Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 139
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Brief Bioinform ; 25(4)2024 May 23.
Artículo en Inglés | MEDLINE | ID: mdl-38975892

RESUMEN

Understanding the biological functions and processes of genes, particularly those not yet characterized, is crucial for advancing molecular biology and identifying therapeutic targets. The hypothesis guiding this study is that the 3D proximity of genes correlates with their functional interactions and relevance in prokaryotes. We introduced 3D-GeneNet, an innovative software tool that utilizes high-throughput sequencing data from chromosome conformation capture techniques and integrates topological metrics to construct gene association networks. Through a series of comparative analyses focused on spatial versus linear distances, we explored various dimensions such as topological structure, functional enrichment levels, distribution patterns of linear distances among gene pairs, and the area under the receiver operating characteristic curve by utilizing model organism Escherichia coli K-12. Furthermore, 3D-GeneNet was shown to maintain good accuracy compared to multiple algorithms (neighbourhood, co-occurrence, coexpression, and fusion) across multiple bacteria, including E. coli, Brucella abortus, and Vibrio cholerae. In addition, the accuracy of 3D-GeneNet's prediction of long-distance gene interactions was identified by bacterial two-hybrid assays on E. coli K-12 MG1655, where 3D-GeneNet not only increased the accuracy of linear genomic distance tripled but also achieved 60% accuracy by running alone. Finally, it can be concluded that the applicability of 3D-GeneNet will extend to various bacterial forms, including Gram-negative, Gram-positive, single-, and multi-chromosomal bacteria through Hi-C sequencing and analysis. Such findings highlight the broad applicability and significant promise of this method in the realm of gene association network. 3D-GeneNet is freely accessible at https://github.com/gaoyuanccc/3D-GeneNet.


Asunto(s)
Redes Reguladoras de Genes , Programas Informáticos , Algoritmos , Biología Computacional/métodos , Secuenciación de Nucleótidos de Alto Rendimiento/métodos , Escherichia coli K12/genética , Escherichia coli K12/metabolismo , Escherichia coli/genética , Escherichia coli/metabolismo
2.
Nucleic Acids Res ; 51(7): 3240-3260, 2023 04 24.
Artículo en Inglés | MEDLINE | ID: mdl-36840716

RESUMEN

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.


Asunto(s)
Actinobacillus pleuropneumoniae , Variación de la Fase , Animales , Porcinos , Actinobacillus pleuropneumoniae/genética , Actinobacillus pleuropneumoniae/metabolismo , Metilasas de Modificación del ADN/genética , Metilasas de Modificación del ADN/metabolismo , Metilación de ADN , Metiltransferasas/genética , Metiltransferasas/metabolismo , Bacterias/genética , ADN/metabolismo
3.
Vet Res ; 54(1): 42, 2023 May 26.
Artículo en Inglés | MEDLINE | ID: mdl-37237397

RESUMEN

Actinobacillus pleuropneumoniae is an important swine respiratory pathogen. Previous studies have suggested that growth as a biofilm is a natural state of A. pleuropneumoniae infection. To understand the survival features involved in the biofilm state, the growth features, morphology and gene expression profiles of planktonic and biofilm A. pleuropneumoniae were compared. A. pleuropneumoniae in biofilms showed reduced viability but maintained the presence of extracellular polymeric substances (EPS) after late log-phase. Under the microscope, bacteria in biofilms formed dense aggregated structures that were connected by abundant EPS, with reduced condensed chromatin. By construction of Δpga and ΔdspB mutants, polymeric ß-1,6-linked N-acetylglucosamine and dispersin B were confirmed to be critical for normal biofilm formation. RNA-seq analysis indicated that, compared to their planktonic counterparts, A. pleuropneumoniae in biofilms had an extensively altered transcriptome. Carbohydrate metabolism, energy metabolism and translation were significantly repressed, while fermentation and genes contributing to EPS synthesis and translocation were up-regulated. The regulators Fnr (HlyX) and Fis were found to be up-regulated and their binding motifs were identified in the majority of the differentially expressed genes, suggesting their coordinated global role in regulating biofilm metabolism. By comparing the transcriptome of wild-type biofilm and Δpga, the utilization of oligosaccharides, iron and sulfur and fermentation were found to be important in adhesion and aggregation during biofilm formation. Additionally, when used as inocula, biofilm bacteria showed reduced virulence in mouse, compared with planktonic grown cells. Thus, these results have identified new facets of A. pleuropneumoniae biofilm maintenance and regulation.


Asunto(s)
Infecciones por Actinobacillus , Actinobacillus pleuropneumoniae , Enfermedades de los Porcinos , Animales , Porcinos , Ratones , Actinobacillus pleuropneumoniae/genética , Biopelículas , Transcriptoma , Virulencia , Infecciones por Actinobacillus/veterinaria , Infecciones por Actinobacillus/microbiología , Enfermedades de los Porcinos/microbiología
4.
Microb Cell Fact ; 22(1): 159, 2023 Aug 18.
Artículo en Inglés | MEDLINE | ID: mdl-37596672

RESUMEN

Conjugate vaccines produced either by chemical or biologically conjugation have been demonstrated to be safe and efficacious in protection against several deadly bacterial diseases. However, conjugate vaccine assembly and production have several shortcomings which hinders their wider availability. Here, we developed a tool, Mobile-element Assisted Glycoconjugation by Insertion on Chromosome, MAGIC, a novel biotechnological platform that overcomes the limitations of the current conjugate vaccine design method(s). As a model, we focused our design on a leading bioconjugation method using N-oligosaccharyltransferase (OTase), PglB. The installation of MAGIC led to at least twofold increase in glycoconjugate yield via MAGIC when compared to conventional N-OTase based bioconjugation method(s). Then, we improved MAGIC to (a) allow rapid installation of glycoengineering component(s), (b) omit the usage of antibiotics, (c) reduce the dependence on protein induction agents. Furthermore, we show the modularity of the MAGIC platform in performing glycoengineering in bacterial species that are less genetically tractable than the commonly used Escherichia coli. The MAGIC system promises a rapid, robust and versatile method to develop vaccines against serious bacterial pathogens. We anticipate the utility of the MAGIC platform could enhance vaccines production due to its compatibility with virtually any bioconjugation method, thus expanding vaccine biopreparedness toolbox.


Asunto(s)
Antibacterianos , Biotecnología , Vacunas Conjugadas , Escherichia coli/genética , Desarrollo de Vacunas
5.
J Bacteriol ; 204(2): e0032621, 2022 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-34807725

RESUMEN

Bacteria have evolved a variety of enzymes to eliminate endogenous or host-derived oxidative stress factors. The Dps protein, first identified in Escherichia coli, contains a ferroxidase center, and protects bacteria from reactive oxygen species damage. Little is known of the role of Dps-like proteins in bacterial pathogenesis. Actinobacillus pleuropneumoniae causes pleuropneumonia, a respiratory disease of swine. The A. pleuropneumoniae ftpA gene is upregulated during shifts to anaerobiosis, in biofilms and, as found in this study, in the presence of H2O2. An A. pleuropneumoniae ftpA deletion mutant (ΔftpA) had increased H2O2 sensitivity, decreased intracellular viability in macrophages, and decreased virulence in a mouse infection model. Expression of ftpA in an E. coli dps mutant restored wild-type H2O2 resistance. FtpA possesses a conserved ferritin domain containing a ferroxidase site. Recombinant rFtpA bound and oxidized Fe2+ reversibly. Under aerobic conditions, the viability of an ΔftpA mutant was reduced compared with the wild-type strain after extended culture, upon transition from anaerobic to aerobic conditions, and upon supplementation with Fenton reaction substrates. Under anaerobic conditions, the addition of H2O2 resulted in a more severe growth defect of ΔftpA than it did under aerobic conditions. Therefore, by oxidizing and mineralizing Fe2+, FtpA alleviates the oxidative damage mediated by intracellular Fenton reactions. Furthermore, by mutational analysis, two residues were confirmed to be critical for Fe2+ binding and oxidization, as well as for A. pleuropneumoniae H2O2 resistance. Taken together, the results of this study demonstrate that A. pleuropneumoniae FtpA is a Dps-like protein, playing critical roles in oxidative stress resistance and virulence. IMPORTANCE As a ferroxidase, Dps of Escherichia coli can protect bacteria from reactive oxygen species damage, but its role in bacterial pathogenesis has received little attention. In this study, FtpA of the swine respiratory pathogen A. pleuropneumoniae was identified as a new Dps-like protein. It facilitated A. pleuropneumoniae resistance to H2O2, survival in macrophages, and infection in vivo. FtpA could bind and oxidize Fe2+ through two important residues in its ferroxidase site and protected the bacteria from oxidative damage mediated by the intracellular Fenton reaction. These findings provide new insights into the role of the FtpA-based antioxidant system in the pathogenesis of A. pleuropneumoniae, and the conserved Fe2+ binding ligands in Dps/FtpA provide novel drug target candidates for disease prevention.


Asunto(s)
Actinobacillus pleuropneumoniae/genética , Actinobacillus pleuropneumoniae/metabolismo , Proteínas Bacterianas/metabolismo , Oxidación-Reducción , Estrés Fisiológico/genética , Actinobacillus pleuropneumoniae/química , Animales , Proteínas de la Membrana Bacteriana Externa/genética , Proteínas Bacterianas/genética , Proteínas Bacterianas/aislamiento & purificación , Proteínas de Escherichia coli/genética , Femenino , Hierro/metabolismo , Ratones , Especies Reactivas de Oxígeno , Virulencia/genética
6.
Infect Immun ; 90(9): e0023922, 2022 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-35938858

RESUMEN

Nitrate metabolism is an adaptation mechanism used by many bacteria for survival in anaerobic environments. As a by-product of inflammation, nitrate is used by the intestinal bacterial pathogens to enable gut infection. However, the responses of bacterial respiratory pathogens to nitrate are less well understood. Actinobacillus pleuropneumoniae is an important bacterial respiratory pathogen of swine. Previous studies have suggested that adaptation of A. pleuropneumoniae to anaerobiosis is important for infection. In this work, A. pleuropneumoniae growth and pathogenesis in response to the nitrate were investigated. Nitrate significantly promoted A. pleuropneumoniae growth under anaerobic conditions in vitro and lethality in mice. By using narQ and narP deletion mutants and single-residue-mutated complementary strains of ΔnarQ, the two-component system NarQ/P was confirmed to be critical for nitrate-induced growth, with Arg50 in NarQ as an essential functional residue. Transcriptome analysis showed that nitrate upregulated multiple energy-generating pathways, including nitrate metabolism, mannose and pentose metabolism, and glycerolipid metabolism via the regulation of NarQ/P. Furthermore, narQ, narP, and its target gene encoding the nitrate reductase Nap contributed to the pathogenicity of A. pleuropneumoniae. The Nap inhibitor tungstate significantly reduced the survival of A. pleuropneumoniae in vivo, suggesting that Nap is a potential drug target. These results give new insights into how the respiratory pathogen A. pleuropneumoniae utilizes the alternative electron acceptor nitrate to overcome the hypoxia microenvironment, which can occur in the inflammatory or necrotic infected tissues.


Asunto(s)
Infecciones por Actinobacillus , Actinobacillus pleuropneumoniae , Actinobacillus pleuropneumoniae/genética , Actinobacillus pleuropneumoniae/metabolismo , Animales , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Regulación Bacteriana de la Expresión Génica , Manosa/metabolismo , Ratones , Nitrato Reductasas/genética , Nitrato Reductasas/metabolismo , Nitratos/metabolismo , Pentosas/metabolismo , Porcinos , Virulencia
7.
Vet Res ; 52(1): 30, 2021 Feb 22.
Artículo en Inglés | MEDLINE | ID: mdl-33618766

RESUMEN

Host proteins interacting with pathogens are receiving more attention as potential therapeutic targets in molecular medicine. Streptococcus suis serotype 2 (SS2) is an important cause of meningitis in both humans and pigs worldwide. SS2 Enolase (Eno) has previously been identified as a virulence factor with a role in altering blood brain barrier (BBB) integrity, but the host cell membrane receptor of Eno and The mechanism(s) involved are unclear. This study identified that SS2 Eno binds to 40S ribosomal protein SA (RPSA) on the surface of porcine brain microvascular endothelial cells leading to activation of intracellular p38/ERK-eIF4E signalling, which promotes intracellular expression of HSPD1 (heat-shock protein family D member 1), and initiation of host-cell apoptosis, and increased BBB permeability facilitating bacterial invasion. This study reveals novel functions for the host-interactional molecules RPSA and HSPD1 in BBB integrity, and provides insight for new therapeutic strategies in meningitis.


Asunto(s)
Barrera Hematoencefálica , Células Endoteliales/metabolismo , Fosfopiruvato Hidratasa/metabolismo , Proteínas Ribosómicas/metabolismo , Infecciones Estreptocócicas/veterinaria , Streptococcus suis/metabolismo , Animales , Apoptosis , Técnicas de Cocultivo , Células Endoteliales/microbiología , Factor 4E Eucariótico de Iniciación/genética , Factor 4E Eucariótico de Iniciación/metabolismo , Quinasas MAP Reguladas por Señal Extracelular/genética , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Regulación de la Expresión Génica , Proteínas de Choque Térmico/genética , Proteínas de Choque Térmico/metabolismo , Ratones , Unión Proteica , Serogrupo , Infecciones Estreptocócicas/microbiología , Infecciones Estreptocócicas/patología , Streptococcus suis/patogenicidad , Porcinos , Enfermedades de los Porcinos/microbiología , Proteínas Quinasas p38 Activadas por Mitógenos/genética , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
8.
J Bacteriol ; 202(8)2020 03 26.
Artículo en Inglés | MEDLINE | ID: mdl-32015147

RESUMEN

The (p)ppGpp-mediated stringent response (SR) is a highly conserved regulatory mechanism in bacterial pathogens, enabling adaptation to adverse environments, and is linked to pathogenesis. Actinobacillus pleuropneumoniae can cause damage to the lungs of pigs, its only known natural host. Pig lungs are known to have a low concentration of free branched-chain amino acids (BCAAs) compared to the level in plasma. We had investigated the role for (p)ppGpp in viability and biofilm formation of A. pleuropneumoniae Now, we sought to determine whether (p)ppGpp was a trigger signal for the SR in A. pleuropneumoniae in the absence of BCAAs. Combining transcriptome and phenotypic analyses of the wild type (WT) and an relA spoT double mutant [which does not produce (p)ppGpp], we found that (p)ppGpp could repress de novo purine biosynthesis and activate antioxidant pathways. There was a positive correlation between GTP and endogenous hydrogen peroxide content. Furthermore, the growth, viability, morphology, and virulence were altered by the inability to produce (p)ppGpp. Genes involved in the biosynthesis of BCAAs were constitutively upregulated, regardless of the existence of BCAAs, without accumulation of (p)ppGpp beyond a basal level. Collectively, our study shows that the absence of BCAAs was not a sufficient signal to trigger the SR in A. pleuropneumoniae (p)ppGpp-mediated regulation in A. pleuropneumoniae is different from that described for the model organism Escherichia coli Further work will establish whether the (p)ppGpp-dependent SR mechanism in A. pleuropneumoniae is conserved among other veterinary pathogens, especially those in the Pasteurellaceae family.IMPORTANCE (p)ppGpp is a key player in reprogramming transcriptomes to respond to nutritional challenges. Here, we present transcriptional and phenotypic differences of A. pleuropneumoniae grown in different chemically defined media in the absence of (p)ppGpp. We show that the deprivation of branched-chain amino acids (BCAAs) does not elicit a change in the basal-level (p)ppGpp, but this level is sufficient to regulate the expression of BCAA biosynthesis. The mechanism found in A. pleuropneumoniae is different from that of the model organism Escherichia coli but similar to that found in some Gram-positive bacteria. This study not only broadens the research scope of (p)ppGpp but also further validates the complexity and multiplicity of (p)ppGpp regulation in microorganisms that occupy different biological niches.


Asunto(s)
Actinobacillus pleuropneumoniae/metabolismo , Aminoácidos de Cadena Ramificada/metabolismo , Guanosina Pentafosfato/metabolismo , Guanosina Trifosfato/metabolismo , Actinobacillus pleuropneumoniae/crecimiento & desarrollo , Peróxido de Hidrógeno/metabolismo
9.
Infect Immun ; 88(5)2020 04 20.
Artículo en Inglés | MEDLINE | ID: mdl-32094250

RESUMEN

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.


Asunto(s)
Haemophilus parasuis/genética , Animales , Biopelículas , Infecciones por Haemophilus/microbiología , Macrófagos Alveolares/microbiología , Fagocitosis/fisiología , Porcinos , Enfermedades de los Porcinos/microbiología , Virulencia/genética
10.
Clin Infect Dis ; 70(10): 2045-2053, 2020 05 06.
Artículo en Inglés | MEDLINE | ID: mdl-31504285

RESUMEN

BACKGROUND: Neisseria meningitidis (Nm) is a nasopharyngeal commensal carried by healthy individuals. However, invasive infections occurs in a minority of individuals, with devastating consequences. There is evidence that common polymorphisms are associated with invasive meningococcal disease (IMD), but the contributions of rare variants other than those in the complement system have not been determined. METHODS: We identified familial cases of IMD in the UK meningococcal disease study and the European Union Life-Threatening Infectious Disease Study. Candidate genetic variants were identified by whole-exome sequencing of 2 patients with familial IMD. Candidate variants were further validated by in vitro assays. RESULTS: Exomes of 2 siblings with IMD identified a novel heterozygous missense mutation in BPIFA1/SPLUNC1. Sequencing of 186 other nonfamilial cases identified another unrelated IMD patient with the same mutation. SPLUNC1 is an innate immune defense protein expressed in the nasopharyngeal epithelia; however, its role in invasive infections is unknown. In vitro assays demonstrated that recombinant SPLUNC1 protein inhibits biofilm formation by Nm, and impedes Nm adhesion and invasion of human airway cells. The dominant negative mutant recombinant SPLUNC1 (p.G22E) showed reduced antibiofilm activity, increased meningococcal adhesion, and increased invasion of cells, compared with wild-type SPLUNC1. CONCLUSIONS: A mutation in SPLUNC1 affecting mucosal attachment, biofilm formation, and invasion of mucosal epithelial cells is a new genetic cause of meningococcal disease.


Asunto(s)
Glicoproteínas/genética , Infecciones Meningocócicas/genética , Infecciones Meningocócicas/microbiología , Neisseria meningitidis , Fosfoproteínas/genética , Proteínas del Sistema Complemento , Células Epiteliales , Humanos , Mutación , Neisseria meningitidis/genética
11.
BMC Vet Res ; 16(1): 167, 2020 May 27.
Artículo en Inglés | MEDLINE | ID: mdl-32460764

RESUMEN

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.


Asunto(s)
Infecciones por Haemophilus/veterinaria , Haemophilus parasuis/inmunología , Proteínas Recombinantes/inmunología , Enfermedades de los Porcinos/prevención & control , Animales , Anticuerpos Antibacterianos/sangre , Proteínas Bacterianas/inmunología , Vacunas Bacterianas/inmunología , Infecciones por Haemophilus/inmunología , Infecciones por Haemophilus/prevención & control , Vacunas contra Haemophilus/inmunología , Haemophilus parasuis/genética , Serogrupo , Sus scrofa , Porcinos , Enfermedades de los Porcinos/inmunología , Enfermedades de los Porcinos/microbiología , Técnicas de Cultivo de Tejidos/veterinaria , Vacunación/veterinaria , Vacunas de Subunidad/administración & dosificación , Vacunas de Subunidad/inmunología
12.
J Clin Microbiol ; 57(7)2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-30944194

RESUMEN

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.


Asunto(s)
Portador Sano/veterinaria , Infecciones Estreptocócicas/veterinaria , Streptococcus suis/aislamiento & purificación , Streptococcus suis/patogenicidad , Enfermedades de los Porcinos/microbiología , Animales , Portador Sano/microbiología , Inglaterra , Marcadores Genéticos/genética , Genoma Bacteriano/genética , Técnicas de Diagnóstico Molecular , Reacción en Cadena de la Polimerasa Multiplex , Tonsila Palatina/microbiología , Infecciones Estreptocócicas/microbiología , Streptococcus suis/genética , Porcinos , Virulencia/genética , Gales
13.
Infect Immun ; 86(3)2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-29203546

RESUMEN

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.


Asunto(s)
Proteínas Bacterianas/inmunología , Infecciones Estreptocócicas/veterinaria , Vacunas Estreptocócicas/administración & dosificación , Streptococcus suis/inmunología , Enfermedades de los Porcinos/prevención & control , Animales , Anticuerpos Antibacterianos/inmunología , Proteínas Bacterianas/administración & dosificación , Proteínas Bacterianas/genética , Protección Cruzada , Femenino , Genómica , Masculino , Infecciones Estreptocócicas/inmunología , Infecciones Estreptocócicas/microbiología , Infecciones Estreptocócicas/prevención & control , Vacunas Estreptocócicas/genética , Vacunas Estreptocócicas/inmunología , Streptococcus suis/química , Streptococcus suis/genética , Streptococcus suis/patogenicidad , Porcinos , Enfermedades de los Porcinos/inmunología , Enfermedades de los Porcinos/microbiología , Vacunas de Subunidad/administración & dosificación , Vacunas de Subunidad/genética , Vacunas de Subunidad/inmunología , Virulencia
14.
RNA ; 22(9): 1373-85, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-27402897

RESUMEN

Bacterial regulatory small RNAs (sRNAs) play important roles in gene regulation and are frequently connected to the expression of virulence factors in diverse bacteria. Only a few sRNAs have been described for Pasteurellaceae pathogens and no in-depth analysis of sRNAs has been described for Actinobacillus pleuropneumoniae, the causative agent of porcine pleuropneumonia, responsible for considerable losses in the swine industry. To search for sRNAs in A. pleuropneumoniae, we developed a strategy for the computational analysis of the bacterial genome by using four algorithms with different approaches, followed by experimental validation. The coding strand and expression of 17 out of 23 RNA candidates were confirmed by Northern blotting, RT-PCR, and RNA sequencing. Among them, two are likely riboswitches, three are housekeeping regulatory RNAs, two are the widely studied GcvB and 6S sRNAs, and 10 are putative novel trans-acting sRNAs, never before described for any bacteria. The latter group has several potential mRNA targets, many of which are involved with virulence, stress resistance, or metabolism, and connect the sRNAs in a complex gene regulatory network. The sRNAs identified are well conserved among the Pasteurellaceae that are evolutionarily closer to A. pleuropneumoniae and/or share the same host. Our results show that the combination of newly developed computational programs can be successfully utilized for the discovery of novel sRNAs and indicate an intricate system of gene regulation through sRNAs in A. pleuropneumoniae and in other Pasteurellaceae, thus providing clues for novel aspects of virulence that will be explored in further studies.


Asunto(s)
Actinobacillus pleuropneumoniae/genética , Algoritmos , ARN Pequeño no Traducido/genética , Análisis de Secuencia de ARN/métodos , Actinobacillus pleuropneumoniae/patogenicidad , Genoma Bacteriano , ARN Pequeño no Traducido/química , Programas Informáticos , Transcriptoma
15.
J Antimicrob Chemother ; 73(1): 57-65, 2018 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-29029160

RESUMEN

OBJECTIVES: To characterize ICEApl2, an SXT-related integrative and conjugative element (ICE) found in a clinical isolate of the porcine pathogen Actinobacillus pleuropneumoniae, and analyse the functional nature of the encoded FloR. METHODS: ICEApl2 was identified in the genome of A. pleuropneumoniae MIDG3553. Functional analysis was done using conjugal transfer experiments. MIDG3553 was tested for susceptibility to the antimicrobials for which resistance genes are present in ICEApl2. Lack of florfenicol/chloramphenicol resistance conferred by the encoded FloR protein was investigated by cloning and site-directed mutagenesis experiments in Escherichia coli. RESULTS: ICEApl2 is 92660 bp and contains 89 genes. Comparative sequence analysis indicated that ICEApl2 is a member of the SXT/R391 ICE family. Conjugation experiments showed that, although ICEApl2 is capable of excision from the chromosome, it is not self-transmissible. ICEApl2 encodes the antimicrobial resistance genes floR, strAB, sul2 and dfrA1, and MIDG3553 is resistant to streptomycin, sulfisoxazole and trimethoprim, but not florfenicol or chloramphenicol. Cloning and site-directed mutagenesis of the floR gene revealed the importance of the nature of the hydrophobic amino acid residues at positions 160 and 228 in FloR for determining resistance to florfenicol and chloramphenicol. CONCLUSIONS: Our results indicate that the nature of hydrophobic residues at positions 160 and 228 of FloR contribute dynamically to specific efflux of florfenicol and chloramphenicol, although some differences in resistance levels may depend on the bacterial host species. This is also, to our knowledge, the first description of an SXT/R391 ICE in A. pleuropneumoniae or any member of the Pasteurellaceae.


Asunto(s)
Actinobacillus pleuropneumoniae/efectos de los fármacos , Actinobacillus pleuropneumoniae/genética , Proteínas Bacterianas/genética , Transporte Biológico/genética , Cloranfenicol/farmacología , Farmacorresistencia Bacteriana Múltiple/genética , Secuencias Repetitivas Esparcidas/genética , Enfermedades de los Porcinos/microbiología , Tianfenicol/análogos & derivados , Actinobacillus pleuropneumoniae/aislamiento & purificación , Animales , Cloranfenicol/metabolismo , Conjugación Genética/genética , Interacciones Hidrofóbicas e Hidrofílicas , Pruebas de Sensibilidad Microbiana , Neumonía/microbiología , Neumonía/veterinaria , Porcinos , Tianfenicol/metabolismo , Tianfenicol/farmacología
16.
Antonie Van Leeuwenhoek ; 111(12): 2371-2384, 2018 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-30008077

RESUMEN

Actinobacillus pleuropneumoniae is the cause of porcine pleuropneumonia, for which the mortality rate is high. Host peripheral blood is a body site for the immune clearance of pathogens mediated by release of inflammatory factors. However, "out of control" inflammatory factor release can contribute to host death. To further understand the changes in the transcription level of immune-related effectors, samples of peripheral blood mononuclear cells (PBMCs) collected from piglets at different stages of infection (0, 24 and 120 h) were sequenced on an Illumina HiSeq™ 4000 platform. We found 3818 differentially expressed genes (DEGs) in the 24 h-infection group compared to the 0 h-infection group (Pb24-Vs-Pb0). DEGs mainly involved in the Gene ontology and KEGG pathways that included nucleic acid metabolism regulation, cell growth, cell differentiation, and organ morphological maintenance were not significantly enriched (P > 0.05). However, DEGs associated with protein kinase activity, receptor activation, metabolism, local adhesion and immune inflammatory responses were significantly enriched in Pb120-Vs-Pb24 (P < 0.05), as were those related to the T cell receptor signalling pathway, with most being down-regulated compared to the preceding stage (Pb24-Vs-Pb0). In PBMCs there were some changes in glucose metabolism, local adhesion and the immune inflammatory response (Pb120-Vs-Pb0). In addition, up-regulated DEGs, such as IL8, IL1ß, and CCL2, and were significantly enriched in immune-inflammatory related pathways compared to the uninfected stage, although they began to decline after 24 h.


Asunto(s)
Infecciones por Actinobacillus/veterinaria , Actinobacillus pleuropneumoniae/fisiología , Leucocitos Mononucleares/inmunología , Pleuroneumonía/veterinaria , Enfermedades de los Porcinos/genética , Infecciones por Actinobacillus/genética , Infecciones por Actinobacillus/inmunología , Infecciones por Actinobacillus/microbiología , Animales , Femenino , Perfilación de la Expresión Génica , Leucocitos Mononucleares/microbiología , Masculino , Pleuroneumonía/genética , Pleuroneumonía/inmunología , Pleuroneumonía/microbiología , Porcinos , Enfermedades de los Porcinos/inmunología , Enfermedades de los Porcinos/microbiología
17.
Infect Immun ; 85(5)2017 05.
Artículo en Inglés | MEDLINE | ID: mdl-28264906

RESUMEN

Neisseria meningitidis is a commensal microbe that colonizes the human nasopharynx but occasionally invades the bloodstream to cause life-threatening infection. N. meningitidis MC58 NMB0419 encodes a Sel1-like repeat (SLR)-containing protein, previously implicated in invasion of epithelial cells. A gene-regulatory function was revealed in Escherichia coli expressing plasmid-borne NMB0419 and showing significantly increased epithelial adherence compared to the wild type, due to increased expression of mannose-sensitive type 1 pili. While a meningococcal NMB0419 mutant did not have altered epithelial adherence, in a transcriptome-wide comparison of the wild type and an NMB0419 mutant, a large proportion of genes differentially regulated in the mutant were involved in iron acquisition and metabolism. Fifty-one percent and 38% of genes, respectively, up- and downregulated in the NMB0419 mutant had previously been identified as being induced and repressed by meningococcal Fur. An in vitro growth defect of the NMB0419 mutant under iron restriction was consistent with the downregulation of tbpAB and hmbR, while an intraepithelial replication defect was consistent with the downregulation of tonB, exbB, and exbD, based on a known phenotype of a meningococcal tonB mutant. Disruption of the N-terminal NMB0419 signal peptide, predicted to export the protein beyond the cytoplasmic membrane, resulted in loss of functional traits in N. meningitidis and E. coli Our study indicates that the expression of NMB0419 is associated with transcriptional changes counterbalancing the regulatory function of Fur, offering a new perspective on regulatory mechanisms involved in meningococcal interaction with epithelial cells, and suggests new insights into the roles of SLR-containing genes in other bacteria.


Asunto(s)
Proteínas Bacterianas/metabolismo , Células Epiteliales/microbiología , Neisseria meningitidis/crecimiento & desarrollo , Neisseria meningitidis/genética , Regulón , Proteínas Represoras/metabolismo , Factores de Transcripción/metabolismo , Adhesión Bacteriana , Escherichia coli/genética , Escherichia coli/metabolismo , Expresión Génica , Perfilación de la Expresión Génica , Regulación Bacteriana de la Expresión Génica , Técnicas de Inactivación de Genes , Hierro/metabolismo , Plásmidos , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Factores de Transcripción/genética
18.
N Engl J Med ; 370(18): 1712-1723, 2014 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-24785206

RESUMEN

BACKGROUND: Improved diagnostic tests for tuberculosis in children are needed. We hypothesized that transcriptional signatures of host blood could be used to distinguish tuberculosis from other diseases in African children who either were or were not infected with the human immunodeficiency virus (HIV). METHODS: The study population comprised prospective cohorts of children who were undergoing evaluation for suspected tuberculosis in South Africa (655 children), Malawi (701 children), and Kenya (1599 children). Patients were assigned to groups according to whether the diagnosis was culture-confirmed tuberculosis, culture-negative tuberculosis, diseases other than tuberculosis, or latent tuberculosis infection. Diagnostic signatures distinguishing tuberculosis from other diseases and from latent tuberculosis infection were identified from genomewide analysis of RNA expression in host blood. RESULTS: We identified a 51-transcript signature distinguishing tuberculosis from other diseases in the South African and Malawian children (the discovery cohort). In the Kenyan children (the validation cohort), a risk score based on the signature for tuberculosis and for diseases other than tuberculosis showed a sensitivity of 82.9% (95% confidence interval [CI], 68.6 to 94.3) and a specificity of 83.6% (95% CI, 74.6 to 92.7) for the diagnosis of culture-confirmed tuberculosis. Among patients with cultures negative for Mycobacterium tuberculosis who were treated for tuberculosis (those with highly probable, probable, or possible cases of tuberculosis), the estimated sensitivity was 62.5 to 82.3%, 42.1 to 80.8%, and 35.3 to 79.6%, respectively, for different estimates of actual tuberculosis in the groups. In comparison, the sensitivity of the Xpert MTB/RIF assay for molecular detection of M. tuberculosis DNA in cases of culture-confirmed tuberculosis was 54.3% (95% CI, 37.1 to 68.6), and the sensitivity in highly probable, probable, or possible cases was an estimated 25.0 to 35.7%, 5.3 to 13.3%, and 0%, respectively; the specificity of the assay was 100%. CONCLUSIONS: RNA expression signatures provided data that helped distinguish tuberculosis from other diseases in African children with and those without HIV infection. (Funded by the European Union Action for Diseases of Poverty Program and others).


Asunto(s)
Mycobacterium tuberculosis/genética , ARN Bacteriano/sangre , Transcriptoma , Tuberculosis/diagnóstico , África , Algoritmos , Técnicas Bacteriológicas , Niño , Preescolar , Diagnóstico Diferencial , Infecciones por VIH/complicaciones , Humanos , Lactante , Tuberculosis Latente/diagnóstico , Masculino , Mycobacterium tuberculosis/aislamiento & purificación , Análisis de Secuencia por Matrices de Oligonucleótidos , Riesgo , Sensibilidad y Especificidad , Tuberculosis/complicaciones , Tuberculosis/genética
19.
J Clin Microbiol ; 55(3): 902-907, 2017 03.
Artículo en Inglés | MEDLINE | ID: mdl-28053219

RESUMEN

Actinobacillus pleuropneumoniae causes pleuropneumonia, an economically significant lung disease of pigs. Recently, isolates of A. pleuropneumoniae that were serologically distinct from the previously characterized 15 serovars were described, and a proposal was put forward that they comprised a new serovar, serovar 16. Here we used whole-genome sequencing of the proposed serovar 16 reference strain A-85/14 to confirm the presence of a unique capsular polysaccharide biosynthetic locus. For molecular diagnostics, primers were designed from the capsule locus of strain A-85/14, and a PCR was formulated that differentiated serovar 16 isolates from all 15 known serovars and other common respiratory pathogenic/commensal bacteria of pigs. Analysis of the capsule locus of strain A-85/14 combined with the previous serological data show the existence of a sixteenth serovar-designated serovar 16-of A. pleuropneumoniae.


Asunto(s)
Infecciones por Actinobacillus/veterinaria , Actinobacillus pleuropneumoniae/clasificación , Cápsulas Bacterianas/genética , Sitios Genéticos , Reacción en Cadena de la Polimerasa/métodos , Serogrupo , Enfermedades de los Porcinos/diagnóstico , Infecciones por Actinobacillus/diagnóstico , Actinobacillus pleuropneumoniae/genética , Animales , Cartilla de ADN/genética , ADN Bacteriano/química , ADN Bacteriano/genética , Genoma Bacteriano , Técnicas de Diagnóstico Molecular/métodos , Pleuroneumonía/microbiología , Pleuroneumonía/veterinaria , Análisis de Secuencia de ADN , Porcinos
20.
J Clin Microbiol ; 55(9): 2617-2628, 2017 09.
Artículo en Inglés | MEDLINE | ID: mdl-28615466

RESUMEN

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.


Asunto(s)
Infecciones por Haemophilus/diagnóstico , Infecciones por Haemophilus/veterinaria , Haemophilus parasuis/genética , Haemophilus parasuis/patogenicidad , Técnicas de Diagnóstico Molecular/métodos , Enfermedades de los Porcinos/diagnóstico , Animales , Genoma/genética , Infecciones por Haemophilus/microbiología , Haemophilus parasuis/aislamiento & purificación , Reacción en Cadena de la Polimerasa Multiplex , Porcinos , Enfermedades de los Porcinos/microbiología , Virulencia/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA