Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 52
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Proc Natl Acad Sci U S A ; 120(47): e2307773120, 2023 Nov 21.
Artigo em Inglês | MEDLINE | ID: mdl-37963246

RESUMO

The expansion and intensification of livestock production is predicted to promote the emergence of pathogens. As pathogens sometimes jump between species, this can affect the health of humans as well as livestock. Here, we investigate how livestock microbiota can act as a source of these emerging pathogens through analysis of Streptococcus suis, a ubiquitous component of the respiratory microbiota of pigs that is also a major cause of disease on pig farms and an important zoonotic pathogen. Combining molecular dating, phylogeography, and comparative genomic analyses of a large collection of isolates, we find that several pathogenic lineages of S. suis emerged in the 19th and 20th centuries, during an early period of growth in pig farming. These lineages have since spread between countries and continents, mirroring trade in live pigs. They are distinguished by the presence of three genomic islands with putative roles in metabolism and cell adhesion, and an ongoing reduction in genome size, which may reflect their recent shift to a more pathogenic ecology. Reconstructions of the evolutionary histories of these islands reveal constraints on pathogen emergence that could inform control strategies, with pathogenic lineages consistently emerging from one subpopulation of S. suis and acquiring genes through horizontal transfer from other pathogenic lineages. These results shed light on the capacity of the microbiota to rapidly evolve to exploit changes in their host population and suggest that the impact of changes in farming on the pathogenicity and zoonotic potential of S. suis is yet to be fully realized.


Assuntos
Infecções Estreptocócicas , Streptococcus suis , Doenças dos Suínos , Animais , Humanos , Suínos , Infecções Estreptocócicas/veterinária , Fazendas , Doenças dos Suínos/epidemiologia , Virulência/genética , Streptococcus suis/genética , Gado
2.
Nucleic Acids Res ; 51(7): 3240-3260, 2023 04 24.
Artigo em Inglês | MEDLINE | ID: mdl-36840716

RESUMO

Actinobacillus pleuropneumoniae is the cause of porcine pleuropneumonia, a severe respiratory tract infection that is responsible for major economic losses to the swine industry. Many host-adapted bacterial pathogens encode systems known as phasevarions (phase-variable regulons). Phasevarions result from variable expression of cytoplasmic DNA methyltransferases. Variable expression results in genome-wide methylation differences within a bacterial population, leading to altered expression of multiple genes via epigenetic mechanisms. Our examination of a diverse population of A. pleuropneumoniae strains determined that Type I and Type III DNA methyltransferases with the hallmarks of phase variation were present in this species. We demonstrate that phase variation is occurring in these methyltransferases, and show associations between particular Type III methyltransferase alleles and serovar. Using Pacific BioSciences Single-Molecule, Real-Time (SMRT) sequencing and Oxford Nanopore sequencing, we demonstrate the presence of the first ever characterised phase-variable, cytosine-specific Type III DNA methyltransferase. Phase variation of distinct Type III DNA methyltransferase in A. pleuropneumoniae results in the regulation of distinct phasevarions, and in multiple phenotypic differences relevant to pathobiology. Our characterisation of these newly described phasevarions in A. pleuropneumoniae will aid in the selection of stably expressed antigens, and direct and inform development of a rationally designed subunit vaccine against this major veterinary pathogen.


Assuntos
Actinobacillus pleuropneumoniae , Variação de Fase , Animais , Suínos , Actinobacillus pleuropneumoniae/genética , Actinobacillus pleuropneumoniae/metabolismo , Metilases de Modificação do DNA/genética , Metilases de Modificação do DNA/metabolismo , Metilação de DNA , Metiltransferases/genética , Metiltransferases/metabolismo , Bactérias/genética , DNA/metabolismo
3.
PLoS Genet ; 18(3): e1009776, 2022 03.
Artigo em Inglês | MEDLINE | ID: mdl-35286304

RESUMO

Shotgun metagenomics is a powerful tool to identify antimicrobial resistance (AMR) genes in microbiomes but has the limitation that extrachromosomal DNA, such as plasmids, cannot be linked with the host bacterial chromosome. Here we present a comprehensive laboratory and bioinformatics pipeline HAM-ART (Hi-C Assisted Metagenomics for Antimicrobial Resistance Tracking) optimised for the generation of metagenome-assembled genomes including both chromosomal and extrachromosomal AMR genes. We demonstrate the performance of the pipeline in a study comparing 100 pig faecal microbiomes from low- and high-antimicrobial use pig farms (organic and conventional farms). We found significant differences in the distribution of AMR genes between low- and high-antimicrobial use farms including a plasmid-borne lincosamide resistance gene exclusive to high-antimicrobial use farms in three species of Lactobacilli. The bioinformatics pipeline code is available at https://github.com/lkalmar/HAM-ART.


Assuntos
Anti-Infecciosos , Microbiota , Animais , Antibacterianos , Anti-Infecciosos/farmacologia , Farmacorresistência Bacteriana/genética , Metagenômica , Suínos
4.
PLoS Genet ; 17(11): e1009864, 2021 11.
Artigo em Inglês | MEDLINE | ID: mdl-34748531

RESUMO

Mutation rates vary both within and between bacterial species, and understanding what drives this variation is essential for understanding the evolutionary dynamics of bacterial populations. In this study, we investigate two factors that are predicted to influence the mutation rate: ecology and genome size. We conducted mutation accumulation experiments on eight strains of the emerging zoonotic pathogen Streptococcus suis. Natural variation within this species allows us to compare tonsil carriage and invasive disease isolates, from both more and less pathogenic populations, with a wide range of genome sizes. We find that invasive disease isolates have repeatedly evolved mutation rates that are higher than those of closely related carriage isolates, regardless of variation in genome size. Independent of this variation in overall rate, we also observe a stronger bias towards G/C to A/T mutations in isolates from more pathogenic populations, whose genomes tend to be smaller and more AT-rich. Our results suggest that ecology is a stronger correlate of mutation rate than genome size over these timescales, and that transitions to invasive disease are consistently accompanied by rapid increases in mutation rate. These results shed light on the impact that ecology can have on the adaptive potential of bacterial pathogens.


Assuntos
Adaptação Biológica/genética , Doenças Transmissíveis Emergentes/microbiologia , Taxa de Mutação , Infecções Estreptocócicas/microbiologia , Streptococcus suis/genética , Zoonoses/microbiologia , Animais , Ecologia , Streptococcus suis/isolamento & purificação , Streptococcus suis/patogenicidade , Virulência/genética
5.
BMC Vet Res ; 19(1): 135, 2023 Aug 28.
Artigo em Inglês | MEDLINE | ID: mdl-37641044

RESUMO

BACKGROUND: Glaesserella parasuis is the causative agent of Glässer's disease in pigs. Serotyping is the most common method used to type G. parasuis isolates. However, the high number of non-typables (NT) and low discriminatory power make serotyping problematic. In this study, 218 field clinical isolates and 15 G. parasuis reference strains were whole-genome sequenced (WGS). Multilocus sequence types (MLST), serotypes, core-genome phylogeny, antimicrobial resistance (AMR) genes, and putative virulence gene information was extracted. RESULTS: In silico WGS serotyping identified 11 of 15 serotypes. The most frequently detected serotypes were 7, 13, 4, and 2. MLST identified 72 sequence types (STs), of which 66 were novel. The most predominant ST was ST454. Core-genome phylogeny depicted 3 primary lineages (LI, LII, and LIII), with LIIIA sublineage isolates lacking all vtaA genes, based on the structure of the phylogenetic tree and the number of virulence genes. At least one group 1 vtaA virulence genes were observed in most isolates (97.2%), except for serotype 8 (ST299 and ST406), 15 (ST408 and ST552) and NT (ST448). A few group 1 vtaA genes were significantly associated with certain serotypes or STs. The putative virulence gene lsgB, was detected in 8.3% of the isolates which were predominantly of serotype 5/12. While most isolates carried the bcr, ksgA, and bacA genes, the following antimicrobial resistant genes were detected in lower frequency;  blaZ (6.9%), tetM (3.7%), spc (3.7%), tetB (2.8%), bla-ROB-1 (1.8%), ermA (1.8%), strA (1.4%), qnrB (0.5%), and aph3''Ia (0.5%).   CONCLUSION: This study showed the use of WGS to type G. parasuis isolates and can be considered an alternative to the more labor-intensive and traditional serotyping and standard MLST. Core-genome phylogeny provided the best strain discrimination. These findings will lead to a better understanding of the molecular epidemiology and virulence in G. parasuis that can be applied to the future development of diagnostic tools, autogenous vaccines, evaluation of antibiotic use, prevention, and disease control.


Assuntos
Haemophilus parasuis , Animais , Suínos , Tipagem de Sequências Multilocus/veterinária , Filogenia , Sorogrupo , Sorotipagem/veterinária , Haemophilus parasuis/genética , América do Norte
6.
Mol Biol Evol ; 38(4): 1570-1579, 2021 04 13.
Artigo em Inglês | MEDLINE | ID: mdl-33313861

RESUMO

Emerging bacterial pathogens threaten global health and food security, and so it is important to ask whether these transitions to pathogenicity have any common features. We present a systematic study of the claim that pathogenicity is associated with genome reduction and gene loss. We compare broad-scale patterns across all bacteria, with detailed analyses of Streptococcus suis, an emerging zoonotic pathogen of pigs, which has undergone multiple transitions between disease and carriage forms. We find that pathogenicity is consistently associated with reduced genome size across three scales of divergence (between species within genera, and between and within genetic clusters of S. suis). Although genome reduction is also found in mutualist and commensal bacterial endosymbionts, genome reduction in pathogens cannot be solely attributed to the features of their ecology that they share with these species, that is, host restriction or intracellularity. Moreover, other typical correlates of genome reduction in endosymbionts (reduced metabolic capacity, reduced GC content, and the transient expansion of nonfunctional elements) are not consistently observed in pathogens. Together, our results indicate that genome reduction is a consistent correlate of pathogenicity in bacteria.


Assuntos
Bactérias/patogenicidade , Evolução Biológica , Tamanho do Genoma , Genoma Bacteriano , Animais , Bactérias/genética , Simbiose
7.
Infect Immun ; 88(5)2020 04 20.
Artigo em Inglês | MEDLINE | ID: mdl-32094250

RESUMO

Glaesserella (Haemophilus) parasuis is a commensal bacterium of the upper respiratory tract in pigs and also the causative agent of Glässer's disease, which causes significant morbidity and mortality in pigs worldwide. Isolates are characterized into 15 serovars by their capsular polysaccharide, which has shown a correlation with isolate pathogenicity. To investigate the role the capsule plays in G. parasuis virulence and host interaction, a capsule mutant of the serovar 5 strain HS069 was generated (HS069Δcap) through allelic exchange following natural transformation. HS069Δcap was unable to cause signs of systemic disease during a pig challenge study and had increased sensitivity to complement killing and phagocytosis by alveolar macrophages. Compared with the parent strain, HS069Δcap produced more robust biofilm and adhered equivalently to 3D4/31 cells; however, it was unable to persistently colonize the nasal cavity of inoculated pigs, with all pigs clearing HS069Δcap by 5 days postchallenge. Our results indicate the importance of the capsular polysaccharide to G. parasuis virulence as well as nasal colonization in pigs.


Assuntos
Haemophilus parasuis/genética , Animais , Biofilmes , Infecções por Haemophilus/microbiologia , Macrófagos Alveolares/microbiologia , Fagocitose/fisiologia , Suínos , Doenças dos Suínos/microbiologia , Virulência/genética
8.
BMC Vet Res ; 16(1): 167, 2020 May 27.
Artigo em Inglês | MEDLINE | ID: mdl-32460764

RESUMO

BACKGROUND: Glaesserella parasuis, the causative agent of Glӓsser's disease, is widespread in swine globally resulting in significant economic losses to the swine industry. Prevention of Glӓsser's disease in pigs has been plagued with an inability to design broadly protective vaccines, as many bacterin based platforms generate serovar or strain specific immunity. Subunit vaccines are of interest to provide protective immunity to multiple strains of G. parasuis. Selected proteins for subunit vaccination should be widespread, highly conserved, and surface exposed. RESULTS: Two candidate proteins for subunit vaccination (RlpB and VacJ) against G. parasuis were identified using random mutagenesis and an in vitro organ culture system. Pigs were vaccinated with recombinant RlpB and VacJ, outer membrane proteins with important contributions to cellular function and viability. Though high antibody titers to the recombinant proteins and increased interferon-γ producing cells were found in subunit vaccinated animals, the pigs were not protected from developing systemic disease. CONCLUSIONS: It appears there may be insufficient RlpB and VacJ exposed on the bacterial surface for antibody to bind, preventing high RlpB and VacJ specific antibody titers from protecting animals from G. parasuis. Additionally, this work confirms the importance of utilizing the natural host species when assessing the efficacy of vaccine candidates.


Assuntos
Infecções por Haemophilus/veterinária , Haemophilus parasuis/imunologia , Proteínas Recombinantes/imunologia , Doenças dos Suínos/prevenção & controle , Animais , Anticorpos Antibacterianos/sangue , Proteínas de Bactérias/imunologia , Vacinas Bacterianas/imunologia , Infecções por Haemophilus/imunologia , Infecções por Haemophilus/prevenção & controle , Vacinas Anti-Haemophilus/imunologia , Haemophilus parasuis/genética , Sorogrupo , Sus scrofa , Suínos , Doenças dos Suínos/imunologia , Doenças dos Suínos/microbiologia , Técnicas de Cultura de Tecidos/veterinária , Vacinação/veterinária , Vacinas de Subunidades Antigênicas/administração & dosagem , Vacinas de Subunidades Antigênicas/imunologia
9.
Nucleic Acids Res ; 46(21): 11466-11476, 2018 11 30.
Artigo em Inglês | MEDLINE | ID: mdl-30304532

RESUMO

Streptococcus suis is a major pathogen of swine, responsible for a number of chronic and acute infections, and is also emerging as a major zoonotic pathogen, particularly in South-East Asia. Our study of a diverse population of S. suis shows that this organism contains both Type I and Type III phase-variable methyltransferases. In all previous examples, phase-variation of methyltransferases results in genome wide methylation differences, and results in differential regulation of multiple genes, a system known as the phasevarion (phase-variable regulon). We hypothesized that each variant in the Type I and Type III systems encoded a methyltransferase with a unique specificity, and could therefore control a distinct phasevarion, either by recombination-driven shuffling between different specificities (Type I) or by biphasic on-off switching via simple sequence repeats (Type III). Here, we present the identification of the target specificities for each Type III allelic variant from S. suis using single-molecule, real-time methylome analysis. We demonstrate phase-variation is occurring in both Type I and Type III methyltransferases, and show a distinct association between methyltransferase type and presence, and population clades. In addition, we show that the phase-variable Type I methyltransferase was likely acquired at the origin of a highly virulent zoonotic sub-population.


Assuntos
Regulação Bacteriana da Expressão Gênica , Metiltransferases/genética , Regulon , Streptococcus suis/enzimologia , Alelos , Animais , Metilação de DNA , Metilases de Modificação do DNA/metabolismo , DNA Bacteriano/metabolismo , Epigênese Genética , Escherichia coli , Variação Genética , Genoma Bacteriano , Repetições de Microssatélites , Oligonucleotídeos/genética , Fenótipo , Infecções Estreptocócicas/microbiologia , Infecções Estreptocócicas/veterinária , Streptococcus suis/genética , Suínos
10.
J Clin Microbiol ; 57(7)2019 07.
Artigo em Inglês | MEDLINE | ID: mdl-30944194

RESUMO

Streptococcus suis is one of the most important zoonotic bacterial pathogens of pigs, causing significant economic losses to the global swine industry. S. suis is also a very successful colonizer of mucosal surfaces, and commensal strains can be found in almost all pig populations worldwide, making detection of the S. suis species in asymptomatic carrier herds of little practical value in predicting the likelihood of future clinical relevance. The value of future molecular tools for surveillance and preventative health management lies in the detection of strains that genetically have increased potential to cause disease in presently healthy animals. Here we describe the use of genome-wide association studies to identify genetic markers associated with the observed clinical phenotypes (i) invasive disease and (ii) asymptomatic carriage on the palatine tonsils of pigs on UK farms. Subsequently, we designed a multiplex PCR to target three genetic markers that differentiated 115 S. suis isolates into disease-associated and non-disease-associated groups, that performed with a sensitivity of 0.91, a specificity of 0.79, a negative predictive value of 0.91, and a positive predictive value of 0.79 in comparison to observed clinical phenotypes. We describe evaluation of our pathotyping tool, using an out-of-sample collection of 50 previously uncharacterized S. suis isolates, in comparison to existing methods used to characterize and subtype S. suis isolates. In doing so, we show our pathotyping approach to be a competitive method to characterize S. suis isolates recovered from pigs on UK farms and one that can easily be updated to incorporate global strain collections.


Assuntos
Portador Sadio/veterinária , Infecções Estreptocócicas/veterinária , Streptococcus suis/isolamento & purificação , Streptococcus suis/patogenicidade , Doenças dos Suínos/microbiologia , Animais , Portador Sadio/microbiologia , Inglaterra , Marcadores Genéticos/genética , Genoma Bacteriano/genética , Técnicas de Diagnóstico Molecular , Reação em Cadeia da Polimerase Multiplex , Tonsila Palatina/microbiologia , Infecções Estreptocócicas/microbiologia , Streptococcus suis/genética , Suínos , Virulência/genética , País de Gales
11.
Vet Res ; 50(1): 21, 2019 Mar 07.
Artigo em Inglês | MEDLINE | ID: mdl-30845993

RESUMO

Brachyspira (B.) hyodysenteriae is widespread globally, and can cause mucohaemorrhagic colitis (swine dysentery, SD) with severe economic impact in infected herds. Typical strains of B. hyodysenteriae are strongly haemolytic on blood agar, and the haemolytic activity is believed to contribute to virulence in vivo. However, recently there have been reports of atypical weakly haemolytic isolates of B. hyodysenteriae (whBh). In this study, 34 European whBh and 82 strongly haemolytic isolates were subjected to comparative genomic analysis. A phylogenetic tree constructed using core single nucleotide polymorphisms showed that the whBh formed a distinct sub-clade. All eight genes previously associated with haemolysis in B. hyodysenteriae were present in the whBh. No consistent patterns of amino acid substitutions for all whBh were found in these genes. In contrast, a genome region containing six coding sequences (CDSs) had consistent nucleotide sequence differences between strongly and whBh isolates. Two CDSs were predicted to encode ABC transporter proteins, and a TolC family protein, which may have a role in the export of haemolysins from B. hyodysenteriae. Another difference in this region was the presence of three CDSs in whBh that are pseudogenes in strongly haemolytic isolates. One of the intact CDSs from whBh encoded a predicted PadR-like transcriptional repressor that may play a role in repression of haemolysis functions. In summary, a sub-clade of whBh isolates has emerged in Europe, and several genomic differences, that potentially explain the weakly haemolytic phenotype, were identified. These markers may provide targets for discriminatory molecular tests needed in SD surveillance.


Assuntos
Brachyspira hyodysenteriae/genética , Infecções por Bactérias Gram-Negativas/veterinária , Doenças dos Suínos/microbiologia , Animais , Genes Bacterianos/genética , Genes Bacterianos/fisiologia , Genoma Bacteriano/genética , Infecções por Bactérias Gram-Negativas/epidemiologia , Infecções por Bactérias Gram-Negativas/microbiologia , Proteínas Hemolisinas/genética , Hemólise/genética , Tipagem de Sequências Multilocus/veterinária , Fenótipo , Filogenia , Análise de Sequência de DNA/veterinária , Suínos , Doenças dos Suínos/epidemiologia
12.
Infect Immun ; 86(3)2018 03.
Artigo em Inglês | MEDLINE | ID: mdl-29203546

RESUMO

Streptococcus suis is a bacterium that is commonly carried in the respiratory tract and that is also one of the most important invasive pathogens of swine, commonly causing meningitis, arthritis, and septicemia. Due to the existence of many serotypes and a wide range of immune evasion capabilities, efficacious vaccines are not readily available. The selection of S. suis protein candidates for inclusion in a vaccine was accomplished by identifying fitness genes through a functional genomics screen and selecting conserved predicted surface-associated proteins. Five candidate proteins were selected for evaluation in a vaccine trial and administered both intranasally and intramuscularly with one of two different adjuvant formulations. Clinical protection was evaluated by subsequent intranasal challenge with virulent S. suis While subunit vaccination with the S. suis proteins induced IgG antibodies to each individual protein and a cellular immune response to the pool of proteins and provided substantial protection from challenge with virulent S. suis, the immune response elicited and the degree of protection were dependent on the parenteral adjuvant given. Subunit vaccination induced IgG reactive against different S. suis serotypes, indicating a potential for cross protection.


Assuntos
Proteínas de Bactérias/imunologia , Infecções Estreptocócicas/veterinária , Vacinas Estreptocócicas/administração & dosagem , Streptococcus suis/imunologia , Doenças dos Suínos/prevenção & controle , Animais , Anticorpos Antibacterianos/imunologia , Proteínas de Bactérias/administração & dosagem , Proteínas de Bactérias/genética , Proteção Cruzada , Feminino , Genômica , Masculino , Infecções Estreptocócicas/imunologia , Infecções Estreptocócicas/microbiologia , Infecções Estreptocócicas/prevenção & controle , Vacinas Estreptocócicas/genética , Vacinas Estreptocócicas/imunologia , Streptococcus suis/química , Streptococcus suis/genética , Streptococcus suis/patogenicidade , Suínos , Doenças dos Suínos/imunologia , Doenças dos Suínos/microbiologia , Vacinas de Subunidades Antigênicas/administração & dosagem , Vacinas de Subunidades Antigênicas/genética , Vacinas de Subunidades Antigênicas/imunologia , Virulência
13.
J Clin Microbiol ; 55(9): 2617-2628, 2017 09.
Artigo em Inglês | MEDLINE | ID: mdl-28615466

RESUMO

Haemophilus parasuis is a diverse bacterial species that is found in the upper respiratory tracts of pigs and can also cause Glässer's disease and pneumonia. A previous pangenome study of H. parasuis identified 48 genes that were associated with clinical disease. Here, we describe the development of a generalized linear model (termed a pathotyping model) to predict the potential virulence of isolates of H. parasuis based on a subset of 10 genes from the pangenome. A multiplex PCR (mPCR) was constructed based on these genes, the results of which were entered into the pathotyping model to yield a prediction of virulence. This new diagnostic mPCR was tested on 143 field isolates of H. parasuis that had previously been whole-genome sequenced and a further 84 isolates from the United Kingdom from cases of H. parasuis-related disease in pigs collected between 2013 and 2014. The combination of the mPCR and the pathotyping model predicted the virulence of an isolate with 78% accuracy for the original isolate collection and 90% for the additional isolate collection, providing an overall accuracy of 83% (81% sensitivity and 93% specificity) compared with that of the "current standard" of detailed clinical metadata. This new pathotyping assay has the potential to aid surveillance and disease control in addition to serotyping data.


Assuntos
Infecções por Haemophilus/diagnóstico , Infecções por Haemophilus/veterinária , Haemophilus parasuis/genética , Haemophilus parasuis/patogenicidade , Técnicas de Diagnóstico Molecular/métodos , Doenças dos Suínos/diagnóstico , Animais , Genoma/genética , Infecções por Haemophilus/microbiologia , Haemophilus parasuis/isolamento & purificação , Reação em Cadeia da Polimerase Multiplex , Suínos , Doenças dos Suínos/microbiologia , Virulência/genética
14.
J Clin Microbiol ; 53(12): 3812-21, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26424843

RESUMO

Haemophilus parasuis causes Glässer's disease and pneumonia in pigs. Indirect hemagglutination (IHA) is typically used to serotype this bacterium, distinguishing 15 serovars with some nontypeable isolates. The capsule loci of the 15 reference strains have been annotated, and significant genetic variation was identified between serovars, with the exception of serovars 5 and 12. A capsule locus and in silico serovar were identified for all but two nontypeable isolates in our collection of >200 isolates. Here, we describe the development of a multiplex PCR, based on variation within the capsule loci of the 15 serovars of H. parasuis, for rapid molecular serotyping. The multiplex PCR (mPCR) distinguished between all previously described serovars except 5 and 12, which were detected by the same pair of primers. The detection limit of the mPCR was 4.29 × 10(5) ng/µl bacterial genomic DNA, and high specificity was indicated by the absence of reactivity against closely related commensal Pasteurellaceae and other bacterial pathogens of pigs. A subset of 150 isolates from a previously sequenced H. parasuis collection was used to validate the mPCR with 100% accuracy compared to the in silico results. In addition, the two in silico-nontypeable isolates were typeable using the mPCR. A further 84 isolates were analyzed by mPCR and compared to the IHA serotyping results with 90% concordance (excluding those that were nontypeable by IHA). The mPCR was faster, more sensitive, and more specific than IHA, enabling the differentiation of 14 of the 15 serovars of H. parasuis.


Assuntos
Técnicas de Genotipagem/métodos , Haemophilus parasuis/classificação , Haemophilus parasuis/genética , Reação em Cadeia da Polimerase Multiplex/métodos , Sorotipagem/métodos , Animais , Cápsulas Bacterianas/genética , Loci Gênicos , Infecções por Haemophilus/diagnóstico , Infecções por Haemophilus/microbiologia , Infecções por Haemophilus/veterinária , Haemophilus parasuis/isolamento & purificação , Sensibilidade e Especificidade , Suínos , Doenças dos Suínos/diagnóstico , Doenças dos Suínos/microbiologia , Fatores de Tempo
15.
J Antimicrob Chemother ; 70(8): 2217-22, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-25957382

RESUMO

OBJECTIVES: The objective of this study was to determine the distribution and genetic basis of trimethoprim resistance in Actinobacillus pleuropneumoniae isolates from pigs in England. METHODS: Clinical isolates collected between 1998 and 2011 were tested for resistance to trimethoprim and sulphonamide. The genetic basis of trimethoprim resistance was determined by shotgun WGS analysis and the subsequent isolation and sequencing of plasmids. RESULTS: A total of 16 (out of 106) A. pleuropneumoniae isolates were resistant to both trimethoprim (MIC >32 mg/L) and sulfisoxazole (MIC ≥256 mg/L), and a further 32 were resistant only to sulfisoxazole (MIC ≥256 mg/L). Genome sequence data for the trimethoprim-resistant isolates revealed the presence of the dfrA14 dihydrofolate reductase gene. The distribution of plasmid sequences in multiple contigs suggested the presence of two distinct dfrA14-containing plasmids in different isolates, which was confirmed by plasmid isolation and sequencing. Both plasmids encoded mobilization genes, the sulphonamide resistance gene sul2, as well as dfrA14 inserted into strA, a streptomycin-resistance-associated gene, although the gene order differed between the two plasmids. One of the plasmids further encoded the strB streptomycin-resistance-associated gene. CONCLUSIONS: This is the first description of mobilizable plasmids conferring trimethoprim resistance in A. pleuropneumoniae and, to our knowledge, the first report of dfrA14 in any member of the Pasteurellaceae. The identification of dfrA14 conferring trimethoprim resistance in A. pleuropneumoniae isolates will facilitate PCR screens for resistance to this important antimicrobial.


Assuntos
Infecções por Actinobacillus/veterinária , Actinobacillus pleuropneumoniae/efeitos dos fármacos , Plasmídeos , Doenças dos Suínos/microbiologia , Tetra-Hidrofolato Desidrogenase/genética , Resistência a Trimetoprima , Infecções por Actinobacillus/microbiologia , Actinobacillus pleuropneumoniae/enzimologia , Actinobacillus pleuropneumoniae/genética , Actinobacillus pleuropneumoniae/isolamento & purificação , Animais , Anti-Infecciosos/farmacologia , Inglaterra , Genoma Bacteriano , Testes de Sensibilidade Microbiana , Dados de Sequência Molecular , Análise de Sequência de DNA , Sulfisoxazol/farmacologia , Suínos , Trimetoprima/farmacologia
16.
Vet Res ; 46: 102, 2015 Sep 23.
Artigo em Inglês | MEDLINE | ID: mdl-26395877

RESUMO

Haemophilus parasuis is a common inhabitant of the upper respiratory tract of pigs, and the causative agent of Glässer's disease. This disease is characterized by polyserositis and arthritis, produced by the severe inflammation caused by the systemic spread of the bacterium. After an initial colonization of the upper respiratory tract, H. parasuis enters the lung during the early stages of pig infection. In order to study gene expression at this location, we sequenced the ex vivo and in vivo H. parasuis Nagasaki transcriptome in the lung using a metatranscriptomic approach. Comparison of gene expression under these conditions with that found in conventional plate culture showed generally reduced expression of genes associated with anabolic and catabolic pathways, coupled with up-regulation of membrane-related genes involved in carbon acquisition, iron binding and pathogenesis. Some of the up-regulated membrane genes, including ABC transporters, virulence-associated autotransporters (vtaAs) and several hypothetical proteins, were only present in virulent H. parasuis strains, highlighting their significance as markers of disease potential. Finally, the analysis also revealed the presence of numerous antisense transcripts with possible roles in gene regulation. In summary, this data sheds some light on the scarcely studied in vivo transcriptome of H. parasuis, revealing nutritional virulence as an adaptive strategy for host survival, besides induction of classical virulence factors.


Assuntos
Infecções por Haemophilus/veterinária , Haemophilus parasuis/genética , Haemophilus parasuis/patogenicidade , Pneumopatias/veterinária , Doenças dos Suínos/genética , Transcriptoma , Animais , Infecções por Haemophilus/genética , Infecções por Haemophilus/microbiologia , Haemophilus parasuis/metabolismo , Pneumopatias/genética , Pneumopatias/microbiologia , Análise de Sequência de DNA/veterinária , Suínos , Doenças dos Suínos/microbiologia , Regulação para Cima , Virulência , Fatores de Virulência
17.
BMC Genomics ; 15: 1179, 2014 Dec 24.
Artigo em Inglês | MEDLINE | ID: mdl-25539682

RESUMO

BACKGROUND: Haemophilus parasuis is the etiologic agent of Glässer's disease in pigs and causes devastating losses to the farming industry. Whilst some hyper-virulent isolates have been described, the relationship between genetics and disease outcome has been only partially established. In particular, there is weak correlation between serovar and disease phenotype. We sequenced the genomes of 212 isolates of H. parasuis and have used this to describe the pan-genome and to correlate this with clinical and carrier status, as well as with serotype. RESULTS: Recombination and population structure analyses identified five groups with very high rates of recombination, separated into two clades of H. parasuis with no signs of recombination between them. We used genome-wide association methods including discriminant analysis of principal components (DAPC) and generalised linear modelling (glm) to look for genetic determinants of this population partition, serovar and pathogenicity. We were able to identify genes from the accessory genome that were significantly associated with phenotypes such as potential serovar specific genes including capsule genes, and 48 putative virulence factors that were significantly different between the clinical and non-clinical isolates. We also show that the presence of many previously suggested virulence factors is not an appropriate marker of virulence. CONCLUSIONS: These genes will inform the generation of new molecular diagnostics and vaccines, and refinement of existing typing schemes and show the importance of the accessory genome of a diverse species when investigating the relationship between genotypes and phenotypes.


Assuntos
Estudo de Associação Genômica Ampla , Haemophilus parasuis/patogenicidade , Animais , Genoma Viral , Haemophilus parasuis/classificação , Haemophilus parasuis/genética , Recombinação Genética , Suínos/virologia , Virulência/genética
18.
J Clin Microbiol ; 52(7): 2380-5, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24759717

RESUMO

An improved multiplex PCR, using redesigned primers targeting the serovar 3 capsule locus, which differentiates serovars 3, 6, and 8 Actinobacillus pleuropneumoniae isolates, is described. The new primers eliminate an aberrant serovar 3-indicative amplicon found in some serovar 6 clinical isolates. Furthermore, we have developed a new multiplex PCR for the detection of serovars 1 to 3, 5 to 8, 10, and 12 along with apxIV, thus extending the utility of this diagnostic PCR to cover a broader range of isolates.


Assuntos
Actinobacillus pleuropneumoniae/classificação , Actinobacillus pleuropneumoniae/genética , Reação em Cadeia da Polimerase Multiplex/métodos , Infecções por Actinobacillus/microbiologia , Actinobacillus pleuropneumoniae/isolamento & purificação , Animais , Primers do DNA/genética , DNA Bacteriano/química , DNA Bacteriano/genética , Humanos , Dados de Sequência Molecular , Análise de Sequência de DNA , Sorogrupo
19.
Vet Res ; 45: 104, 2014 Nov 27.
Artigo em Inglês | MEDLINE | ID: mdl-25428823

RESUMO

Haemophilus parasuis is a commensal bacterium of the upper respiratory tract of healthy pigs. It is also the etiological agent of Glässer's disease, a systemic disease characterized by polyarthritis, fibrinous polyserositis and meningitis, which causes high morbidity and mortality in piglets. The aim of this study was to evaluate biofilm formation by well-characterized virulent and non-virulent strains of H. parasuis. We observed that non-virulent strains isolated from the nasal cavities of healthy pigs formed significantly (p < 0.05) more biofilms than virulent strains isolated from lesions of pigs with Glässer's disease. These differences were observed when biofilms were formed in microtiter plates under static conditions or formed in the presence of shear force in a drip-flow apparatus or a microfluidic system. Confocal laser scanning microscopy using different fluorescent probes on a representative subset of strains indicated that the biofilm matrix contains poly-N-acetylglucosamine, proteins and eDNA. The biofilm matrix was highly sensitive to degradation by proteinase K. Comparison of transcriptional profiles of biofilm and planktonic cells of the non-virulent H. parasuis F9 strain revealed a significant number of up-regulated membrane-related genes in biofilms, and genes previously identified in Actinobacillus pleuropneumoniae biofilms. Our data indicate that non-virulent strains of H. parasuis have the ability to form robust biofilms in contrast to virulent, systemic strains. Biofilm formation might therefore allow the non-virulent strains to colonize and persist in the upper respiratory tract of pigs. Conversely, the planktonic state of the virulent strains might allow them to disseminate within the host.


Assuntos
Biofilmes/crescimento & desenvolvimento , Infecções por Haemophilus/veterinária , Haemophilus parasuis/fisiologia , Haemophilus parasuis/patogenicidade , Doenças dos Suínos/microbiologia , Traqueia/microbiologia , Animais , Infecções por Haemophilus/microbiologia , Haemophilus parasuis/genética , Haemophilus parasuis/crescimento & desenvolvimento , Microscopia Confocal/veterinária , Dados de Sequência Molecular , Análise de Sequência de DNA/veterinária , Suínos , Virulência
20.
J Bacteriol ; 195(18): 4264-73, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-23873912

RESUMO

Haemophilus parasuis is the causative agent of Glässer's disease, a systemic disease of pigs, and is also associated with pneumonia. H. parasuis can be classified into 15 different serovars. Here we report, from the 15 serotyping reference strains, the DNA sequences of the loci containing genes for the biosynthesis of the group 1 capsular polysaccharides, which are potential virulence factors of this bacterium. We contend that these loci contain genes for polysaccharide capsule structures, and not a lipopolysaccharide O antigen, supported by the fact that they contain genes such as wza, wzb, and wzc, which are associated with the export of polysaccharide capsules in the current capsule classification system. A conserved region at the 3' end of the locus, containing the wza, ptp, wzs, and iscR genes, is consistent with the characteristic export region 1 of the model group 1 capsule locus. A potential serovar-specific region (region 2) has been found by comparing the predicted coding sequences (CDSs) in all 15 loci for synteny and homology. The region is unique to each reference strain with the exception of those in serovars 5 and 12, which are identical in terms of gene content. The identification and characterization of this locus among the 15 serovars is the first step in understanding the genetic, molecular, and structural bases of serovar specificity in this poorly studied but important pathogen and opens up the possibility of developing an improved molecular serotyping system, which would greatly assist diagnosis and control of Glässer's disease.


Assuntos
Proteínas de Bactérias/genética , Variação Genética , Haemophilus parasuis/classificação , Haemophilus parasuis/genética , Polissacarídeos Bacterianos/biossíntese , Doenças dos Suínos/microbiologia , Animais , Proteínas de Bactérias/metabolismo , Técnicas de Tipagem Bacteriana , Infecções por Haemophilus/microbiologia , Infecções por Haemophilus/veterinária , Haemophilus parasuis/metabolismo , Polissacarídeos Bacterianos/química , Polissacarídeos Bacterianos/genética , Análise de Sequência de DNA , Sorotipagem , Especificidade da Espécie , Sus scrofa , Suínos , Virulência , Fatores de Virulência/biossíntese , Fatores de Virulência/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA