Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
Vet Res ; 54(1): 76, 2023 Sep 13.
Article in English | MEDLINE | ID: mdl-37705063

ABSTRACT

Due to the increase in bacterial resistance, improving the anti-infectious immunity of the host is rapidly becoming a new strategy for the prevention and treatment of bacterial pneumonia. However, the specific lung immune responses and key immune cell subsets involved in bacterial infection are obscure. Actinobacillus pleuropneumoniae (APP) can cause porcine pleuropneumonia, a highly contagious respiratory disease that has caused severe economic losses in the swine industry. Here, using high-dimensional mass cytometry, the major immune cell repertoire in the lungs of mice with APP infection was profiled. Various phenotypically distinct neutrophil subsets and Ly-6C+ inflammatory monocytes/macrophages accumulated post-infection. Moreover, a linear differentiation trajectory from inactivated to activated to apoptotic neutrophils corresponded with the stages of uninfected, onset, and recovery of APP infection. CD14+ neutrophils, which mainly increased in number during the recovery stage of infection, were revealed to have a stronger ability to produce cytokines, especially IL-10 and IL-21, than their CD14- counterparts. Importantly, MHC-II+ neutrophils with antigen-presenting cell features were identified, and their numbers increased in the lung after APP infection. Similar results were further confirmed in the lungs of piglets infected with APP and Klebsiella pneumoniae infection by using a single-cell RNA-seq technique. Additionally, a correlation analysis between cluster composition and the infection process yielded a dynamic and temporally associated immune landscape where key immune clusters, including previously unrecognized ones, marked various stages of infection. Thus, these results reveal the characteristics of key neutrophil clusters and provide a detailed understanding of the immune response to bacterial pneumonia.


Subject(s)
Actinobacillus Infections , Actinobacillus pleuropneumoniae , Ascomycota , Mycoplasma Infections , Pleuropneumonia , Pneumonia , Swine Diseases , Animals , Mice , Swine , Neutrophils , Pneumonia/veterinary , Pleuropneumonia/veterinary , Mycoplasma Infections/veterinary , Actinobacillus Infections/veterinary , Lung
2.
Vet Microbiol ; 269: 109438, 2022 Jun.
Article in English | MEDLINE | ID: mdl-35468400

ABSTRACT

Interleukin 5 (IL-5) regulates the maturation, activation, proliferation and function of immune cells, and plays an important role in the inflammatory response induced by an allergy. However, its anti-pathogen effect is poorly understood currently, especially on pneumonia. Here, this study was designed to elucidate the immunological role of IL-5 in the infection of mice with Actinobacillus pleuropneumoniae (APP). We established an acute lung infection model of APP in IL-5 knockout mice (IL-5-/-) and wild-type mice (WT) through nasal infusion or intraperitoneal injection, compared the survival rate, clinical symptoms, lung bacterial load, proportion of various immune cells, immune molecular expression, and neutrophil germicidal ability through flow cytometry, RT-qPCR, ELISA and immunofluorescence. Compared to WT mice, the IL-5-/- mice had a lower survival rate, more severe clinical symptoms, significantly increased bacterial load, and inflammatory cell infiltration in the lung after APP infection. In an uninfected state, IL-5 deficiency decreased the number of M1 interstitial macrophages and CD14- monocytes, while after infection, IL-5 deficiency significantly reduced the M2 alveolar macrophages, and increased PMN-II cells in the lung. Furthermore, the expression of IL-10, IL-4, IL-33, TNF-α, iNOS in the lung was lower in IL-5-/- mice under an uninfected condition, and the secretion of IL-18 was significantly increased after infection. In addition, IL-5 deficiency decreased bactericidal ability by inhibiting the formation of neutrophil extracellular traps (NETs). Collectively, these results provide evidence that IL-5 can enhance the resistance of APP infection, and its anti-infection mechanism, implying new targets and ideas for APP or similar respiratory agents' prevention and treatment.


Subject(s)
Actinobacillus Infections , Actinobacillus pleuropneumoniae , Actinobacillus , Extracellular Traps , Mycoplasma Infections , Mycoplasma , Pleuropneumonia , Rodent Diseases , Actinobacillus Infections/veterinary , Animals , Extracellular Traps/metabolism , Interleukin-5/genetics , Interleukin-5/metabolism , Lung/microbiology , Mice , Mice, Knockout , Mycoplasma Infections/veterinary , Pleuropneumonia/microbiology , Pleuropneumonia/veterinary
3.
AMB Express ; 11(1): 175, 2021 Dec 24.
Article in English | MEDLINE | ID: mdl-34952961

ABSTRACT

Porcine infectious pleuropneumonia is characterized by a high-rate of carriage and mixed infection with other pathogens. The host immune response induced by Actinobacillus pleuropneumoniae (APP) is the basis for elucidating pathogenesis and controlling disease. However, there is currently no comprehensive and dynamic data characterising the host immune response. In this study, piglets were infected with APP and differentially expressed proteins of bronchoalveolar lavage fluid (BALF) and peripheral serum were identified by iTRAQ-LC-MS/MS, and differentially expressed genes of peripheral blood mononuclear cells (PBMC) by RNA-seq. The results of the integrated analysis of serum, BALF and PBMC showed significant metabolism and local immune responses in BALF, the general immune response in PBMC mainly involves cytokines, while that in serum mainly involves biosynthesis, phagosome, and complement and coagulation cascades. Furthermore, immune responses in PBMCs and serum were rapid and maintained compared to the lung where metabolism and cell adhesion activities were enriched. Some innate immunity pathways of the cellular response to ROS, neutrophil mediated immunity, granulocyte activation and leukocyte cell-cell adhesion were identified as central points, connecting multiple signaling pathways to form an integrated large network. At 24 h post-infection, 14 molecules were up regulated in BALF, 10 of which were shared with PBMC, but at 120 h, 20 down-regulated molecules were identified in BALF, 11 of them still up- regulated in PBMC. We conclude that, the immune response in the lung is different from that in blood, but there is a similarity in response in PBMC and serum.

4.
Infect Immun ; 89(6)2021 05 17.
Article in English | MEDLINE | ID: mdl-33685942

ABSTRACT

Porcine pleuropneumonia is a common infectious disease of pigs caused by Actinobacillus pleuropneumoniae Interferon gamma (IFN-γ) expression increases in the lung of pigs after A. pleuropneumoniae infection, but the role of IFN-γ during the infection is still obscure. In this study, an IFN-γ-/- mouse infection model was established, and bacterial load, levels of inflammatory cytokines, and types of neutrophils in the lungs were studied at different times post-A. pleuropneumoniae infection. We found that wild-type (WT) mice were more susceptible to A. pleuropneumoniae than IFN-γ-/- mice. At 6 h postinfection (hpi), the expression of interleukin 18 (IL-18) and IL-1ß in the lungs of IFN-γ-/- mice was significantly increased compared to WT mice. The bacterial load and levels of inflammatory cytokines (IL-1ß and IL-6) of IFN-γ-/- mice were significantly reduced at 12 hpi compared to WT mice. After an initial loss, the numbers of lung polymorphonuclear (PMN)-I cells dramatically increased in the lungs of IFN-γ-/- but not WT mice, whereas PMN-II cells continually decreased. Finally, in vivo administration of IL-18 significantly reduced clinical scores and bacterial load in the lungs of A. pleuropneumoniae-infected mice. This study identifies IFN-γ as a target for regulating the inflammatory response in the lung and provides a basis for understanding the course of clinical bacterial pneumonia and for the formulation of treatment protocols.


Subject(s)
Actinobacillus Infections/immunology , Actinobacillus Infections/metabolism , Actinobacillus pleuropneumoniae/immunology , Host-Pathogen Interactions , Interleukin-18/metabolism , Neutrophils/immunology , Neutrophils/metabolism , Actinobacillus Infections/microbiology , Actinobacillus Infections/pathology , Animals , Disease Models, Animal , Host-Pathogen Interactions/genetics , Host-Pathogen Interactions/immunology , Lung/metabolism , Lung/microbiology , Lung/pathology , Mice , Mice, Knockout , Neutrophil Infiltration , Neutrophils/pathology
5.
Vet Microbiol ; 254: 108981, 2021 Mar.
Article in English | MEDLINE | ID: mdl-33445055

ABSTRACT

Streptococcus suis serotype 2 (SS2) is an important zoonotic pathogen that causes meningitis. The ubiquitously expressed 40S ribosome protein SA (RPSA) is a multifunctional protein involved in the pathogenesis of multiple pathogens, especially those causing meningitis. However, the role of RPSA in SS2-induced meningitis is not clear. In this study, immunofluorescence staining revealed that SS2 infection promoted the intracellular transfer of RPSA to the surface of human cerebral microvascular endothelial cells (HCMECs). Moreover, SS2 infection promoted the accumulation of caveolin 1 (CAV1) and the formation of membrane bulges where RPSA enveloped CAV1 on the cell surface. SS2 infection also caused dynamic changes in the localization of RPSA and CAV1 on the cell surface which could be eliminated by disruption of caveolae/rafts by addition of methyl-ß-cyclodextrin (MßCD). Co-immunoprecipitation analysis demonstrated that α-enolase (ENO), a key virulence factor of SS2, interacted with RPSA, and promoted the interaction between RPSA and CAV1. Immunofluorescence staining, western blotting and flow cytometry analyses showed that damaged caveolae/rafts significantly enhanced ENO adhesion to HCMECs, promoted the "destruction" of RPSA by ENO, and enhanced the toxic effect of ENO on HCMECs. Importantly, these effects could be relieved upon the addition of cholesterol. We conclude that caveolae/rafts weaken the toxic effect of SS2 ENO on RPSA-mediated events in HCMECs. Our study has led to better understanding of the roles of RPSA and caveolae/rafts upon SS2 infection, and a new pathological role for RPSA in infection.


Subject(s)
Caveolae/metabolism , Caveolin 1/metabolism , Endothelial Cells/microbiology , Phosphopyruvate Hydratase/metabolism , Ribosome Subunits, Small, Eukaryotic/metabolism , Streptococcus suis/pathogenicity , Animals , Cell Line , Fluorescent Antibody Technique , HEK293 Cells , Humans , Phosphopyruvate Hydratase/genetics , Ribosome Subunits, Small, Eukaryotic/genetics , Serogroup , Streptococcus suis/classification , Streptococcus suis/enzymology , Virulence Factors
6.
Microb Pathog ; 152: 104609, 2021 Mar.
Article in English | MEDLINE | ID: mdl-33217534

ABSTRACT

AIMS: 40S ribosomal protein SA (RPSA), a component of the small ribosomal subunit, is a high-affinity receptor of laminin that is widely expressed in cells and involves in many biological processes. However, it hasn't been reported which tissues and cells may be targeted by RPSA-mediated pathogen regulation. Therefore, in this study, a gram-positive bacterium Streptococcus suis Type 2 (SS2), gram-negative bacterium Actinobacillus pleuropneumoniae (A.pleuropneumoniae), and porcine circovirus Type 2 (PCV2) were used to infect ICR mice. METHODS AND RESULTS: The effects of infection with the three pathogens on expression levels of RPSA in mouse tissues and peripheral blood immune cells were analysed by immunohistochemistry and flow cytometry. The results suggested that the pathological changes in mice infected with SS2 were mainly manifested as congestion and inflammatory infiltration in the meninges, lungs, hearts and livers. The mice infected with A.pleuropneumoniae or PCV2 showed lung lesions and mild hepatocyte degeneration, respectively. In uninfected mice, RPSA protein was expressed to various degrees in all tissues except the spleen. After SS2 infection for 3 d, the expression of RPSA in the liver and brain increased, while decreased significantly in the heart and duodenum. These results were corroborated on examining the correlation between RPSA expression and the process of SS2 infection, except that there was no significant difference between the expression levels in the heart at 1 d and 3 d. After A.pleuropneumoniae and PCV2 infection for 3 d, the expression of RPSA decreased in the heart, and brain, respectively. Additionally, under physiological conditions, RPSA expression in CD4+ T cells, CD8+ T cells, neutrophils, and macrophages in the peripheral blood of mice was higher than that in B cells and NK cells. After SS2 infection for 3 d, RPSA expression increased significantly in CD4+ T cells and CD8+ T cells but decreased significantly in macrophages. The expression of RPSA after A.pleuropneumoniae and PCV2 infection were similar, and RPSA expression decreased only in macrophages. CONCLUSIONS: The results revealed that RPSA showed different expression levels in tissues and immune cells due to different pathogens causing disease courses, suggesting different target tissues and target cells in RPSA-mediated pathogenesis after infection, which supports the systematic study of the pathogenesis of RPSA in infectious diseases.


Subject(s)
Circoviridae Infections , Circovirus , Streptococcus suis , Swine Diseases , Animals , CD8-Positive T-Lymphocytes , Mice , Mice, Inbred ICR , Ribosomal Proteins , Swine
7.
AMB Express ; 10(1): 123, 2020 Jul 08.
Article in English | MEDLINE | ID: mdl-32642871

ABSTRACT

Actinobacillus pleuropneumoniae (A. pleuropneumoniae/APP) is the pathogen that causes porcine contagious pleuropneumonia. Actinobacillus pleuropneumoniae is divided into 18 serovars, and the cross protection efficacy of epitopes is debatable, which has resulted in the slow development of a vaccine. Consequently, epitope-based vaccines conferring Actinobacillus pleuropneumoniae cross protection have rarely been reported. In this study, B cell epitopes in the head domain of trimeric autotransporter adhesin were predicted, and 6 epitopes were selected. Then, the predicted epitopes (Ba1, Bb5, C1, PH1 and PH2) were connected by linkers to construct a recombinant tandem antigen (rta) gene. The RTA protein encoded by the recombinant rta gene was expressed, and finally the ICR mice were immunized with the RTA protein with or without inactivated Actinobacillus pleuropneumoniae (serovars 1 and 5b) and challenged with Actinobacillus pleuropneumoniae to evaluate the protective effect of the epitope-based vaccine and combined vaccine. The mice in the RTA-immunized group and RTA plus inactivated Actinobacillus pleuropneumoniae vaccine group had a significant improvement in clinical symptoms and a higher level of antibody in the serum than those in the control group. The RTA immune group had a 40% survival rate after Actinobacillus pleuropneumoniae infection, whereas the combination of RTA and inactivated Actinobacillus pleuropneumoniae produced very strong cross immune protection in mice, at least 50% (RTA IB1 + C5) and at most 100% (RTA IB5 + C1), whereas no cross immunoprotection was found in the solo Actinobacillus pleuropneumoniae immune group. Overall, the combination of the RTA protein and inactivated bacteria significantly enhanced the cross protection effects. This implies that RTA protein in combination with a suitable inactivated Actinobacillus pleuropneumoniae strain could be a candidate vaccine for porcine contagious pleuropneumonia.

8.
AMB Express ; 10(1): 121, 2020 Jul 06.
Article in English | MEDLINE | ID: mdl-32632500

ABSTRACT

Porcine pleuropneumonia caused by Actinobacillus pleuropneumoniae (APP) is a swine respiratory disease with an important impact around the world either as a single infection or part of the porcine respiratory disease complex. The data of interaction between hosts and pathogens has becoming more crucial for exploration of the mechanism. However, up to now, comparatively little information is available on the systemic and dynamic changes that occur in pig serum in response to APP infection. This study used iTRAQ to identify differentially expressed proteins (DEPs) in pig serum in response to APP infection. Compared with the APP un-infected group (S0),there were 137 up-regulated and 68 down-regulated proteins at 24 h (S24), and 81 up-regulated and 107 down-regulated proteins at 120 h (S120). At 24 h, the immune response was not significantly enriched, but cell adhesion, cytosol, Golgi apparatus, GTP and ATP binding and regulation of cell cycle were extremely active, implying host preparation of immune response starting. Subsequently, innate immune response, negative regulation of apoptotic process, immunological synapse, adaptive immune response, the regulation of inflammatory response, positive regulation of T cell proliferation were more enhanced at 120 h then that of 24 h, representing innate immunity transferring to the adaptive, while endocytosis, cell adhesion and platelet aggregation showed obvious decline. The pathways of T cell receptor signaling pathway, cytokine-cytokine receptor interaction, complement and coagulation cascades, leukocyte transendothelial migration were active remarkably during all infection period, and more pathways could connect to form innate immune defense networks. Surprisingly, the pathways like amoebiasis, rheumatoid arthritis and malaria had been found up-regulated. As a conclusion, APP could delay host inflammatory response to the infection at early stage, and induced innate immunity to convert from adhesion, interaction into complement activation, proteasome digestion, bacterial invasion at later stage. This would increase our understanding of the porcine distinct response to APP infection.

9.
Vet Microbiol ; 247: 108755, 2020 Aug.
Article in English | MEDLINE | ID: mdl-32686648

ABSTRACT

Excessive cytokine production is an important component of the acute respiratory distress syndrome and multiple organ failure. Pneumonia can lead to an overexpression of cytokines, although comparatively little is known about the relevance and differences in cytokines between blood and lung. In this study, piglets were experimentally infected intranasally with Actinobacillus pleuropneumoniae (APP), and transcriptomes of lung tissue and peripheral blood mononuclear cells determined. In addition, the levels of 30 cytokines in broncheoalveolar lavage fluid (BALF) and sera were determined by ELISA. Post infection, there was an early increase in lung monocytes, and a later rise in inflammatory cytokines in BALF. Blood lymphocytes increased early in infection and there was a rise in inflammatory cytokines in the peripheral blood of infected piglets. Genes involved in cytokine production, leukocyte migration and differentiation, lymphocyte activation, and cytokine-mediated signaling pathways in the transcriptomes of lung tissue were significantly down-regulated early in infection. At this early phase of APP infection (0-6 h), the cytokines IL-1ß, MCP-1, and IL-5 in sera increased rapidly and significantly, while many cytokines in BALF decreased. At 48 h post-infection, cytokines in sera were no longer significantly increased, although some were up-regulated in BALF, and there was aggravated pathological damage in the lungs at this time. The data indicate there are substantial differences between immune cells and cytokines in the lung and peripheral blood of APP infected piglets at equivalent time points. The results increase our understanding of pig-APP host interactive biology, and will be important in formulating future therapeutic and preventative strategies to prevent disease caused by APP.


Subject(s)
Actinobacillus Infections/blood , Actinobacillus Infections/veterinary , Immunity , Lung/microbiology , Respiratory System/microbiology , Actinobacillus pleuropneumoniae/immunology , Animals , Bronchoalveolar Lavage Fluid/microbiology , Chemokines/immunology , Cytokines/immunology , Leukocytes, Mononuclear/immunology , Lung/pathology , Lymphocytes/immunology , Specific Pathogen-Free Organisms , Swine , Transcriptome
10.
Front Immunol ; 11: 585399, 2020.
Article in English | MEDLINE | ID: mdl-33603733

ABSTRACT

Streptococcus suis serotype 2 (SS2), an important zoonotic pathogen that causes septicemia, arthritis, and irreversible meningitis in pigs and humans, can be transmitted to humans from pigs. S. suis causes huge economic losses to the swine industry and poses a serious threat to public health. Previously, we found that the brain tissues of mice with SS2-induced meningitis showed disrupted structural integrity and significantly enhanced polymorphonuclear neutrophil (PMN) infiltration. We showed that the brain tissues of SS2-infected mice had increased ribosomal protein SA (RPSA)-positive PMN counts. However, the inflammatory responses of RPSA+ PMNs to SS2 and their effects on the blood-brain barrier (BBB) remain unclear. Therefore, in studying the pathogenesis of SS2-induced meningitis, it is essential that we explore the functions of RPSA+ PMNs and their effects on the BBB. Herein, using flow cytometry and immunofluorescence microscopy analyses, we found that RPSA expression enhances PMN-induced phagocytosis and PMN-induced formation of neutrophil extracellular traps (NETs), which facilitate further elimination of bacteria. PMN surface expression of RPSA also alleviates local inflammation and tissue injuries by inhibiting secretion of the pro-inflammatory cytokines, TNF-α and IL-6. Moreover, the single-cell BBB model showed that RPSA disrupts BBB integrity by downregulating expression of tight junction-associated membrane proteins on PMNs. Taken together, our data suggest that PMN-surface expression of RPSA is a double-edged sword. RPSA+ PMN owns a stronger ability of bacterial cleaning and weakens inflammatory cytokines release which are useful to anti-infection, but does hurt BBB. Partly, RPSA+ PMN may be extremely useful to control the infection as a therapeutic cellular population, following novel insights into the special PMN population.


Subject(s)
Extracellular Traps/immunology , Meningitis, Pneumococcal/immunology , Neutrophils/immunology , Phagocytosis/immunology , Receptors, Laminin/immunology , Ribosomal Proteins/immunology , Animals , Blood-Brain Barrier/immunology , Blood-Brain Barrier/pathology , Cytokines/immunology , Meningitis, Pneumococcal/pathology , Mice , Mice, Inbred C57BL , Neutrophils/metabolism , Receptors, Laminin/metabolism , Ribosomal Proteins/metabolism , Streptococcus suis/immunology
11.
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
12.
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
13.
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
14.
Immunol Res ; 65(6): 1124-1129, 2017 Dec.
Article in English | MEDLINE | ID: mdl-28929313

ABSTRACT

Porcine contagious pleuropneumonia is a highly fatal respiratory disease that is caused by Actinobacillus pleuropneumoniae (APP) and results in tremendous economic losses for the pig breeding industry worldwide. Previous studies have demonstrated that Propionibacterium acnes (PA) could effectively prevent APP infection in mice and pigs. The humoral immune response played a primary role during this process and anti-PA antibody could mediate macrophages to kill the bacteria. However, the role of neutrophils in this process is currently unknown. In this study, mice were injected with cyclophosphamide to deplete neutrophils and then passively immunized with anti-PA serum or negative serum. Mice were subsequently challenged with APP serotype 1. The results showed that the mice exhibited less bacterial colonization, less lung damage, and a high survival rate, which were immunized with the anti-PA antibody whether neutrophils were depleted or not. Worse still, the presence of neutrophils increased the damage to the mice after challenge. These results suggest that the activity of the anti-PA antibody against APP infection was independent of neutrophils. These findings have important significance for understanding the mechanisms of humoral immunity conferred by heterologous immunization and lay a good foundation for preventing APP infection.


Subject(s)
Actinobacillus Infections/immunology , Actinobacillus pleuropneumoniae/immunology , Antibodies, Bacterial/metabolism , Lung/pathology , Neutrophils/immunology , Pleuropneumonia, Contagious/immunology , Propionibacterium acnes/physiology , Animals , Cyclophosphamide/administration & dosage , Female , Immunity, Heterologous , Immunity, Humoral , Immunization, Passive , Leukapheresis , Mice , Mice, Inbred BALB C , Swine
15.
Mol Immunol ; 81: 108-117, 2017 01.
Article in English | MEDLINE | ID: mdl-27940254

ABSTRACT

Actinobacillus pleuropneumoniae is the causative pathogen of porcine pleuropneumonia, which results in large economic losses in the pig industry worldwide. There are, however, no effective subunit vaccines are available in the market owing to the various serotypes and the absence of cross-protection against this pathogen. Therefore, the selection of protective components is of great significance for vaccine development. We previously showed that trimeric autotransporter adhesins are important virulence factors of A. pleuropneumoniae. To determine the potential role in vaccine development of the functional head domain (Apa2H1) of Apa2, a trimeric autotransporter adhesin found in A. pleuropneumoniae, we obtained nature-like trimeric Apa2H1 using a prokaryotic expression system and co-culture of Apa2H1 with bone marrow derived dendritic cells (BMDCs) in vitro resulted in maturation of BMDCs, characterised by the up-regulation of CD83, MHC-II, CCR7, ICAM-I and the increased expression of factors related to B lymphoid cells stimulation, such as proliferation-inducing ligand (APRIL), B lymphocyte stimulator (BLyS) and B cell activating factor (BAFF). The in vivo results showed that vaccination with Apa2H1 resulted in the robust production of antigen-specific antibodies, modestly induced mixed Th1 and Th2 immunity, impaired bacterial colonization and dissemination, and improved mouse survival rates. This study is the first to show that Apa2H1 is antigenic and can be used as a component of a subunit vaccine against A. pleuropneumoniae infection, providing valuable reference material for the development of an effective vaccine against A. pleuropneumoniae.


Subject(s)
Actinobacillus Infections/immunology , Adhesins, Bacterial/immunology , Bacterial Vaccines/immunology , Bone Marrow Cells/immunology , Dendritic Cells/immunology , Actinobacillus pleuropneumoniae/pathogenicity , Animals , Blotting, Western , Disease Models, Animal , Female , Flow Cytometry , Mice , Mice, Inbred BALB C , Protein Domains , Real-Time Polymerase Chain Reaction , Vaccination , Vaccines, Subunit/immunology , Virulence Factors/immunology
16.
Sci Rep ; 6: 24058, 2016 Apr 05.
Article in English | MEDLINE | ID: mdl-27046446

ABSTRACT

Members of the Trimeric Autotransporter Adhesin (TAA) family play a crucial role in the adhesion of Gram-negative pathogens to host cells, but the immunopathogenesis of TAAs remains unknown. Our previous studies demonstrated that Adh from Actinobacillus pleuropneumoniae (A. pleuropneumoniae) is required for full bacterial pathogenicity. Alveolar macrophages are the first line of defense against respiratory infections. This study compared the interactions between porcine alveolar macrophages (PAMs) and wild-type A. pleuropneumoniae (5b WT) or an Adh-deletion strain (5b ΔAdh) via gene microarray, immunoprecipitation and other technologies. We found that Adh was shown to interact with the PAMs membrane protein OR5M11, an olfactory receptor, resulting in the high-level secretion of IL-8 by activation of p38 MAPK signaling pathway. Subsequently, PAMs apoptosis via the activation of the Fax and Bax signaling pathways was observed, followed by activation of caspases 8, 9, and 3. The immunological pathogenic roles of Adh were also confirmed in both murine and piglets infectious models in vivo. These results identify a novel immunological strategy for TAAs to boost the pathogenicity of A. pleuropneumoniae. Together, these datas reveal the high versatility of the Adh protein as a virulence factor and provide novel insight into the immunological pathogenic role of TAAs.


Subject(s)
Actinobacillus pleuropneumoniae/pathogenicity , Apoptosis , Bacterial Proteins/metabolism , Interleukin-8/metabolism , Receptors, Odorant/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism , Adhesins, Bacterial/metabolism , Animals , Caspase 9/metabolism , Cytokines/metabolism , Flow Cytometry , Gene Deletion , Gene Expression Profiling , HEK293 Cells , Humans , Inflammation , Lymphocytes/cytology , Macrophages, Alveolar/metabolism , Membrane Potential, Mitochondrial , Mice , Mice, Inbred BALB C , Microarray Analysis , Oligonucleotide Array Sequence Analysis , Phosphorylation , Receptors, G-Protein-Coupled/metabolism , Signal Transduction , Swine , Virulence , Virulence Factors/metabolism , bcl-2-Associated X Protein/metabolism , fas Receptor/metabolism
17.
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
SELECTION OF CITATIONS
SEARCH DETAIL
...