bacterial , Mice, Inbred BALB C , Mice, Transgenic , Proof of Concept Study
4.
Nature ; 595(7866): 272-277, 2021 07.
Article in English | MEDLINE | ID: mdl-34163067

ABSTRACT

Diet is a major factor that shapes the gut microbiome1, but the consequences of diet-induced changes in the microbiome for host pathophysiology remain poorly understood. We conducted a randomized human intervention study using a very-low-calorie diet (NCT01105143). Although metabolic health was improved, severe calorie restriction led to a decrease in bacterial abundance and restructuring of the gut microbiome. Transplantation of post-diet microbiota to mice decreased their body weight and adiposity relative to mice that received pre-diet microbiota. Weight loss was associated with impaired nutrient absorption and enrichment in Clostridioides difficile, which was consistent with a decrease in bile acids and was sufficient to replicate metabolic phenotypes in mice in a toxin-dependent manner. These results emphasize the importance of diet-microbiome interactions in modulating host energy balance and the need to understand the role of diet in the interplay between pathogenic and beneficial symbionts.


Subject(s)
Bacteria/isolation & purification , Bacteria/metabolism , Caloric Restriction , Diet, Reducing , Gastrointestinal Microbiome/physiology , Adiposity , Animals , Bacteria/growth & development , Bacteria/pathogenicity , Bacterial Toxins/metabolism , Bile Acids and Salts/metabolism , Body Weight , Clostridioides difficile/growth & development , Clostridioides difficile/isolation & purification , Clostridioides difficile/metabolism , Energy Metabolism , Humans , Intestinal Absorption , Male , Mice , Nutrients/metabolism , Symbiosis , Weight Loss
5.
Acc Chem Res ; 54(9): 2065-2075, 2021 05 04.
Article in English | MEDLINE | ID: mdl-33877820

ABSTRACT

Rifamycin antibiotics include the WHO essential medicines rifampin, rifabutin, and rifapentine. These are semisynthetic derivatives of the natural product rifamycins, originally isolated from the soil bacterium Amycolatopsis rifamycinica. These antibiotics are primarily used to treat mycobacterial infections, including tuberculosis. Rifamycins act by binding to the ß-subunit of bacterial RNA polymerase, inhibiting transcription, which results in cell death. These antibiotics consist of a naphthalene core spanned by a polyketide ansa bridge. This structure presents a unique 3D configuration that engages RNA polymerase through a series of hydrogen bonds between hydroxyl groups linked to the naphthalene core and C21 and C23 of the ansa bridge. This binding occurs not in the enzyme active site where template-directed RNA synthesis occurs but instead in the RNA exit tunnel, thereby blocking productive formation of full-length RNA. In their clinical use to treat tuberculosis, resistance to rifamycin antibiotics arises principally from point mutations in RNA polymerase that decrease the antibiotic's affinity for the binding site in the RNA exit tunnel. In contrast, the rifamycin resistome of environmental mycobacteria and actinomycetes is much richer and diverse. In these organisms, rifamycin resistance includes many different enzymatic mechanisms that modify and alter the antibiotic directly, thereby inactivating it. These enzymes include ADP ribosyltransferases, glycosyltransferases, phosphotransferases, and monooxygenases.ADP ribosyltransferases catalyze group transfer of ADP ribose from the cofactor NAD+, which is more commonly deployed for metabolic redox reactions. ADP ribose is transferred to the hydroxyl linked to C23 of the antibiotic, thereby sterically blocking productive interaction with RNA polymerase. Like ADP ribosyltransferases, rifamycin glycosyl transferases also modify the hydroxyl of position C23 of rifamycins, transferring a glucose moiety from the donor molecule UDP-glucose. Unlike other antibiotic resistance kinases that transfer the γ-phosphate of ATP to inactivate antibiotics such as aminoglycosides or macrolides, rifamycin phosphotransferases are ATP-dependent dikinases. These enzymes transfer the ß-phosphate of ATP to the C21 hydroxyl of the rifamycin ansa bridge. The result is modification of a critical RNA polymerase binding group that blocks productive complex formation. On the other hand, rifamycin monooxygenases are FAD-dependent enzymes that hydroxylate the naphthoquinone core. The result of this modification is untethering of the ansa chain from the naphthyl moiety, disrupting the essential 3D shape necessary for productive RNA polymerase binding and inhibition that leads to cell death.All of these enzymes have homologues in bacterial metabolism that either are their direct precursors or share common ancestors to the resistance enzyme. The diversity of these resistance mechanisms, often redundant in individual bacterial isolates, speaks to the importance of protecting RNA polymerase from these compounds and validates this enzyme as a critical antibiotic target.


Subject(s)
Anti-Bacterial Agents/metabolism , RNA-Dependent RNA Polymerase/metabolism , Rifamycins/metabolism , Amycolatopsis/chemistry , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/pharmacology , Drug Resistance, Bacterial/drug effects , Rifamycins/chemistry , Rifamycins/pharmacology
6.
Proc Natl Acad Sci U S A ; 117(27): 16009-16018, 2020 07 07.
Article in English | MEDLINE | ID: mdl-32571913

ABSTRACT

Food and drug products contain diverse and abundant small-molecule additives (excipients) with unclear impacts on human physiology, drug safety, and response. Here, we evaluate their potential impact on intestinal drug absorption. By screening 136 unique compounds for inhibition of the key intestinal transporter OATP2B1 we identified and validated 24 potent OATP2B1 inhibitors, characterized by higher molecular weight and hydrophobicity compared to poor or noninhibitors. OATP2B1 inhibitors were also enriched for dyes, including 8 azo (R-N=N-R') dyes. Pharmacokinetic studies in mice confirmed that FD&C Red No. 40, a common azo dye excipient and a potent inhibitor of OATP2B1, decreased the plasma level of the OATP2B1 substrate fexofenadine, suggesting that FD&C Red No. 40 has the potential to block drug absorption through OATP2B1 inhibition in vivo. However, the gut microbiomes of multiple unrelated healthy individuals as well as diverse human gut bacterial isolates were capable of inactivating the identified azo dye excipients, producing metabolites that no longer inhibit OATP2B1 transport. These results support a beneficial role for the microbiome in limiting the unintended effects of food and drug additives in the intestine and provide a framework for the data-driven selection of excipients. Furthermore, the ubiquity and genetic diversity of gut bacterial azoreductases coupled to experiments in conventionally raised and gnotobiotic mice suggest that variations in gut microbial community structure may be less important to consider relative to the high concentrations of azo dyes in food products, which have the potential to saturate gut bacterial enzymatic activity.


Subject(s)
Bacteria/metabolism , Excipients/metabolism , Food Additives/metabolism , Food , Gastrointestinal Microbiome/physiology , Intestinal Absorption/physiology , Organic Anion Transporters/metabolism , ATP Binding Cassette Transporter, Subfamily B/genetics , Animals , Anti-Allergic Agents/metabolism , Anti-Allergic Agents/pharmacokinetics , Azo Compounds , Bacteria/isolation & purification , Excipients/pharmacokinetics , Female , Food Additives/pharmacokinetics , Histamine H1 Antagonists, Non-Sedating/metabolism , Histamine H1 Antagonists, Non-Sedating/pharmacokinetics , Humans , Intestinal Absorption/drug effects , Male , Mice , Mice, Inbred BALB C , Mice, Knockout , Terfenadine/analogs & derivatives , ATP-Binding Cassette Sub-Family B Member 4
7.
Nat Microbiol ; 5(1): 56-66, 2020 01.
Article in English | MEDLINE | ID: mdl-31686027

ABSTRACT

Plant-derived lignans, consumed daily by most individuals, are thought to protect against cancer and other diseases1; however, their bioactivity requires gut bacterial conversion to enterolignans2. Here, we dissect a four-species bacterial consortium sufficient for all five reactions in this pathway. A single enzyme (benzyl ether reductase, encoded by the gene ber) was sufficient for the first two biotransformations, variable between strains of Eggerthella lenta, critical for enterolignan production in gnotobiotic mice and unique to Coriobacteriia. Transcriptional profiling (RNA sequencing) independently identified ber and genomic loci upregulated by each of the remaining substrates. Despite their low abundance in gut microbiomes and restricted phylogenetic range, all of the identified genes were detectable in the distal gut microbiomes of most individuals living in northern California. Together, these results emphasize the importance of considering strain-level variations and bacterial co-occurrence to gain a mechanistic understanding of the bioactivation of plant secondary metabolites by the human gut microbiome.


Subject(s)
Actinobacteria/genetics , Gastrointestinal Microbiome/genetics , Gene Expression Profiling , Lignans/metabolism , Actinobacteria/classification , Actinobacteria/metabolism , Animals , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Biotransformation , Genome, Bacterial/genetics , Humans , Lignans/chemistry , Metabolic Networks and Pathways/genetics , Mice , Microbial Consortia/genetics , Phylogeny , Species Specificity
8.
Nat Microbiol ; 4(12): 2052-2063, 2019 12.
Article in English | MEDLINE | ID: mdl-31570867

ABSTRACT

Diet is a critical determinant of variation in gut microbial structure and function, outweighing even host genetics1-3. Numerous microbiome studies have compared diets with divergent ingredients1-5, but the everyday practice of cooking remains understudied. Here, we show that a plant diet served raw versus cooked reshapes the murine gut microbiome, with effects attributable to improvements in starch digestibility and degradation of plant-derived compounds. Shifts in the gut microbiota modulated host energy status, applied across multiple starch-rich plants, and were detectable in humans. Thus, diet-driven host-microbial interactions depend on the food as well as its form. Because cooking is human-specific, ubiquitous and ancient6,7, our results prompt the hypothesis that humans and our microbiomes co-evolved under unique cooking-related pressures.


Subject(s)
Bacteria/classification , Cooking , Diet , Food , Gastrointestinal Microbiome , Raw Foods/analysis , Adult , Animals , Feces/microbiology , Female , Genetic Variation , Germ-Free Life , Hot Temperature , Humans , Male , Metabolomics , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , RNA, Ribosomal, 16S/genetics , Transcriptome , Young Adult
9.
Drug Metab Dispos ; 46(11): 1588-1595, 2018 11.
Article in English | MEDLINE | ID: mdl-30111623

ABSTRACT

With a paradigm shift occurring in health care toward personalized and precision medicine, understanding the numerous environmental factors that impact drug disposition is of paramount importance. The highly diverse and variant nature of the human microbiome is now recognized as a factor driving interindividual variation in therapeutic outcomes. The purpose of this review is to provide a practical guide on methodology that can be applied to study the effects of microbes on the absorption, distribution, metabolism, and excretion of drugs. We also highlight recent examples of how these methods have been successfully applied to help build the basis for researching the intersection of the microbiome and pharmacology. Although in vitro and in vivo preclinical models are highlighted, these methods are also relevant in late-phase drug development or even as a part of routine after-market surveillance. These approaches will aid in filling major knowledge gaps for both current and upcoming therapeutics with the long-term goal of achieving a new type of knowledge-based medicine that integrates data on the host and the microbiome.


Subject(s)
Gastrointestinal Microbiome/physiology , Pharmaceutical Preparations/metabolism , Animals , Drug Discovery/methods , Humans , Inactivation, Metabolic/physiology
11.
Nat Commun ; 7: 11343, 2016 Apr 22.
Article in English | MEDLINE | ID: mdl-27103605

ABSTRACT

Rifampin (RIF) phosphotransferase (RPH) confers antibiotic resistance by conversion of RIF and ATP, to inactive phospho-RIF, AMP and Pi. Here we present the crystal structure of RPH from Listeria monocytogenes (RPH-Lm), which reveals that the enzyme is comprised of three domains: two substrate-binding domains (ATP-grasp and RIF-binding domains); and a smaller phosphate-carrying His swivel domain. Using solution small-angle X-ray scattering and mutagenesis, we reveal a mechanism where the swivel domain transits between the spatially distinct substrate-binding sites during catalysis. RPHs are previously uncharacterized dikinases that are widespread in environmental and pathogenic bacteria. These enzymes are members of a large unexplored group of bacterial enzymes with substrate affinities that have yet to be fully explored. Such an enzymatically complex mechanism of antibiotic resistance augments the spectrum of strategies used by bacteria to evade antimicrobial compounds.


Subject(s)
Anti-Bacterial Agents/metabolism , Bacterial Proteins/chemistry , Listeria monocytogenes/enzymology , Phosphotransferases/chemistry , Rifampin/metabolism , Adenosine Triphosphate/chemistry , Adenosine Triphosphate/metabolism , Amino Acid Sequence , Anti-Bacterial Agents/pharmacology , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Binding Sites , Biotransformation , Crystallography, X-Ray , Drug Resistance, Bacterial , Escherichia coli/genetics , Escherichia coli/metabolism , Gene Expression , Listeria monocytogenes/classification , Listeria monocytogenes/drug effects , Listeria monocytogenes/genetics , Models, Molecular , Molecular Sequence Data , Phosphotransferases/genetics , Phosphotransferases/metabolism , Phylogeny , Protein Binding , Protein Structure, Secondary , Protein Structure, Tertiary , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Rifampin/pharmacology , Sequence Alignment
12.
Nat Rev Microbiol ; 14(5): 273-87, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26972811

ABSTRACT

Although the importance of human genetic polymorphisms in therapeutic outcomes is well established, the role of our 'second genome' (the microbiome) has been largely overlooked. In this Review, we highlight recent studies that have shed light on the mechanisms that link the human gut microbiome to the efficacy and toxicity of xenobiotics, including drugs, dietary compounds and environmental toxins. Continued progress in this area could enable more precise tools for predicting patient responses and for the development of a new generation of therapeutics based on, or targeted at, the gut microbiome. Indeed, the admirable goal of precision medicine may require us to first understand the microbial pharmacists within.


Subject(s)
Gastrointestinal Microbiome , Pharmaceutical Preparations/metabolism , Xenobiotics/metabolism , Animals , Diet , Drug Therapy , Gastrointestinal Microbiome/physiology , Humans , Immune System/physiology , Metabolome , Metagenome , Pharmacogenetics
13.
Proc Natl Acad Sci U S A ; 111(19): 7102-7, 2014 May 13.
Article in English | MEDLINE | ID: mdl-24778229

ABSTRACT

Many environmental bacteria are multidrug-resistant and represent a reservoir of ancient antibiotic resistance determinants, which have been linked to genes found in pathogens. Exploring the environmental antibiotic resistome, therefore, reveals the diversity and evolution of antibiotic resistance and also provides insight into the vulnerability of clinically used antibiotics. In this study, we describe the identification of a highly conserved regulatory motif, the rifampin (RIF) -associated element (RAE), which is found upstream of genes encoding RIF-inactivating enzymes from a diverse collection of actinomycetes. Using gene expression assays, we confirmed that the RAE is involved in RIF-responsive regulation. By using the RAE as a probe for new RIF-associated genes in several actinomycete genomes, we identified a heretofore unknown RIF resistance gene, RIF phosphotransferase (rph). The RPH enzyme is a RIF-inactivating phosphotransferase and represents a new protein family in antibiotic resistance. RPH orthologs are widespread and found in RIF-sensitive bacteria, including Bacillus cereus and the pathogen Listeria monocytogenes. Heterologous expression and in vitro enzyme assays with purified RPHs from diverse bacterial genera show that these enzymes are capable of conferring high-level resistance to a variety of clinically used rifamycin antibiotics. This work identifies a new antibiotic resistance protein family and reinforces the fact that the study of resistance in environmental organisms can serve to identify resistance elements with relevance to pathogens.


Subject(s)
Bacterial Proteins/metabolism , Drug Resistance, Bacterial/genetics , Listeria monocytogenes/enzymology , Phosphotransferases/metabolism , Rifamycins/pharmacology , Streptomycetaceae/enzymology , Actinobacteria/genetics , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/pharmacology , Bacillus cereus/enzymology , Bacillus cereus/genetics , Bacillus cereus/pathogenicity , Bacterial Proteins/genetics , Base Sequence , Conserved Sequence , Drug Design , Listeria monocytogenes/genetics , Listeria monocytogenes/pathogenicity , Molecular Sequence Data , Phosphotransferases/genetics , Rifamycins/chemistry , Soil Microbiology , Streptomycetaceae/genetics , Streptomycetaceae/pathogenicity
14.
Nat Biotechnol ; 31(10): 922-7, 2013 Oct.
Article in English | MEDLINE | ID: mdl-24056948

ABSTRACT

Microbially derived natural products are major sources of antibiotics and other medicines, but discovering new antibiotic scaffolds and increasing the chemical diversity of existing ones are formidable challenges. We have designed a screen to exploit the self-protection mechanism of antibiotic producers to enrich microbial libraries for producers of selected antibiotic scaffolds. Using resistance as a discriminating criterion we increased the discovery rate of producers of both glycopeptide and ansamycin antibacterial compounds by several orders of magnitude in comparison with historical hit rates. Applying a phylogeny-based screening filter for biosynthetic genes enabled the binning of producers of distinct scaffolds and resulted in the discovery of a glycopeptide antibacterial compound, pekiskomycin, with an unusual peptide scaffold. This strategy provides a means to readily sample the chemical diversity available in microbes and offers an efficient strategy for rapid discovery of microbial natural products and their associated biosynthetic enzymes.


Subject(s)
Anti-Bacterial Agents/biosynthesis , Drug Evaluation, Preclinical/methods , Drug Resistance, Microbial , Actinobacteria/chemistry , Actinobacteria/drug effects , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/isolation & purification , Anti-Bacterial Agents/pharmacology , Biosynthetic Pathways/drug effects , Drug Resistance, Microbial/drug effects , Glycopeptides/biosynthesis , Glycopeptides/chemistry , Glycopeptides/isolation & purification , Glycopeptides/pharmacology , Phylogeny , Reproducibility of Results , Rifampin/chemistry , Rifampin/pharmacology , Vancomycin/chemistry , Vancomycin/isolation & purification , Vancomycin/pharmacology
15.
Biochem J ; 454(2): 191-200, 2013 Sep 01.
Article in English | MEDLINE | ID: mdl-23758273

ABSTRACT

Activity of the aminoglycoside phosphotransferase APH(3')-Ia leads to resistance to aminoglycoside antibiotics in pathogenic Gram-negative bacteria, and contributes to the clinical obsolescence of this class of antibiotics. One strategy to rescue compromised antibiotics such as aminoglycosides is targeting the enzymes that confer resistance with small molecules. We demonstrated previously that ePK (eukaryotic protein kinase) inhibitors could inhibit APH enzymes, owing to the structural similarity between these two enzyme families. However, limited structural information of enzyme-inhibitor complexes hindered interpretation of the results. In addition, cross-reactivity of compounds between APHs and ePKs represents an obstacle to their use as aminoglycoside adjuvants to rescue aminoglycoside antibiotic activity. In the present study, we structurally and functionally characterize inhibition of APH(3')-Ia by three diverse chemical scaffolds, anthrapyrazolone, 4-anilinoquinazoline and PP (pyrazolopyrimidine), and reveal distinctions in the binding mode of anthrapyrazolone and PP compounds to APH(3')-Ia compared with ePKs. Using this observation, we identify PP derivatives that select against ePKs, attenuate APH(3')-Ia activity and rescue aminoglycoside antibiotic activity against a resistant Escherichia coli strain. The structures described in the present paper and the inhibition studies provide an important opportunity for structure-based design of compounds to target aminoglycoside phosphotransferases for inhibition, potentially overcoming this form of antibiotic resistance.


Subject(s)
Aminoglycosides/pharmacology , Anti-Bacterial Agents/pharmacology , Bacterial Proteins/antagonists & inhibitors , Drug Design , Drug Resistance, Bacterial/drug effects , Kanamycin Kinase/antagonists & inhibitors , Protein Kinase Inhibitors/pharmacology , Acinetobacter baumannii/enzymology , Anthracenes/chemistry , Anthracenes/metabolism , Anthracenes/pharmacology , Bacterial Proteins/chemistry , Bacterial Proteins/metabolism , Binding Sites , Escherichia coli/drug effects , Escherichia coli/growth & development , Escherichia coli/metabolism , Isoenzymes/antagonists & inhibitors , Isoenzymes/chemistry , Isoenzymes/genetics , Isoenzymes/metabolism , Kanamycin/chemistry , Kanamycin/metabolism , Kanamycin/pharmacology , Kanamycin Kinase/chemistry , Kanamycin Kinase/genetics , Kanamycin Kinase/metabolism , Microbial Sensitivity Tests , Molecular Conformation , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/metabolism , Pyrazoles/chemistry , Pyrazoles/metabolism , Pyrazoles/pharmacology , Pyrimidines/chemistry , Pyrimidines/metabolism , Pyrimidines/pharmacology , Quinazolines/chemistry , Quinazolines/metabolism , Quinazolines/pharmacology , Recombinant Proteins/antagonists & inhibitors , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Structure-Activity Relationship
16.
Antimicrob Agents Chemother ; 57(7): 3348-57, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23650175

ABSTRACT

The field of antibiotic drug discovery and the monitoring of new antibiotic resistance elements have yet to fully exploit the power of the genome revolution. Despite the fact that the first genomes sequenced of free living organisms were those of bacteria, there have been few specialized bioinformatic tools developed to mine the growing amount of genomic data associated with pathogens. In particular, there are few tools to study the genetics and genomics of antibiotic resistance and how it impacts bacterial populations, ecology, and the clinic. We have initiated development of such tools in the form of the Comprehensive Antibiotic Research Database (CARD; http://arpcard.mcmaster.ca). The CARD integrates disparate molecular and sequence data, provides a unique organizing principle in the form of the Antibiotic Resistance Ontology (ARO), and can quickly identify putative antibiotic resistance genes in new unannotated genome sequences. This unique platform provides an informatic tool that bridges antibiotic resistance concerns in health care, agriculture, and the environment.


Subject(s)
Anti-Infective Agents , Databases, Genetic , Drug Resistance, Microbial/genetics , Genes, Bacterial , Base Sequence , Computational Biology , Genome, Bacterial , Internet , User-Computer Interface
17.
Antimicrob Agents Chemother ; 56(10): 5061-9, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22802246

ABSTRACT

Identifying and understanding the collection of all antibiotic resistance determinants presented in the global microbiota, the antibiotic resistome, provides insight into the evolution of antibiotic resistance and critical information for the development of future antimicrobials. The rifamycins are broad-spectrum antibiotics that target bacterial transcription by inhibition of RNA polymerase. Although mutational alteration of the drug target is the predominant mechanism of resistance to this family of antibiotics in the clinic, a number of diverse inactivation mechanisms have also been reported. In this report, we investigate a subset of environmental rifampin-resistant actinomycete isolates and identify a diverse collection of rifampin inactivation mechanisms. We describe a single isolate, WAC1438, capable of inactivating rifampin by glycosylation. A draft genome sequence of WAC1438 (most closely related to Streptomyces speibonae, according to a 16S rRNA gene comparison) was assembled, and the associated rifampin glycosyltransferase open reading frame, rgt1438, was identified. The role of rgt1438 in rifampin resistance was confirmed by its disruption in the bacterial chromosome, resulting in a loss of antibiotic inactivation and a 4-fold decrease in MIC. Interestingly, examination of the RNA polymerase ß-subunit sequence of WAC1438 suggests that it harbors a resistant target and thus possesses dual mechanisms of rifamycin resistance. Using an in vitro assay with purified enzyme, Rgt1438 could inactivate a variety of rifamycin antibiotics with comparable steady-state kinetics constants. Our results identify rgt1438 as a rifampin resistance determinant from WAC1438 capable of inactivating an assortment of rifamycins, adding a new element to the rifampin resistome.


Subject(s)
Actinobacteria/enzymology , Actinobacteria/metabolism , Glycosyltransferases/metabolism , Rifampin/metabolism , Rifampin/pharmacology , Actinobacteria/drug effects , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Genome, Bacterial/genetics , Glycosyltransferases/genetics , Molecular Sequence Data , Open Reading Frames/genetics , Phylogeny , Streptomyces/drug effects , Streptomyces/enzymology , Streptomyces/metabolism
18.
Cell Mol Life Sci ; 67(3): 419-31, 2010 Feb.
Article in English | MEDLINE | ID: mdl-19862477

ABSTRACT

Resistance to tetracycline emerged soon after its discovery six decades ago. Extensive clinical and non-clinical uses of this class of antibiotic over the years have combined to select for a large number of resistant determinants, collectively termed the tetracycline resistome. In order to impart resistance, microbes use different molecular mechanisms including target protection, active efflux, and enzymatic degradation. A deeper understanding of the structure, mechanism, and regulation of the genes and proteins associated with tetracycline resistance will contribute to the development of tetracycline derivatives that overcome resistance. Newer generations of tetracyclines derived from engineering of biosynthetic genetic programs, semi-synthesis, and in particular recent developments in their chemical synthesis, together with a growing understanding of resistance, will serve to retain this class of antibiotic to combat pathogens.


Subject(s)
Anti-Bacterial Agents/chemistry , Tetracycline Resistance , Tetracycline/chemistry , Anti-Bacterial Agents/biosynthesis , Anti-Bacterial Agents/metabolism , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Molecular Conformation , Ribosomes/metabolism , Tetracycline/biosynthesis , Tetracycline/metabolism , Tetracycline Resistance/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...