Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 207
Filter
Add more filters

Publication year range
1.
Mamm Genome ; 31(1-2): 54-67, 2020 02.
Article in English | MEDLINE | ID: mdl-31960078

ABSTRACT

Actinobacillus (A.) pleuropneumoniae is one of the most important respiratory pathogens in global pig production. Antimicrobial treatment and vaccination provide only limited protection, but genetic disease resistance is a very promising alternative for sustainable prophylaxis. Previous studies have discovered multiple QTL that may explain up to 30% of phenotypic variance. Based on these findings, the aim of the present study was to use genomic sequencing to identify genetic markers for resistance to pleuropneumonia in a segregating commercial German Landrace line. 163 pigs were infected with A. pleuropneumoniae Serotype 7 through a standardized aerosol infection method. Phenotypes were accurately defined on a clinical, pathological and microbiological basis. The 58 pigs with the most extreme phenotypes were genotyped by sequencing (next-generation sequencing). SNPs were used in a genome-wide association study. The study identified genome-wide associated SNPs on three chromosomes, two of which were chromosomes of QTL which had been mapped in a recent experiment. Each variant explained up to 20% of the total phenotypic variance. Combined, the three variants explained 52.8% of the variance. The SNPs are located in genes involved in the pathomechanism of pleuropneumonia. This study confirms the genetic background for the host's resistance to pleuropneumonia and indicates a potential role of three candidates on SSC2, SSC12 and SSC15. Favorable gene variants are segregating in commercial populations. Further work is needed to verify the results in a controlled study and to identify the functional QTN.


Subject(s)
Disease Resistance/genetics , Pleuropneumonia/veterinary , Quantitative Trait Loci/genetics , Swine Diseases/immunology , Actinobacillus Infections/immunology , Actinobacillus Infections/microbiology , Actinobacillus Infections/veterinary , Actinobacillus pleuropneumoniae/pathogenicity , Animals , Breeding , Chromosome Mapping/veterinary , Genetic Markers , Genetic Variation , Genome-Wide Association Study/veterinary , Genotype , Phenotype , Pleuropneumonia/immunology , Pleuropneumonia/microbiology , Polymorphism, Single Nucleotide , Swine , Swine Diseases/microbiology
2.
Molecules ; 25(23)2020 Dec 03.
Article in English | MEDLINE | ID: mdl-33287430

ABSTRACT

Respiratory infections are a real threat for humans, and therefore the pig model is of interest for studies. As one of a case for studies, Actinobacillus pleuropneumoniae (APP) caused infections and still worries many pig breeders around the world. To better understand the influence of pathogenic effect of APP on a respiratory system-lungs and tracheobronchial lymph nodes (TBLN), we aimed to employ matrix-assisted laser desorption/ionization time-of-flight mass spectrometry imaging (MALDI-TOF MSI). In this study, six pigs were intranasally infected by APP and two were used as non-infected control, and 48 cryosections have been obtained. MALDI-TOF MSI and immunohistochemistry (IHC) were used to study spatial distribution of infectious markers, especially interleukins, in cryosections of porcine tissues of lungs (necrotic area, marginal zone) and tracheobronchial lymph nodes (TBLN) from pigs infected by APP. CD163, interleukin 1ß (IL­1ß) and a protegrin-4 precursor were successfully detected based on their tryptic fragments. CD163 and IL­1ß were confirmed also by IHC. The protegrin-4 precursor was identified by MALDI-TOF/TOF directly on the tissue cryosections. CD163, IL­1ß and protegrin­4 precursor were all significantly (p < 0.001) more expressed in necrotic areas of lungs infected by APP than in marginal zone, TBLN and in control lungs.


Subject(s)
Biomarkers/metabolism , Bronchi/metabolism , Lung/metabolism , Lymph Nodes/metabolism , Respiratory Tract Infections/metabolism , Actinobacillus Infections/metabolism , Actinobacillus Infections/microbiology , Actinobacillus pleuropneumoniae/pathogenicity , Animals , Antigens, CD/metabolism , Antigens, Differentiation, Myelomonocytic/metabolism , Antimicrobial Cationic Peptides/metabolism , Interleukin-1beta/metabolism , Receptors, Cell Surface/metabolism , Respiratory Tract Infections/microbiology , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization/methods , Swine
3.
Infect Immun ; 87(9)2019 09.
Article in English | MEDLINE | ID: mdl-31285248

ABSTRACT

Actinobacillus pleuropneumoniae is a capnophilic pathogen of the porcine respiratory tract lacking enzymes of the oxidative branch of the tricarboxylic acid (TCA) cycle. We previously claimed that A. pleuropneumoniae instead uses the reductive branch in order to generate energy and metabolites. Here, we show that bicarbonate and oxaloacetate supported anaerobic growth of A. pleuropneumoniae Isotope mass spectrometry revealed heterotrophic fixation of carbon from stable isotope-labeled bicarbonate by A. pleuropneumoniae, which was confirmed by nano-scale secondary ion mass spectrometry at a single-cell level. By gas chromatography-combustion-isotope ratio mass spectrometry we could further show that the labeled carbon atom is mainly incorporated into the amino acids aspartate and lysine, which are derived from the TCA metabolite oxaloacetate. We therefore suggest that carbon fixation occurs at the interface of glycolysis and the reductive branch of the TCA cycle. The heme precursor δ-aminolevulinic acid supported growth of A. pleuropneumoniae, similar to bicarbonate, implying that anaplerotic carbon fixation is needed for heme synthesis. However, deletion of potential carbon-fixing enzymes, including PEP-carboxylase (PEPC), PEP-carboxykinase (PEPCK), malic enzyme, and oxaloacetate decarboxylase, as well as various combinations thereof, did not affect carbon fixation. Interestingly, generation of a deletion mutant lacking all four enzymes was not possible, suggesting that carbon fixation in A. pleuropneumoniae is an essential metabolic pathway controlled by a redundant set of enzymes. A double deletion mutant lacking PEPC and PEPCK was not impaired in carbon fixation in vitro but showed reduction of virulence in a pig infection model.


Subject(s)
Actinobacillus Infections/metabolism , Actinobacillus pleuropneumoniae , Carbon Cycle/physiology , Pleuropneumonia/metabolism , Virulence/physiology , Actinobacillus pleuropneumoniae/metabolism , Actinobacillus pleuropneumoniae/pathogenicity , Animals , Disease Models, Animal , Swine
4.
Microb Pathog ; 126: 310-317, 2019 Jan.
Article in English | MEDLINE | ID: mdl-30419341

ABSTRACT

Pili have been demonstrated to contribute to the pathogenicity of many bacterial pathogens. Flp pilus encoded by the tad locus belongs to the type IVb pilus. Our previous study has revealed that the intact tad locus is essential for Flp pilus formation in Actinobacillus pleuropneumoniae, a very important porcine respiratory pathogen. To further investigate the functions of Flp pilus in A. pleuropneumoniae pathogenesis, the flp1 and tadD single deletion mutants were constructed by homologous recombination. Both of the mutant strains lost pilus on their cell surfaces. The abilities of biofilm formation, cell adhesion, resistance to phagocytosis, survival in swine whole blood, and in vivo colonization of the two mutants were significantly reduced compared with those of the parental strain. The corresponding complemented strains recovered the phenotypes. These results demonstrated that flp1 and tadD were essential for the biosynthesis of Flp pilus and that the pilus played important roles during infection of A. pleuropneumoniae.


Subject(s)
Actinobacillus Infections/microbiology , Actinobacillus pleuropneumoniae/metabolism , Actinobacillus pleuropneumoniae/pathogenicity , Bacterial Proteins/metabolism , Fimbriae Proteins/metabolism , Fimbriae, Bacterial/metabolism , Actinobacillus Infections/blood , Actinobacillus pleuropneumoniae/growth & development , Animals , Bacterial Adhesion/genetics , Bacterial Proteins/genetics , Biofilms/growth & development , Cell Line , Disease Models, Animal , Female , Fimbriae Proteins/genetics , Fimbriae, Bacterial/genetics , Gene Expression Regulation, Bacterial , Genes, Bacterial , Homologous Recombination , Mice, Inbred BALB C , Microbial Viability , Phagocytosis , Phenotype , Sequence Deletion , Virulence
5.
Microb Pathog ; 128: 381-389, 2019 Mar.
Article in English | MEDLINE | ID: mdl-30664928

ABSTRACT

Actinobacillus pleuropneumoniae (APP) causes porcine pleuropneumonia, a disease responsible for substantial losses in the worldwide pig industry. In this study, outbred Kunming (KM) and Institute of Cancer Research (ICR) mice were evaluated as alternative mice models for APP research. After intranasal infection of serotype 5 reference strain L20, there was less lung damage and a lower clinical sign score in ICR compared to KM mice. However, ICR mice showed more obvious changes in body weight loss, the amount of immune cells (such as neutrophils and lymphocytes) and cytokines (such as IL-6, IL-1ß and TNF-α) in blood and bronchoalveolar lavage fluid (BALF). The immunological changes observed in ICR mice closely mimicked those found in piglets infected with L20. While both ICR and KM mice are susceptible to APP and induce pathological lesions, we suggest that ICR and KM mice are more suitable for immunological and pathogenesis studies, respectively. The research lays the theoretical basis for determine that mice could replace pigs as the APP infection model and it is of significance for the study of APP infection in the laboratory.


Subject(s)
Actinobacillus Infections , Actinobacillus pleuropneumoniae/pathogenicity , Disease Models, Animal , Pleuropneumonia , Actinobacillus Infections/blood , Actinobacillus Infections/immunology , Actinobacillus Infections/microbiology , Actinobacillus Infections/pathology , Animals , Bacterial Load , Body Weight , Bronchoalveolar Lavage Fluid , Cytokines/blood , Female , Lung/microbiology , Lung/pathology , Lung Injury/microbiology , Lung Injury/pathology , Lymphocytes , Mice , Neutrophils , Pleuropneumonia/blood , Pleuropneumonia/immunology , Pleuropneumonia/microbiology , Pleuropneumonia/pathology , Serogroup , Survival Rate , Swine , Swine Diseases/microbiology
6.
Microb Pathog ; 134: 103596, 2019 Sep.
Article in English | MEDLINE | ID: mdl-31212036

ABSTRACT

To establish infection in the host, pathogens have evolved sophisticated systems to cope with environmental conditions and to protect cells against host immunity. TolC is the outer membrane channel component of type 1 secretion systems and multidrug efflux pumps that plays critical roles during the infection process in many pathogens. However, little is known about the exact roles of TolC1 in the pathogenicity of A. pleuropneumoniae, an etiological agent of the porcine contagious pleuropneumoniae that causes severe respiratory disease. In this study, deletion of tolC1 causes apparent ultrastructural defects in A. pleuropneumoniae cell examined by transmission electron microscopy. The tolC1 mutant is hypersensitivity to oxidative, osmotic and acid challenges by in vitro stress assays. Analysis on secreted proteins shows that the excretion of ApxIIA and an ApxIVA-like protein, ApxIVA-S, is abolished in the absence of TolC1. This result confirms the essential role of TolC1 in the secretion of Apx toxins and this is the first identification of an ApxIVA-like protein in in vitro culture of A. pleuropneumoniae. Besides, disruption of TolC1 leads to a significant attenuation of virulence in mice by an intraperitoneal route of A. pleuropneumoniae. The basis for the attenuation is further investigated using a mouse intranasal infection model, which reveals an impaired ability to colonize and induce lesions in the lungs for the loss of TolC1 of A. pleuropneumoniae. In conclusion, our findings demonstrate significant roles of TolC1 in facilitating bacterial survival in hostile conditions, maximum colonization as well as pathogenicity during the infection of A. pleuropneumoniae.


Subject(s)
Actinobacillus Infections/microbiology , Actinobacillus pleuropneumoniae/physiology , Bacterial Outer Membrane Proteins/chemistry , Bacterial Outer Membrane Proteins/metabolism , Bacterial Proteins/chemistry , Bacterial Proteins/metabolism , Virulence Factors/metabolism , Actinobacillus Infections/pathology , Actinobacillus pleuropneumoniae/cytology , Actinobacillus pleuropneumoniae/pathogenicity , Animals , Bacterial Outer Membrane Proteins/classification , Bacterial Outer Membrane Proteins/genetics , Bacterial Proteins/classification , Bacterial Proteins/genetics , Disease Models, Animal , Gene Deletion , Genes, MDR , Hemolysin Proteins/genetics , Hemolysin Proteins/metabolism , Host-Pathogen Interactions/physiology , Lung/microbiology , Lung/pathology , Mice , Osmotic Pressure , Oxidative Stress , Proteome/analysis , Proteome/isolation & purification , Recombinant Proteins , Stress, Physiological , Transcriptome , Type I Secretion Systems/chemistry , Type I Secretion Systems/genetics , Type I Secretion Systems/metabolism , Virulence , Virulence Factors/genetics
7.
RNA ; 22(9): 1373-85, 2016 09.
Article in English | MEDLINE | ID: mdl-27402897

ABSTRACT

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.


Subject(s)
Actinobacillus pleuropneumoniae/genetics , Algorithms , RNA, Small Untranslated/genetics , Sequence Analysis, RNA/methods , Actinobacillus pleuropneumoniae/pathogenicity , Genome, Bacterial , RNA, Small Untranslated/chemistry , Software , Transcriptome
8.
Transgenic Res ; 27(5): 409-422, 2018 10.
Article in English | MEDLINE | ID: mdl-30003470

ABSTRACT

Use of huge amounts of antibiotics in farm animal production has promoted the prevalence of antibiotic-resistant bacteria, which poses a serious threat to public health. Therefore, alternative approaches are needed to reduce or replace antibiotic usage in the food animal industry. PR-39 is a pig-derived proline-rich antimicrobial peptide that has a broad spectrum of antibacterial activity and a low propensity for development of resistance by microorganisms. To test whether ubiquitous expression of PR-39 in transgenic (TG) mice can increase resistance against bacterial infection, we generated TG mice that ubiquitously express a pig-derived antimicrobial peptide PR-39 and analyzed their growth and resistance to infection of the highly pathogenic Actinobacillus pleuropneumoniae (APP) isolated from swine. The growth performance was significantly increased in TG mice compared with their wild-type (WT) littermates. After the APP challenge, TG mice exhibited a significantly higher survival rate and significantly lower tissue bacterial load than WT littermates. Furthermore, the tissue lesion severity that resulted from APP infection was milder in TG mice than that in their WT littermates. This study provides a good foundation for the development of PR-39-expressing TG animals, which could reduce the use of antibiotics in the farm animal industry.


Subject(s)
Actinobacillus Infections/genetics , Antimicrobial Cationic Peptides/genetics , Disease Resistance/genetics , Mice, Transgenic , Actinobacillus Infections/microbiology , Actinobacillus Infections/mortality , Actinobacillus pleuropneumoniae/pathogenicity , Animals , Antimicrobial Cationic Peptides/metabolism , Bacterial Load , Female , Gene Expression , HEK293 Cells , Humans , Male , Mice, Inbred C57BL , Mice, Transgenic/growth & development , Mice, Transgenic/microbiology , Promoter Regions, Genetic , Swine
9.
BMC Microbiol ; 17(1): 193, 2017 Sep 12.
Article in English | MEDLINE | ID: mdl-28899359

ABSTRACT

BACKGROUND: The complexity of the pathogenic mechanism underlying the host immune response to Actinobacillus pleuropneumonia (App) makes the use of preventive measures difficult, and a more global view of the host-pathogen interactions and new insights into this process are urgently needed to reveal the pathogenic and immune mechanisms underlying App infection. Here, we infected specific pathogen-free Mus musculus with App serotype 7 by intranasal inoculation to construct an acute hemorrhagic pneumonia infection model and isolated the infected lungs for analysis of the interactions by dual RNA-seq. RESULTS: Four cDNA libraries were constructed, and 2428 differentially expressed genes (DEGs) of the host and 333 DEGs of App were detected. The host DEGs were mainly enriched in inflammatory signaling pathways, such as the TLR, NLR, RLR, BCR and TCR signaling pathways, resulting in large-scale cytokine up-regulation and thereby yielding a cytokine cascade for anti-infection and lung damage. The majority of the up-regulated cytokines are involved in the IL-23/IL-17 cytokine-regulated network, which is crucial for host defense against bacterial infection. The DEGs of App were mainly related to the transport and metabolism of energy and materials. Most of these genes are metabolic genes involved in anaerobic metabolism and important for challenging the host and adapting to the anaerobic stress conditions observed in acute hemorrhagic pneumonia. Some of these genes, such as adhE, dmsA, and aspA, might be potential virulence genes. In addition, the up-regulation of genes associated with peptidoglycan and urease synthesis and the restriction of major virulence genes might be immune evasion strategies of App. The regulation of metabolic genes and major virulence genes indicate that the dominant antigens might differ during the infection process and that vaccines based on these antigens might allow establishment of a precise and targeted immune response during the early phase of infection. CONCLUSION: Through an analysis of transcriptional data by dual RNA-seq, our study presents a novel global view of the interactions of App with its host and provides a basis for further study.


Subject(s)
Actinobacillus Infections/immunology , Actinobacillus pleuropneumoniae/immunology , Actinobacillus pleuropneumoniae/pathogenicity , Gene Expression Profiling , Host-Pathogen Interactions/immunology , Sequence Analysis, RNA/methods , Serogroup , Transcriptome , Actinobacillus Infections/microbiology , Actinobacillus Infections/pathology , Actinobacillus pleuropneumoniae/genetics , Actinobacillus pleuropneumoniae/metabolism , Adaptive Immunity , Amino Acids/metabolism , Animals , Antigens, Bacterial/immunology , Base Sequence , Carbohydrate Metabolism , Chromosome Mapping , Cytokines/metabolism , Disease Models, Animal , Gene Expression Regulation , Host-Pathogen Interactions/genetics , Immune Evasion , Immunity, Innate , Interleukin-17/metabolism , Interleukin-23/metabolism , Lung/microbiology , Lung/pathology , Mice , Signal Transduction , Transcriptome/genetics , Up-Regulation , Virulence/genetics
10.
BMC Vet Res ; 13(1): 14, 2017 Jan 06.
Article in English | MEDLINE | ID: mdl-28061786

ABSTRACT

BACKGROUND: Actinobacillus pleuropneumoniae is the etiological agent of porcine pleuropneumonia, which leads to large economic losses to the swine industry worldwide. In this study, S-8△clpP△apxIIC, a double-deletion mutant of A. pleuropneumoniae was constructed, and its safety and protective efficacy were evaluated in pigs. RESULTS: The S-8△clpP△apxIIC mutant exhibited attenuated virulence in a murine (BALB/c) model, and caused no detrimental effects on pigs even at a dose of up to 1.0 × 109 CFU. Furthermore, the S-8△clpP△apxIIC mutant was able to induce a strong immune response in pigs, which included high levels of IgG1 and IgG2, stimulated gamma interferon (IFN-γ), interleukin 12 (IL-12), and interleukin 4 (IL-4) production, and conferred effective protection against the lethal challenge with A. pleuropneumoniae serovars 7 or 5a. The pigs in the S-8△clpP△apxIIC immunized groups have no lesions and reduced bacterial loads in the lung tissue after challenge. CONCLUSIONS: The data obtained in this study suggest that the S-8△clpP△apxIIC mutant can serve as a highly immunogenic and potential live attenuated vaccine candidate against A. pleuropneumoniae infection.


Subject(s)
Actinobacillus Infections/veterinary , Actinobacillus pleuropneumoniae/genetics , Actinobacillus pleuropneumoniae/immunology , Bacterial Vaccines/immunology , Swine Diseases/prevention & control , Actinobacillus Infections/microbiology , Actinobacillus Infections/prevention & control , Actinobacillus pleuropneumoniae/metabolism , Actinobacillus pleuropneumoniae/pathogenicity , Animals , Gene Deletion , Mice , Mice, Inbred BALB C , Swine , Virulence
11.
Antonie Van Leeuwenhoek ; 110(12): 1647-1657, 2017 Dec.
Article in English | MEDLINE | ID: mdl-28733844

ABSTRACT

Actinobacillus pleuropneumoniae is the cause of porcine contagious pleuropneumonia, which is one of the most important respiratory diseases in swine and causes huge economic losses in the swine industry. PotD, a polyamine-binding protein, has been well characterised in many pathogens of humans and animals. In this study, a ΔpotD2 mutant of A. pleuropneumoniae strain MS71 (serovar 1) was constructed successfully by homologous recombination. Growth curves of different strains showed that the growth of the ΔpotD2 mutant was affected greatly in the logarithmic phase compared with that of parental strain. In vitro stress assays revealed that the viability of ΔpotD2 mutant strain was significantly impaired under multiple environmental stresses, including high temperature, oxidation and hyperosmosis. Additionally, the ΔpotD2 mutant caused significantly decreased mortality in a mouse model. Taken together, the findings in this study suggest an important role of PotD2 in the growth, stress tolerance and virulence of A. pleuropneumoniae.


Subject(s)
Actinobacillus pleuropneumoniae/physiology , Adaptation, Biological/genetics , Bacterial Proteins/genetics , Stress, Physiological , Actinobacillus pleuropneumoniae/pathogenicity , Animals , Bacterial Proteins/chemistry , Bacterial Proteins/immunology , Computational Biology/methods , Genetic Complementation Test , Immune Sera/immunology , Mice , Mutation , Recombinant Proteins , Virulence/genetics
12.
Infect Immun ; 84(1): 127-37, 2016 01.
Article in English | MEDLINE | ID: mdl-26483403

ABSTRACT

Actinobacillus pleuropneumoniae is a Gram-negative bacterium belonging to the Pasteurellaceae family and the causative agent of porcine pleuropneumonia, a highly contagious lung disease causing important economic losses. Surface polysaccharides, including lipopolysaccharides (LPS) and capsular polysaccharides (CPS), are implicated in the adhesion and virulence of A. pleuropneumoniae, but their role in biofilm formation is still unclear. In this study, we investigated the requirement for these surface polysaccharides in biofilm formation by A. pleuropneumoniae serotype 1. Well-characterized mutants were used: an O-antigen LPS mutant, a truncated core LPS mutant with an intact O antigen, a capsule mutant, and a poly-N-acetylglucosamine (PGA) mutant. We compared the amount of biofilm produced by the parental strain and the isogenic mutants using static and dynamic systems. Compared to the findings for the biofilm of the parental or other strains, the biofilm of the O antigen and the PGA mutants was dramatically reduced, and it had less cell-associated PGA. Real-time PCR analyses revealed a significant reduction in the level of pgaA, cpxR, and cpxA mRNA in the biofilm cells of the O-antigen mutant compared to that in the biofilm cells of the parental strain. Specific binding between PGA and LPS was consistently detected by surface plasmon resonance, but the lack of O antigen did not abolish these interactions. In conclusion, the absence of the O antigen reduces the ability of A. pleuropneumoniae to form a biofilm, and this is associated with the reduced expression and production of PGA.


Subject(s)
Actinobacillus pleuropneumoniae/pathogenicity , Bacterial Adhesion/genetics , Biofilms/growth & development , Lipopolysaccharides/genetics , O Antigens/genetics , Actinobacillus Infections/microbiology , Actinobacillus pleuropneumoniae/genetics , Animals , Bacterial Capsules/genetics , Bacterial Capsules/immunology , Bacterial Outer Membrane Proteins/genetics , Bacterial Outer Membrane Proteins/metabolism , Bacterial Proteins/genetics , Lipopolysaccharides/metabolism , O Antigens/immunology , Protein Binding , Protein Kinases/genetics , Swine , Swine Diseases/microbiology
13.
BMC Microbiol ; 16(1): 128, 2016 06 27.
Article in English | MEDLINE | ID: mdl-27349384

ABSTRACT

BACKGROUND: Actinobacillus pleuropneumoniae is the etiologic agent of porcine contagious pleuropneumonia, which causes important worldwide economic losses in the swine industry. Several respiratory tract infections are associated with biofilm formation, and A. pleuropneumoniae has the ability to form biofilms in vitro. Biofilms are structured communities of bacterial cells enclosed in a self-produced polymer matrix that are attached to an abiotic or biotic surface. Virtually all bacteria can grow as a biofilm, and multi-species biofilms are the most common form of microbial growth in nature. The goal of this study was to determine the ability of A. pleuropneumoniae to form multi-species biofilms with other bacteria frequently founded in pig farms, in the absence of pyridine compounds (nicotinamide mononucleotide [NMN], nicotinamide riboside [NR] or nicotinamide adenine dinucleotide [NAD]) that are essential for the growth of A. pleuropneumoniae. RESULTS: For the biofilm assay, strain 719, a field isolate of A. pleuropneumoniae serovar 1, was mixed with swine isolates of Streptococcus suis, Bordetella bronchiseptica, Pasteurella multocida, Staphylococcus aureus or Escherichia coli, and deposited in 96-well microtiter plates. Based on the CFU results, A. pleuropneumoniae was able to grow with every species tested in the absence of pyridine compounds in the culture media. Interestingly, A. pleuropneumoniae was also able to form strong biofilms when mixed with S. suis, B. bronchiseptica or S. aureus. In the presence of E. coli, A. pleuropneumoniae only formed a weak biofilm. The live and dead populations, and the matrix composition of multi-species biofilms were also characterized using fluorescent markers and enzyme treatments. The results indicated that poly-N-acetyl-glucosamine remains the primary component responsible for the biofilm structure. CONCLUSIONS: In conclusion, A. pleuropneumoniae apparently is able to satisfy the requirement of pyridine compounds through of other swine pathogens by cross-feeding, which enables A. pleuropneumoniae to grow and form multi-species biofilms.


Subject(s)
Actinobacillus Infections/veterinary , Actinobacillus pleuropneumoniae/growth & development , Actinobacillus pleuropneumoniae/metabolism , Biofilms/growth & development , NAD/deficiency , Acetylglucosamine/metabolism , Actinobacillus Infections/microbiology , Actinobacillus pleuropneumoniae/isolation & purification , Actinobacillus pleuropneumoniae/pathogenicity , Animals , Biofilms/drug effects , Bordetella bronchiseptica/growth & development , Bordetella bronchiseptica/metabolism , Culture Media , Deoxyribonuclease I/pharmacology , Endopeptidase K/pharmacology , Escherichia coli/growth & development , Escherichia coli/metabolism , In Situ Hybridization, Fluorescence , Microscopy, Confocal , Niacinamide/analogs & derivatives , Niacinamide/deficiency , Nicotinamide Mononucleotide/deficiency , Pasteurella multocida/growth & development , Pasteurella multocida/metabolism , Pyridines/metabolism , Pyridinium Compounds , Species Specificity , Staphylococcus aureus/growth & development , Staphylococcus aureus/metabolism , Stem Cells , Streptococcus suis/growth & development , Streptococcus suis/metabolism , Swine , Swine Diseases/microbiology
14.
Antonie Van Leeuwenhoek ; 109(1): 51-70, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26494209

ABSTRACT

Actinobacillus pleuropneumoniae is an important pathogen that causes respiratory disease in pigs. Trimeric autotransporter adhesin (TAA) is a recently discovered bacterial virulence factor that mediates bacterial adhesion and colonization. Two TAA coding genes have been found in the genome of A. pleuropneumoniae strain 5b L20, but whether they contribute to bacterial pathogenicity is unclear. In this study, we used homologous recombination to construct a double-gene deletion mutant, ΔTAA, in which both TAA coding genes were deleted and used it in in vivo and in vitro studies to confirm that TAAs participate in bacterial auto-aggregation, biofilm formation, cell adhesion and virulence in mice. A microarray analysis was used to determine whether TAAs can regulate other A. pleuropneumoniae genes during interactions with porcine primary alveolar macrophages. The results showed that deletion of both TAA coding genes up-regulated 36 genes, including ene1514, hofB and tbpB2, and simultaneously down-regulated 36 genes, including lgt, murF and ftsY. These data illustrate that TAAs help to maintain full bacterial virulence both directly, through their bioactivity, and indirectly by regulating the bacterial type II and IV secretion systems and regulating the synthesis or secretion of virulence factors. This study not only enhances our understanding of the role of TAAs but also has significance for those studying A. pleuropneumoniae pathogenesis.


Subject(s)
Actinobacillus pleuropneumoniae/pathogenicity , Adhesins, Bacterial/genetics , Gene Expression Regulation, Bacterial , Macrophages, Alveolar/microbiology , Actinobacillus Infections/microbiology , Actinobacillus pleuropneumoniae/genetics , Actinobacillus pleuropneumoniae/metabolism , Actinobacillus pleuropneumoniae/physiology , Adhesins, Bacterial/metabolism , Animals , Bacterial Adhesion/genetics , Bacterial Adhesion/physiology , Female , Gene Deletion , Gene Expression Profiling , Genes, Bacterial , Mice , Mice, Inbred BALB C , Oligonucleotide Array Sequence Analysis , Primary Cell Culture , Swine , Type V Secretion Systems/genetics , Type V Secretion Systems/metabolism , Virulence Factors/biosynthesis , Virulence Factors/genetics
15.
BMC Genomics ; 16: 417, 2015 May 28.
Article in English | MEDLINE | ID: mdl-26018580

ABSTRACT

BACKGROUND: Actinobacillus pleuropneumoniae causes pleuropneumonia in pigs, a disease which is associated with high morbidity and mortality, as well as impaired animal welfare. To obtain in-depth understanding of this infection, the interplay between virulence factors of the pathogen and defense mechanisms of the porcine host needs to be elucidated. However, research has traditionally focused on either bacteriology or immunology; an unbiased picture of the transcriptional responses can be obtained by investigating both organisms in the same biological sample. RESULTS: Host and pathogen responses in pigs experimentally infected with A. pleuropneumoniae were analyzed by high-throughput RT-qPCR. This approach allowed concurrent analysis of selected genes encoding proteins known or hypothesized to be important in the acute phase of this infection. The expression of 17 bacterial and 31 porcine genes was quantified in lung samples obtained within the first 48 hours of infection. This provided novel insight into the early time course of bacterial genes involved in synthesis of pathogen-associated molecular patterns (lipopolysaccharide, peptidoglycan, lipoprotein) and genes involved in pattern recognition (TLR4, CD14, MD2, LBP, MYD88) in response to A. pleuropneumoniae. Significant up-regulation of proinflammatory cytokines such as IL1B, IL6, and IL8 was observed, correlating with protein levels, infection status and histopathological findings. Host genes encoding proteins involved in iron metabolism, as well as bacterial genes encoding exotoxins, proteins involved in adhesion, and iron acquisition were found to be differentially expressed according to disease progression. By applying laser capture microdissection, porcine expression of selected genes could be confirmed in the immediate surroundings of the invading pathogen. CONCLUSIONS: Microbial pathogenesis is the product of interactions between host and pathogen. Our results demonstrate the applicability of high-throughput RT-qPCR for the elucidation of dual-organism gene expression analysis during infection. We showed differential expression of 12 bacterial and 24 porcine genes during infection and significant correlation of porcine and bacterial gene expression. This is the first study investigating the concurrent transcriptional response of both bacteria and host at the site of infection during porcine respiratory infection.


Subject(s)
Actinobacillus Infections/veterinary , Actinobacillus pleuropneumoniae/genetics , Host-Pathogen Interactions , Lung/microbiology , Pleuropneumonia/veterinary , Swine Diseases/genetics , Actinobacillus Infections/genetics , Actinobacillus Infections/microbiology , Actinobacillus Infections/pathology , Actinobacillus pleuropneumoniae/pathogenicity , Animals , Bacterial Proteins/genetics , Gene Expression Profiling , Gene Expression Regulation, Bacterial , Pleuropneumonia/genetics , Pleuropneumonia/microbiology , Pleuropneumonia/pathology , RNA, Bacterial/analysis , Swine , Swine Diseases/microbiology , Swine Diseases/pathology , Virulence Factors/genetics
16.
Microbiology (Reading) ; 161(Pt 2): 387-400, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25414045

ABSTRACT

Actinobacillus pleuropneumoniae is responsible for swine pleuropneumonia, a respiratory disease that causes significant global economic loss. Its virulence depends on many factors, such as capsular polysaccharides, RTX toxins and iron-acquisition systems. Analysis of virulence may require easy-to-use models that approximate mammalian infection and avoid ethical issues. Here, we investigate the potential use of the wax moth Galleria mellonella as an informative model for A. pleuropneumoniae infection. Genotypically distinct A. pleuropneumoniae clinical isolates were able to kill larvae at 37 °C but had different LD50 values, ranging from 10(4) to 10(7) c.f.u. per larva. The most virulent isolate (1022) was able to persist and replicate within the insect, while the least virulent (780) was rapidly cleared. We observed a decrease in haemocyte concentration, aggregation and DNA damage post-infection with isolate 1022. Melanization points around bacterial cells were observed in the fat body and pericardial tissues of infected G. mellonella, indicating vigorous cell and humoral immune responses close to the larval dorsal vessel. As found in pigs, an A. pleuropneumoniae hfq mutant was significantly attenuated for infection in the G. mellonella model. Additionally, the model could be used to assess the effectiveness of several antimicrobial agents against A. pleuropneumoniae in vivo. G. mellonella is a suitable inexpensive alternative infection model that can be used to study the virulence of A. pleuropneumoniae, as well as assess the effectiveness of antimicrobial agents against this pathogen.


Subject(s)
Actinobacillus Infections/microbiology , Actinobacillus pleuropneumoniae/physiology , Disease Models, Animal , Moths/microbiology , Actinobacillus pleuropneumoniae/genetics , Actinobacillus pleuropneumoniae/pathogenicity , Animals , Humans , Larva/microbiology , Virulence
17.
Biometals ; 27(5): 891-903, 2014 Oct.
Article in English | MEDLINE | ID: mdl-24878848

ABSTRACT

Actinobacillus pleuropneumoniae (App) is a Gram-negative bacterium that causes porcine pleuropneumonia, leading to economic losses in the swine industry. Due to bacterial resistance to antibiotics, new treatments for this disease are currently being sought. Lactoferrin (Lf) is an innate immune system glycoprotein of mammals that is microbiostatic and microbicidal and affects several bacterial virulence factors. The aim of this study was to investigate whether bovine iron-free Lf (BapoLf) has an effect on the growth and virulence of App. Two serotype 1 strains (reference strain S4074 and the isolate BC52) and a serotype 7 reference strain (WF83) were analyzed. First, the ability of App to grow in iron-charged BLf was discarded because in vivo, BapoLf sequesters iron and could be a potential source of this element favoring the infection. The minimum inhibitory concentration of BapoLf was 14.62, 11.78 and 10.56 µM for the strain BC52, S4074 and WF83, respectively. A subinhibitory concentration (0.8 µM) was tested by assessing App adhesion to porcine buccal epithelial cells, biofilm production, and the secretion and function of toxins and proteases. Decrease in adhesion (24-42 %) was found in the serotype 1 strains. Biofilm production decreased (27 %) for only the strain 4074 of serotype 1. Interestingly, biofilm was decreased (60-70 %) in the three strains by BholoLf. Hemolysis of erythrocytes and toxicity towards HeLa cells were not affected by BapoLf. In contrast, proteolytic activity in all strains was suppressed in the presence of BapoLf. Finally, oxytetracycline produced synergistic effect with BapoLf against App. Our results suggest that BapoLf affects the growth and several of the virulence factors in App.


Subject(s)
Actinobacillus pleuropneumoniae/growth & development , Actinobacillus pleuropneumoniae/pathogenicity , Apoproteins/physiology , Lactoferrin/physiology , Actinobacillus Infections/etiology , Actinobacillus Infections/veterinary , Actinobacillus pleuropneumoniae/physiology , Animals , Anti-Bacterial Agents/administration & dosage , Antimicrobial Cationic Peptides/administration & dosage , Antimicrobial Cationic Peptides/immunology , Antimicrobial Cationic Peptides/physiology , Apoproteins/administration & dosage , Apoproteins/immunology , Bacterial Adhesion , Bacterial Toxins/biosynthesis , Biofilms/drug effects , Biofilms/growth & development , Cattle , Drug Synergism , HeLa Cells , Humans , Iron/metabolism , Lactoferrin/administration & dosage , Lactoferrin/immunology , Oxytetracycline/administration & dosage , Pleuropneumonia/etiology , Pleuropneumonia/veterinary , Swine , Swine Diseases/etiology , Virulence
18.
Gene ; 931: 148875, 2024 Dec 30.
Article in English | MEDLINE | ID: mdl-39173979

ABSTRACT

Porcine pleuropneumonia is one of the respiratory diseases that pigs are susceptible to Actinobacillus pleuropneumoniae (A. pleuropneumoniae), poses a great threat to the global pig industry. Glutathione (GSH) is an important sulfur source, cellular antioxidant and virulence determinant of many pathogenic bacteria. In this study, roles of two HbpA-like proteins HbpA1 and HbpA2 of A. pleuropneumoniae were analyzed. A. pleuropneumoniae mutants without HbpA2 were basically unable to grow in chemically defined medium (CDM) with GSH as the sole sulfur source and had significantly reduced oxidative tolerance; whereas mutation in hbpA1 led to reduced survival under low-temperature environments. Neither HbpA1 nor HbpA2 affects utilization of heme. These two HbpA-like proteins are not associated with the virulence of A. pleuropneumoniae. Our results reveal the correlation of A. pleuropneumoniae HbpA1 and HbpA2 in GSH utilization, highlight the roles of HbpA1 in the cold stress resistance and HbpA2 in the anti-oxidative response. GSH limitation is not a way to attenuate colonization and pathogenicity of A. pleuropneumoniae.


Subject(s)
Actinobacillus pleuropneumoniae , Bacterial Proteins , Glutathione , Oxidative Stress , Actinobacillus pleuropneumoniae/pathogenicity , Actinobacillus pleuropneumoniae/genetics , Actinobacillus pleuropneumoniae/metabolism , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Animals , Virulence , Glutathione/metabolism , Sulfur/metabolism , Swine , Cold Temperature , Actinobacillus Infections/microbiology , Actinobacillus Infections/veterinary , Swine Diseases/microbiology
19.
Infect Immun ; 81(8): 2952-61, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23732171

ABSTRACT

Actinobacillus pleuropneumoniae is the etiological agent of porcine pleuropneumonia, an economically important disease of pigs. The hfq gene in A. pleuropneumoniae, encoding the RNA chaperone and posttranscriptional regulator Hfq, is upregulated during infection of porcine lungs. To investigate the role of this in vivo-induced gene in A. pleuropneumoniae, an hfq mutant strain was constructed. The hfq mutant was defective in biofilm formation on abiotic surfaces. The level of pgaC transcript, encoding the biosynthesis of poly-ß-1,6-N-acetylglucosamine (PNAG), a major biofilm matrix component, was lower and PNAG content was 10-fold lower in the hfq mutant than in the wild-type strain. When outer membrane proteins were examined, cysteine synthase, implicated in resistance to oxidative stress and tellurite, was not found at detectable levels in the absence of Hfq. The hfq mutant displayed enhanced sensitivity to superoxide generated by methyl viologen and tellurite. These phenotypes were readily reversed by complementation with the hfq gene expressed from its native promoter. The role of Hfq in the fitness of A. pleuropneumoniae was assessed in a natural host infection model. The hfq mutant failed to colonize porcine lungs and was outcompeted by the wild-type strain (median competitive index of 2 × 10(-5)). Our data demonstrate that the in vivo-induced gene hfq is involved in the regulation of PNAG-dependent biofilm formation, resistance to superoxide stress, and the fitness and virulence of A. pleuropneumoniae in pigs and begin to elucidate the role of an in vivo-induced gene in the pathogenesis of pleuropneumonia.


Subject(s)
Actinobacillus Infections/metabolism , Actinobacillus pleuropneumoniae/physiology , Actinobacillus pleuropneumoniae/pathogenicity , Host Factor 1 Protein/metabolism , Actinobacillus Infections/genetics , Actinobacillus Infections/veterinary , Amino Acid Sequence , Animals , Base Sequence , Biofilms/growth & development , Electrophoresis, Polyacrylamide Gel , Host Factor 1 Protein/genetics , Molecular Sequence Data , Pleuropneumonia/genetics , Pleuropneumonia/metabolism , Pleuropneumonia/veterinary , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Swine , Swine Diseases/genetics , Virulence/physiology , beta-Glucans
20.
Vet Res ; 44: 2, 2013 Jan 11.
Article in English | MEDLINE | ID: mdl-23311865

ABSTRACT

A better understanding of the variation in infectivity and its relation with clinical signs may help to improve measures to control and prevent (clinical) outbreaks of diseases. Here we investigated the role of disease severity on infectivity and transmission of Actinobacillus pleuropneumoniae, a bacterium causing respiratory problems in pig farms. We carried out transmission experiments with 10 pairs of caesarean-derived, colostrum-deprived pigs. In each pair, one pig was inoculated intranasally with 5×10(6) CFUs of A. pleuropneumoniae strain 1536 and housed together with a contact pig. Clinical signs were scored and the course of infection was observed by bacterial examination and qPCR analysis of tonsillar brush and nasal swab samples. In 6 out of 10 pairs transmission to contact pigs was observed, but disease scores in contact infected pigs were low compared to the score in inoculated pigs. Whereas disease score was positively associated with bacterial load in inoculated pigs and bacterial load with the transmission rate, the disease score had a negative association with transmission. These findings indicate that in pigs with equal bacterial load, those with higher clinical scores transmit A. pleuropneumoniae less efficiently. Finally, the correlation between disease score in inoculated pigs and in positive contact pigs was low. Although translation of experimental work towards farm level has limitations, our results suggest that clinical outbreaks of A. pleuropneumoniae are unlikely to be caused only by spread of the pathogen by clinically diseased pigs, but may rather be the result of development of clinical signs in already infected pigs.


Subject(s)
Actinobacillus Infections/veterinary , Actinobacillus pleuropneumoniae/physiology , Actinobacillus pleuropneumoniae/pathogenicity , Swine Diseases/transmission , Actinobacillus Infections/microbiology , Actinobacillus Infections/transmission , Animals , Feces/microbiology , Nasal Mucosa/microbiology , Palatine Tonsil/microbiology , Polymerase Chain Reaction/veterinary , Random Allocation , Swine , Swine Diseases/microbiology
SELECTION OF CITATIONS
SEARCH DETAIL