Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 206
Filter
1.
Arq. Ciênc. Vet. Zool. UNIPAR (Online) ; 26(1cont): 226-238, jan.-jun. 2023.
Article in Portuguese | LILACS, VETINDEX | ID: biblio-1443234

ABSTRACT

As doenças respiratórias são um problema significativo na produção suína e podem levar à condenação de carcaças no abate. Entre os agentes causadores dessas doenças destacam-se o Actinobacillus pleuropneumoniae, Mycoplasma hyopneumoniae e a Pasteurella multocida. O Actinobacillus pleuropneumoniae é um patógeno altamente contagioso, que ocasiona hemorragia, pleuropneumonia purulenta e fibrosa. A Pleuropneumonia é amplamente distribuída e gera graves prejuízos para a suinocultura. O Mycoplasma hyopneumoniae ocasionador da pneumonia por micoplasma, doença respiratória crônica. As infecções originadas podem regular negativamente o sistema imunológico do hospedeiro e aumentar a infecção e assim a replicação de outros patógenos. A Pasteurella multocida é o agente causador de uma ampla gama de infecções levando a alto impacto econômico. Patógeno comensal e oportunista da boca, nasofaringe e trato respiratório superior. A identificação precoce e o manejo adequado desses agentes causadores de doenças respiratórias são fundamentais para minimizar a incidência de carcaças suínas. A adoção de medidas preventivas, como a vacinação e práticas de manejo adequadas, pode ajudar a prevenir a propagação dessas doenças e garantir a produção de carne suína segura e de alta qualidade para o consumo humano.(AU)


Respiratory diseases are a significant problem in pork production and can lead to condemnation of carcasses at slaughter. Among the causative agents of these diseases are Actinobacillus pleuropneumoniae, Mycoplasma hyopneumoniae and Pasteurella multocida. Actinobacillus pleuropneumoniae is a highly contagious pathogen that causes hemorrhage, purulent and fibrous pleuropneumonia. Pleuropneumonia is widely distributed and causes serious damage to pig farming. Mycoplasma hyopneumoniae causes mycoplasma pneumonia, a chronic respiratory disease. Originating infections can down-regulate the host's immune system and increase infection and thus replication of other pathogens. Pasteurella multocida is the causative agent of a wide range of infections leading to high economic impact. Commensal and opportunistic pathogen of the mouth, nasopharynx and upper respiratory tract. Early identification and proper management of these agents that cause respiratory diseases are essential to minimize the incidence of swine carcasses. Adopting preventive measures, such as vaccination and proper management practices, can help prevent the spread of these diseases and ensure the production of safe, high-quality pork for human consumption.(AU)


Las enfermedades respiratorias son un problema importante en la producción porcina y pueden provocar el decomiso de las canales en el matadero. Entre los agentes causantes de estas enfermedades se encuentran Actinobacillus pleuropneumoniae, Mycoplasma hyopneumoniae y Pasteurella multocida. Actinobacillus pleuropneumoniae es un patógeno altamente contagioso que causa hemorragia, pleuroneumonía purulenta y fibrosa. La pleuroneumonía está ampliamente distribuida y causa graves daños a la cría de cerdos. Mycoplasma hyopneumoniae causa neumonía por micoplasma, una enfermedad respiratoria crónica. Las infecciones que se originan pueden regular a la baja el sistema inmunitario del huésped y aumentar la infección y, por lo tanto, la replicación de otros patógenos. Pasteurella multocida es el agente causal de una amplia gama de infecciones que tienen un alto impacto económico. Patógeno comensal y oportunista de la boca, nasofaringe y tracto respiratorio superior. La identificación temprana y el manejo adecuado de estos agentes causantes de enfermedades respiratorias son fundamentales para minimizar la incidencia de las canales porcinas. La adopción de medidas preventivas, como la vacunación y prácticas de manejo adecuadas, puede ayudar a prevenir la propagación de estas enfermedades y garantizar la producción de carne de cerdo segura y de alta calidad para el consumo humano.(AU)


Subject(s)
Animals , Pasteurella Infections/diagnosis , Swine/physiology , Actinobacillus Infections/diagnosis , Animal Culling/methods , Pork Meat/analysis , Mycoplasma Infections/diagnosis , Respiratory Tract Diseases/veterinary , Pasteurella multocida/pathogenicity , Actinobacillus pleuropneumoniae/pathogenicity , Mycoplasma hyopneumoniae/pathogenicity
2.
Front Immunol ; 12: 688294, 2021.
Article in English | MEDLINE | ID: mdl-34394082

ABSTRACT

Polarization of macrophages to different functional states is important for mounting responses against pathogen infections. Macrophages are the major target cells of porcine circovirus type 2 (PCV2), which is the primary causative agent of porcine circovirus-associated disease (PCVAD) leading to immense economic losses in the global swine industry. Clinically, PCV2 is often found to increase risk of other pathogenic infections yet the underlying mechanisms remain to be elusive. Here we found that PCV2 infection skewed macrophages toward a M1 status through reprogramming expression of a subset of M1-associated genes and M2-associated genes. Mechanistically, induction of M1-associated genes by PCV2 infection is dependent on activation of nuclear factor kappa B (NF-κB) and c-jun N-terminal kinase (JNK) signaling pathways whereas suppression of M2-associated genes by PCV2 is via inhibiting expression of jumonji domain containing-3 (JMJD3), a histone 3 Lys27 (H3K27) demethylase that regulates M2 activation of macrophages. Finally, we identified that PCV2 capsid protein (Cap) directly inhibits JMJD3 transcription to restrain expression of interferon regulatory factor (IRF4) that controls M2 macrophage polarization. Consequently, sustained infection of PCV2 facilitates bacterial infection in vitro. In summary, these findings showed that PCV2 infection functionally modulated M1 macrophage polarization via targeting canonical signals and epigenetic histone modification, which contributes to bacterial coinfection and virial pathogenesis.


Subject(s)
Actinobacillus Infections/microbiology , Actinobacillus pleuropneumoniae/pathogenicity , Circoviridae Infections/virology , Circovirus/pathogenicity , Coinfection , Macrophages/microbiology , Macrophages/virology , Salmonella Infections/microbiology , Salmonella typhimurium/pathogenicity , Actinobacillus Infections/immunology , Actinobacillus Infections/metabolism , Actinobacillus pleuropneumoniae/immunology , Animals , Cells, Cultured , Chromatin Assembly and Disassembly , Circoviridae Infections/immunology , Circovirus/immunology , Disease Models, Animal , Epigenesis, Genetic , Host-Pathogen Interactions , Interferon Regulatory Factors/metabolism , JNK Mitogen-Activated Protein Kinases/metabolism , Jumonji Domain-Containing Histone Demethylases/metabolism , Macrophages/immunology , Macrophages/metabolism , Mice, Inbred C57BL , Mice, Knockout , Myeloid Differentiation Factor 88/genetics , Myeloid Differentiation Factor 88/metabolism , NF-kappa B/metabolism , Phenotype , Salmonella Infections/immunology , Salmonella Infections/metabolism , Salmonella typhimurium/immunology , Signal Transduction
3.
mBio ; 12(3): e0089721, 2021 06 29.
Article in English | MEDLINE | ID: mdl-34076489

ABSTRACT

Capsule polymers are crucial virulence factors of pathogenic bacteria and are used as antigens in glycoconjugate vaccine formulations. Some Gram-negative pathogens express poly(glycosylglycerol phosphate) capsule polymers that resemble Gram-positive wall teichoic acids and are synthesized by TagF-like capsule polymerases. So far, the biotechnological use of these enzymes for vaccine developmental studies was restricted by the unavailability of enantiopure CDP-glycerol, one of the donor substrates required for polymer assembly. Here, we use CTP:glycerol-phosphate cytidylyltransferases (GCTs) and TagF-like polymerases to synthesize the poly(glycosylglycerol phosphate) capsule polymer backbones of the porcine pathogen Actinobacillus pleuropneumoniae, serotypes 3 and 7 (App3 and App7). GCT activity was confirmed by high-performance liquid chromatography, and polymers were analyzed using comprehensive nuclear magnetic resonance studies. Solid-phase synthesis protocols were established to allow potential scale-up of polymer production. In addition, one-pot reactions exploiting glycerol-kinase allowed us to start the reaction from inexpensive, widely available substrates. Finally, this study highlights that multidomain TagF-like polymerases can be transformed by mutagenesis of active site residues into single-action transferases, which in turn can act in trans to build-up structurally new polymers. Overall, our protocols provide enantiopure, nature-identical capsule polymer backbones from App2, App3, App7, App9, and App11, Neisseria meningitidis serogroup H, and Bibersteinia trehalosi serotypes T3 and T15. IMPORTANCE Economic synthesis platforms for the production of animal vaccines could help reduce the overuse and misuse of antibiotics in animal husbandry, which contributes greatly to the increase of antibiotic resistance. Here, we describe a highly versatile, easy-to-use mix-and-match toolbox for the generation of glycerol-phosphate-containing capsule polymers that can serve as antigens in glycoconjugate vaccines against Actinobacillus pleuropneumoniae and Bibersteinia trehalosi, two pathogens causing considerable economic loss in the swine, sheep, and cattle industries. We have established scalable protocols for the exploitation of a versatile enzymatic cascade with modular architecture, starting with the preparative-scale production of enantiopure CDP-glycerol, a precursor for a multitude of bacterial surface structures. Thereby, our approach not only allows the synthesis of capsule polymers but might also be exploitable for the (chemo)enzymatic synthesis of other glycerol-phosphate-containing structures such as Gram-positive wall teichoic acids or lipoteichoic acids.


Subject(s)
Actinobacillus pleuropneumoniae/chemistry , Bacterial Capsules/chemistry , Glycerophosphates/biosynthesis , Neisseria meningitidis/chemistry , Pasteurellaceae/chemistry , Polymers/chemistry , Actinobacillus pleuropneumoniae/pathogenicity , Animals , Bacterial Vaccines/chemistry , Cattle , Glycerophosphates/analysis , Glycerophosphates/metabolism , Sheep , Swine
4.
Vet Microbiol ; 258: 109122, 2021 Jul.
Article in English | MEDLINE | ID: mdl-34052743

ABSTRACT

Actinobacillus pleuropneumoniae is a Gram-negative bacterium causing porcine pleuropneumonia and severe economic losses in the global swine industry. The toxic trace element copper is required for many physiological and pathological processes in organisms. However, CopA, one of the most well-characterized P-type ATPases contributing to copper resistance, has not been characterized in A. pleuropneumoniae. We used quantitative PCR analysis to examine expression of the copA gene in A. pleuropneumoniae and investigated sequence conservation among serotypes and other Gram-negative bacteria. Growth characteristics were determined using growth curve analyses and spot dilution assays of the wild-type strain and a △copA mutant. We also used flame atomic absorption spectrophotometry to determine intracellular copper content and examined the virulence of the △copA mutant in a mouse model. The copA expression was induced by copper, and its nucleotide sequence was highly conserved among different serotypes of A. pleuropneumoniae. The amino acid sequence of CopA shared high identity with CopA sequences reported from several Gram-negative bacteria. Furthermore, the △copA mutant exhibited impaired growth and had higher intracellular copper content compared with the wild-type strain when supplemented with copper. The mouse model revealed that CopA had no influence on the virulence of A. pleuropneumoniae. In conclusion, these results demonstrated that CopA is required for resistance of A. pleuropneumoniae to copper and protects A. pleuropneumoniae against copper toxicity via copper efflux.


Subject(s)
Actinobacillus Infections/microbiology , Actinobacillus pleuropneumoniae/drug effects , Bacterial Proteins/metabolism , Copper/pharmacology , Gene Expression Regulation, Bacterial/drug effects , Actinobacillus pleuropneumoniae/genetics , Actinobacillus pleuropneumoniae/metabolism , Actinobacillus pleuropneumoniae/pathogenicity , Animals , Bacterial Proteins/genetics , Computational Biology , Mice , Mice, Inbred BALB C , Up-Regulation/drug effects , Virulence
5.
Sci Rep ; 11(1): 1753, 2021 01 18.
Article in English | MEDLINE | ID: mdl-33462305

ABSTRACT

ApxI exotoxin is an important virulence factor derived from Actinobacillus pleuropneumoniae that causes pleuropneumonia in swine. Here, we investigate the role of lymphocyte function-associated antigen 1 (LFA-1, CD11a/CD18), a member of the ß2 integrin family, and the involvement of the integrin signaling molecules focal adhesion kinase (FAK) and Akt in ApxI cytotoxicity. Using Western blot analysis, we found that ApxI downregulated the activity of FAK and Akt in porcine alveolar macrophages (AMs). Preincubation of porcine AMs with an antibody specific for porcine CD18 reduced ApxI-induced cytotoxicity as measured by a lactate dehydrogenase release assay and decreased ApxI-induced FAK and Akt attenuation, as shown by Western blot analysis. Pretreatment with the chemical compounds PMA and SC79, which activate FAK and Akt, respectively, failed to overcome the ApxI-induced attenuation of FAK and Akt and death of porcine AMs. Notably, the transfection experiments revealed that ectopic expression of porcine LFA-1 (pLFA-1) conferred susceptibility to ApxI in ApxI-insensitive cell lines, including human embryonic kidney 293T cells and FAK-deficient mouse embryonic fibroblasts (MEFs). Furthermore, ectopic expression of FAK significantly reduced ApxI cytotoxicity in pLFA-1-cotransfected FAK-deficient MEFs. These findings show for the first time that pLFA-1 renders cells susceptible to ApxI and ApxI-mediated attenuation of FAK activity via CD18, thereby contributing to subsequent cell death.


Subject(s)
Actinobacillus Infections/pathology , Actinobacillus pleuropneumoniae/metabolism , Bacterial Proteins/metabolism , Focal Adhesion Kinase 1/antagonists & inhibitors , Hemolysin Proteins/metabolism , Lymphocyte Function-Associated Antigen-1/metabolism , Swine Diseases/pathology , Actinobacillus Infections/metabolism , Actinobacillus Infections/microbiology , Actinobacillus pleuropneumoniae/isolation & purification , Actinobacillus pleuropneumoniae/pathogenicity , Animals , Cell Death/physiology , Cells, Cultured , Focal Adhesion Kinase 1/metabolism , Humans , Macrophages, Alveolar/metabolism , Macrophages, Alveolar/pathology , Mice , Proto-Oncogene Proteins c-akt/metabolism , Swine , Swine Diseases/metabolism , Swine Diseases/microbiology
6.
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
7.
Pathog Dis ; 78(9)2020 11 23.
Article in English | MEDLINE | ID: mdl-33095236

ABSTRACT

The RNA chaperone Hfq regulates diverse processes in numerous bacteria. In this study, we compared phenotypes (growth rate, adherence, response to different stress conditions and virulence in Galleria mellonella) of wild-type (WT) and isogenic hfq mutants of three serovars (1, 8 and 15) of the porcine pathogen Actinobacillus pleuropneumoniae. Similar growth in rich broth was seen for all strains except Ap1∆hfq, which showed slightly reduced growth throughout the 24 h time course, and the complemented Ap8∆hfqC mutant had a prolonged lag phase. Differences were seen between the three serovar WT strains regarding adherence, stress response and virulence in G. mellonella, and deletion of hfq affected some, but not all of these phenotypes, depending on serovar. Complementation by expression of cloned hfq from an endogenous promoter only restored some WT phenotypes, indicating that complex regulatory networks may be involved, and that levels of Hfq may be as important as presence/absence of the protein regarding its contribution to gene regulation. Our results support that Hfq is a pleiotropic global regulator in A. pleuropneumoniae, but serovar-related differences exist. These results highlight the importance of testing multiple strains/serovars within a given species when determining contributions of global regulators, such as Hfq, to expression of complex phenotypes.


Subject(s)
Actinobacillus pleuropneumoniae/pathogenicity , Bacterial Adhesion , Host Factor 1 Protein/metabolism , Stress, Physiological , Virulence , Actinobacillus Infections/microbiology , Actinobacillus pleuropneumoniae/classification , Animals , Disease Models, Animal , Gene Deletion , Gene Expression Regulation, Bacterial , Genetic Complementation Test , Host Factor 1 Protein/genetics , Larva/microbiology , Moths/microbiology , Phenotype , Promoter Regions, Genetic , Serogroup , Swine
8.
Vet Microbiol ; 245: 108704, 2020 Jun.
Article in English | MEDLINE | ID: mdl-32456813

ABSTRACT

Actinobacillus pleuropneumoniae is a Gram-negative pathogen that causes porcine pleuropneumonia, an infectious disease responsible for significant losses in the pig industry. Sulfur is an essential nutrient that is widely required by microorganisms; however, the mechanism involved in A. pleuropneumoniae sulfur transport is unknown. In this study, we showed that a periplasmic protein predicted to be involved in sulfur acquisition (sulfate-binding protein (Sbp)), is required for A. pleuropneumoniae growth in chemically defined medium (CDM) containing sulfate or methionine as the sole sulfur sources. However, utilization of glutathione and cysteine was not affected in the sbp-deletion mutant. The virulence of A. pleuropneumoniae in mice was not affected by the absence of Sbp. Moreover, we demonstrated that Sbp was not essential for the in vivo colonization of A. pleuropneumoniae in mice or pigs. Collectively, these findings reveal that A. pleuropneumoniae Sbp plays an important role in the acquisition of the sulfur nutrients, sulfate and methionine. The presence of other sulfur uptake systems suggests A. pleuropneumoniae has multiple functionally redundant pathways ensuring uptake of important nutrients during infection.


Subject(s)
Actinobacillus pleuropneumoniae/metabolism , Methionine/metabolism , Periplasmic Binding Proteins/metabolism , Sulfates/metabolism , Actinobacillus pleuropneumoniae/pathogenicity , Animals , Female , Mice , Mice, Inbred BALB C , Periplasmic Binding Proteins/genetics , Sequence Deletion , Swine , Virulence
9.
Vet Microbiol ; 243: 108634, 2020 Apr.
Article in English | MEDLINE | ID: mdl-32273013

ABSTRACT

Actinobacillus pleuropneumoniae is the causative agent of highly contagious and fatal respiratory infections, causing substantial economic losses to the global pig industry. Due to increased antibiotic resistance, there is an urgent need to find new antibiotic alternatives for treating A. pleuropneumoniae infections. MPX is obtained from wasp venom and has a killing effect on various bacteria. This study found that MPX had a good killing effect on A. pleuropneumoniae and that the minimum inhibitory concentration (MIC) was 16 µg/mL. The bacterial density of A. pleuropneumoniae decreased 1000 times after MPX (1 × MIC) treatment for 1 h, and the antibacterial activity was not affected by pH or temperature. Fluorescence microscopy showed that MPX (1 × MIC) destroyed the bacterial cell membrane after treatment for 0.5 h, increasing membrane permeability and releasing bacterial proteins and Ca2+, Na+ and other cations. In addition, MPX (1 × MIC) treatment significantly reduced the formation of bacterial biofilms. Quantitative RT-PCR results showed that MPX treatment significantly upregulated the expression of the PurC virulence gene and downregulated that of ApxI, ApxII, and Apa1. In addition, the Sap A gene was found to play an important role in the tolerance of A. pleuropneumoniae to antimicrobial peptides. Therapeutic evaluation in a murine model showed that MPX protects mice from a lethal dose of A. pleuropneumoniae and relieves lung inflammation. This study reports the use of MPX to treat A. pleuropneumonia infections, laying the foundation for the development of new drugs for bacterial infections.


Subject(s)
Actinobacillus Infections/drug therapy , Actinobacillus pleuropneumoniae/drug effects , Actinobacillus pleuropneumoniae/pathogenicity , Antimicrobial Cationic Peptides/pharmacology , Animals , Bacterial Proteins/genetics , Biofilms/drug effects , Biofilms/growth & development , Cell Membrane/drug effects , Female , Lung/drug effects , Lung/microbiology , Mice , Microbial Sensitivity Tests , Peptide Synthases/genetics , Swine , Swine Diseases/microbiology , Virulence/drug effects
10.
mBio ; 11(2)2020 03 17.
Article in English | MEDLINE | ID: mdl-32184239

ABSTRACT

The posttranslational Ca2+-dependent "clip-and-link" activity of large repeat-in-toxin (RTX) proteins starts by Ca2+-dependent structural rearrangement of a highly conserved self-processing module (SPM). Subsequently, an internal aspartate-proline (Asp-Pro) peptide bond at the N-terminal end of SPM breaks, and the liberated C-terminal aspartyl residue can react with a free ε-amino group of an adjacent lysine residue to form a new isopeptide bond. Here, we report a solution structure of the calcium-loaded SPM (Ca-SPM) derived from the FrpC protein of Neisseria meningitidis The Ca-SPM structure defines a unique protein architecture and provides structural insight into the autocatalytic cleavage of the Asp-Pro peptide bond through a "twisted-amide" activation. Furthermore, in-frame deletion of the SPM domain from the ApxIVA protein of Actinobacillus pleuropneumoniae attenuated the virulence of this porcine pathogen in a pig respiratory challenge model. We hypothesize that the Ca2+-dependent clip-and-link activity represents an unconventional strategy for Gram-negative pathogens to adhere to the host target cell surface.IMPORTANCE The Ca2+-dependent clip-and-link activity of large repeat-in-toxin (RTX) proteins is an exceptional posttranslational process in which an internal domain called a self-processing module (SPM) mediates Ca2+-dependent processing of a highly specific aspartate-proline (Asp-Pro) peptide bond and covalent linkage of the released aspartyl to an adjacent lysine residue through an isopeptide bond. Here, we report the solution structures of the Ca2+-loaded SPM (Ca-SPM) defining the mechanism of the autocatalytic cleavage of the Asp414-Pro415 peptide bond of the Neisseria meningitidis FrpC exoprotein. Moreover, deletion of the SPM domain in the ApxIVA protein, the FrpC homolog of Actinobacillus pleuropneumoniae, resulted in attenuation of virulence of the bacterium in a pig infection model, indicating that the Ca2+-dependent clip-and-link activity plays a role in the virulence of Gram-negative pathogens.


Subject(s)
Bacterial Proteins/chemistry , Bacterial Toxins/chemistry , Calcium/metabolism , Membrane Proteins/chemistry , Protein Processing, Post-Translational , Actinobacillus Infections/veterinary , Actinobacillus pleuropneumoniae/chemistry , Actinobacillus pleuropneumoniae/pathogenicity , Animals , Bacterial Proteins/genetics , Neisseria meningitidis/chemistry , Swine , Virulence
11.
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
12.
Cell Death Dis ; 10(9): 657, 2019 09 10.
Article in English | MEDLINE | ID: mdl-31506432

ABSTRACT

Actinobacillus pleuropneumoniae (A.pp) causes severe pneumonia associated with enormous economic loss in pigs. Peracute diseased pigs die in <24 h with pneumonia. Neutrophils are the prominent innate immune cell in this infection that massively infiltrate the infected lung. Here we show that neutrophils release neutrophil extracellular traps (NETs) as response to A.pp infection. Numerous NET-markers were identified in bronchoalveolar lavage fluid (BALF) of A.pp-infected piglets in vivo, however, most NET fibers are degraded. Importantly, A.pp is able to enhance its growth rate in the presence of NETs that have been degraded by nucleases efficiently. A.pp itself releases no nuclease, but we identified host nucleases as sources that degrade NETs after A.pp infection. Furthermore, the nucleases of co-infecting pathogens like Streptococcus suis increase growth of A.pp in presence of porcine NETs. Thus, A.pp is not only evading the antimicrobial activity of NETs, A.pp is rather additionally using parts of NETs as growth factor thereby taking advantage of host nucleases as DNase1 or nucleases of co-infecting bacteria, which degrade NETs. This effect can be diminished by inhibiting the bacterial adenosine synthase indicating that degraded NETs serve as a source for NAD, which is required by A.pp for its growth. A similar phenotype was found for the human pathogen Haemophilus (H.) influenzae and its growth in the presence of human neutrophils. H. influenzae benefits from host nucleases in the presence of neutrophils. These data shed light on the detrimental effects of NETs during host immune response against certain bacterial species that require and/or efficiently take advantage of degraded DNA material, which has been provided by host nuclease or nucleases of other co-infecting bacteria, as growth source.


Subject(s)
Actinobacillus Infections/metabolism , Actinobacillus pleuropneumoniae/growth & development , Extracellular Traps/metabolism , Lung/metabolism , Pneumonia, Bacterial/metabolism , Swine Diseases/metabolism , Actinobacillus Infections/pathology , Actinobacillus Infections/veterinary , Actinobacillus pleuropneumoniae/pathogenicity , Animals , Lung/microbiology , Lung/pathology , Pneumonia, Bacterial/microbiology , Pneumonia, Bacterial/pathology , Pneumonia, Bacterial/veterinary , Swine , Swine Diseases/microbiology , Swine Diseases/pathology
13.
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
14.
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
15.
J Vet Med Sci ; 81(3): 480-485, 2019 Mar 30.
Article in English | MEDLINE | ID: mdl-30713216

ABSTRACT

Six atypical Actinobacillus pleuropneumoniae serovar 15 strains were isolated from pneumonic lesions of naturally infected dead pigs from the same farm in Japan. Genetic analyses of apx genes revealed that the atypical isolates contained the toxin-associated genes apxIBD, apxIIICA, apxIIIBD, and apxIVA, but not apxIICA. Coinciding with the result of the atypical gene profile, analyses of toxin protein production revealed that these atypical isolates expressed only ApxIII but not ApxII. A mouse pathogenicity test showed that the atypical isolate tested seemed to be less virulent than the typical isolates. This is the first report describing the emergence of atypical A. pleuropneumoniae serovar 15, which does not produce ApxII due to the absence of apxIICA genes, in Japan.


Subject(s)
Actinobacillus Infections/veterinary , Actinobacillus pleuropneumoniae/isolation & purification , Bacterial Proteins/genetics , Genes, Bacterial , Hemolysin Proteins/genetics , Swine Diseases/microbiology , Actinobacillus Infections/microbiology , Actinobacillus pleuropneumoniae/genetics , Actinobacillus pleuropneumoniae/pathogenicity , Animals , Female , Gene Deletion , Japan , Mice , Swine , Transcriptome , Virulence/genetics
16.
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
17.
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
18.
J Microbiol Biotechnol ; 28(12): 2095-2105, 2018 Dec 28.
Article in English | MEDLINE | ID: mdl-30380823

ABSTRACT

In our previous studies, we have identified several in vivo-induced antigens and evaluated their potential as subunit vaccine candidates in a murine model, in which the recombinant protein GalT showed the most potent immunogenicity and immunoprotective efficacy against Actinobacillus pleuropneumoniae. To exploit a more efficient way of delivering GalT proteins, in this study, we employed the widely studied E. coli outer membrane vesicles (OMVs) as a platform to deliver GalT protein and performed the vaccine trial using the recombinant GalT-OMVs in the murine model. Results revealed that GalT-OMVs could elicit a highly-specific, IgG antibody titer that was comparable with the adjuvant GalT group. Significantly higher lymphocyte proliferation and cytokines secretion levels were observed in the GalT-OMVs group. 87.5% and 50% of mice were protected from a lethal dose challenge using A. pleuropneumoniae in active or passive immunization, respectively. Histopathologic and immunohistochemical analyses showed remarkably reduced pathological changes and infiltration of neutrophils in the lungs of mice immunized with GalT-OMVs after the challenge. Taken together, these findings confirm that OMVs can be used as a platform to deliver GalT protein and enhance its immunogenicity to induce both humoral and cellular immune responses in mice.


Subject(s)
Actinobacillus Infections/immunology , Actinobacillus Infections/prevention & control , Actinobacillus pleuropneumoniae/drug effects , Bacterial Vaccines/immunology , Escherichia coli/metabolism , Immunization , Protein Transport/immunology , UTP-Hexose-1-Phosphate Uridylyltransferase/immunology , Actinobacillus Infections/pathology , Actinobacillus pleuropneumoniae/immunology , Actinobacillus pleuropneumoniae/pathogenicity , Adjuvants, Immunologic , Animals , Antibodies, Bacterial , Bacterial Outer Membrane Proteins/immunology , Bacterial Vaccines/genetics , Cell Proliferation , Cytokines/metabolism , Disease Models, Animal , Escherichia coli/genetics , Female , Immunity, Cellular , Immunity, Humoral , Immunoglobulin G , Lethal Dose 50 , Lung/pathology , Lymphocytes , Mice , Mice, Inbred BALB C , Neutrophils/pathology , Protein Engineering , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/immunology , UTP-Hexose-1-Phosphate Uridylyltransferase/genetics , UTP-Hexose-1-Phosphate Uridylyltransferase/metabolism , Vaccination
19.
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
20.
Genes Genomics ; 40(8): 847-856, 2018 08.
Article in English | MEDLINE | ID: mdl-30047117

ABSTRACT

Porcine pleuropneumonia caused by Actinobacillus pleuropneumoniae has led to severe economic losses in the pig industry worldwide. A. pleuropneumoniae displays various levels of antimicrobial resistance, leading to the dire need to identify new drug targets. Protein-protein interaction (PPI) network can aid the identification of drug targets by discovering essential proteins during the life of bacteria. The aim of this study is to identify drug target candidates of A. pleuropneumoniae from essential proteins in PPI network. The homologous protein mapping method (HPM) was utilized to construct A. pleuropneumoniae PPI network. Afterwards, the subnetwork centered with H-NS was selected to verify the PPI network using bacterial two-hybrid assays. Drug target candidates were identified from the hub proteins by analyzing the topology of the network using interaction degree and homologous comparison with the pig proteome. An A. pleuropneumoniae PPI network containing 2737 non-redundant interaction pairs among 533 proteins was constructed. These proteins were distributed in 21 COG functional categories and 28 KEGG metabolic pathways. The A. pleuropneumoniae PPI network was scale free and the similar topological tendencies were found when compared with other bacteria PPI network. Furthermore, 56.3% of the H-NS subnetwork interactions were validated. 57 highly connected proteins (hub proteins) were identified from the A. pleuropneumoniae PPI network. Finally, 9 potential drug targets were identified from the hub proteins, with no homologs in swine. This study provides drug target candidates, which are promising for further investigations to explore lead compounds against A. pleuropneumoniae.


Subject(s)
Actinobacillus Infections/genetics , Actinobacillus pleuropneumoniae/genetics , Pleuropneumonia/genetics , Protein Interaction Maps/genetics , Actinobacillus Infections/microbiology , Actinobacillus Infections/veterinary , Actinobacillus pleuropneumoniae/pathogenicity , Animals , Drug Delivery Systems/methods , Metabolic Networks and Pathways/genetics , Molecular Targeted Therapy , Pleuropneumonia/microbiology , Pleuropneumonia/veterinary , Swine/genetics , Swine/microbiology , Swine Diseases/genetics , Swine Diseases/microbiology
SELECTION OF CITATIONS
SEARCH DETAIL
...