Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 37
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Microbiology (Reading) ; 167(4)2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33871329

RESUMEN

Membrane vesicles (MVs) are membrane-bound spherical nanostructures that prevail in all three domains of life. In Gram-negative bacteria, MVs are thought to be produced through blebbing of the outer membrane and are often referred to as outer membrane vesicles (OMVs). We have recently described another mechanism of MV formation in Pseudomonas aeruginosa that involves explosive cell-lysis events, which shatters cellular membranes into fragments that rapidly anneal into MVs. Interestingly, MVs are often observed within preparations of lytic bacteriophage, however the source of these MVs and their association with bacteriophage infection has not been explored. In this study we aimed to determine if MV formation is associated with lytic bacteriophage infection. Live super-resolution microscopy demonstrated that explosive cell lysis of Escherichia coli cells infected with either bacteriophage T4 or T7, resulted in the formation of MVs derived from shattered membrane fragments. Infection by either bacteriophage was also associated with the formation of membrane blebs on intact bacteria. TEM revealed multiple classes of MVs within phage lysates, consistent with multiple mechanisms of MV formation. These findings suggest that bacteriophage infection may be a major contributor to the abundance of bacterial MVs in nature.


Asunto(s)
Bacteriófagos/fisiología , Membrana Celular/virología , Escherichia coli/virología , Vesículas Extracelulares/metabolismo , Membrana Celular/genética , Membrana Celular/metabolismo , Escherichia coli/genética , Escherichia coli/metabolismo , Vesículas Extracelulares/genética
2.
3.
Proc Natl Acad Sci U S A ; 111(15): 5694-9, 2014 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-24706808

RESUMEN

Escherichia coli sequence type 131 (ST131) is a globally disseminated, multidrug resistant (MDR) clone responsible for a high proportion of urinary tract and bloodstream infections. The rapid emergence and successful spread of E. coli ST131 is strongly associated with several factors, including resistance to fluoroquinolones, high virulence gene content, the possession of the type 1 fimbriae FimH30 allele, and the production of the CTX-M-15 extended spectrum ß-lactamase (ESBL). Here, we used genome sequencing to examine the molecular epidemiology of a collection of E. coli ST131 strains isolated from six distinct geographical locations across the world spanning 2000-2011. The global phylogeny of E. coli ST131, determined from whole-genome sequence data, revealed a single lineage of E. coli ST131 distinct from other extraintestinal E. coli strains within the B2 phylogroup. Three closely related E. coli ST131 sublineages were identified, with little association to geographic origin. The majority of single-nucleotide variants associated with each of the sublineages were due to recombination in regions adjacent to mobile genetic elements (MGEs). The most prevalent sublineage of ST131 strains was characterized by fluoroquinolone resistance, and a distinct virulence factor and MGE profile. Four different variants of the CTX-M ESBL-resistance gene were identified in our ST131 strains, with acquisition of CTX-M-15 representing a defining feature of a discrete but geographically dispersed ST131 sublineage. This study confirms the global dispersal of a single E. coli ST131 clone and demonstrates the role of MGEs and recombination in the evolution of this important MDR pathogen.


Asunto(s)
Farmacorresistencia Bacteriana Múltiple/genética , Escherichia coli/genética , Genoma Bacteriano/genética , Filogenia , Secuencia de Bases , Biología Computacional , Fluoroquinolonas , Funciones de Verosimilitud , Modelos Genéticos , Datos de Secuencia Molecular , Filogeografía , Polimorfismo de Nucleótido Simple/genética , Alineación de Secuencia , Análisis de Secuencia de ADN , Especificidad de la Especie , beta-Lactamasas/metabolismo
4.
Proc Natl Acad Sci U S A ; 111(18): 6768-73, 2014 May 06.
Artículo en Inglés | MEDLINE | ID: mdl-24753568

RESUMEN

The genus Yersinia has been used as a model system to study pathogen evolution. Using whole-genome sequencing of all Yersinia species, we delineate the gene complement of the whole genus and define patterns of virulence evolution. Multiple distinct ecological specializations appear to have split pathogenic strains from environmental, nonpathogenic lineages. This split demonstrates that contrary to hypotheses that all pathogenic Yersinia species share a recent common pathogenic ancestor, they have evolved independently but followed parallel evolutionary paths in acquiring the same virulence determinants as well as becoming progressively more limited metabolically. Shared virulence determinants are limited to the virulence plasmid pYV and the attachment invasion locus ail. These acquisitions, together with genomic variations in metabolic pathways, have resulted in the parallel emergence of related pathogens displaying an increasingly specialized lifestyle with a spectrum of virulence potential, an emerging theme in the evolution of other important human pathogens.


Asunto(s)
Evolución Molecular , Virulencia/genética , Yersinia/genética , Yersinia/patogenicidad , Genoma Bacteriano , Humanos , Redes y Vías Metabólicas/genética , Filogenia , Especificidad de la Especie , Yersinia/metabolismo , Yersinia enterocolitica/genética , Yersinia enterocolitica/metabolismo , Yersinia enterocolitica/patogenicidad
5.
Infect Immun ; 84(3): 775-81, 2016 Jan 04.
Artículo en Inglés | MEDLINE | ID: mdl-26729762

RESUMEN

Enterohemorrhagic Escherichia coli (EHEC) is an important cause of diarrhea and hemolytic-uremic syndrome (HUS) worldwide. Australia's worst outbreak of HUS occurred in Adelaide in 1995 and was one of the first major HUS outbreaks attributed to a non-O157 Shiga-toxigenic E. coli (STEC) strain. Molecular analyses conducted at the time suggested that the outbreak was caused by an O111:H(-) clone, with strains from later in the outbreak harboring an extra copy of the genes encoding the potent Shiga toxin 2 (Stx2). Two decades later, we have used next-generation sequencing to compare two isolates from early and late in this important outbreak. We analyzed genetic content, single-nucleotide polymorphisms (SNPs), and prophage insertion sites; for the latter, we demonstrate how paired-end sequence data can be leveraged to identify such insertion sites. The two strains are genetically identical except for six SNP differences and the presence of not one but two additional Stx2-converting prophages in the later isolate. Isolates from later in the outbreak were associated with higher levels of morbidity, suggesting that the presence of the additional Stx2-converting prophages is significant in terms of the virulence of this clone.


Asunto(s)
Escherichia coli Enterohemorrágica/genética , Infecciones por Escherichia coli/microbiología , Síndrome Hemolítico-Urémico/microbiología , Australia/epidemiología , Brotes de Enfermedades , Escherichia coli Enterohemorrágica/clasificación , Escherichia coli Enterohemorrágica/aislamiento & purificación , Infecciones por Escherichia coli/epidemiología , Proteínas de Escherichia coli/química , Proteínas de Escherichia coli/genética , Proteínas de Escherichia coli/metabolismo , Genoma Bacteriano , Genómica , Humanos , Filogenia
6.
Antimicrob Agents Chemother ; 60(7): 4082-8, 2016 07.
Artículo en Inglés | MEDLINE | ID: mdl-27114281

RESUMEN

blaNDM genes confer carbapenem resistance and have been identified on transferable plasmids belonging to different incompatibility (Inc) groups. Here we present the complete sequences of four plasmids carrying a blaNDM gene, pKP1-NDM-1, pEC2-NDM-3, pECL3-NDM-1, and pEC4-NDM-6, from four clinical samples originating from four different patients. Different plasmids carry segments that align to different parts of the blaNDM region found on Acinetobacter plasmids. pKP1-NDM-1 and pEC2-NDM-3, from Klebsiella pneumoniae and Escherichia coli, respectively, were identified as type 1 IncA/C2 plasmids with almost identical backbones. Different regions carrying blaNDM are inserted in different locations in the antibiotic resistance island known as ARI-A, and ISCR1 may have been involved in the acquisition of blaNDM-3 by pEC2-NDM-3. pECL3-NDM-1 and pEC4-NDM-6, from Enterobacter cloacae and E. coli, respectively, have similar IncFIIY backbones, but different regions carrying blaNDM are found in different locations. Tn3-derived inverted-repeat transposable elements (TIME) appear to have been involved in the acquisition of blaNDM-6 by pEC4-NDM-6 and the rmtC 16S rRNA methylase gene by IncFIIY plasmids. Characterization of these plasmids further demonstrates that even very closely related plasmids may have acquired blaNDM genes by different mechanisms. These findings also illustrate the complex relationships between antimicrobial resistance genes, transposable elements, and plasmids and provide insights into the possible routes for transmission of blaNDM genes among species of the Enterobacteriaceae family.


Asunto(s)
Plásmidos/genética , Acinetobacter/efectos de los fármacos , Acinetobacter/genética , Antibacterianos/farmacología , Carbapenémicos/farmacología , Elementos Transponibles de ADN/genética , Farmacorresistencia Bacteriana Múltiple/genética , Enterobacter cloacae/efectos de los fármacos , Enterobacter cloacae/genética , Escherichia coli/efectos de los fármacos , Escherichia coli/genética , Klebsiella pneumoniae/efectos de los fármacos , Klebsiella pneumoniae/genética , Pruebas de Sensibilidad Microbiana , ARN Ribosómico 16S/genética , beta-Lactamasas/genética
7.
Infect Immun ; 83(4): 1384-95, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25624355

RESUMEN

The evolution of pathogenic bacteria is a multifaceted and complex process, which is strongly influenced by the horizontal acquisition of genetic elements and their subsequent expression in their new hosts. A well-studied example is the RegA regulon of the enteric pathogen Citrobacter rodentium. The RegA regulatory protein is a member of the AraC/XylS superfamily, which coordinates the expression of a gene repertoire that is necessary for full pathogenicity of this murine pathogen. Upon stimulation by an exogenous, gut-associated signal, namely, bicarbonate ions, RegA activates the expression of a series of genes, including virulence factors, such as autotransporters, fimbriae, a dispersin-like protein, and the grlRA operon on the locus of enterocyte effacement pathogenicity island. Interestingly, the genes encoding RegA homologues are distributed across the genus Escherichia, encompassing pathogenic and nonpathogenic subtypes. In this study, we carried out a series of bioinformatic, transcriptional, and functional analyses of the RegA regulons of these bacteria. Our results demonstrated that regA has been horizontally transferred to Escherichia spp. and C. rodentium. Comparative studies of two RegA homologues, namely, those from C. rodentium and E. coli SMS-3-5, a multiresistant environmental strain of E. coli, showed that the two regulators acted similarly in vitro but differed in terms of their abilities to activate the virulence of C. rodentium in vivo, which evidently was due to their differential activation of grlRA. Our data indicate that RegA from C. rodentium has strain-specific adaptations that facilitate infection of its murine host. These findings shed new light on the development of virulence by C. rodentium and on the evolution of virulence-regulatory genes of bacterial pathogens in general.


Asunto(s)
Factor de Transcripción de AraC/genética , Proteínas Bacterianas/genética , Citrobacter rodentium/genética , Citrobacter rodentium/patogenicidad , Proteínas de Escherichia coli/genética , Escherichia coli/genética , Animales , Evolución Biológica , Escherichia coli/patogenicidad , Regulación Bacteriana de la Expresión Génica , Transferencia de Gen Horizontal , Masculino , Ratones , Ratones Endogámicos C57BL , Fosfoproteínas/genética , Filogenia , Proteínas Represoras/genética , Factores de Virulencia/genética
8.
Infect Immun ; 83(5): 1749-64, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25667270

RESUMEN

Urinary tract infections (UTIs) are among the most common infectious diseases of humans, with Escherichia coli responsible for >80% of all cases. One extreme of UTI is asymptomatic bacteriuria (ABU), which occurs as an asymptomatic carrier state that resembles commensalism. To understand the evolution and molecular mechanisms that underpin ABU, the genome of the ABU E. coli strain VR50 was sequenced. Analysis of the complete genome indicated that it most resembles E. coli K-12, with the addition of a 94-kb genomic island (GI-VR50-pheV), eight prophages, and multiple plasmids. GI-VR50-pheV has a mosaic structure and contains genes encoding a number of UTI-associated virulence factors, namely, Afa (afimbrial adhesin), two autotransporter proteins (Ag43 and Sat), and aerobactin. We demonstrated that the presence of this island in VR50 confers its ability to colonize the murine bladder, as a VR50 mutant with GI-VR50-pheV deleted was attenuated in a mouse model of UTI in vivo. We established that Afa is the island-encoded factor responsible for this phenotype using two independent deletion (Afa operon and AfaE adhesin) mutants. E. coli VR50afa and VR50afaE displayed significantly decreased ability to adhere to human bladder epithelial cells. In the mouse model of UTI, VR50afa and VR50afaE displayed reduced bladder colonization compared to wild-type VR50, similar to the colonization level of the GI-VR50-pheV mutant. Our study suggests that E. coli VR50 is a commensal-like strain that has acquired fitness factors that facilitate colonization of the human bladder.


Asunto(s)
Adaptación Biológica , Bacteriuria/microbiología , Portador Sano/microbiología , Infecciones por Escherichia coli/microbiología , Escherichia coli/genética , Evolución Molecular , Sistema Urinario/microbiología , Adulto , Animales , Adhesión Bacteriana , Línea Celular , ADN Bacteriano/química , ADN Bacteriano/genética , Células Epiteliales/microbiología , Escherichia coli/aislamiento & purificación , Femenino , Genoma Bacteriano , Humanos , Ratones Endogámicos C57BL , Modelos Animales , Datos de Secuencia Molecular , Análisis de Secuencia de ADN
9.
BMC Genomics ; 15: 389, 2014 May 21.
Artículo en Inglés | MEDLINE | ID: mdl-24885207

RESUMEN

BACKGROUND: Salmonella enterica subsp. enterica serovar Virchow has been recognized as a significant health burden in Asia, Australia and Europe. In addition to its global distribution, S. Virchow is clinically significant due to the frequency at which it causes invasive infections and its association with outbreaks arising from food-borne transmission. Here, we examine the genome of an invasive isolate of S. Virchow SVQ1 (phage type 8) from an outbreak in southeast Queensland, Australia. In addition to identifying new potential genotyping targets that could be used for discriminating between S. Virchow strains in outbreak scenarios, we also aimed to carry out a comprehensive comparative analysis of the S. Virchow genomes. RESULTS: Genome comparisons between S. Virchow SVQ1 and S. Virchow SL491, a previously published strain, identified a high degree of genomic similarity between the two strains with fewer than 200 single nucleotide differences. Clustered Regularly Interspaced Palindromic Repeats (CRISPR) regions were identified as a highly variable region that could be used to discriminate between S. Virchow isolates. We amplified and sequenced the CRISPR regions of fifteen S. Virchow isolates collected from seven different outbreaks across Australia. We observed three allelic types of the CRISPR region from these isolates based on the presence/absence of the spacers and were able to discriminate S. Virchow phage type 8 isolates originating from different outbreaks. A comparison with 27 published Salmonella genomes found that the S. Virchow SVQ1 genome encodes 11 previously described Salmonella Pathogenicity Islands (SPI), as well as additional genomic islands including a remnant integrative conjugative element that is distinct from SPI-7. In addition, the S. Virchow genome possesses a novel prophage that encodes the Type III secretion system effector protein SopE, a key Salmonella virulence factor. The prophage shares very little similarity to the SopE prophages found in other Salmonella serovars suggesting an independent acquisition of sopE. CONCLUSIONS: The availability of this genome will serve as a genome template and facilitate further studies on understanding the virulence and global distribution of the S. Virchow serovar, as well as the development of genotyping methods for outbreak investigations.


Asunto(s)
Repeticiones Palindrómicas Cortas Agrupadas y Regularmente Espaciadas/genética , Genoma Bacteriano , Salmonella enterica/genética , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Secuencia de Bases , Mapeo Cromosómico , Análisis por Conglomerados , Genotipo , Datos de Secuencia Molecular , Salmonella enterica/aislamiento & purificación
10.
Appl Environ Microbiol ; 80(7): 2216-28, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24487530

RESUMEN

Pseudomonas syringae pv. actinidiae is a reemerging pathogen which causes bacterial canker of kiwifruit (Actinidia sp.). Since 2008, a global outbreak of P. syringae pv. actinidiae has occurred, and in 2010 this pathogen was detected in New Zealand. The economic impact and the development of resistance in P. syringae pv. actinidiae and other pathovars against antibiotics and copper sprays have led to a search for alternative management strategies. We isolated 275 phages, 258 of which were active against P. syringae pv. actinidiae. Extensive host range testing on P. syringae pv. actinidiae, other pseudomonads, and bacteria isolated from kiwifruit orchards showed that most phages have a narrow host range. Twenty-four were analyzed by electron microscopy, pulse-field gel electrophoresis, and restriction digestion. Their suitability for biocontrol was tested by assessing stability and the absence of lysogeny and transduction. A detailed host range was performed, phage-resistant bacteria were isolated, and resistance to other phages was examined. The phages belonged to the Caudovirales and were analyzed based on morphology and genome size, which showed them to be representatives of Myoviridae, Podoviridae, and Siphoviridae. Twenty-one Myoviridae members have similar morphologies and genome sizes yet differ in restriction patterns, host range, and resistance, indicating a closely related group. Nine of these Myoviridae members were sequenced, and each was unique. The most closely related sequenced phages were a group infecting Pseudomonas aeruginosa and characterized by phages JG004 and PAK_P1. In summary, this study reports the isolation and characterization of P. syringae pv. actinidiae phages and provides a framework for the intelligent formulation of phage biocontrol agents against kiwifruit bacterial canker.


Asunto(s)
Caudovirales/aislamiento & purificación , ADN Viral/genética , Especificidad del Huésped , Fagos Pseudomonas/aislamiento & purificación , Pseudomonas syringae/virología , Virión/ultraestructura , Actinidia/microbiología , Caudovirales/genética , Caudovirales/fisiología , Caudovirales/ultraestructura , ADN Viral/química , Electroforesis en Gel de Campo Pulsado , Datos de Secuencia Molecular , Nueva Zelanda , Enfermedades de las Plantas/microbiología , Polimorfismo de Longitud del Fragmento de Restricción , Fagos Pseudomonas/genética , Fagos Pseudomonas/fisiología , Fagos Pseudomonas/ultraestructura , Análisis de Secuencia de ADN
11.
PLoS Pathog ; 7(8): e1002191, 2011 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-21876672

RESUMEN

The genus Salmonella contains two species, S. bongori and S. enterica. Compared to the well-studied S. enterica there is a marked lack of information regarding the genetic makeup and diversity of S. bongori. S. bongori has been found predominantly associated with cold-blooded animals, but it can infect humans. To define the phylogeny of this species, and compare it to S. enterica, we have sequenced 28 isolates representing most of the known diversity of S. bongori. This cross-species analysis allowed us to confidently differentiate ancestral functions from those acquired following speciation, which include both metabolic and virulence-associated capacities. We show that, although S. bongori inherited a basic set of Salmonella common virulence functions, it has subsequently elaborated on this in a different direction to S. enterica. It is an established feature of S. enterica evolution that the acquisition of the type III secretion systems (T3SS-1 and T3SS-2) has been followed by the sequential acquisition of genes encoding secreted targets, termed effectors proteins. We show that this is also true of S. bongori, which has acquired an array of novel effector proteins (sboA-L). All but two of these effectors have no significant S. enterica homologues and instead are highly similar to those found in enteropathogenic Escherichia coli (EPEC). Remarkably, SboH is found to be a chimeric effector protein, encoded by a fusion of the T3SS-1 effector gene sopA and a gene highly similar to the EPEC effector nleH from enteropathogenic E. coli. We demonstrate that representatives of these new effectors are translocated and that SboH, similarly to NleH, blocks intrinsic apoptotic pathways while being targeted to the mitochondria by the SopA part of the fusion. This work suggests that S. bongori has inherited the ancestral Salmonella virulence gene set, but has adapted by incorporating virulence determinants that resemble those employed by EPEC.


Asunto(s)
Evolución Biológica , Salmonella/genética , Animales , Escherichia coli Enteropatógena/genética , Genes Bacterianos , Islas Genómicas/genética , Humanos , Filogenia , Salmonella enterica/genética , Análisis de Secuencia de ADN , Translocación Genética , Virulencia/genética , Factores de Virulencia/genética
12.
PLoS Pathog ; 7(4): e1002018, 2011 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21490962

RESUMEN

Citrobacter rodentium is a natural mouse pathogen that causes attaching and effacing (A/E) lesions. It shares a common virulence strategy with the clinically significant human A/E pathogens enteropathogenic E. coli (EPEC) and enterohaemorrhagic E. coli (EHEC) and is widely used to model this route of pathogenesis. We previously reported the complete genome sequence of C. rodentium ICC168, where we found that the genome displayed many characteristics of a newly evolved pathogen. In this study, through PFGE, sequencing of isolates showing variation, whole genome transcriptome analysis and examination of the mobile genetic elements, we found that, consistent with our previous hypothesis, the genome of C. rodentium is unstable as a result of repeat-mediated, large-scale genome recombination and because of active transposition of mobile genetic elements such as the prophages. We sequenced an additional C. rodentium strain, EX-33, to reveal that the reference strain ICC168 is representative of the species and that most of the inactivating mutations were common to both isolates and likely to have occurred early on in the evolution of this pathogen. We draw parallels with the evolution of other bacterial pathogens and conclude that C. rodentium is a recently evolved pathogen that may have emerged alongside the development of inbred mice as a model for human disease.


Asunto(s)
Citrobacter rodentium/genética , Citrobacter rodentium/patogenicidad , Genoma Bacteriano , Animales , Citrobacter rodentium/clasificación , ADN Bacteriano/genética , Modelos Animales de Enfermedad , Electroforesis en Gel de Campo Pulsado , Infecciones por Enterobacteriaceae/patología , Femenino , Perfilación de la Expresión Génica , Reordenamiento Génico , Inestabilidad Genómica , Humanos , Secuencias Repetitivas Esparcidas , Ratones , Ratones Endogámicos C57BL , Plásmidos/genética , Profagos/genética , Análisis de Secuencia de ADN , Virulencia
14.
Bioinformatics ; 27(7): 1009-10, 2011 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-21278367

RESUMEN

UNLABELLED: Easyfig is a Python application for creating linear comparison figures of multiple genomic loci with an easy-to-use graphical user interface. BLAST comparisons between multiple genomic regions, ranging from single genes to whole prokaryote chromosomes, can be generated, visualized and interactively coloured, enabling a rapid transition between analysis and the preparation of publication quality figures. AVAILABILITY: Easyfig is freely available (under a GPL license) for download (for Mac OS X, Unix and Microsoft Windows) from the SourceForge web site: http://easyfig.sourceforge.net/.


Asunto(s)
Gráficos por Computador , Genómica/métodos , Programas Informáticos , Genoma
15.
BMC Genomics ; 12: 402, 2011 Aug 08.
Artículo en Inglés | MEDLINE | ID: mdl-21824423

RESUMEN

BACKGROUND: Visualisation of genome comparisons is invaluable for helping to determine genotypic differences between closely related prokaryotes. New visualisation and abstraction methods are required in order to improve the validation, interpretation and communication of genome sequence information; especially with the increasing amount of data arising from next-generation sequencing projects. Visualising a prokaryote genome as a circular image has become a powerful means of displaying informative comparisons of one genome to a number of others. Several programs, imaging libraries and internet resources already exist for this purpose, however, most are either limited in the number of comparisons they can show, are unable to adequately utilise draft genome sequence data, or require a knowledge of command-line scripting for implementation. Currently, there is no freely available desktop application that enables users to rapidly visualise comparisons between hundreds of draft or complete genomes in a single image. RESULTS: BLAST Ring Image Generator (BRIG) can generate images that show multiple prokaryote genome comparisons, without an arbitrary limit on the number of genomes compared. The output image shows similarity between a central reference sequence and other sequences as a set of concentric rings, where BLAST matches are coloured on a sliding scale indicating a defined percentage identity. Images can also include draft genome assembly information to show read coverage, assembly breakpoints and collapsed repeats. In addition, BRIG supports the mapping of unassembled sequencing reads against one or more central reference sequences. Many types of custom data and annotations can be shown using BRIG, making it a versatile approach for visualising a range of genomic comparison data. BRIG is readily accessible to any user, as it assumes no specialist computational knowledge and will perform all required file parsing and BLAST comparisons automatically. CONCLUSIONS: There is a clear need for a user-friendly program that can produce genome comparisons for a large number of prokaryote genomes with an emphasis on rapidly utilising unfinished or unassembled genome data. Here we present BRIG, a cross-platform application that enables the interactive generation of comparative genomic images via a simple graphical-user interface. BRIG is freely available for all operating systems at http://sourceforge.net/projects/brig/.


Asunto(s)
Gráficos por Computador , Genómica/métodos , Programas Informáticos , Genoma Arqueal/genética , Genoma Bacteriano/genética , Interfaz Usuario-Computador
16.
J Bacteriol ; 192(21): 5746-54, 2010 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-20817773

RESUMEN

A number of bacteriophages have been identified that target the Vi capsular antigen of Salmonella enterica serovar Typhi. Here we show that these Vi phages represent a remarkably diverse set of phages belonging to three phage families, including Podoviridae and Myoviridae. Genome analysis facilitated the further classification of these phages and highlighted aspects of their independent evolution. Significantly, a conserved protein domain carrying an acetyl esterase was found to be associated with at least one tail fiber gene for all Vi phages, and the presence of this domain was confirmed in representative phage particles by mass spectrometric analysis. Thus, we provide a simple explanation and paradigm of how a diverse group of phages target a single key virulence antigen associated with this important human-restricted pathogen.


Asunto(s)
Acetilesterasa/metabolismo , Regulación Viral de la Expresión Génica/fisiología , Polisacáridos Bacterianos/fisiología , Fagos de Salmonella/metabolismo , Salmonella typhi/metabolismo , Acetilesterasa/genética , Genoma Viral , Datos de Secuencia Molecular , Estructura Terciaria de Proteína , Fagos de Salmonella/genética , Sintenía , Proteínas Virales/genética , Proteínas Virales/metabolismo
17.
J Bacteriol ; 192(22): 6001-16, 2010 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-20833813

RESUMEN

Legionella pneumophila is a ubiquitous inhabitant of environmental water reservoirs. The bacteria infect a wide variety of protozoa and, after accidental inhalation, human alveolar macrophages, which can lead to severe pneumonia. The capability to thrive in phagocytic hosts is dependent on the Dot/Icm type IV secretion system (T4SS), which translocates multiple effector proteins into the host cell. In this study, we determined the draft genome sequence of L. pneumophila strain 130b (Wadsworth). We found that the 130b genome encodes a unique set of T4SSs, namely, the Dot/Icm T4SS, a Trb-1-like T4SS, and two Lvh T4SS gene clusters. Sequence analysis substantiated that a core set of 107 Dot/Icm T4SS effectors was conserved among the sequenced L. pneumophila strains Philadelphia-1, Lens, Paris, Corby, Alcoy, and 130b. We also identified new effector candidates and validated the translocation of 10 novel Dot/Icm T4SS effectors that are not present in L. pneumophila strain Philadelphia-1. We examined the prevalence of the new effector genes among 87 environmental and clinical L. pneumophila isolates. Five of the new effectors were identified in 34 to 62% of the isolates, while less than 15% of the strains tested positive for the other five genes. Collectively, our data show that the core set of conserved Dot/Icm T4SS effector proteins is supplemented by a variable repertoire of accessory effectors that may partly account for differences in the virulences and prevalences of particular L. pneumophila strains.


Asunto(s)
ADN Bacteriano/química , Genoma Bacteriano , Legionella pneumophila/genética , Proteínas de Transporte de Membrana/genética , Factores de Virulencia/genética , Secuencia Conservada , ADN Bacteriano/genética , Microbiología Ambiental , Humanos , Legionella pneumophila/aislamiento & purificación , Legionella pneumophila/metabolismo , Legionella pneumophila/patogenicidad , Enfermedad de los Legionarios/microbiología , Proteínas de Transporte de Membrana/metabolismo , Datos de Secuencia Molecular , Análisis de Secuencia de ADN , Homología de Secuencia de Aminoácido , Factores de Virulencia/metabolismo
18.
J Bacteriol ; 192(2): 525-38, 2010 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19897651

RESUMEN

Citrobacter rodentium (formally Citrobacter freundii biotype 4280) is a highly infectious pathogen that causes colitis and transmissible colonic hyperplasia in mice. In common with enteropathogenic and enterohemorrhagic Escherichia coli (EPEC and EHEC, respectively), C. rodentium exploits a type III secretion system (T3SS) to induce attaching and effacing (A/E) lesions that are essential for virulence. Here, we report the fully annotated genome sequence of the 5.3-Mb chromosome and four plasmids harbored by C. rodentium strain ICC168. The genome sequence revealed key information about the phylogeny of C. rodentium and identified 1,585 C. rodentium-specific (without orthologues in EPEC or EHEC) coding sequences, 10 prophage-like regions, and 17 genomic islands, including the locus for enterocyte effacement (LEE) region, which encodes a T3SS and effector proteins. Among the 29 T3SS effectors found in C. rodentium are all 22 of the core effectors of EPEC strain E2348/69. In addition, we identified a novel C. rodentium effector, named EspS. C. rodentium harbors two type VI secretion systems (T6SS) (CTS1 and CTS2), while EHEC contains only one T6SS (EHS). Our analysis suggests that C. rodentium and EPEC/EHEC have converged on a common host infection strategy through access to a common pool of mobile DNA and that C. rodentium has lost gene functions associated with a previous pathogenic niche.


Asunto(s)
Citrobacter rodentium/genética , Escherichia coli/genética , Evolución Molecular , Genoma Bacteriano/genética , Animales , Citrobacter rodentium/clasificación , Biología Computacional , Humanos , Masculino , Ratones , Datos de Secuencia Molecular , Filogenia
19.
J Bacteriol ; 192(21): 5822-31, 2010 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-20802035

RESUMEN

In most cases, Escherichia coli exists as a harmless commensal organism, but it may on occasion cause intestinal and/or extraintestinal disease. Enterotoxigenic E. coli (ETEC) is the predominant cause of E. coli-mediated diarrhea in the developing world and is responsible for a significant portion of pediatric deaths. In this study, we determined the complete genomic sequence of E. coli H10407, a prototypical strain of enterotoxigenic E. coli, which reproducibly elicits diarrhea in human volunteer studies. We performed genomic and phylogenetic comparisons with other E. coli strains, revealing that the chromosome is closely related to that of the nonpathogenic commensal strain E. coli HS and to those of the laboratory strains E. coli K-12 and C. Furthermore, these analyses demonstrated that there were no chromosomally encoded factors unique to any sequenced ETEC strains. Comparison of the E. coli H10407 plasmids with those from several ETEC strains revealed that the plasmids had a mosaic structure but that several loci were conserved among ETEC strains. This study provides a genetic context for the vast amount of experimental and epidemiological data that have been published.


Asunto(s)
Escherichia coli Enterotoxigénica/clasificación , Escherichia coli Enterotoxigénica/genética , Proteínas de Escherichia coli/metabolismo , Genoma Bacteriano , Secuencia de Aminoácidos , Cromosomas Bacterianos , Proteínas de Escherichia coli/genética , Proteínas Fimbrias , Perfilación de la Expresión Génica , Regulación Bacteriana de la Expresión Génica/fisiología , Datos de Secuencia Molecular , Plásmidos/genética
20.
BMC Genomics ; 11: 214, 2010 Mar 30.
Artículo en Inglés | MEDLINE | ID: mdl-20353581

RESUMEN

BACKGROUND: Our understanding of the dynamics of genome stability versus gene flux within bacteriophage lineages is limited. Recently, there has been a renewed interest in the use of bacteriophages as 'therapeutic' agents; a prerequisite for their use in such therapies is a thorough understanding of their genetic complement, genome stability and their ecology to avoid the dissemination or mobilisation of phage or bacterial virulence and toxin genes. Campylobacter, a food-borne pathogen, is one of the organisms for which the use of bacteriophage is being considered to reduce human exposure to this organism. RESULTS: Sequencing and genome analysis was performed for two Campylobacter bacteriophages. The genomes were extremely similar at the nucleotide level (> or = 96%) with most differences accounted for by novel insertion sequences, DNA methylases and an approximately 10 kb contiguous region of metabolic genes that were dissimilar at the sequence level but similar in gene function between the two phages. Both bacteriophages contained a large number of radical S-adenosylmethionine (SAM) genes, presumably involved in boosting host metabolism during infection, as well as evidence that many genes had been acquired from a wide range of bacterial species. Further bacteriophages, from the UK Campylobacter typing set, were screened for the presence of bacteriophage structural genes, DNA methylases, mobile genetic elements and regulatory genes identified from the genome sequences. The results indicate that many of these bacteriophages are related, with 10 out of 15 showing some relationship to the sequenced genomes. CONCLUSIONS: Two large virulent Campylobacter bacteriophages were found to show very high levels of sequence conservation despite separation in time and place of isolation. The bacteriophages show adaptations to their host and possess genes that may enhance Campylobacter metabolism, potentially advantaging both the bacteriophage and its host. Genetic conservation has been shown to extend to other Campylobacter bacteriophages, forming a highly conserved lineage of bacteriophages that predate upon campylobacters and indicating that highly adapted bacteriophage genomes can be stable over prolonged periods of time.


Asunto(s)
Bacteriófagos/genética , Campylobacter/virología , Bacteriófagos/patogenicidad , Secuencia Conservada , Genoma Viral , Análisis de Secuencia , Proteínas Estructurales Virales/genética , Virulencia
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA