Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 91
Filter
1.
Res Vet Sci ; 152: 1-9, 2022 Dec 20.
Article in English | MEDLINE | ID: mdl-35901636

ABSTRACT

Distinct monocyte subpopulations have been previously described in healthy pigs and pigs experimentally infected with Actinobacillus pleuropneumoniae (APP). The CD163+ subpopulation of bone marrow (BM), peripheral blood (PB) and lung monocytes was found to play an important role in the inflammatory process. The inflammation is accompanied by elevation of inflammatory cytokines. The aim of the study was to evaluate the contribution of CD163+ monocytes and macrophages to cytokine production during APP-induced lung inflammation. Cytokine production was assessed by flow cytometry (FC) and quantitative PCR (qPCR) in CD163+ monocytes and by qPCR, immunohistochemistry/fluorescence in lungs and tracheobronchial lymph nodes (TBLN). Despite the systemic inflammatory response after APP infection, BM and PB CD163+ monocytes did not express elevated levels of a wide range of cytokines compared to control pigs. In contrast, significant amounts of IL-1ß, IL-6, IL-8 and TNF-α were produced in lung lesions and IL-1ß in the TBLN. At the protein level, TNF-α was expressed by both CD163+ monocytes and macrophages in lung lesions, whereas IL-1ß, IL-6 and IL-8 expression was found only in CD163+ monocytes; no CD163+ macrophages were found to produce these cytokines. Furthermore, the quantification of CD163+ monocytes expressing the two cytokines IL-1ß and IL-8 that were most elevated was performed. In lung lesions, 36.5% IL-1ß positive CD163+ monocytes but only 18.3% IL-8 positive CD163+ monocytes were found. In conclusion, PB and BM CD163+ monocytes do not appear to contribute to the elevated cytokine levels in plasma. On the other hand, CD163+ monocytes contribute to inflammatory cytokine expression, especially IL-1ß at the site of inflammation during the inflammatory process.


Subject(s)
Actinobacillus Infections , Actinobacillus pleuropneumoniae , Swine , Animals , Actinobacillus pleuropneumoniae/physiology , Monocytes/metabolism , Cytokines , Tumor Necrosis Factor-alpha/metabolism , Interleukin-8/metabolism , Interleukin-6/metabolism , Actinobacillus Infections/veterinary , Inflammation/metabolism , Inflammation/veterinary
2.
Acta Vet Scand ; 62(1): 55, 2020 Sep 17.
Article in English | MEDLINE | ID: mdl-32943077

ABSTRACT

BACKGROUND: Penicillin is important for treatment of pigs, but data on its absorption and disposition in pigs are sparse. This is reflected by the variation in recommended dosages in the literature. Inadequate dosage may lead to treatment failure and selection of resistant bacteria. To optimize treatment regimens, plasma exposure to benzylpenicillin for two sustained release formulations of procaine benzylpenicillin for intramuscular administration was studied in growing pigs by means of tandem mass spectrometry (UPLC-MS/MS). One formulation was an aqueous suspension, Ethacilin® vet (ETH), and the other an oily suspension, Ultrapen vet (UPA). Benzylpenicillin exposure after intravenous administration of potassium benzylpenicillin was also explored. Exposure profiles were first studied after single administrations of the approved dosages in healthy pigs and then after repeated administration of different dosages in pigs inoculated intranasally with an Actinobacillus pleuropneumoniae serotype 2 strain. RESULTS: After intravenous administration of benzylpenicillin (n = 6), maximum plasma concentration (Cmax), 1860-9318 µg/L, was observed after 15 min. At four h, plasma concentrations decreased to 15-76 µg/L. After intramuscular administration of ETH (n = 6) Cmax, 1000-4270 µg/L, was observed within one h (tmax) in 5 pigs but at four h in one pig. Cmax for UPA (n = 6), 910-3220 µg/L, was observed within one h in three pigs, but at four or 24 h in three pigs. For both ETH and UPA, the terminal phase was characterized by slow decline compared with intravenous administration. Repeated administration of different dosages of ETH and UPA in pigs inoculated with A. pleuropneumoniae (n = 54) showed that the approved dose for UPA (30 mg/kg, qd) but not for ETH (20 mg/kg, qd) gave adequate plasma exposure for bacteria with a penicillin MIC of 500 µg/L. However, more frequent dosing of ETH (bid) or increased dosage gave an adequate exposure. CONCLUSIONS: The approved dosage of ETH provided insufficient plasma exposure for adequate therapy of infections caused by A. pleuropneumoniae or other bacteria with a penicillin MIC of 500 µg/L. More frequent ETH dosing (bid) or an increased dosage would improve exposure. The approved dosage of UPA however provided adequate exposure.


Subject(s)
Anti-Bacterial Agents/pharmacokinetics , Penicillin G/pharmacokinetics , Sus scrofa/metabolism , Actinobacillus Infections/drug therapy , Actinobacillus Infections/veterinary , Actinobacillus pleuropneumoniae/drug effects , Actinobacillus pleuropneumoniae/physiology , Animals , Dose-Response Relationship, Drug , Female , Injections, Intramuscular/veterinary , Male
3.
J Vet Diagn Invest ; 32(4): 581-584, 2020 Jul.
Article in English | MEDLINE | ID: mdl-32517629

ABSTRACT

Two Actinobacillus pleuropneumoniae isolates from clinical cases of porcine pleuropneumonia in Japan were positive in the capsular serovar 15-specific PCR assay, but nontypeable (NT) in the agar gel precipitation (AGP) test. Nucleotide sequence analysis of gene clusters involved in the biosynthesis of capsular polysaccharide (CPS) and lipopolysaccharide O-polysaccharide (O-PS) revealed that both clusters contained transposable element ISApl1 of A. pleuropneumoniae belonging to the IS30 family. Immunoblot analysis revealed that these 2 isolates could not produce O-PS. We conclude that the ISApl1 of A. pleuropneumoniae can interfere in the biosynthesis of both CPS and O-PS.


Subject(s)
Actinobacillus Infections/veterinary , Actinobacillus pleuropneumoniae/physiology , DNA Transposable Elements , Pleuropneumonia/veterinary , Polysaccharides/analysis , Swine Diseases/diagnosis , Actinobacillus Infections/diagnosis , Actinobacillus Infections/microbiology , Actinobacillus pleuropneumoniae/isolation & purification , Animals , Genes, Bacterial , Immunoblotting/veterinary , Multigene Family , Pleuropneumonia/diagnosis , Pleuropneumonia/microbiology , Polymerase Chain Reaction/veterinary , Swine , Swine Diseases/microbiology
4.
J Vet Sci ; 21(2): e20, 2020 Mar.
Article in English | MEDLINE | ID: mdl-32233129

ABSTRACT

Actinobacillus pleuropneumoniae (APP) causes a form of porcine pleuropneumonia that leads to significant economic losses in the swine industry worldwide. The apxIBD gene is responsible for the secretion of the ApxI and ApxII toxins and the pnp gene is responsible for the adaptation of bacteria to cold temperature and a virulence factor. The apxIBD and pnp genes were deleted successfully from APP serotype 1 and 5 by transconjugation and sucrose counter-selection. The APP1ΔapxIBDΔpnp and APP5ΔapxIBDΔpnp mutants lost hemolytic activity and could not secrete ApxI and ApxII toxins outside the bacteria because both mutants lost the ApxI- and ApxII-secreting proteins by deletion of the apxIBD gene. Besides, the growth of these mutants was defective at low temperatures resulting from the deletion of pnp. The APP1ΔapxIBDΔpnp and APP5ΔapxIBDΔpnp mutants were significantly attenuated compared with wild-type ones. However, mice vaccinated intraperitoneally with APP5ΔapxIBDΔpnp did not provide any protection when challenged with a 10-times 50% lethal dose of virulent homologous (APP5) and heterologous (APP1) bacterial strains, while mice vaccinated with APP1ΔapxIBDΔpnp offered 75% protection against a homologous challenge. The ΔapxIBDΔpnp mutants were significantly attenuated and gave different protection rate against homologous virulent wild-type APP challenging.


Subject(s)
Actinobacillus pleuropneumoniae/physiology , Gene Deletion , Genes, Bacterial , Actinobacillus Infections/microbiology , Animals , Female , Mice , Mice, Inbred BALB C , Serogroup , Vaccination
5.
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
6.
Vet Microbiol ; 233: 93-101, 2019 Jun.
Article in English | MEDLINE | ID: mdl-31176418

ABSTRACT

Actinobacillus pleuropneumoniae (APP) and porcine circovirus type 2 (PCV2) are both important pathogens of the porcine respiratory disease complex (PRDC), which results in significant worldwide economic losses. Recently, PCV2 and APP coinfection has been described in the worldwide pork industry, and represents an extremely complex situation in veterinary medicine. However, the mechanism of their coinfection has not been investigated. In this study, we found that PCV2 promoted APP adhesion to and invasion of porcine alveolar macrophages (PAMs) during coinfection. Additionally, PCV2 suppressed reactive oxygen species (ROS) production by inhibiting cytomembrane NADPH oxidase activity, which was beneficial for APP survival in PAMs in vitro. During coinfection, PCV2 weakened the inflammatory response and macrophage antigen presentation by decreasing TNF-α, IFN-γ and IL-4 expression, and reduced clearance of the invading bacteria. The host-cell experimental results were verified in a mouse model. The findings provide a deeper and novel understanding of porcine coinfection, and will be extremely helpful for the design of strategies for PRDC control.


Subject(s)
Actinobacillus pleuropneumoniae/physiology , Circovirus/physiology , Coinfection/veterinary , Macrophages, Alveolar/microbiology , Macrophages, Alveolar/virology , Reactive Oxygen Species/metabolism , Actinobacillus Infections/immunology , Actinobacillus Infections/veterinary , Animals , Antibodies, Viral/immunology , Antigen Presentation , Bacterial Adhesion , Circoviridae Infections/immunology , Circoviridae Infections/veterinary , Cytokines/genetics , Cytokines/immunology , Female , Inflammation , Male , Mice , Mice, Inbred ICR , Microbial Viability , NADPH Oxidases/metabolism , Swine
7.
Pathog Dis ; 77(1)2019 02 01.
Article in English | MEDLINE | ID: mdl-30395241

ABSTRACT

Actinobacillus pleuropneumoniae is a respiratory pathogen that causes porcine pleuropneumonia, a fatal respiratory disease responsible for high economic losses in the swine industry worldwide. With the objective to better understand the interactions between A. pleuropneumoniae and the porcine respiratory epithelium, we investigated the capacity of this pathogen to damage the epithelial barrier and induce an inflammatory response. We showed that A. pleuropneumoniae, even at a multiplicity of infection of 10, is able to break the tracheal epithelial barrier integrity as determined by monitoring the transepithelial electrical resistance and fluorescein-isothiocyanate-dextran transport. Immunofluorescence staining analysis suggested that A. pleuropneumoniae is affecting two important tight junction proteins (occludin, zonula occludens-1). As a consequence of the breakdown of the epithelial barrier integrity, A. pleuropneumoniae can translocate across a cell monolayer. We also showed that tracheal epithelial cells secrete pro-inflammatory cytokines (IL-8, IL-6, TNF-α) in response to a stimulation with this pathogen. In summary, A. pleuropneumoniae is able to induce damage to the porcine respiratory epithelial barrier. Challenging the epithelial cells with A. pleuropneumoniae was also associated with the secretion of pro-inflammatory cytokines. This better knowledge of the interactions between A. pleuropneumoniae and the epithelial cells may help to design novel strategies to prevent epithelium invasion by this bacterium along with other swine respiratory pathogens.


Subject(s)
Actinobacillus Infections/veterinary , Actinobacillus pleuropneumoniae/physiology , Respiratory Mucosa/metabolism , Respiratory Mucosa/microbiology , Swine Diseases/metabolism , Swine Diseases/microbiology , Animals , Biomarkers , Cell Survival , Cytokines/metabolism , Epithelial Cells/metabolism , Inflammation Mediators , Respiratory Mucosa/pathology , Swine , Swine Diseases/pathology , Tight Junctions/metabolism
8.
Vet Res ; 49(1): 97, 2018 Sep 25.
Article in English | MEDLINE | ID: mdl-30253796

ABSTRACT

Actinobacillus (A.) pleuropneumoniae is normally considered strictly adapted to the respiratory tract of swine. Despite this, scattered case reports of arthritis, osteomyelitis, hepatitis, meningitis or nephritis exist, in which A. pleuropneumoniae remained the only detectable pathogen. Therefore, the aim of this study was to investigate whether spreading to other organs than the lungs is incidental or may occur more frequently. For this, organ samples (blood, liver, spleen, kidney, tarsal and carpal joints, meninges, pleural and pericardial fluids) from weaners (n = 47) infected experimentally with A. pleuropneumoniae serovar 7 by aerosol infection (infection dose: 10.9 × 103 cfu/animal) were examined by culture during the first week after infection. In addition, tissue samples of eight weaners were examined by histology and immunohistochemistry (IHC). A. pleuropneumoniae was isolated in all examined sample sites (86.7% pleural fluids, 73.3% pericardial fluids, 50.0% blood, 61.7% liver, 51.1% spleen, 55.3% kidney, 14.9% tarsal joints, 12.8% carpal joints, 27.7% meninges). These results were also obtained from animals with only mild clinical symptoms. IHC detection confirmed these findings in all locations except carpal joints. Histological examination revealed purulent hepatitis (n = 2), nephritis (n = 1) and beginning meningitis (n = 2). Isolation results were significantly correlated (p < 0.001) with the degree of lung colonization and, to a lower extent, with the severity of disease. Detection of A. pleuropneumoniae in peripheral tissues was significantly correlated to spleen colonization. In conclusion, multi-organ spreading of A. pleuropneumoniae serovar 7 strain AP 76 seems to occur more frequently during acute infection following effective lung colonization than previously thought.


Subject(s)
Actinobacillus Infections/veterinary , Actinobacillus pleuropneumoniae/isolation & purification , Swine Diseases/physiopathology , Actinobacillus Infections/physiopathology , Actinobacillus Infections/virology , Actinobacillus pleuropneumoniae/genetics , Actinobacillus pleuropneumoniae/physiology , Animals , Serogroup , Swine , Swine Diseases/virology , Weaning
9.
Antonie Van Leeuwenhoek ; 111(12): 2371-2384, 2018 Dec.
Article in English | MEDLINE | ID: mdl-30008077

ABSTRACT

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


Subject(s)
Actinobacillus Infections/veterinary , Actinobacillus pleuropneumoniae/physiology , Leukocytes, Mononuclear/immunology , Pleuropneumonia/veterinary , Swine Diseases/genetics , Actinobacillus Infections/genetics , Actinobacillus Infections/immunology , Actinobacillus Infections/microbiology , Animals , Female , Gene Expression Profiling , Leukocytes, Mononuclear/microbiology , Male , Pleuropneumonia/genetics , Pleuropneumonia/immunology , Pleuropneumonia/microbiology , Swine , Swine Diseases/immunology , Swine Diseases/microbiology
10.
Res Vet Sci ; 118: 498-501, 2018 Jun.
Article in English | MEDLINE | ID: mdl-29758533

ABSTRACT

Porcine pleuropneumonia is an important cause of lowered productivity and economic loss in the pig industry worldwide, associated primarily with Actinobacillus pleuropneumoniae infection. Its colonization and persistence within the upper respiratory tract of affected pigs depends upon interactions between a number of genetically controlled virulence factors, such as pore-forming repeats-in-toxin exoproteins, biofilm formation, and antimicrobial resistance. This study investigated correlations between biofilm-forming capacity, antimicrobial resistance, and virulence of A. pleuropneumoniae obtained from clinical outbreaks of disease, using a Galleria mellonella alternative infection model. Results suggest that virulence is diverse amongst the 21 strains of A. pleuropneumoniae examined and biofilm formation correlated with genetic control of antimicrobial resistance.


Subject(s)
Actinobacillus Infections/microbiology , Actinobacillus pleuropneumoniae/drug effects , Actinobacillus pleuropneumoniae/physiology , Anti-Bacterial Agents/pharmacology , Biofilms/growth & development , Drug Resistance, Bacterial , Swine Diseases/microbiology , Actinobacillus Infections/drug therapy , Animals , Swine , Swine Diseases/drug therapy , Virulence
11.
PLoS One ; 13(1): e0191286, 2018.
Article in English | MEDLINE | ID: mdl-29373591

ABSTRACT

Actinobacillus pleuropneumoniae (APP) causes serious economic losses in the swine industry, and is the etiologic agent of porcine pleuropneumonia. In this study we have engineered a trivalent Apx fusion protein enclosed in outer membrane vesicles (Apxr-OMV) and studied its immunoprotective efficacy against APP serotypes 1 and 7 challenge in mice. The results showed that the IgG levels in the Apxr-OMVs immune group were significantly higher than those of the negative control (P < 0.05). Up-regulation of both Th1 (IFN-γ, IL-2) and Th2 (IL-4) cytokines were detected in splenocytes of Apxr-OMVs immune group. The survival rates 87.5% and 62.5% were observed against APP strain 1516 of serotype 7 and APP strain 2701 of serotype 1 in the groups of Apxr-OMVs immune group, respectively. Histopathological lesions of the pulmonary structure alveoli were found to be minimal in APX-OMV group challenged with APP serotypes 1 and 7. These results strongly indicated that engineered OMVs could effectively induce specific humoral or cellular immune responses. Moreover, Apxr-OMVs used as novel vaccine provides cross-protective immunity against different serotype 1 and 7 of APP infection in a mouse model. In contrast, the OMV-empty and PBS as negative controls or inactivated strain of APP-2701 and APP-1516 as positive controls for the animal study cannot provide protection or cross-protection.


Subject(s)
Actinobacillus pleuropneumoniae/physiology , Bacterial Vaccines/immunology , Cell Membrane/metabolism , Escherichia coli/cytology , Escherichia coli/genetics , Protein Engineering , Recombinant Fusion Proteins/immunology , Actinobacillus pleuropneumoniae/immunology , Animals , Bacterial Vaccines/genetics , Cell Proliferation , Cytokines/metabolism , Female , Immunity, Cellular , Lymphocytes/cytology , Mice , Mice, Inbred BALB C , Recombinant Fusion Proteins/genetics
12.
J Med Microbiol ; 67(2): 249-264, 2018 Feb.
Article in English | MEDLINE | ID: mdl-29293082

ABSTRACT

PURPOSE: Recently, the strong antiviral activity of an Actinobacillus pleuropneumoniae (App) culture supernatant against porcine reproductive and respiratory syndrome virus (PRRSV) was discovered. Following this finding, the objective of the present study was to understand how the App culture supernatant inhibits PRRSV replication in its natural targeted host cells, i.e. porcine alveolar macrophages (PAMs). METHODOLOGY: Several assays were conducted with App culture supernatant-treated PRRSV-infected cell lines, such as PAM, St-Jude porcine lung and MARC-145 cells. RT-qPCR assays were used to determine the expression levels of type I and II IFN mRNAs, viral genomic (gRNA) and sub-genomic RNAs (sgRNAs). Proteomic, Western blot and immunofluorescence assays were conducted to determine the involvement of actin filaments in the App culture supernatant antiviral effect.Results/Key findings. Type I and II IFN mRNA expressions were not upregulated by the App culture supernatant. Time courses of gRNA and sgRNA expression levels demonstrated that the App culture supernatant inhibits PRRSV infection before the first viral transcription cycle. Western blot experiments confirmed an increase in the expression of cofilin (actin cytoskeleton dynamics regulator) and immunofluorescence also demonstrated a significant decrease of actin filaments in App culture supernatant-treated PRRSV-infected PAM cells. App culture supernatant antiviral activity was also demonstrated against other PRRSV strains of genotypes I and II. CONCLUSION: App culture supernatant antiviral effect against PRRSV takes place early during PRRSV infection. Results suggest that App culture supernatant antiviral effect may take place via the activation of cofilin, which induces actin depolymerization and subsequently, probably affects PRRSV endocytosis. Other experiments are needed to fully validate this latest hypothesis.


Subject(s)
Actinobacillus pleuropneumoniae/physiology , Actins/metabolism , Antiviral Agents/pharmacology , Porcine respiratory and reproductive syndrome virus/physiology , Virus Replication/drug effects , Animals , Antibiosis , Cell Line , Culture Media/chemistry , Genome, Viral , Interferon Type I/genetics , Macrophages, Alveolar/drug effects , Macrophages, Alveolar/virology , Porcine respiratory and reproductive syndrome virus/genetics , Real-Time Polymerase Chain Reaction , Swine , Transcription, Genetic/drug effects
13.
Infect Immun ; 86(3)2018 03.
Article in English | MEDLINE | ID: mdl-29229731

ABSTRACT

Actinobacillus pleuropneumoniae is the causative agent of porcine contagious pleuropneumonia. Overproduction of proinflammatory cytokines, like interleukin-1ß (IL-1ß), IL-6, tumor necrosis factor alpha, and resistin, in the lung is an important feature of A. pleuropneumoniae infection. These proinflammatory cytokines enhance inflammatory and immunological responses. However, the mechanism that leads to cytokine production remains unclear. As a major virulence factor of A. pleuropneumoniae, lipopolysaccharide (LPS) may act as a potent stimulator of Toll-like receptor 4 (TLR4), triggering a number of intracellular signaling pathways that lead to the synthesis of proinflammatory cytokines. Porcine alveolar macrophages (PAMs) are the first line of defense against pathogenic microbes during pathogen invasion. The results of the present study demonstrate that A. pleuropneumoniae LPS induces PAMs to produce inflammatory cytokines in time- and dose-dependent manners. Moreover, PAMs were activated by A. pleuropneumoniae LPS, resulting in upregulation of signaling molecules, including TLR4, MyD88, TRIF-related adaptor molecule, and NF-κB. In contrast, the activation effects of A. pleuropneumoniae LPS on PAMs could be suppressed by specific inhibitors, like small interfering RNA and Bay11-7082. Taken together, our data indicate that A. pleuropneumoniae LPS can induce PAMs to produce proinflammatory cytokines via the TLR4/NF-κB-mediated pathway. These findings partially reveal the mechanism of the overproduction of proinflammatory cytokines in the lungs of swine with A. pleuropneumoniae infection and may provide targets for the prevention of A. pleuropneumoniae-induced pneumonia. All the data could be used as a reference for the pathogenesis of respiratory infection.


Subject(s)
Actinobacillus Infections/veterinary , Actinobacillus pleuropneumoniae/physiology , Lipopolysaccharides/immunology , Macrophages, Alveolar/immunology , Pleuropneumonia/veterinary , Swine Diseases/immunology , Toll-Like Receptor 4/immunology , Tumor Necrosis Factor-alpha/immunology , Actinobacillus Infections/genetics , Actinobacillus Infections/immunology , Actinobacillus Infections/microbiology , Actinobacillus pleuropneumoniae/genetics , Actinobacillus pleuropneumoniae/immunology , Animals , Cytokines/genetics , Cytokines/immunology , Macrophages, Alveolar/microbiology , Pleuropneumonia/genetics , Pleuropneumonia/immunology , Pleuropneumonia/microbiology , Signal Transduction , Swine , Swine Diseases/genetics , Swine Diseases/microbiology , Toll-Like Receptor 4/genetics , Tumor Necrosis Factor-alpha/genetics
14.
Anim Health Res Rev ; 19(1): 17-30, 2018 06.
Article in English | MEDLINE | ID: mdl-29110751

ABSTRACT

Actinobacillus pleuropneumoniae is a Gram-negative bacterium that belongs to the family Pasteurellaceae. It is the causative agent of porcine pleuropneumonia, a highly contagious respiratory disease that is responsible for major economic losses in the global pork industry. The disease may present itself as a chronic or an acute infection characterized by severe pathology, including hemorrhage, fibrinous and necrotic lung lesions, and, in the worst cases, rapid death. A. pleuropneumoniae is transmitted via aerosol route, direct contact with infected pigs, and by the farm environment. Many virulence factors associated with this bacterium are well characterized. However, much less is known about the role of biofilm, a sessile mode of growth that may have a critical impact on A. pleuropneumoniae pathogenicity. Here we review the current knowledge on A. pleuropneumoniae biofilm, factors associated with biofilm formation and dispersion, and the impact of biofilm on the pathogenesis A. pleuropneumoniae. We also provide an overview of current vaccination strategies against A. pleuropneumoniae and consider the possible role of biofilms vaccines for controlling the disease.


Subject(s)
Actinobacillus Infections/veterinary , Actinobacillus pleuropneumoniae/physiology , Bacterial Vaccines/immunology , Biofilms/growth & development , Swine Diseases/prevention & control , Actinobacillus Infections/microbiology , Actinobacillus Infections/prevention & control , Animals , Swine , Swine Diseases/microbiology
15.
Vet Res ; 48(1): 75, 2017 11 10.
Article in English | MEDLINE | ID: mdl-29126442

ABSTRACT

Housing of pigs in barren, stimulus-poor housing conditions may influence their immune status, including antibody responses to (auto-)antigens, and thus affect immune protection, which will influence the onset and outcome of infection. In the present study, we investigated the effects of environmental enrichment versus barren housing on the level of natural (auto-)antibodies (NA(A)b) and their isotypes (IgM and IgG) binding keyhole limpet hemocyanin (KLH), myelin basic protein (MBP), and phosphorycholine conjugated to bovine serum albumin (PC-BSA) in pigs co-infected with porcine reproductive and respiratory syndrome virus (PRRSV) and Actinobacillus pleuropneumoniae (A. pleuropneumoniae). Pigs (n = 56) were housed in either barren or enriched pens from birth to 54 days of age. They were infected with PRRSV on 44 days of age, and with A. pleuropneumoniae 8 days later. Blood samples were taken on 7 different sampling days. Housing significantly affected the overall serum levels of NA(A)b binding KLH, MBP and PC-BSA, and before infection barren housed pigs had significantly higher levels of NA(A)b than enriched housed pigs, except for KLH-IgM and PC-BSA-IgG. Infection only affected the IgM, but not the IgG isotype. Moreover, changes in MBP-IgM and PC-BSA-IgM following infection were different for enriched and barren housed pigs. These results suggest that the effect of infection on NA(A)b is influenced by housing conditions and that NA(A)b, especially IgM may be affected by infection.


Subject(s)
Actinobacillus Infections/veterinary , Antibodies, Bacterial/immunology , Antibodies, Viral/immunology , Autoantibodies/immunology , Housing, Animal , Porcine Reproductive and Respiratory Syndrome/immunology , Swine Diseases/immunology , Actinobacillus Infections/immunology , Actinobacillus Infections/virology , Actinobacillus pleuropneumoniae/physiology , Animals , Coinfection/immunology , Coinfection/virology , Female , Male , Porcine Reproductive and Respiratory Syndrome/virology , Porcine respiratory and reproductive syndrome virus/physiology , Sus scrofa/physiology , Swine , Swine Diseases/virology
16.
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
17.
PLoS One ; 12(4): e0176374, 2017.
Article in English | MEDLINE | ID: mdl-28448619

ABSTRACT

Pyridoxal 5'-phosphate (PLP) is an essential cofactor for numerous enzymes involved in a diversity of cellular processes in living organisms. Previous analysis of the Actinobacillus pleuropneumoniae S-8 genome sequence revealed the presence of pdxS and pdxT genes, which are implicated in deoxyxylulose 5-phosphate (DXP)-independent pathway of PLP biosynthesis; however, little is known about their roles in A. pleuropneumoniae pathogenicity. Our data demonstrated that A. pleuropneumoniae could synthesize PLP by PdxS and PdxT enzymes. Disruption of the pdxS and pdxT genes rendered the pathogen auxotrophic for PLP, and the defective growth as a result of these mutants was chemically compensated by the addition of PLP, suggesting the importance of PLP production for A. pleuropneumoniae growth and viability. Additionally, the pdxS and pdxT deletion mutants displayed morphological defects as indicated by irregular and aberrant shapes in the absence of PLP. The reduced growth of the pdxS and pdxT deletion mutants under osmotic and oxidative stress conditions suggests that the PLP synthases PdxS/PdxT are associated with the stress tolerance of A. pleuropneumoniae. Furthermore, disruption of the PLP biosynthesis pathway led to reduced colonization and attenuated virulence of A. pleuropneumoniae in the BALB/c mouse model. The data presented in this study reveal the critical role of PLP synthases PdxS/PdxT in viability, stress tolerance, and virulence of A. pleuropneumoniae.


Subject(s)
Actinobacillus pleuropneumoniae/enzymology , Actinobacillus pleuropneumoniae/physiology , Ligases/metabolism , Microbial Viability , Pyridoxal Phosphate/biosynthesis , Stress, Physiological , Actinobacillus pleuropneumoniae/genetics , Actinobacillus pleuropneumoniae/pathogenicity , Animals , Female , Gene Knockout Techniques , Hydrogen Peroxide/pharmacology , Ligases/deficiency , Ligases/genetics , Mice , Mice, Inbred BALB C , Mutation , Sodium Chloride/pharmacology , Virulence
18.
Microb Biotechnol ; 10(4): 756-760, 2017 07.
Article in English | MEDLINE | ID: mdl-27790837

ABSTRACT

Actinobacillus pleuropneumoniae causes porcine pleuropneumonia and forms biofilms in vitro on abiotic surfaces; however, presence of biofilms during infections has not been documented. The aim of this study was to use a species-specific fluorescent oligonucleotide probe and confocal microscopy to localize A. pleuropneumoniae in the lungs of two naturally infected pigs. Actinobacillus pleuropneumoniae was detected by fluorescence in situ hybridization and observed to grow as aggregates (~30-45 µm) during a natural infection. As the A. pleuropneumoniae aggregates observed in porcine lungs differed from the biofilms grown on a solid surface obtained in vitro, we designed a new biofilm assay using agarose, a porous substrate, favouring the formation of aggregates. In this study, we described for the first time the mode of growth of A. pleuropneumoniae during a natural infection in pigs. We also propose an in vitro biofilm assay for A. pleuropneumoniae using a porous substrate which allows the formation of aggregates. This assay might be more representative of the in vivo situation, at least in terms of the size of the bacterial aggregates and the presence of a porous matrix, and could potentially be used to test the susceptibility of A. pleuropneumoniae aggregates to antibiotics and disinfectants.


Subject(s)
Actinobacillus Infections/veterinary , Actinobacillus pleuropneumoniae/physiology , Bacterial Adhesion , Biofilms/growth & development , Lung/microbiology , Pleuropneumonia/veterinary , Swine Diseases/microbiology , Actinobacillus Infections/microbiology , Actinobacillus Infections/pathology , Actinobacillus pleuropneumoniae/growth & development , Animals , Bacteriological Techniques , In Situ Hybridization, Fluorescence , Lung/pathology , Microscopy, Confocal , Pleuropneumonia/microbiology , Pleuropneumonia/pathology , Swine , Swine Diseases/pathology
19.
Acta Vet Scand ; 58(1): 71, 2016 Oct 04.
Article in English | MEDLINE | ID: mdl-27716292

ABSTRACT

BACKGROUND: Respiratory illness is traditionally regarded as the disease of the growing pig, and has historically mainly been associated to bacterial infections with focus on Mycoplasma hyopneumoniae and Actinobacillus pleuropneumoniae. These bacteria still are of great importance, but continuously increasing herd sizes have complicated the scenario and the influence of secondary invaders may have been increased. The aim of this study was to evaluate the presence of A. pleuropneumoniae and M. hyopneumoniae, as well as that of the secondary invaders Pasteurella multocida and Streptococcus suis by serology in four pig herds (A-D) using age segregated rearing systems with high incidences of pleuritic lesions at slaughter. RESULTS: Pleuritic lesions registered at slaughter ranged from 20.5 to 33.1 % in the four herds. In herd A, the levels of serum antibodies to A. pleuropneumoniae exceeded A450 > 1.5, but not to any other microbe searched for. The seroconversion took place early during the fattening period. Similar levels of serum antibodies to A. pleuropneumoniae were also recorded in herd B, with a subsequent increase in levels of antibodies to P. multocida. Pigs seroconverted to both agents during the early phase of the fattening period. In herd C, pigs seroconverted to P. multocida during the early phase of the fattening period and thereafter to A. pleuropneumoniae. In herd D, the levels of antibodies to P. multocida exceeded A450 > 1.0 in absence (A450 < 0.5) of antibodies to A. pleuropneumoniae. The levels of serum antibodies to M. hyopneumoniae and to S. suis remained below A450 < 1.0 in all four herds. Pigs seroconverted to M. hyopneumoniae late during the rearing period (herd B-D), or not at all (herd A). CONCLUSION: Different serological patterns were found in the four herds with high levels of serum antibodies to A. pleuropneumoniae and P. multocida, either alone or in combination with each other. Seroconversion to M. hyopneumoniae late during the rearing period or not at all, confirmed the positive effect of age segregated rearing in preventing or delaying infections with M. hyopneumoniae. The results obtained highlight the necessity of diagnostic investigations to define the true disease pattern in herds with a high incidence of pleuritic lesions.


Subject(s)
Antibodies, Bacterial/blood , Bacterial Infections/veterinary , Pleurisy/veterinary , Swine Diseases/diagnosis , Swine Diseases/microbiology , Actinobacillus pleuropneumoniae/physiology , Animal Husbandry , Animals , Bacterial Infections/blood , Bacterial Infections/microbiology , Bacterial Infections/pathology , Mycoplasma hyopneumoniae/physiology , Pasteurella multocida/physiology , Pleurisy/blood , Pleurisy/microbiology , Pleurisy/pathology , Seroconversion , Streptococcus suis/physiology , Swine , Swine Diseases/blood , Swine Diseases/pathology , Time Factors
20.
PLoS One ; 11(9): e0161832, 2016.
Article in English | MEDLINE | ID: mdl-27606818

ABSTRACT

Until today, anti-microbial drugs have been the therapy of choice to combat bacterial diseases. Resistance against antibiotics is of growing concern in man and animals. Stress, caused by demanding environmental conditions, can reduce immune protection in the host, influencing the onset and outcome of infectious diseases. Therefore psychoneuro-immunological intervention may prove to be a successful approach to diminish the impact of diseases and antibiotics use. This study was designed to investigate the effect of social and environmental enrichment on the impact of disease, referred to as "disease susceptibility", in pigs using a co-infection model of PRRSV and A. pleuropneumoniae. Twenty-eight pigs were raised in four pens under barren conditions and twenty-eight other pigs were raised in four pens under enriched conditions. In the enriched pens a combination of established social and environmental enrichment factors were introduced. Two pens of the barren (BH) and two pens of the enriched housed (EH) pigs were infected with PRRSV followed by A. pleuropneumoniae, the other two pens in each housing treatment served as control groups. We tested if differences in disease susceptibility in terms of pathological and clinical outcome were related to the different housing regimes and if this was reflected in differences in behavioural and immunological states of the animals. Enriched housed pigs showed a faster clearance of viral PRRSV RNA in blood serum (p = 0.014) and histologically 2.8 fold less interstitial pneumonia signs in the lungs (p = 0.014). More barren housed than enriched housed pigs developed lesions in the lungs (OR = 19.2, p = 0.048) and the lesions in the barren housed pigs showed a higher total pathologic tissue damage score (p<0.001) than those in enriched housed pigs. EH pigs showed less stress-related behaviour and differed immunologically and clinically from BH pigs. We conclude that enriched housing management reduces disease susceptibility to co-infection of PRRSV and A. pleuropneumoniae in pigs. Enrichment positively influences behavioural state, immunological response and clinical outcome in pigs.


Subject(s)
Actinobacillus pleuropneumoniae/physiology , Coinfection/microbiology , Coinfection/virology , Housing, Animal , Porcine respiratory and reproductive syndrome virus/physiology , Swine Diseases/microbiology , Swine Diseases/virology , Actinobacillus Infections/blood , Actinobacillus Infections/complications , Actinobacillus Infections/virology , Animals , Antibodies/metabolism , Behavior, Animal , Biomarkers/metabolism , Body Temperature , Bronchoalveolar Lavage Fluid/cytology , Coinfection/blood , Disease Susceptibility , Female , Flow Cytometry , Leukocyte Count , Lung/microbiology , Lung/pathology , Lung/virology , Male , Phenotype , Porcine Reproductive and Respiratory Syndrome/blood , Porcine Reproductive and Respiratory Syndrome/virology , RNA, Viral/blood , Skin/microbiology , Skin/pathology , Skin/virology , Sus scrofa , Swine
SELECTION OF CITATIONS
SEARCH DETAIL
...