Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 63
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Cell ; 185(16): 2936-2951.e19, 2022 08 04.
Artículo en Inglés | MEDLINE | ID: mdl-35931021

RESUMEN

We studied the prevalent cytotoxic CD8 T cell response mounted against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) Spike glycoprotein269-277 epitope (sequence YLQPRTFLL) via the most frequent human leukocyte antigen (HLA) class I worldwide, HLA A∗02. The Spike P272L mutation that has arisen in at least 112 different SARS-CoV-2 lineages to date, including in lineages classified as "variants of concern," was not recognized by the large CD8 T cell response seen across cohorts of HLA A∗02+ convalescent patients and individuals vaccinated against SARS-CoV-2, despite these responses comprising of over 175 different individual T cell receptors. Viral escape at prevalent T cell epitopes restricted by high frequency HLAs may be particularly problematic when vaccine immunity is focused on a single protein such as SARS-CoV-2 Spike, providing a strong argument for inclusion of multiple viral proteins in next generation vaccines and highlighting the need for monitoring T cell escape in new SARS-CoV-2 variants.


Asunto(s)
COVID-19 , SARS-CoV-2 , Linfocitos T CD8-positivos , Epítopos de Linfocito T , Antígenos HLA-A , Antígenos de Histocompatibilidad Clase I , Humanos
2.
Cell ; 184(20): 5179-5188.e8, 2021 09 30.
Artículo en Inglés | MEDLINE | ID: mdl-34499854

RESUMEN

We present evidence for multiple independent origins of recombinant SARS-CoV-2 viruses sampled from late 2020 and early 2021 in the United Kingdom. Their genomes carry single-nucleotide polymorphisms and deletions that are characteristic of the B.1.1.7 variant of concern but lack the full complement of lineage-defining mutations. Instead, the remainder of their genomes share contiguous genetic variation with non-B.1.1.7 viruses circulating in the same geographic area at the same time as the recombinants. In four instances, there was evidence for onward transmission of a recombinant-origin virus, including one transmission cluster of 45 sequenced cases over the course of 2 months. The inferred genomic locations of recombination breakpoints suggest that every community-transmitted recombinant virus inherited its spike region from a B.1.1.7 parental virus, consistent with a transmission advantage for B.1.1.7's set of mutations.


Asunto(s)
COVID-19/epidemiología , COVID-19/transmisión , Pandemias , Recombinación Genética , SARS-CoV-2/genética , Secuencia de Bases/genética , COVID-19/virología , Biología Computacional/métodos , Frecuencia de los Genes , Genoma Viral , Genotipo , Humanos , Mutación , Filogenia , Polimorfismo de Nucleótido Simple , Reino Unido/epidemiología , Secuenciación Completa del Genoma/métodos
3.
Cell ; 184(1): 64-75.e11, 2021 01 07.
Artículo en Inglés | MEDLINE | ID: mdl-33275900

RESUMEN

Global dispersal and increasing frequency of the SARS-CoV-2 spike protein variant D614G are suggestive of a selective advantage but may also be due to a random founder effect. We investigate the hypothesis for positive selection of spike D614G in the United Kingdom using more than 25,000 whole genome SARS-CoV-2 sequences. Despite the availability of a large dataset, well represented by both spike 614 variants, not all approaches showed a conclusive signal of positive selection. Population genetic analysis indicates that 614G increases in frequency relative to 614D in a manner consistent with a selective advantage. We do not find any indication that patients infected with the spike 614G variant have higher COVID-19 mortality or clinical severity, but 614G is associated with higher viral load and younger age of patients. Significant differences in growth and size of 614G phylogenetic clusters indicate a need for continued study of this variant.


Asunto(s)
Sustitución de Aminoácidos , COVID-19/transmisión , COVID-19/virología , SARS-CoV-2/genética , SARS-CoV-2/patogenicidad , Glicoproteína de la Espiga del Coronavirus/genética , Ácido Aspártico/análisis , Ácido Aspártico/genética , COVID-19/epidemiología , Genoma Viral , Glicina/análisis , Glicina/genética , Humanos , Mutación , SARS-CoV-2/crecimiento & desarrollo , Reino Unido/epidemiología , Virulencia , Secuenciación Completa del Genoma
4.
Nat Rev Genet ; 23(9): 547-562, 2022 09.
Artículo en Inglés | MEDLINE | ID: mdl-35459859

RESUMEN

Determining the transmissibility, prevalence and patterns of movement of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infections is central to our understanding of the impact of the pandemic and to the design of effective control strategies. Phylogenies (evolutionary trees) have provided key insights into the international spread of SARS-CoV-2 and enabled investigation of individual outbreaks and transmission chains in specific settings. Phylodynamic approaches combine evolutionary, demographic and epidemiological concepts and have helped track virus genetic changes, identify emerging variants and inform public health strategy. Here, we review and synthesize studies that illustrate how phylogenetic and phylodynamic techniques were applied during the first year of the pandemic, and summarize their contributions to our understanding of SARS-CoV-2 transmission and control.


Asunto(s)
COVID-19 , Pandemias , Humanos , Pandemias/prevención & control , Filogenia , SARS-CoV-2/genética
5.
Cell ; 148(4): 780-91, 2012 Feb 17.
Artículo en Inglés | MEDLINE | ID: mdl-22341448

RESUMEN

The Tasmanian devil (Sarcophilus harrisii), the largest marsupial carnivore, is endangered due to a transmissible facial cancer spread by direct transfer of living cancer cells through biting. Here we describe the sequencing, assembly, and annotation of the Tasmanian devil genome and whole-genome sequences for two geographically distant subclones of the cancer. Genomic analysis suggests that the cancer first arose from a female Tasmanian devil and that the clone has subsequently genetically diverged during its spread across Tasmania. The devil cancer genome contains more than 17,000 somatic base substitution mutations and bears the imprint of a distinct mutational process. Genotyping of somatic mutations in 104 geographically and temporally distributed Tasmanian devil tumors reveals the pattern of evolution and spread of this parasitic clonal lineage, with evidence of a selective sweep in one geographical area and persistence of parallel lineages in other populations.


Asunto(s)
Neoplasias Faciales/veterinaria , Inestabilidad Genómica , Marsupiales/genética , Mutación , Animales , Evolución Clonal , Especies en Peligro de Extinción , Neoplasias Faciales/epidemiología , Neoplasias Faciales/genética , Neoplasias Faciales/patología , Femenino , Estudio de Asociación del Genoma Completo , Masculino , Datos de Secuencia Molecular , Tasmania/epidemiología
6.
Mol Biol Evol ; 40(10)2023 Oct 04.
Artículo en Inglés | MEDLINE | ID: mdl-37738143

RESUMEN

The RNA-dependent RNA polymerase of the severe acute respiratory syndrome coronavirus 2 virus is error prone, with errors being corrected by the exonuclease (NSP14) proofreading mechanism. However, the mutagenesis and subsequent evolutionary trajectory of the virus is mediated by the delicate interplay of replicase fidelity and environmental pressures. Here, we have shown that a single, distal mutation (F60S) in NSP14 can have a profound impact upon proofreading with an increased accumulation of mutations and elevated evolutionary rate being observed. Understanding the implications of these changes is crucial, as these underlying mutational processes may have important implications for understanding the population-wide evolution of the virus. This study underscores the urgent need for continued research into the replicative mechanisms of this virus to combat its continued impact on global health, through the re-emergence of immuno-evasive variants.

7.
Microbiology (Reading) ; 169(8)2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-37526960

RESUMEN

Burkholderia multivorans is the dominant Burkholderia pathogen recovered from lung infection in people with cystic fibrosis. However, as an understudied pathogen there are knowledge gaps in relation to its population biology, phenotypic traits and useful model strains. A phylogenomic study of B. multivorans was undertaken using a total of 283 genomes, of which 73 were sequenced and 49 phenotypically characterized as part of this study. Average nucleotide identity analysis (ANI) and phylogenetic alignment of core genes demonstrated that the B. multivorans population separated into two distinct evolutionary clades, defined as lineage 1 (n=58 genomes) and lineage 2 (n=221 genomes). To examine the population biology of B. multivorans, a representative subgroup of 77 B. multivorans genomes (28 from the reference databases and the 49 novel short-read genome sequences) were selected based on multilocus sequence typing (MLST), isolation source and phylogenetic placement criteria. Comparative genomics was used to identify B. multivorans lineage-specific genes - ghrB_1 in lineage 1 and glnM_2 in lineage 2 - and diagnostic PCRs targeting them were successfully developed. Phenotypic analysis of 49 representative B. multivorans strains showed considerable inter-strain variance, but the majority of the isolates tested were motile and capable of biofilm formation. A striking absence of B. multivorans protease activity in vitro was observed, but no lineage-specific phenotypic differences were demonstrated. Using phylogenomic and phenotypic criteria, three model B. multivorans CF strains were identified, BCC0084 (lineage 1), BCC1272 (lineage 2a) and BCC0033 lineage 2b, and their complete genome sequences determined. B. multivorans CF strains BCC0033 and BCC0084, and the environmental reference strain, ATCC 17616, were all capable of short-term survival within a murine lung infection model. By mapping the population biology, identifying lineage-specific PCRs and model strains, we provide much needed baseline resources for future studies of B. multivorans.


Asunto(s)
Infecciones por Burkholderia , Burkholderia , Fibrosis Quística , Filogenia , Animales , Ratones , Burkholderia/clasificación , Burkholderia/genética , Infecciones por Burkholderia/complicaciones , Infecciones por Burkholderia/microbiología , Fibrosis Quística/complicaciones , Fibrosis Quística/microbiología , Tipificación de Secuencias Multilocus , Genoma Bacteriano/genética , Ratones Endogámicos BALB C , Femenino
8.
Bioinformatics ; 36(6): 1681-1688, 2020 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-31693070

RESUMEN

MOTIVATION: Influenza viruses represent a global public health burden due to annual epidemics and pandemic potential. Due to a rapidly evolving RNA genome, inter-species transmission, intra-host variation, and noise in short-read data, reads can be lost during mapping, and de novo assembly can be time consuming and result in misassembly. We assessed read loss during mapping and designed a graph-based classifier, VAPOR, for selecting mapping references, assembly validation and detection of strains of non-human origin. RESULTS: Standard human reference viruses were insufficient for mapping diverse influenza samples in simulation. VAPOR retrieved references for 257 real whole-genome sequencing samples with a mean of >99.8% identity to assemblies, and increased the proportion of mapped reads by up to 13.3% compared to standard references. VAPOR has the potential to improve the robustness of bioinformatics pipelines for surveillance and could be adapted to other RNA viruses. AVAILABILITY AND IMPLEMENTATION: VAPOR is available at https://github.com/connor-lab/vapor. SUPPLEMENTARY INFORMATION: Supplementary data are available at Bioinformatics online.


Asunto(s)
Secuenciación de Nucleótidos de Alto Rendimiento , Gripe Humana , Algoritmos , Genoma , Humanos , Análisis de Secuencia de ADN , Programas Informáticos
9.
J Invertebr Pathol ; 186: 107438, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-32652083

RESUMEN

In 1998 a nomenclature for the growing list of pesticidal proteins from Bacillus thuringiensis (Bt) was derived based solely on protein sequence comparisons. This nomenclature was widely adopted and provided a robust framework for the naming and classification of the proteins. The success of these proteins in integrated pest management schemes prompted an increased effort to find others with improved or more diverse activities. These discovery activities led to the characterization of proteins from a wider range of bacteria and with a variety of different protein folds. Since most of these new proteins were grouped together as Cry proteins it became apparent that the existing nomenclature had limitations in representing the diverse range of proteins that had been identified. This revised nomenclature retains the basic principles of the 1998 version but provides specific mnemonics to represent different structural groups. For the purposes of consistency, the vast majority of the proteins have either retained their name or have a new name that clearly references the previous one. Other pesticidal proteins not previously included in the nomenclature have been incorporated into this version.


Asunto(s)
Bacillus thuringiensis/química , Proteínas Bacterianas/clasificación , Agentes de Control Biológico/química , Insecticidas/clasificación , Terminología como Asunto , Control Biológico de Vectores
10.
J Immunol ; 197(3): 971-82, 2016 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-27307560

RESUMEN

The nonclassical HLA molecule MHC-related protein 1 (MR1) presents metabolites of the vitamin B synthesis pathways to mucosal-associated invariant T (MAIT) cells and other MR1-restricted T cells. This new class of Ags represents a variation on the classical paradigm of self/non-self discrimination because these T cells are activated through their TCR by small organic compounds generated during microbial vitamin B2 synthesis. Beyond the fundamental significance, the invariant nature of MR1 across the human population is a tantalizing feature for the potential development of universal immune therapeutic and diagnostic tools. However, many aspects of MR1 Ag presentation and MR1-restricted T cell biology remain unknown, and the ubiquitous expression of MR1 across tissues and cell lines can be a confounding factor for experimental purposes. In this study, we report the development of a novel CRISPR/Cas9 genome editing lentiviral system and its use to efficiently disrupt MR1 expression in A459, THP-1, and K562 cell lines. We generated isogenic MR1(-/-) clonal derivatives of the A549 lung carcinoma and THP-1 monocytic cell lines and used these to study T cell responses to intracellular pathogens. We confirmed that MAIT cell clones were unable to respond to MR1(-/-) clones infected with bacteria whereas Ag presentation by classical and other nonclassical HLAs was unaffected. This system represents a robust and efficient method to disrupt the expression of MR1 and should facilitate investigations into the processing and presentation of MR1 Ags as well as into the biology of MAIT cells.


Asunto(s)
Presentación de Antígeno/inmunología , Edición Génica/métodos , Antígenos de Histocompatibilidad Clase I/inmunología , Activación de Linfocitos/inmunología , Antígenos de Histocompatibilidad Menor/inmunología , Linfocitos T/inmunología , Línea Celular , Repeticiones Palindrómicas Cortas Agrupadas y Regularmente Espaciadas/genética , Citometría de Flujo , Vectores Genéticos , Humanos , Lentivirus , Mutagénesis Sitio-Dirigida , Reacción en Cadena de la Polimerasa , Subgrupos de Linfocitos T/inmunología
11.
Proc Natl Acad Sci U S A ; 112(3): 863-8, 2015 Jan 20.
Artículo en Inglés | MEDLINE | ID: mdl-25535353

RESUMEN

Many bacterial pathogens are specialized, infecting one or few hosts, and this is often associated with more acute disease presentation. Specific genomes show markers of this specialization, which often reflect a balance between gene acquisition and functional gene loss. Within Salmonella enterica subspecies enterica, a single lineage exists that includes human and animal pathogens adapted to cause infection in different hosts, including S. enterica serovar Enteritidis (multiple hosts), S. Gallinarum (birds), and S. Dublin (cattle). This provides an excellent evolutionary context in which differences between these pathogen genomes can be related to host range. Genome sequences were obtained from ∼ 60 isolates selected to represent the known diversity of this lineage. Examination and comparison of the clades within the phylogeny of this lineage revealed signs of host restriction as well as evolutionary events that mark a path to host generalism. We have identified the nature and order of events for both evolutionary trajectories. The impact of functional gene loss was predicted based upon position within metabolic pathways and confirmed with phenotyping assays. The structure of S. Enteritidis is more complex than previously known, as a second clade of S. Enteritidis was revealed that is distinct from those commonly seen to cause disease in humans or animals, and that is more closely related to S. Gallinarum. Isolates from this second clade were tested in a chick model of infection and exhibited a reduced colonization phenotype, which we postulate represents an intermediate stage in pathogen-host adaptation.


Asunto(s)
Adaptación Fisiológica , Evolución Molecular , Genoma Bacteriano , Salmonella/genética , Cromosomas Bacterianos , Seudogenes
12.
Proc Natl Acad Sci U S A ; 112(27): E3574-81, 2015 Jul 07.
Artículo en Inglés | MEDLINE | ID: mdl-26100894

RESUMEN

Klebsiella pneumoniae is now recognized as an urgent threat to human health because of the emergence of multidrug-resistant strains associated with hospital outbreaks and hypervirulent strains associated with severe community-acquired infections. K. pneumoniae is ubiquitous in the environment and can colonize and infect both plants and animals. However, little is known about the population structure of K. pneumoniae, so it is difficult to recognize or understand the emergence of clinically important clones within this highly genetically diverse species. Here we present a detailed genomic framework for K. pneumoniae based on whole-genome sequencing of more than 300 human and animal isolates spanning four continents. Our data provide genome-wide support for the splitting of K. pneumoniae into three distinct species, KpI (K. pneumoniae), KpII (K. quasipneumoniae), and KpIII (K. variicola). Further, for K. pneumoniae (KpI), the entity most frequently associated with human infection, we show the existence of >150 deeply branching lineages including numerous multidrug-resistant or hypervirulent clones. We show K. pneumoniae has a large accessory genome approaching 30,000 protein-coding genes, including a number of virulence functions that are significantly associated with invasive community-acquired disease in humans. In our dataset, antimicrobial resistance genes were common among human carriage isolates and hospital-acquired infections, which generally lacked the genes associated with invasive disease. The convergence of virulence and resistance genes potentially could lead to the emergence of untreatable invasive K. pneumoniae infections; our data provide the whole-genome framework against which to track the emergence of such threats.


Asunto(s)
Variación Genética , Genoma Bacteriano/genética , Infecciones por Klebsiella/microbiología , Klebsiella pneumoniae/genética , Animales , Antiinfecciosos/farmacología , Proteínas Bacterianas/clasificación , Proteínas Bacterianas/genética , Infección Hospitalaria/microbiología , Farmacorresistencia Bacteriana Múltiple/genética , Genes Bacterianos/genética , Genómica/métodos , Humanos , Klebsiella pneumoniae/clasificación , Klebsiella pneumoniae/patogenicidad , Filogenia , Dinámica Poblacional , Salud Pública/estadística & datos numéricos , Salud Pública/tendencias , Análisis de Secuencia de ADN , Especificidad de la Especie , Virulencia/genética
13.
Nature ; 477(7365): 462-5, 2011 Aug 24.
Artículo en Inglés | MEDLINE | ID: mdl-21866102

RESUMEN

Vibrio cholerae is a globally important pathogen that is endemic in many areas of the world and causes 3-5 million reported cases of cholera every year. Historically, there have been seven acknowledged cholera pandemics; recent outbreaks in Zimbabwe and Haiti are included in the seventh and ongoing pandemic. Only isolates in serogroup O1 (consisting of two biotypes known as 'classical' and 'El Tor') and the derivative O139 can cause epidemic cholera. It is believed that the first six cholera pandemics were caused by the classical biotype, but El Tor has subsequently spread globally and replaced the classical biotype in the current pandemic. Detailed molecular epidemiological mapping of cholera has been compromised by a reliance on sub-genomic regions such as mobile elements to infer relationships, making El Tor isolates associated with the seventh pandemic seem superficially diverse. To understand the underlying phylogeny of the lineage responsible for the current pandemic, we identified high-resolution markers (single nucleotide polymorphisms; SNPs) in 154 whole-genome sequences of globally and temporally representative V. cholerae isolates. Using this phylogeny, we show here that the seventh pandemic has spread from the Bay of Bengal in at least three independent but overlapping waves with a common ancestor in the 1950s, and identify several transcontinental transmission events. Additionally, we show how the acquisition of the SXT family of antibiotic resistance elements has shaped pandemic spread, and show that this family was first acquired at least ten years before its discovery in V. cholerae.


Asunto(s)
Cólera/epidemiología , Cólera/transmisión , Pandemias/estadística & datos numéricos , Vibrio cholerae/genética , Vibrio cholerae/aislamiento & purificación , Cólera/microbiología , Genoma Bacteriano/genética , Haití/epidemiología , Humanos , Funciones de Verosimilitud , Epidemiología Molecular , Filogenia , Polimorfismo de Nucleótido Simple/genética , Vibrio cholerae/clasificación , Zimbabwe/epidemiología
14.
Nucleic Acids Res ; 43(3): e15, 2015 Feb 18.
Artículo en Inglés | MEDLINE | ID: mdl-25414349

RESUMEN

The emergence of new sequencing technologies has facilitated the use of bacterial whole genome alignments for evolutionary studies and outbreak analyses. These datasets, of increasing size, often include examples of multiple different mechanisms of horizontal sequence transfer resulting in substantial alterations to prokaryotic chromosomes. The impact of these processes demands rapid and flexible approaches able to account for recombination when reconstructing isolates' recent diversification. Gubbins is an iterative algorithm that uses spatial scanning statistics to identify loci containing elevated densities of base substitutions suggestive of horizontal sequence transfer while concurrently constructing a maximum likelihood phylogeny based on the putative point mutations outside these regions of high sequence diversity. Simulations demonstrate the algorithm generates highly accurate reconstructions under realistically parameterized models of bacterial evolution, and achieves convergence in only a few hours on alignments of hundreds of bacterial genome sequences. Gubbins is appropriate for reconstructing the recent evolutionary history of a variety of haploid genotype alignments, as it makes no assumptions about the underlying mechanism of recombination. The software is freely available for download at github.com/sanger-pathogens/Gubbins, implemented in Python and C and supported on Linux and Mac OS X.


Asunto(s)
Algoritmos , Bacterias/clasificación , Genoma Bacteriano , Filogenia , Recombinación Genética , Bacterias/genética , Análisis de Secuencia
15.
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
16.
Infect Immun ; 84(12): 3618-3628, 2016 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-27736780

RESUMEN

Salmonella species utilize type III secretion systems (T3SSs) to translocate effectors into the cytosol of mammalian host cells, subverting cell signaling and facilitating the onset of gastroenteritis. In this study, we compared a draft genome assembly of Salmonella enterica subsp. salamae strain 3588/07 against the genomes of S. enterica subsp. enterica serovar Typhimurium strain LT2 and Salmonella bongori strain 12419. S. enterica subsp. salamae encodes the Salmonella pathogenicity island 1 (SPI-1), SPI-2, and the locus of enterocyte effacement (LEE) T3SSs. Though several key S Typhimurium effector genes are missing (e.g., avrA, sopB, and sseL), S. enterica subsp. salamae invades HeLa cells and contains homologues of S. bongori sboK and sboC, which we named seoC SboC and SeoC are homologues of EspJ from enteropathogenic and enterohemorrhagic Escherichia coli (EPEC and EHEC, respectively), which inhibit Src kinase-dependent phagocytosis by ADP-ribosylation. By screening 73 clinical and environmental Salmonella isolates, we identified EspJ homologues in S. bongori, S. enterica subsp. salamae, and Salmonella enterica subsp. arizonae The ß-lactamase TEM-1 reporter system showed that SeoC is translocated by the SPI-1 T3SS. All the Salmonella SeoC/SboC homologues ADP-ribosylate Src E310 in vitro Ectopic expression of SeoC/SboC inhibited phagocytosis of IgG-opsonized beads into Cos-7 cells stably expressing green fluorescent protein (GFP)-FcγRIIa. Concurrently, S. enterica subsp. salamae infection of J774.A1 macrophages inhibited phagocytosis of beads, in a seoC-dependent manner. These results show that S. bongori, S. enterica subsp. salamae, and S. enterica subsp. arizonae share features of the infection strategy of extracellular pathogens EPEC and EHEC and shed light on the complexities of the T3SS effector repertoires of Enterobacteriaceae.


Asunto(s)
Proteínas Bacterianas/metabolismo , Regulación Bacteriana de la Expresión Génica/fisiología , Genoma Bacteriano , Salmonella enterica/clasificación , Sistemas de Secreción Tipo III/fisiología , Secuencia de Aminoácidos , Animales , Proteínas Bacterianas/genética , Células COS , Chlorocebus aethiops , Células HeLa , Humanos , Prevalencia , Receptores de IgG/genética , Receptores de IgG/metabolismo , Salmonella enterica/metabolismo
17.
Emerg Infect Dis ; 22(9): 1545-53, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-27532625

RESUMEN

Shigellae are sensitive indicator species for studying trends in the international transmission of antimicrobial-resistant Enterobacteriaceae. Orthodox Jewish communities (OJCs) are a known risk group for shigellosis; Shigella sonnei is cyclically epidemic in OJCs in Israel, and sporadic outbreaks occur in OJCs elsewhere. We generated whole-genome sequences for 437 isolates of S. sonnei from OJCs and non-OJCs collected over 22 years in Europe (the United Kingdom, France, and Belgium), the United States, Canada, and Israel and analyzed these within a known global genomic context. Through phylogenetic and genomic analysis, we showed that strains from outbreaks in OJCs outside of Israel are distinct from strains in the general population and relate to a single multidrug-resistant sublineage of S. sonnei that prevails in Israel. Further Bayesian phylogenetic analysis showed that this strain emerged approximately 30 years ago, demonstrating the speed at which antimicrobial drug-resistant pathogens can spread widely through geographically dispersed, but internationally connected, communities.


Asunto(s)
Antibacterianos/farmacología , Infecciones Comunitarias Adquiridas/epidemiología , Infecciones Comunitarias Adquiridas/transmisión , Farmacorresistencia Bacteriana Múltiple , Disentería Bacilar/epidemiología , Disentería Bacilar/transmisión , Judíos , Shigella sonnei/efectos de los fármacos , Viaje , Antibacterianos/uso terapéutico , Infecciones Comunitarias Adquiridas/historia , Infecciones Comunitarias Adquiridas/microbiología , Brotes de Enfermedades , Disentería Bacilar/historia , Disentería Bacilar/microbiología , Genes Bacterianos , Genoma Bacteriano , Salud Global , Historia del Siglo XX , Historia del Siglo XXI , Humanos , Pruebas de Sensibilidad Microbiana , Vigilancia de la Población , Factores de Riesgo , Shigella sonnei/clasificación , Shigella sonnei/genética , Shigella sonnei/aislamiento & purificación , Secuenciación Completa del Genoma
18.
Antimicrob Agents Chemother ; 59(2): 923-9, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25421466

RESUMEN

The blaNDM-1 gene is associated with extensive drug resistance in Gram-negative bacteria. This probably spread to Enterobacteriaceae from Acinetobacter spp., and we characterized plasmids associated with blaNDM-1 in Acinetobacter spp. to gain insight into their role in this dissemination. Four clinical NDM-1-producing Acinetobacter species strains from India and Pakistan were investigated. A plasmid harboring blaNDM-1, pNDM-40-1, was characterized by whole-genome sequencing of Acinetobacter bereziniae CHI-40-1 and comparison with related plasmids. The presence of similar plasmids in strains from Pakistan was sought by PCR and sequencing of amplicons. Conjugation frequency was tested and stability of pNDM-40-1 investigated by real-time PCR of isolates passaged with and without antimicrobial selection pressure. A. bereziniae and Acinetobacter haemolyticus strains contained plasmids similar to the pNDM-BJ01-like plasmids identified in Acinetobacter spp. in China. The backbone of pNDM-40-1 was almost identical to that of pNDM-BJ01-like plasmids, but the transposon harboring blaNDM-1, Tn125, contained two short deletions. Escherichia coli and Acinetobacter pittii transconjugants were readily obtained. Transconjugants retained pNDM-40-1 after a 14-day passage experiment, although stability was greater with meropenem selection. Fragments of pNDM-BJ01-like plasmid backbones are found near blaNDM-1 in some genetic contexts from Enterobacteriaceae, suggesting that cross-genus transfer has occurred. pNDM-BJ01-like plasmids have been described in isolates originating from a wide geographical region in southern Asia. In vitro data on plasmid transfer and stability suggest that these plasmids could have contributed to the spread of blaNDM-1 into Enterobacteriaceae.


Asunto(s)
Acinetobacter/genética , Farmacorresistencia Bacteriana Múltiple/genética , Plásmidos/genética , Acinetobacter/efectos de los fármacos , Antibacterianos/farmacología , India , Pruebas de Sensibilidad Microbiana , Pakistán
19.
Mol Biol Evol ; 30(5): 1224-8, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23408797

RESUMEN

Phylogeographical analyses have become commonplace for a myriad of organisms with the advent of cheap DNA sequencing technologies. Bayesian model-based clustering is a powerful tool for detecting important patterns in such data and can be used to decipher even quite subtle signals of systematic differences in molecular variation. Here, we introduce two upgrades to the Bayesian Analysis of Population Structure (BAPS) software, which enable 1) spatially explicit modeling of variation in DNA sequences and 2) hierarchical clustering of DNA sequence data to reveal nested genetic population structures. We provide a direct interface to map the results from spatial clustering with Google Maps using the portal http://www.spatialepidemiology.net/ and illustrate this approach using sequence data from Borrelia burgdorferi. The usefulness of hierarchical clustering is demonstrated through an analysis of the metapopulation structure within a bacterial population experiencing a high level of local horizontal gene transfer. The tools that are introduced are freely available at http://www.helsinki.fi/bsg/software/BAPS/.


Asunto(s)
Evolución Molecular , Programas Informáticos , Teorema de Bayes , Borrelia burgdorferi/clasificación , Borrelia burgdorferi/genética , Genética de Población , Análisis de Secuencia de ADN/métodos
20.
PLoS Pathog ; 8(10): e1002995, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-23133377

RESUMEN

Relapsing C. difficile disease in humans is linked to a pathological imbalance within the intestinal microbiota, termed dysbiosis, which remains poorly understood. We show that mice infected with epidemic C. difficile (genotype 027/BI) develop highly contagious, chronic intestinal disease and persistent dysbiosis characterized by a distinct, simplified microbiota containing opportunistic pathogens and altered metabolite production. Chronic C. difficile 027/BI infection was refractory to vancomycin treatment leading to relapsing disease. In contrast, treatment of C. difficile 027/BI infected mice with feces from healthy mice rapidly restored a diverse, healthy microbiota and resolved C. difficile disease and contagiousness. We used this model to identify a simple mixture of six phylogenetically diverse intestinal bacteria, including novel species, which can re-establish a health-associated microbiota and clear C. difficile 027/BI infection from mice. Thus, targeting a dysbiotic microbiota with a defined mixture of phylogenetically diverse bacteria can trigger major shifts in the microbial community structure that displaces C. difficile and, as a result, resolves disease and contagiousness. Further, we demonstrate a rational approach to harness the therapeutic potential of health-associated microbial communities to treat C. difficile disease and potentially other forms of intestinal dysbiosis.


Asunto(s)
Clostridioides difficile/patogenicidad , Enterocolitis Seudomembranosa/terapia , Heces/microbiología , Intestinos/microbiología , Interacciones Microbianas , Probióticos/uso terapéutico , Animales , Clostridioides difficile/efectos de los fármacos , Farmacorresistencia Bacteriana , Enterocolitis Seudomembranosa/tratamiento farmacológico , Enterocolitis Seudomembranosa/microbiología , Femenino , Ratones , Ratones Endogámicos C3H , Ratones Endogámicos C57BL , Consorcios Microbianos , Datos de Secuencia Molecular , Recurrencia , Vancomicina/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA