Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
1.
Nature ; 620(7974): 625-633, 2023 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-37495698

RESUMEN

Most bacteria in the biosphere are predicted to be polylysogens harbouring multiple prophages1-5. In studied systems, prophage induction from lysogeny to lysis is near-universally driven by DNA-damaging agents6. Thus, how co-residing prophages compete for cell resources if they respond to an identical trigger is unknown. Here we discover regulatory modules that control prophage induction independently of the DNA-damage cue. The modules bear little resemblance at the sequence level but share a regulatory logic by having a transcription factor that activates the expression of a neighbouring gene that encodes a small protein. The small protein inactivates the master repressor of lysis, which leads to induction. Polylysogens that harbour two prophages exposed to DNA damage release mixed populations of phages. Single-cell analyses reveal that this blend is a consequence of discrete subsets of cells producing one, the other or both phages. By contrast, induction through the DNA-damage-independent module results in cells producing only the phage sensitive to that specific cue. Thus, in the polylysogens tested, the stimulus used to induce lysis determines phage productivity. Considering the lack of potent DNA-damaging agents in natural habitats, additional phage-encoded sensory pathways to lysis likely have fundamental roles in phage-host biology and inter-prophage competition.


Asunto(s)
Bacterias , Bacteriófagos , Lisogenia , Profagos , Proteínas Virales , Bacteriófagos/genética , Bacteriófagos/metabolismo , Lisogenia/genética , Profagos/genética , Profagos/metabolismo , Proteínas Virales/metabolismo , Activación Viral/genética , Bacterias/virología , Daño del ADN , ADN Viral/genética , ADN Viral/metabolismo , Análisis de la Célula Individual , Factores de Transcripción/metabolismo , Interacciones Huésped-Patógeno
2.
J Mol Biol ; 435(7): 167952, 2023 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-36638909

RESUMEN

CRISPR-Cas immune systems in bacteria and archaea protect against viral infection, which has spurred viruses to develop dedicated inhibitors of these systems called anti-CRISPRs (Acrs). Like most host-virus arms races, many diverse examples of these immune and counter-immune proteins are encoded by the genomes of bacteria, archaea, and their viruses. For the case of Acrs, it is almost certain that just a small minority of nature's true diversity has been described. In this review, I discuss the various approaches used to identify these Acrs and speculate on the future for Acr discovery. Because Acrs can determine infection outcomes in nature and regulate CRISPR-Cas activities in applied settings, they have a dual importance to both host-virus conflicts and emerging biotechnologies. Thus, revealing the largely hidden world of Acrs should provide important lessons in microbiology that have the potential to ripple far beyond the field.


Asunto(s)
Archaea , Bacterias , Bacteriófagos , Sistemas CRISPR-Cas , Proteínas Virales , Archaea/genética , Archaea/virología , Bacterias/genética , Bacterias/virología , Bacteriófagos/genética , Bacteriófagos/metabolismo , Sistemas CRISPR-Cas/genética , Proteínas Virales/genética , Proteínas Virales/metabolismo , Interacciones Microbianas/genética
3.
Nucleic Acids Res ; 50(9): 5171-5190, 2022 05 20.
Artículo en Inglés | MEDLINE | ID: mdl-35511079

RESUMEN

Bacteriophage exclusion ('BREX') phage restriction systems are found in a wide range of bacteria. Various BREX systems encode unique combinations of proteins that usually include a site-specific methyltransferase; none appear to contain a nuclease. Here we describe the identification and characterization of a Type I BREX system from Acinetobacter and the effect of deleting each BREX ORF on growth, methylation, and restriction. We identified a previously uncharacterized gene in the BREX operon that is dispensable for methylation but involved in restriction. Biochemical and crystallographic analyses of this factor, which we term BrxR ('BREX Regulator'), demonstrate that it forms a homodimer and specifically binds a DNA target site upstream of its transcription start site. Deletion of the BrxR gene causes cell toxicity, reduces restriction, and significantly increases the expression of BrxC. In contrast, the introduction of a premature stop codon into the BrxR gene, or a point mutation blocking its DNA binding ability, has little effect on restriction, implying that the BrxR coding sequence and BrxR protein play independent functional roles. We speculate that elements within the BrxR coding sequence are involved in cis regulation of anti-phage activity, while the BrxR protein itself plays an additional regulatory role, perhaps during horizontal transfer.


Asunto(s)
Acinetobacter/fisiología , Factores de Restricción Antivirales , Bacteriófagos , Acinetobacter/genética , Acinetobacter/virología , Factores de Restricción Antivirales/genética , Bacteriófagos/fisiología , ADN/metabolismo , Metiltransferasas/genética , Operón
4.
PLoS Biol ; 19(10): e3001428, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34644300

RESUMEN

To overcome CRISPR-Cas defense systems, many phages and mobile genetic elements (MGEs) encode CRISPR-Cas inhibitors called anti-CRISPRs (Acrs). Nearly all characterized Acrs directly bind Cas proteins to inactivate CRISPR immunity. Here, using functional metagenomic selection, we describe AcrIIA22, an unconventional Acr found in hypervariable genomic regions of clostridial bacteria and their prophages from human gut microbiomes. AcrIIA22 does not bind strongly to SpyCas9 but nonetheless potently inhibits its activity against plasmids. To gain insight into its mechanism, we obtained an X-ray crystal structure of AcrIIA22, which revealed homology to PC4-like nucleic acid-binding proteins. Based on mutational analyses and functional assays, we deduced that acrIIA22 encodes a DNA nickase that relieves torsional stress in supercoiled plasmids. This may render them less susceptible to SpyCas9, which uses free energy from negative supercoils to form stable R-loops. Modifying DNA topology may provide an additional route to CRISPR-Cas resistance in phages and MGEs.


Asunto(s)
Proteínas Bacterianas/metabolismo , Proteína 9 Asociada a CRISPR/metabolismo , ADN/metabolismo , Proteínas Bacterianas/química , Mapeo Contig , ADN Superhelicoidal/metabolismo , Genoma Bacteriano , Metagenómica , Plásmidos , Profagos/genética , Multimerización de Proteína
5.
Elife ; 82019 09 10.
Artículo en Inglés | MEDLINE | ID: mdl-31502535

RESUMEN

CRISPR-Cas systems protect bacteria and archaea from phages and other mobile genetic elements, which use small anti-CRISPR (Acr) proteins to overcome CRISPR-Cas immunity. Because Acrs are challenging to identify, their natural diversity and impact on microbial ecosystems are underappreciated. To overcome this discovery bottleneck, we developed a high-throughput functional selection to isolate ten DNA fragments from human oral and fecal metagenomes that inhibit Streptococcus pyogenes Cas9 (SpyCas9) in Escherichia coli. The most potent Acr from this set, AcrIIA11, was recovered from a Lachnospiraceae phage. We found that AcrIIA11 inhibits SpyCas9 in bacteria and in human cells. AcrIIA11 homologs are distributed across diverse bacteria; many distantly-related homologs inhibit both SpyCas9 and a divergent Cas9 from Treponema denticola. We find that AcrIIA11 antagonizes SpyCas9 using a different mechanism than other previously characterized Type II-A Acrs. Our study highlights the power of functional selection to uncover widespread Cas9 inhibitors within diverse microbiomes.


Asunto(s)
Proteínas Bacterianas/metabolismo , Proteína 9 Asociada a CRISPR/antagonistas & inhibidores , Inhibidores Enzimáticos/metabolismo , Microbiota , Proteínas Virales/metabolismo , Proteínas Bacterianas/genética , Proteínas Bacterianas/aislamiento & purificación , Inhibidores Enzimáticos/aislamiento & purificación , Heces/microbiología , Heces/virología , Humanos , Metagenómica , Boca/microbiología , Boca/virología , Proteínas Virales/genética , Proteínas Virales/aislamiento & purificación
6.
Nat Chem Biol ; 15(2): 206, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30591731

RESUMEN

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.

7.
Nat Chem Biol ; 14(6): 556-564, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29713061

RESUMEN

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.


Asunto(s)
Microbiota , Sistemas de Lectura Abierta , Microbiología del Suelo , beta-Lactamas/metabolismo , Amidohidrolasas/metabolismo , Burkholderia , Clonación Molecular , Regulación Bacteriana de la Expresión Génica , Genoma , Hidrolasas/metabolismo , Pruebas de Sensibilidad Microbiana , Operón , Penicilinas/metabolismo , Fenilacetatos/metabolismo , Filogenia , Pseudomonas , Suelo , Transcriptoma , Regulación hacia Arriba , beta-Lactamasas/metabolismo
8.
Curr Biol ; 28(8): R361-R364, 2018 04 23.
Artículo en Inglés | MEDLINE | ID: mdl-29689213

RESUMEN

Bacteria protect themselves against infection using multiple defensive systems that move by horizontal gene transfer and accumulate in genomic 'defense islands'. A recent study exploited these features to uncover ten novel defense systems, substantially expanding the catalog of bacterial defense systems and predicting the discovery of many more.


Asunto(s)
Bacterias/genética , Genoma Bacteriano , Transferencia de Gen Horizontal , Islas Genómicas , Genómica
9.
ACS Infect Dis ; 3(9): 653-665, 2017 09 08.
Artículo en Inglés | MEDLINE | ID: mdl-28756664

RESUMEN

Aminoglycoside N-acetyltransferases (AACs) confer resistance against the clinical use of aminoglycoside antibiotics. The origin of AACs can be traced to environmental microbial species representing a vast reservoir for new and emerging resistance enzymes, which are currently undercharacterized. Here, we performed detailed structural characterization and functional analyses of four metagenomic AAC (meta-AACs) enzymes recently identified in a survey of agricultural and grassland soil microbiomes ( Forsberg et al. Nature 2014 , 509 , 612 ). These enzymes are new members of the Gcn5-Related-N-Acetyltransferase superfamily and confer resistance to the aminoglycosides gentamicin C, sisomicin, and tobramycin. Moreover, the meta-AAC0020 enzyme demonstrated activity comparable with an AAC(3)-I enzyme that serves as a model AAC enzyme identified in a clinical bacterial isolate. The crystal structure of meta-AAC0020 in complex with sisomicin confirmed an unexpected AAC(6') regiospecificity of this enzyme and revealed a drug binding mechanism distinct from previously characterized AAC(6') enzymes. Together, our data highlights the presence of highly active antibiotic-modifying enzymes in the environmental microbiome and reveals unexpected diversity in substrate specificity. These observations of additional AAC enzymes must be considered in the search for novel aminoglycosides less prone to resistance.


Asunto(s)
Acetiltransferasas/química , Acetiltransferasas/metabolismo , Farmacorresistencia Microbiana , Microbiología del Suelo , Acetiltransferasas/genética , Aminoglicósidos/farmacología , Sitios de Unión , Cristalografía por Rayos X , Metagenómica , Modelos Moleculares , Unión Proteica , Conformación Proteica , Especificidad por Sustrato
10.
Genome Announc ; 5(32)2017 Aug 10.
Artículo en Inglés | MEDLINE | ID: mdl-28798166

RESUMEN

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.

11.
Nucleic Acids Res ; 44(5): 2240-54, 2016 Mar 18.
Artículo en Inglés | MEDLINE | ID: mdl-26837573

RESUMEN

Lignin-derived (e.g. phenolic) compounds can compromise the bioconversion of lignocellulosic biomass to fuels and chemicals due to their toxicity and recalcitrance. The lipid-accumulating bacterium Rhodococcus opacus PD630 has recently emerged as a promising microbial host for lignocellulose conversion to value-added products due to its natural ability to tolerate and utilize phenolics. To gain a better understanding of its phenolic tolerance and utilization mechanisms, we adaptively evolved R. opacus over 40 passages using phenol as its sole carbon source (up to 373% growth improvement over wild-type), and extensively characterized two strains from passages 33 and 40. The two adapted strains showed higher phenol consumption rates (∼20 mg/l/h) and ∼2-fold higher lipid production from phenol than the wild-type strain. Whole-genome sequencing and comparative transcriptomics identified highly-upregulated degradation pathways and putative transporters for phenol in both adapted strains, highlighting the important linkage between mechanisms of regulated phenol uptake, utilization, and evolved tolerance. Our study shows that the R. opacus mutants are likely to use their transporters to import phenol rather than export them, suggesting a new aromatic tolerance mechanism. The identified tolerance genes and pathways are promising candidates for future metabolic engineering in R. opacus for improved lignin conversion to lipid-based products.


Asunto(s)
Adaptación Fisiológica/genética , Proteínas Bacterianas/genética , Proteínas Portadoras/genética , Regulación Bacteriana de la Expresión Génica , Fenoles/metabolismo , Rhodococcus/genética , Proteínas Bacterianas/metabolismo , Biotransformación , Carbono/metabolismo , Proteínas Portadoras/metabolismo , Ontología de Genes , Secuenciación de Nucleótidos de Alto Rendimiento , Lipogénesis/genética , Anotación de Secuencia Molecular , Rhodococcus/metabolismo , Transcriptoma
12.
Appl Environ Microbiol ; 82(2): 528-37, 2016 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-26546427

RESUMEN

The production of fuels or chemicals from lignocellulose currently requires thermochemical pretreatment to release fermentable sugars. These harsh conditions also generate numerous small-molecule inhibitors of microbial growth and fermentation, limiting production. We applied small-insert functional metagenomic selections to discover genes that confer microbial tolerance to these inhibitors, identifying both individual genes and general biological processes associated with tolerance to multiple inhibitory compounds. Having screened over 248 Gb of DNA cloned from 16 diverse soil metagenomes, we describe gain-of-function tolerance against acid, alcohol, and aldehyde inhibitors derived from hemicellulose and lignin, demonstrating that uncultured soil microbial communities hold tremendous genetic potential to address the toxicity of pretreated lignocellulose. We recovered genes previously known to confer tolerance to lignocellulosic inhibitors as well as novel genes that confer tolerance via unknown functions. For instance, we implicated galactose metabolism in overcoming the toxicity of lignin monomers and identified a decarboxylase that confers tolerance to ferulic acid; this enzyme has been shown to catalyze the production of 4-vinyl guaiacol, a valuable precursor to vanillin production. These metagenomic tolerance genes can enable the flexible design of hardy microbial catalysts, customized to withstand inhibitors abundant in specific bioprocessing applications.


Asunto(s)
Bacterias/genética , Bacterias/aislamiento & purificación , Lignina/metabolismo , Metagenoma , Microbiología del Suelo , Bacterias/clasificación , Bacterias/metabolismo , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Suelo/química
13.
Chem Biol ; 22(7): 888-97, 2015 Jul 23.
Artículo en Inglés | MEDLINE | ID: mdl-26097034

RESUMEN

Enzymes capable of inactivating tetracycline are paradoxically rare compared with enzymes that inactivate other natural-product antibiotics. We describe a family of flavoenzymes, previously unrecognizable as resistance genes, which are capable of degrading tetracycline antibiotics. From soil functional metagenomic selections, we discovered nine genes that confer high-level tetracycline resistance by enzymatic inactivation. We also demonstrate that a tenth enzyme, an uncharacterized homolog in the human pathogen Legionella longbeachae, similarly inactivates tetracycline. These enzymes catalyze the oxidation of tetracyclines in vitro both by known mechanisms and via previously undescribed activity. Tetracycline-inactivation genes were identified in diverse soil types, encompass substantial sequence diversity, and are adjacent to genes implicated in horizontal gene transfer. Because tetracycline inactivation is scarcely observed in hospitals, these enzymes may fill an empty niche in pathogenic organisms, and should therefore be monitored for their dissemination potential into the clinic.


Asunto(s)
Oxigenasas/genética , Oxigenasas/metabolismo , Resistencia a la Tetraciclina/genética , Tetraciclina/farmacocinética , Activación Metabólica , Escherichia coli/enzimología , Escherichia coli/genética , Datos de Secuencia Molecular , Microbiología del Suelo , Tetraciclina/farmacología
14.
ISME J ; 9(1): 207-16, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25003965

RESUMEN

Antibiotic resistance is a dire clinical problem with important ecological dimensions. While antibiotic resistance in human pathogens continues to rise at alarming rates, the impact of environmental resistance on human health is still unclear. To investigate the relationship between human-associated and environmental resistomes, we analyzed functional metagenomic selections for resistance against 18 clinically relevant antibiotics from soil and human gut microbiota as well as a set of multidrug-resistant cultured soil isolates. These analyses were enabled by Resfams, a new curated database of protein families and associated highly precise and accurate profile hidden Markov models, confirmed for antibiotic resistance function and organized by ontology. We demonstrate that the antibiotic resistance functions that give rise to the resistance profiles observed in environmental and human-associated microbial communities significantly differ between ecologies. Antibiotic resistance functions that most discriminate between ecologies provide resistance to ß-lactams and tetracyclines, two of the most widely used classes of antibiotics in the clinic and agriculture. We also analyzed the antibiotic resistance gene composition of over 6000 sequenced microbial genomes, revealing significant enrichment of resistance functions by both ecology and phylogeny. Together, our results indicate that environmental and human-associated microbial communities harbor distinct resistance genes, suggesting that antibiotic resistance functions are largely constrained by ecology.


Asunto(s)
Antibacterianos/farmacología , Bacterias/efectos de los fármacos , Farmacorresistencia Bacteriana/genética , Ecología , Bacterias/genética , Secuencia de Bases , Humanos , Metagenómica , Filogenia , Microbiología del Suelo
15.
Nature ; 509(7502): 612-6, 2014 May 29.
Artículo en Inglés | MEDLINE | ID: mdl-24847883

RESUMEN

Ancient and diverse antibiotic resistance genes (ARGs) have previously been identified from soil, including genes identical to those in human pathogens. Despite the apparent overlap between soil and clinical resistomes, factors influencing ARG composition in soil and their movement between genomes and habitats remain largely unknown. General metagenome functions often correlate with the underlying structure of bacterial communities. However, ARGs are proposed to be highly mobile, prompting speculation that resistomes may not correlate with phylogenetic signatures or ecological divisions. To investigate these relationships, we performed functional metagenomic selections for resistance to 18 antibiotics from 18 agricultural and grassland soils. The 2,895 ARGs we discovered were mostly new, and represent all major resistance mechanisms. We demonstrate that distinct soil types harbour distinct resistomes, and that the addition of nitrogen fertilizer strongly influenced soil ARG content. Resistome composition also correlated with microbial phylogenetic and taxonomic structure, both across and within soil types. Consistent with this strong correlation, mobility elements (genes responsible for horizontal gene transfer between bacteria such as transposases and integrases) syntenic with ARGs were rare in soil by comparison with sequenced pathogens, suggesting that ARGs may not transfer between soil bacteria as readily as is observed between human pathogens. Together, our results indicate that bacterial community composition is the primary determinant of soil ARG content, challenging previous hypotheses that horizontal gene transfer effectively decouples resistomes from phylogeny.


Asunto(s)
Bacterias/genética , Bacterias/aislamiento & purificación , Farmacorresistencia Microbiana/genética , Ecosistema , Metagenoma/genética , Filogenia , Microbiología del Suelo , Agricultura , Antibacterianos/farmacología , Bacterias/clasificación , Bacterias/efectos de los fármacos , Farmacorresistencia Microbiana/efectos de los fármacos , Fertilizantes , Transferencia de Gen Horizontal/genética , Genes Bacterianos/efectos de los fármacos , Genes Bacterianos/genética , Genoma Bacteriano/efectos de los fármacos , Genoma Bacteriano/genética , Integrasas/genética , Metagenoma/efectos de los fármacos , Metagenómica , Modelos Genéticos , Datos de Secuencia Molecular , Nitrógeno/metabolismo , Nitrógeno/farmacología , Sistemas de Lectura Abierta/genética , Poaceae/crecimiento & desarrollo , ARN Ribosómico 16S/genética , Sintenía/genética , Transposasas/genética
16.
PLoS One ; 8(11): e78822, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24236055

RESUMEN

Emerging antibiotic resistance threatens human health. Gut microbes are an epidemiologically important reservoir of resistance genes (resistome), yet prior studies indicate that the true diversity of gut-associated resistomes has been underestimated. To deeply characterize the pediatric gut-associated resistome, we created metagenomic recombinant libraries in an Escherichia coli host using fecal DNA from 22 healthy infants and children (most without recent antibiotic exposure), and performed functional selections for resistance to 18 antibiotics from eight drug classes. Resistance-conferring DNA fragments were sequenced (Illumina HiSeq 2000), and reads assembled and annotated with the PARFuMS computational pipeline. Resistance to 14 of the 18 antibiotics was found in stools of infants and children. Recovered genes included chloramphenicol acetyltransferases, drug-resistant dihydrofolate reductases, rRNA methyltransferases, transcriptional regulators, multidrug efflux pumps, and every major class of beta-lactamase, aminoglycoside-modifying enzyme, and tetracycline resistance protein. Many resistance-conferring sequences were mobilizable; some had low identity to any known organism, emphasizing cryptic organisms as potentially important resistance reservoirs. We functionally confirmed three novel resistance genes, including a 16S rRNA methylase conferring aminoglycoside resistance, and two tetracycline-resistance proteins nearly identical to a bifidobacterial MFS transporter (B. longum s. longum JDM301). We provide the first report to our knowledge of resistance to folate-synthesis inhibitors conferred by a predicted Nudix hydrolase (part of the folate synthesis pathway). This functional metagenomic survey of gut-associated resistomes, the largest of its kind to date, demonstrates that fecal resistomes of healthy children are far more diverse than previously suspected, that clinically relevant resistance genes are present even without recent selective antibiotic pressure in the human host, and that cryptic gut microbes are an important resistance reservoir. The observed transferability of gut-associated resistance genes to a gram-negative (E. coli) host also suggests that the potential for gut-associated resistomes to threaten human health by mediating antibiotic resistance in pathogens warrants further investigation.


Asunto(s)
Farmacorresistencia Bacteriana/genética , Microbiota/genética , Adolescente , Antibacterianos/farmacología , Bacteroides/genética , Niño , Preescolar , Clostridium/genética , Enterobacter/genética , Heces/microbiología , Femenino , Genes Bacterianos , Humanos , Lactante , Funciones de Verosimilitud , Masculino , Anotación de Secuencia Molecular , Filogenia , Análisis de Secuencia de ADN , Adulto Joven
17.
Front Microbiol ; 4: 145, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23760651

RESUMEN

Rates of infection with antibiotic-resistant bacteria have increased precipitously over the past several decades, with far-reaching healthcare and societal costs. Recent evidence has established a link between antibiotic resistance genes in human pathogens and those found in non-pathogenic, commensal, and environmental organisms, prompting deeper investigation of natural and human-associated reservoirs of antibiotic resistance. Functional metagenomic selections, in which shotgun-cloned DNA fragments are selected for their ability to confer survival to an indicator host, have been increasingly applied to the characterization of many antibiotic resistance reservoirs. These experiments have demonstrated that antibiotic resistance genes are highly diverse and widely distributed, many times bearing little to no similarity to known sequences. Through unbiased selections for survival to antibiotic exposure, functional metagenomics can improve annotations by reducing the discovery of false-positive resistance and by allowing for the identification of previously unrecognizable resistance genes. In this review, we summarize the novel resistance functions uncovered using functional metagenomic investigations of natural and human-impacted resistance reservoirs. Examples of novel antibiotic resistance genes include those highly divergent from known sequences, those for which sequence is entirely unable to predict resistance function, bifunctional resistance genes, and those with unconventional, atypical resistance mechanisms. Overcoming antibiotic resistance in the clinic will require a better understanding of existing resistance reservoirs and the dissemination networks that govern horizontal gene exchange, informing best practices to limit the spread of resistance-conferring genes to human pathogens.

18.
Science ; 337(6098): 1107-11, 2012 Aug 31.
Artículo en Inglés | MEDLINE | ID: mdl-22936781

RESUMEN

Soil microbiota represent one of the ancient evolutionary origins of antibiotic resistance and have been proposed as a reservoir of resistance genes available for exchange with clinical pathogens. Using a high-throughput functional metagenomic approach in conjunction with a pipeline for the de novo assembly of short-read sequence data from functional selections (termed PARFuMS), we provide evidence for recent exchange of antibiotic resistance genes between environmental bacteria and clinical pathogens. We describe multidrug-resistant soil bacteria containing resistance cassettes against five classes of antibiotics (ß-lactams, aminoglycosides, amphenicols, sulfonamides, and tetracyclines) that have perfect nucleotide identity to genes from diverse human pathogens. This identity encompasses noncoding regions as well as multiple mobilization sequences, offering not only evidence of lateral exchange but also a mechanism by which antibiotic resistance disseminates.


Asunto(s)
Antibacterianos/farmacología , Bacterias/efectos de los fármacos , Bacterias/genética , Farmacorresistencia Bacteriana Múltiple/genética , Metagenoma/efectos de los fármacos , Metagenoma/genética , Microbiología del Suelo , Aminoglicósidos/farmacología , Bacterias/patogenicidad , Secuencia de Bases , Ensayos Analíticos de Alto Rendimiento , Humanos , Metagenómica , Datos de Secuencia Molecular , Sulfonamidas/farmacología , Tetraciclinas/farmacología , beta-Lactamas/farmacología
19.
Nat Biotechnol ; 28(10): 1097-105, 2010 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-20852635

RESUMEN

Analysis of DNA methylation patterns relies increasingly on sequencing-based profiling methods. The four most frequently used sequencing-based technologies are the bisulfite-based methods MethylC-seq and reduced representation bisulfite sequencing (RRBS), and the enrichment-based techniques methylated DNA immunoprecipitation sequencing (MeDIP-seq) and methylated DNA binding domain sequencing (MBD-seq). We applied all four methods to biological replicates of human embryonic stem cells to assess their genome-wide CpG coverage, resolution, cost, concordance and the influence of CpG density and genomic context. The methylation levels assessed by the two bisulfite methods were concordant (their difference did not exceed a given threshold) for 82% for CpGs and 99% of the non-CpG cytosines. Using binary methylation calls, the two enrichment methods were 99% concordant and regions assessed by all four methods were 97% concordant. We combined MeDIP-seq with methylation-sensitive restriction enzyme (MRE-seq) sequencing for comprehensive methylome coverage at lower cost. This, along with RNA-seq and ChIP-seq of the ES cells enabled us to detect regions with allele-specific epigenetic states, identifying most known imprinted regions and new loci with monoallelic epigenetic marks and monoallelic expression.


Asunto(s)
Alelos , Metilación de ADN/genética , Epigénesis Genética , Análisis de Secuencia de ADN/métodos , Línea Celular , Islas de CpG/genética , Citosina/metabolismo , Células Madre Embrionarias/metabolismo , Regulación de la Expresión Génica , Humanos , Sulfitos/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA