1
|
Ke CH, Lai PY, Hsu FY, Hsueh PR, Chiou MT, Lin CN. Antimicrobial susceptibility and resistome of Actinobacillus pleuropneumoniae in Taiwan: a next-generation sequencing analysis. Vet Q 2024; 44:1-13. [PMID: 38688482 PMCID: PMC11064736 DOI: 10.1080/01652176.2024.2335947] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Accepted: 03/21/2024] [Indexed: 05/02/2024] Open
Abstract
Actinobacillus pleuropneumoniae infection causes a high mortality rate in porcine animals. Antimicrobial resistance poses global threats to public health. The current study aimed to determine the antimicrobial susceptibilities and probe the resistome of A. pleuropneumoniae in Taiwan. Herein, 133 isolates were retrospectively collected; upon initial screening, 38 samples were subjected to next-generation sequencing (NGS). Over the period 2017-2022, the lowest frequencies of resistant isolates were found for ceftiofur, cephalexin, cephalothin, and enrofloxacin, while the highest frequencies of resistant isolates were found for oxytetracycline, streptomycin, doxycycline, ampicillin, amoxicillin, kanamycin, and florfenicol. Furthermore, most isolates (71.4%) showed multiple drug resistance. NGS-based resistome analysis revealed aminoglycoside- and tetracycline-related genes at the highest prevalence, followed by genes related to beta-lactam, sulfamethoxazole, florphenicol, and macrolide. A plasmid replicon (repUS47) and insertion sequences (IS10R and ISVAp11) were identified in resistant isolates. Notably, the multiple resistance roles of the insertion sequence IS10R were widely proposed in human medicine; however, this is the first time IS10R has been reported in veterinary medicine. Concordance analysis revealed a high consistency of phenotypic and genotypic susceptibility to florphenicol, tilmicosin, doxycycline, and oxytetracycline. The current study reports the antimicrobial characterization of A. pleuropneumoniae for the first time in Taiwan using NGS.
Collapse
Affiliation(s)
- Chiao-Hsu Ke
- Sustainable Swine Research Center, National Pingtung University of Science and Technology, Pingtung, Taiwan
- Animal Disease Diagnostic Center, College of Veterinary Medicine, National Pingtung University of Science and Technology, Pingtung, Taiwan
| | - Pan-Yun Lai
- Department of Veterinary Medicine, College of Veterinary Medicine, National Pingtung University of Science and Technology, Pingtung, Taiwan
| | - Feng-Yang Hsu
- Animal Disease Diagnostic Center, College of Veterinary Medicine, National Pingtung University of Science and Technology, Pingtung, Taiwan
| | - Po-Ren Hsueh
- Department of Laboratory Medicine and Internal Medicine, China Medical University Hospital, School of Medicine, China Medical University, Taichung, Taiwan
- Department of Laboratory Medicine and Internal Medicine, National Taiwan University Hospital, National Taiwan University College of Medicine, Taipei, Taiwan
| | - Ming-Tang Chiou
- Sustainable Swine Research Center, National Pingtung University of Science and Technology, Pingtung, Taiwan
- Animal Disease Diagnostic Center, College of Veterinary Medicine, National Pingtung University of Science and Technology, Pingtung, Taiwan
- Department of Veterinary Medicine, College of Veterinary Medicine, National Pingtung University of Science and Technology, Pingtung, Taiwan
| | - Chao-Nan Lin
- Sustainable Swine Research Center, National Pingtung University of Science and Technology, Pingtung, Taiwan
- Animal Disease Diagnostic Center, College of Veterinary Medicine, National Pingtung University of Science and Technology, Pingtung, Taiwan
- Department of Veterinary Medicine, College of Veterinary Medicine, National Pingtung University of Science and Technology, Pingtung, Taiwan
| |
Collapse
|
2
|
Sun M, Gao AX, Li A, Ledesma-Amaro R, Wang P, Chen W, Bai Z, Liu X. Hyper-production of porcine contagious pleuropneumonia subunit vaccine proteins in Escherichia coli by developing a bicistronic T7 expression system. Biotechnol J 2024; 19:e2300187. [PMID: 38178735 DOI: 10.1002/biot.202300187] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Revised: 12/15/2023] [Accepted: 12/20/2023] [Indexed: 01/06/2024]
Abstract
The ApxII toxin and the outer membrane lipoprotein (Oml) of Actinobacillus pleuropneumoniae are important vaccine antigens against porcine contagious pleuropneumonia (PCP), a prevalent infectious disease affecting the swine industry worldwide. Previous studies have reported the recombinant expression of ApxII and Oml in Escherichia coli; however, their yields were not satisfactory. Here, we aimed to enhance the production of ApxII and Oml by constructing a bicistronic expression system based on the widely used T7 promoter. To create efficient T7 bicistronic expression cassettes, 16 different fore-cistron sequences were introduced downstream of the T7 promoter. The expression of three vaccine antigens Oml1, Oml7, and ApxII in the four strongest bicistronic vectors were enhanced compared to the monocistronic control. Further optimization of the fermentation conditions in micro-well plates (MWP) led to improved production. Finally, the production yields reached unprecedented levels of 2.43 g L-1 of Oml1, 2.59 g L-1 of Oml7, and 1.21 g L-1 of ApxII, in a 5 L bioreactor. These three antigens also demonstrated well-protective immunity against A. pleuropneumoniae infection. In conclusion, this study establishes an efficient bicistronic T7 expression system that can be used to express recombinant proteins in E. coli and achieves the hyper-production of PCP vaccine proteins.
Collapse
Affiliation(s)
- Manman Sun
- National Engineering Research Center of Cereal Fermentation and Food Biomanufacturing, Jiangnan University, Wuxi, China
- Department of Bioengineering and Imperial College Centre for Synthetic Biology, Imperial College London, London, UK
- Key Laboratory of High Magnetic Field and Ion Beam Physical Biology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, China
| | - Alex Xiong Gao
- Division of Life Science, The Hong Kong University of Science and Technology, Hong Kong, China
| | - An Li
- National Engineering Research Center of Cereal Fermentation and Food Biomanufacturing, Jiangnan University, Wuxi, China
| | - Rodrigo Ledesma-Amaro
- Department of Bioengineering and Imperial College Centre for Synthetic Biology, Imperial College London, London, UK
| | - Peng Wang
- Key Laboratory of High Magnetic Field and Ion Beam Physical Biology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, China
| | - Wenchao Chen
- Oil Crops Research Institute of the Chinese Academy of Agricultural Sciences, Wuhan, Hubei, China
| | - Zhonghu Bai
- National Engineering Research Center of Cereal Fermentation and Food Biomanufacturing, Jiangnan University, Wuxi, China
| | - Xiuxia Liu
- National Engineering Research Center of Cereal Fermentation and Food Biomanufacturing, Jiangnan University, Wuxi, China
| |
Collapse
|
3
|
Li N, Zhu J, Chen P, Bao C, Wang J, Abdelaal T, Chen D, Zhu S, Wang W, Mao J, Scicluna BP, Koning F, Li F, Lei L. High-dimensional analysis reveals an immune atlas and novel neutrophil clusters in the lungs of model animals with Actinobacillus pleuropneumoniae-induced pneumonia. Vet Res 2023; 54:76. [PMID: 37705063 PMCID: PMC10500746 DOI: 10.1186/s13567-023-01207-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2023] [Accepted: 07/24/2023] [Indexed: 09/15/2023] Open
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.
Collapse
Affiliation(s)
- Na Li
- State Key Laboratory for Zoonotic Diseases, Key Laboratory of Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, China
| | - Junhui Zhu
- State Key Laboratory for Zoonotic Diseases, Key Laboratory of Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, China
| | - Peiru Chen
- State Key Laboratory for Zoonotic Diseases, Key Laboratory of Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, China
| | - Chuntong Bao
- State Key Laboratory for Zoonotic Diseases, Key Laboratory of Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, China
| | - Jun Wang
- State Key Laboratory for Zoonotic Diseases, Key Laboratory of Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, China
| | - Tamim Abdelaal
- Leiden Computational Biology Center, Leiden University Medical Center, Leiden, The Netherlands
- Department of Pattern Recognition and Bioinformatics Group, Delft University of Technology, Delft, The Netherlands
| | - Dexi Chen
- Beijing Institute of Hepatology, Beijing Youan Hospital, Capital Medical University, Beijing, China
| | - Sibo Zhu
- School of Life Sciences, Fudan University, Shanghai, China
| | - Wenjing Wang
- Beijing Institute of Hepatology, Beijing Youan Hospital, Capital Medical University, Beijing, China
| | - Jiangnan Mao
- School of Life Sciences, Fudan University, Shanghai, China
| | - Brendon P Scicluna
- Department of Applied Biomedical Science, Faculty of Health Sciences, Mater Dei Hospital, University of Malta, Msida, Malta
- Centre for Molecular Medicine and Biobanking, University of Malta, Msida, Malta
| | - Frits Koning
- Department of Immunology, Leiden University Medical Center, Leiden, The Netherlands
| | - Fengyang Li
- State Key Laboratory for Zoonotic Diseases, Key Laboratory of Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, China.
| | - Liancheng Lei
- State Key Laboratory for Zoonotic Diseases, Key Laboratory of Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, China.
- College of Animal Science, Yangtze University, Jingzhou, Hubei, China.
| |
Collapse
|
4
|
Comparative Efficacy in Challenge Dose Models of a Toxin Expressing Whole-Cell Vaccine against Eight Serovars of Actinobacillus pleuropneumoniae in Pigs. Animals (Basel) 2022; 12:ani12233244. [PMID: 36496765 PMCID: PMC9740876 DOI: 10.3390/ani12233244] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2022] [Revised: 11/19/2022] [Accepted: 11/20/2022] [Indexed: 11/24/2022] Open
Abstract
Actinobacillus pleuropneumoniae is a major economically significant bacterial respiratory pig pathogen, and whole cell vaccines are used to prevent disease. However, there is little data available on multi-serovar whole cell vaccine protection. Therefore, we determined the protective efficacies of a whole-cell A. pleuropneumoniae serovar 1 and 2 vaccine comprising ApxI-III toxins (C-vaccine, Coglapix®, Ceva, France) against serovars 1, 2, 4, 5, 6, 7, 9/11, and 13. The infection doses used induced disease representative of endemic field conditions, and standard protocols were used for all studies. Protection against homologous serovars 1 and 2 significantly reduced lung lesion scores (LLS) compared to positive controls: p = 0.00007 and p = 0.00124, respectively. The protection against heterologous serovars 4, 5, 6, 7, 9/11, and 13 also significantly reduced LLS: range p = 2.9 × 10-10 to p = 0.00953. As adjudged by the estimated random effect, reproducibility between studies was high. A highly significant serovar-independent reduction of pathological lung lesions by the C-vaccine was found for all the serovars tested (1, 2, 4, 5, 6, 7, 9/11, and 13). We conclude that the C-vaccine gives high serovar-independent protection against disease and is suitable for this use in the field.
Collapse
|
5
|
Outer Membrane Vesicles of Actinobacillus pleuropneumoniae Exert Immunomodulatory Effects on Porcine Alveolar Macrophages. Microbiol Spectr 2022; 10:e0181922. [PMID: 36040198 PMCID: PMC9602539 DOI: 10.1128/spectrum.01819-22] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Outer membrane vesicles (OMVs) are spontaneously released by Gram-negative bacteria, including Actinobacillus pleuropneumoniae, which causes contagious pleuropneumonia in pigs and leads to considerable economic losses in the swine industry worldwide. A. pleuropneumoniae OMVs have previously been demonstrated to contain Apx toxins and proteases, as well as antigenic proteins. Nevertheless, comprehensive characterizations of their contents and interactions with host immune cells have not been made. Understanding the protein compositions and immunomodulating ability of A. pleuropneumoniae OMVs could help illuminate their biological functions and facilitate the development of OMV-based applications. In the current investigation, we comprehensively characterized the proteome of native A. pleuropneumoniae OMVs. Moreover, we qualitatively and quantitatively compared the OMV proteomes of a wild-type strain and three mutant strains, in which relevant genes were disrupted to increase OMV production and/or produce OMVs devoid of superantigen PalA. Furthermore, the interaction between A. pleuropneumoniae OMVs and porcine alveolar macrophages was also characterized. Our results indicate that native OMVs spontaneously released by A. pleuropneumoniae MIDG2331 appeared to dampen the innate immune responses by porcine alveolar macrophages stimulated by either inactivated or live parent cells. The findings suggest that OMVs may play a role in manipulating the porcine defense during the initial phases of the A. pleuropneumoniae infection. IMPORTANCE Owing to their built-in adjuvanticity and antigenicity, bacterial outer membrane vesicles (OMVs) are gaining increasing attention as potential vaccines for both human and animal use. OMVs released by Actinobacillus pleuropneumoniae, an important respiratory pathogen in pigs, have also been investigated for vaccine development. Our previous studies have shown that A. pleuropneumoniae secretes OMVs containing multiple immunogenic proteins. However, immunization of pigs with these vesicles was not able to relieve the pig lung lesions induced by the challenge with A. pleuropneumoniae, implying the elusive roles that A. pleuropneumoniae OMVs play in host-pathogen interaction. Here, we showed that A. pleuropneumoniae secretes OMVs whose yield and protein content can be altered by the deletion of the nlpI and palA genes. Furthermore, we demonstrate that A. pleuropneumoniae OMVs dampen the immune responses in porcine alveolar macrophages stimulated by A. pleuropneumoniae cells, suggesting a novel mechanism that A. pleuropneumoniae might use to evade host defense.
Collapse
|
6
|
Stancheva SG, Frömbling J, Sassu EL, Hennig-Pauka I, Ladinig A, Gerner W, Grunert T, Ehling-Schulz M. Proteomic and immunoproteomic insights into the exoproteome of Actinobacillus pleuropneumoniae, the causative agent of porcine pleuropneumonia. Microb Pathog 2022; 172:105759. [PMID: 36087692 DOI: 10.1016/j.micpath.2022.105759] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Revised: 08/26/2022] [Accepted: 08/30/2022] [Indexed: 10/31/2022]
Abstract
Porcine pleuropneumonia caused by Actinobacillus pleuropneumoniae affects pig health status and the swine industry worldwide. Despite the extensive number of studies focused on A. pleuropneumoniae infection and vaccine development, a thorough analysis of the A. pleuropneumoniae exoproteome is still missing. Using a complementary approach of quantitative proteomics and immunoproteomics we gained an in-depth insight into the A. pleuropneumoniae serotype 2 exoproteome, which provides the basis for future functional studies. Label-free liquid chromatography-tandem mass spectrometry (LC-MS/MS) revealed 593 exoproteins, of which 104 were predicted to be virulence factors. The RTX toxins ApxIIA and ApxIIIA -were found to be the most abundant proteins in the A. pleuropneumoniae serotype 2 exoproteome. Furthermore, the ApxIVA toxin was one of the proteins showing the highest abundance, although ApxIVA is commonly assumed to be expressed exclusively in vivo. Our study revealed several antigens, including proteins with moonlight functions, such as the elongation factor (EF)-Tu, and proteins linked to specific metabolic traits, such as the maltodextrin-binding protein MalE, that warrant future functional characterization and might present potential targets for novel therapeutics and vaccines. Our Ig-classes specific serological proteome analysis (SERPA) approach allowed us to explore the development of the host humoral immune response over the course of the infection. These SERPAs pinpointed proteins that might play a key role in virulence and persistence and showed that the immune response to the different Apx toxins is distinct. For instance, our results indicate that the ApxIIIA toxin has properties of a thymus-independent antigen, which should be studied in more detail.
Collapse
Affiliation(s)
- Stelli G Stancheva
- Institute of Microbiology, Department for Pathobiology, University of Veterinary Medicine Vienna, Austria
| | - Janna Frömbling
- Institute of Microbiology, Department for Pathobiology, University of Veterinary Medicine Vienna, Austria
| | - Elena L Sassu
- University Clinic for Swine, Department for Farm Animals and Veterinary Public Health, University of Veterinary Medicine Vienna, Austria
| | - Isabel Hennig-Pauka
- Field Station for Epidemiology, University of Veterinary Medicine Hannover, Bakum, Germany
| | - Andrea Ladinig
- University Clinic for Swine, Department for Farm Animals and Veterinary Public Health, University of Veterinary Medicine Vienna, Austria
| | - Wilhelm Gerner
- Institute of Immunology, Department of Pathobiology, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Tom Grunert
- Institute of Microbiology, Department for Pathobiology, University of Veterinary Medicine Vienna, Austria
| | - Monika Ehling-Schulz
- Institute of Microbiology, Department for Pathobiology, University of Veterinary Medicine Vienna, Austria.
| |
Collapse
|
7
|
Chuekwon K, Chu CY, Cheng LT. N-terminus of flagellin enhances vaccine efficacy against Actinobacillus pleuropneumoniae. BMC Vet Res 2022; 18:279. [PMID: 35842618 PMCID: PMC9288005 DOI: 10.1186/s12917-022-03380-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Accepted: 07/12/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Flagellin elicits potent immune response and may serve as a vaccine adjuvant. We previously reported that the N-terminus of flagellin (residues 1-99, nFliC) is sufficient for vaccine efficacy enhancement against Pasteurella multocida challenge in chickens. In this study, we futher tested the adjuvancy of nFliC in a subunit vaccine against the pig pathogen Actinobacillus pleuropneumoniae in a mice model. For vaccine formulation, the antigen ApxIIPF (the pore-forming region of the exotoxin ApxII) was combined with nFliC, either through genetic fusion or simple admixture. RESULTS Immune analysis showed that nFliC, introduced through genetic fusion or admixture, enhanced both humoral (antibody levels) and cellular (T cell response and cytokine production) immunity. In a challenge test, nFliC increased vaccine protective efficacy to 60-80%, vs. 20% for the antigen-only group. Further analysis showed that, even without a supplemental adjuvant such as mineral salt or oil emulsion, genetically linked nFliC still provided significant immune enhancement. CONCLUSIONS We conclude that nFliC is a versatile and potent adjuvant for vaccine formulation.
Collapse
Affiliation(s)
- Kamonpun Chuekwon
- Department of Tropical Agriculture and International Cooperation, International College, National Pingtung University of Science and Technology, 1, Shuefu Road, Neipu, Pingtung, 91201, Taiwan
| | - Chun-Yen Chu
- Graduate Institute of Animal Vaccine Technology, College of Veterinary Medicine, National Pingtung University of Science and Technology, 1, Shuefu Road, Neipu, Pingtung, 91201, Taiwan
| | - Li-Ting Cheng
- Graduate Institute of Animal Vaccine Technology, College of Veterinary Medicine, National Pingtung University of Science and Technology, 1, Shuefu Road, Neipu, Pingtung, 91201, Taiwan.
| |
Collapse
|
8
|
Zhang L, Luo W, Xiong R, Li H, Yao Z, Zhuo W, Zou G, Huang Q, Zhou R. A Combinatorial Vaccine Containing Inactivated Bacterin and Subunits Provides Protection Against Actinobacillus pleuropneumoniae Infection in Mice and Pigs. Front Vet Sci 2022; 9:902497. [PMID: 35747235 PMCID: PMC9212066 DOI: 10.3389/fvets.2022.902497] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Accepted: 04/26/2022] [Indexed: 11/29/2022] Open
Abstract
Actinobacillus pleuropneumoniae (APP) is the etiological agent of porcine contagious pleuropneumonia (PCP) that causes great economic losses in the swine industry. Currently, vaccination is still a commonly used strategy for the prevention of the disease. Commercially available vaccines of this disease, including inactivated bacterins and subunit vaccines, have clinical limitations such as side effects and low cross-protection. In this study, a combinatorial vaccine (Bac-sub) was developed, which contained inactivated bacterial cells of a serovar 1 strain and three recombinant protoxins (rApxIA, rApxIIA, and rApxIIIA). Its side effects, immune protection, and cross-protection were evaluated and compared with a commercial subunit vaccine and a commercial trivalent bacterin in a mouse infection model. The results revealed that the Bac-sub vaccine showed no obvious side effects, and induced higher levels of Apx toxin-specific IgG, IgG1, and IgG2a than the commercial vaccines after booster. After a challenge with virulent strains of serovars 1, 5, and 7, the Bac-sub vaccine provided greater protection (91.76%, 100%, and 100%, respectively) than commercial vaccines. Much lower lung bacterial loads (LBLs) and milder lung lesions were observed in the Bac-sub-vaccinated mice than in those vaccinated with the other two vaccines. The protective efficacy of the Bac-sub vaccine was further evaluated in pigs, which showed that vaccinated pigs displayed significantly milder clinical symptoms and lung lesions than the unvaccinated pigs after the challenge. Taken together, Bac-sub is a safe and effective vaccine that could provide high protection against A. pleuropneumoniae infection in both mice and pigs.
Collapse
Affiliation(s)
- Lijun Zhang
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Wentao Luo
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Ruyue Xiong
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Haotian Li
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Zhiming Yao
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Wenxiao Zhuo
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Geng Zou
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Qi Huang
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
- The Cooperative Innovation Center of Sustainable Pig Production, Wuhan, China
- International Research Center for Animal Diseases, Ministry of Science and Technology (China), Wuhan, China
- *Correspondence: Qi Huang
| | - Rui Zhou
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
- The Cooperative Innovation Center of Sustainable Pig Production, Wuhan, China
- International Research Center for Animal Diseases, Ministry of Science and Technology (China), Wuhan, China
- The HZAU-HVSEN Institute, Huazhong Agricultural University, Wuhan, China
- Rui Zhou
| |
Collapse
|
9
|
Sun M, Gao AX, Ledesma-Amaro R, Li A, Wang R, Nie J, Zheng P, Yang Y, Bai Z, Liu X. Hypersecretion of OmlA antigen in Corynebacterium glutamicum through high-throughput based development process. Appl Microbiol Biotechnol 2022; 106:2953-2967. [PMID: 35435456 DOI: 10.1007/s00253-022-11918-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Revised: 03/24/2022] [Accepted: 04/04/2022] [Indexed: 11/26/2022]
Abstract
Outer membrane lipoprotein A (OmlA) is a vaccine antigen against porcine contagious pleuropneumonia (PCP), a disease severely affecting the swine industry. Here, we aimed to systematically potentiate the secretory production of OmlA in Corynebacterium glutamicum (C. glutamicum), a widely used microorganism in the food industry, by establishing a holistic development process based on our high-throughput culture platform. The expression patterns, expression element combinations, medium composition, and induction conditions were comprehensively screened or optimized in microwell plates (MWPs), followed by fermentation parameter optimization in a 4 × 1 L parallel fermentation system (CUBER4). An unprecedented yield of 1.01 g/L OmlA was ultimately achieved in a 5-L bioreactor following the scaling-up strategy of fixed oxygen mass transfer coefficient (kLa), and the produced OmlA antigen showed well-protective immunity against Actinobacillus pleuropneumoniae challenge. This result provides a rapid and reliable pipeline to achieve the hyper-production of OmlA, and possibly other recombinant vaccines, in C. glutamicum. KEY POINTS: • Established a holistic development process and applied it to potentiate the secretion of OmlA. • The secretion of OmlA reached an unprecedented yield of 1.01 g/L. • The recombinant OmlA antigen induced efficient protective immunity.
Collapse
Affiliation(s)
- Manman Sun
- National Engineering Research Center for Cereal Fermentation and Food Biomanufacturing, Jiangnan University, Wuxi, 214112, China
- Key Laboratory of Industrial Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi, 214122, China
- Jiangsu Provincial Research Center for Bioactive Product Processing Technology, Jiangnan University, Wuxi, 214122, China
| | - Alex Xiong Gao
- Division of Life Science, The Hong Kong University of Science and Technology, Hong Kong, China
| | - Rodrigo Ledesma-Amaro
- Department of Bioengineering and Imperial College Centre for Synthetic Biology, Imperial College London, London, SW7 2AZ, UK
| | - An Li
- National Engineering Research Center for Cereal Fermentation and Food Biomanufacturing, Jiangnan University, Wuxi, 214112, China
- Key Laboratory of Industrial Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi, 214122, China
- Jiangsu Provincial Research Center for Bioactive Product Processing Technology, Jiangnan University, Wuxi, 214122, China
| | - Rongbin Wang
- Department of Life Technologies, University of Turku, 20014, Turku, Finland
| | - Jianqi Nie
- National Engineering Research Center for Cereal Fermentation and Food Biomanufacturing, Jiangnan University, Wuxi, 214112, China
- Key Laboratory of Industrial Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi, 214122, China
- Jiangsu Provincial Research Center for Bioactive Product Processing Technology, Jiangnan University, Wuxi, 214122, China
| | - Pei Zheng
- Tecon Biology CO.Ltd, Urumqi, 830000, China
| | - Yankun Yang
- National Engineering Research Center for Cereal Fermentation and Food Biomanufacturing, Jiangnan University, Wuxi, 214112, China
- Key Laboratory of Industrial Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi, 214122, China
- Jiangsu Provincial Research Center for Bioactive Product Processing Technology, Jiangnan University, Wuxi, 214122, China
| | - Zhonghu Bai
- National Engineering Research Center for Cereal Fermentation and Food Biomanufacturing, Jiangnan University, Wuxi, 214112, China
- Key Laboratory of Industrial Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi, 214122, China
- Jiangsu Provincial Research Center for Bioactive Product Processing Technology, Jiangnan University, Wuxi, 214122, China
| | - Xiuxia Liu
- National Engineering Research Center for Cereal Fermentation and Food Biomanufacturing, Jiangnan University, Wuxi, 214112, China.
- Key Laboratory of Industrial Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi, 214122, China.
- Jiangsu Provincial Research Center for Bioactive Product Processing Technology, Jiangnan University, Wuxi, 214122, China.
| |
Collapse
|
10
|
Donà V, Ramette A, Perreten V. Comparative genomics of 26 complete circular genomes of 18 different serotypes of Actinobacillus pleuropneumoniae. Microb Genom 2022; 8. [PMID: 35196217 PMCID: PMC8942016 DOI: 10.1099/mgen.0.000776] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Actinobacillus pleuropneumoniae is a Gram-negative, rod-shaped bacterium of the family Pasteurellaceae causing pig pleuropneumonia associated with great economic losses worldwide. Nineteen serotypes with distinctive lipopolysaccharide (LPS) and capsular (CPS) compositions have been described so far, yet complete circular genomes are publicly available only for the reference strains of serotypes 1, 4 and 5b, and for field strains of serotypes 1, 3, 7 and 8. We aimed to complete this picture by sequencing the reference strains of 17 different serotypes with the MinION sequencer (Oxford Nanopore Technologies, ONT) and on an Illumina HiSeq (Illumina) platform. We also included two field isolates of serotypes 2 and 3 that were PacBio- and MinION-sequenced, respectively. Genome assemblies were performed following two different strategies, i.e. PacBio- or ONT-only de novo assemblies polished with Illumina reads or a hybrid assembly by directly combining ONT and Illumina reads. Both methods proved successful in obtaining accurate circular genomes with comparable qualities. blast-based genome comparisons and core-genome phylogeny based on core genes, SNP typing and multi-locus sequence typing (cgMLST) of the 26 circular genomes indicated well-conserved genomes across the 18 different serotypes, differing mainly in phage insertions, and CPS, LPS and RTX-toxin clusters, which, consistently, encode serotype-specific antigens. We also identified small antibiotic resistance plasmids, and complete subtype I-F and subtype II-C CRISPR-Cas systems. Of note, highly similar clusters encoding all those serotype-specific traits were also found in other pathogenic and commensal Actinobacillus species. Taken together with the presence of transposable elements surrounding these loci, we speculate a dynamic intra- and interspecies exchange of such virulence-related factors by horizontal gene transfer. In conclusion, our comprehensive genomics analysis provides useful information for diagnostic test and vaccine development, but also for whole-genome-based epidemiological studies, as well as for the surveillance of the evolution of antibiotic resistance and virulence genes in A. pleuropneumoniae.
Collapse
Affiliation(s)
- Valentina Donà
- Institute of Veterinary Bacteriology, Vetsuisse Faculty, University of Bern, Bern, Switzerland
| | - Alban Ramette
- Institute for Infectious Diseases, University of Bern, Bern, Switzerland
| | - Vincent Perreten
- Institute of Veterinary Bacteriology, Vetsuisse Faculty, University of Bern, Bern, Switzerland
| |
Collapse
|
11
|
Zhang L, Xie H, Wang H, Ding H, Zhang G, Hu J. Kill Rate and Evaluation of Ex Vivo PK/PD Integration of Cefquinome Against Actinobacillus pleuropneumoniae. Front Vet Sci 2021; 8:751957. [PMID: 34966804 PMCID: PMC8710486 DOI: 10.3389/fvets.2021.751957] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Accepted: 11/12/2021] [Indexed: 11/13/2022] Open
Abstract
We wished to study the detailed and precise antibacterial activity of cefquinome against Actinobacillus pleuropneumoniae (APP) in vitro and ex vivo. We analyzed the relationships between kill rate and cefquinome concentration in broth and between pharmacokinetic/pharmacodynamic (PK/PD) parameters and antibacterial effect in serum and tissue cage fluid (TCF) of piglets. Cefquinome exhibited time-dependent antibacterial activity against APP according to the kill rate. The maximum kill rate was 0.48 log10 CFU/mL/h at the 0-9-h period in broth. In the ex vivo PK/PD study, the maximum concentration (Cmax), time to reach the maximum concentration (Tmax), terminal half-life (T1/2β), and area under the concentration time curve (AUCinfinity) were 5.65 μg/ml, 0.58 h, 2.24 h, and 18.48 μg·h/ml in serum and 1.13 μg/ml, 2.60 h, 12.22 h, and 20.83 μg·h/ml in TCF, respectively. The values of area under the curve during 24 h/minimum inhibitory concentration (AUC24h/MIC) for bacteriostatic, bactericidal, and bacterial eradication effects were 18.94, 246.8, and 1013.23 h in serum and 4.20, 65.81, and 391.35 h in TCF, respectively. Our findings will provide a valuable basis for optimization of dosage regimens when applying cefquinome to treat APP infection.
Collapse
Affiliation(s)
- Longfei Zhang
- Postdoctoral Research Base, Henan Institute of Science and Technology, Xinxiang, China.,College of Animal Science and Veterinary Medicine, Henan Institute of Science and Technology, Xinxiang, China.,Postdoctoral Research Station, Henan Agriculture University, Zhengzhou, China
| | - Hongbing Xie
- College of Animal Science and Veterinary Medicine, Henan Institute of Science and Technology, Xinxiang, China
| | - Hongjuan Wang
- College of Animal Science and Veterinary Medicine, Henan Institute of Science and Technology, Xinxiang, China
| | - Huanzhong Ding
- Guangdong Key Laboratory for Veterinary Drug Development and Safety Evaluation, South China Agricultural University, Guangzhou, China
| | - Gaiping Zhang
- Postdoctoral Research Station, Henan Agriculture University, Zhengzhou, China
| | - Jianhe Hu
- Postdoctoral Research Base, Henan Institute of Science and Technology, Xinxiang, China.,College of Animal Science and Veterinary Medicine, Henan Institute of Science and Technology, Xinxiang, China
| |
Collapse
|
12
|
Litschko C, Budde I, Berger M, Bethe A, Schulze J, Alcala Orozco EA, Mahour R, Goettig P, Führing JI, Rexer T, Gerardy-Schahn R, Schubert M, Fiebig T. Mix-and-Match System for the Enzymatic Synthesis of Enantiopure Glycerol-3-Phosphate-Containing Capsule Polymer Backbones from Actinobacillus pleuropneumoniae, Neisseria meningitidis, and Bibersteinia trehalosi. mBio 2021; 12:e0089721. [PMID: 34076489 PMCID: PMC8262930 DOI: 10.1128/mbio.00897-21] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Accepted: 04/12/2021] [Indexed: 01/19/2023] Open
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.
Collapse
Affiliation(s)
- Christa Litschko
- Institute of Clinical Biochemistry, Hannover Medical School, Hannover, Germany
| | - Insa Budde
- Institute of Clinical Biochemistry, Hannover Medical School, Hannover, Germany
| | - Monika Berger
- Institute of Clinical Biochemistry, Hannover Medical School, Hannover, Germany
| | - Andrea Bethe
- Institute of Clinical Biochemistry, Hannover Medical School, Hannover, Germany
| | - Julia Schulze
- Institute of Clinical Biochemistry, Hannover Medical School, Hannover, Germany
| | - E. Alberto Alcala Orozco
- Max Planck Institute for Dynamics of Complex Technical Systems, Bioprocess Engineering, Magdeburg, Germany
| | - Reza Mahour
- Max Planck Institute for Dynamics of Complex Technical Systems, Bioprocess Engineering, Magdeburg, Germany
| | - Peter Goettig
- Department of Biosciences, University of Salzburg, Salzburg, Austria
| | - Jana Indra Führing
- Institute of Clinical Biochemistry, Hannover Medical School, Hannover, Germany
- Fraunhofer International Consortium for Anti-Infective Research (iCAIR), Hannover, Germany
| | - Thomas Rexer
- Max Planck Institute for Dynamics of Complex Technical Systems, Bioprocess Engineering, Magdeburg, Germany
| | - Rita Gerardy-Schahn
- Institute of Clinical Biochemistry, Hannover Medical School, Hannover, Germany
| | - Mario Schubert
- Department of Biosciences, University of Salzburg, Salzburg, Austria
| | - Timm Fiebig
- Institute of Clinical Biochemistry, Hannover Medical School, Hannover, Germany
| |
Collapse
|
13
|
A Multivalent Vaccine Containing Actinobacillus pleuropneumoniae and Mycoplasma hyopneumoniae Antigens Elicits Strong Immune Responses and Promising Protection in Pigs. JOURNAL OF PURE AND APPLIED MICROBIOLOGY 2021. [DOI: 10.22207/jpam.15.1.11] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Actinobacillus pleuropneumoniae (App) and Mycoplasma hyopneumoniae (Mhp) cause porcine pleuropneumonia and mycoplasmal pneumonia, respectively, and have serious impacts on the swine industry because they retard the growth of pigs. To protect pigs against these diseases, we have developed a multivalent vaccine consisting of App bacterins, APP RTX toxins (Apx toxins), and Mhp bacterin and adhesin protein. This vaccine induced the production of higher levels of antibodies against App and Mhp than the commercial vaccine (Nisseiken Swine APM Inactivated Vaccine). Furthermore, the vaccine efficiently protected pigs against virulent App challenge, showing promise as an efficient vaccine for the prevention of two important respiratory diseases, porcine pleuropneumonia and mycoplasmal pneumonia.
Collapse
|
14
|
Litschko C, Budde I, Berger M, Fiebig T. Exploitation of Capsule Polymerases for Enzymatic Synthesis of Polysaccharide Antigens Used in Glycoconjugate Vaccines. Methods Mol Biol 2021; 2183:313-330. [PMID: 32959251 DOI: 10.1007/978-1-0716-0795-4_16] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
The exploitation of recombinant enzymes for the synthesis of complex carbohydrates is getting increasing attention. Unfortunately, the analysis of the resulting products often requires advanced methods like nuclear magnetic resonance spectroscopy and mass spectrometry. Here, we use the capsule polymerases Cps4B and Cps11D from Actinobacillus pleuropneumoniae serotypes 4 and 11, respectively, as examples for the in vitro synthesis of capsule polymers similar to those used in glycoconjugate vaccine formulations. We demonstrate how substrate turnover in an enzymatic reaction can be analyzed by HPLC-based anion exchange chromatography and provide the protocol for separation and detection of UV-active polymer. Moreover, we describe how UV-inactive polymer can be separated and visualized using polyacrylamide gel electrophoresis followed by combined alcian blue-silver staining.
Collapse
Affiliation(s)
- Christa Litschko
- Institute of Clinical Biochemistry, Hannover Medical School, Hannover, Germany
| | - Insa Budde
- Institute of Clinical Biochemistry, Hannover Medical School, Hannover, Germany
| | - Monika Berger
- Institute of Clinical Biochemistry, Hannover Medical School, Hannover, Germany
| | - Timm Fiebig
- Institute of Clinical Biochemistry, Hannover Medical School, Hannover, Germany.
| |
Collapse
|
15
|
Antenucci F, Ovsepian A, Wrobel A, Winther-Larsen HC, Bojesen AM. Design and Characterization of a Novel Tool for the Antigenic Enrichment of Actinobacillus pleuropneumoniae Outer Membrane. Pathogens 2020; 9:E1014. [PMID: 33276526 PMCID: PMC7761619 DOI: 10.3390/pathogens9121014] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Revised: 11/30/2020] [Accepted: 11/30/2020] [Indexed: 02/06/2023] Open
Abstract
Production and isolation of recombinant proteins are costly and work-intensive processes, especially in immunology when tens or hundreds of potential immunogens need to be purified for testing. Here we propose an alternative method for fast screening of immunogen candidates, based on genetic engineering of recombinant bacterial strains able to express and expose selected antigens on their outer membrane. In Actinobacillus pleuropneumoniae, a Gram-negative porcine pathogen responsible for extensive economic losses worldwide, we identified a conserved general secretion pathway (GSP) domain in the N-terminal part of the outer membrane protein ApfA (ApfA stem: ApfAs). ApfAs was used as an outer membrane anchor, to which potential immunogens can be attached. To enable confirmation of correct positioning, ApfAs, was cloned in combination with the modified acyl carrier protein (ACP) fluorescent tag ACP mini (ACPm) and the putative immunogen VacJ. The chimeric construct was inserted in the pMK-express vector, subsequently transformed into A. pleuropneumoniae for expression. Flow cytometry, fluorescence imaging and mass spectrometry analysis were employed to demonstrate that the outer membrane of the transformed strain was enriched with the chimeric ApfAs-ACPm-VacJ antigen. Our results confirmed correct positioning of the chimeric ApfAs-ACPm-VacJ antigen and supported this system's potential as platform technology enabling antigenic enrichment of the outer membrane of A. pleuropneumoniae.
Collapse
Affiliation(s)
- Fabio Antenucci
- Department of Veterinary and Animal Sciences, University of Copenhagen, Stigbøjlen 4, 1870 Frederiksberg C, Copenhagen, Denmark; (F.A.); (A.O.)
| | - Armen Ovsepian
- Department of Veterinary and Animal Sciences, University of Copenhagen, Stigbøjlen 4, 1870 Frederiksberg C, Copenhagen, Denmark; (F.A.); (A.O.)
| | - Agnieszka Wrobel
- Section of Pharmaceutical Biosciences, Centre of Integrative Microbial Evolution, Department of Pharmacy, University of Oslo, Sem Sælandsvei 3, 0316 Oslo, Norway; (A.W.); (H.C.W.-L.)
| | - Hanne Cecilie Winther-Larsen
- Section of Pharmaceutical Biosciences, Centre of Integrative Microbial Evolution, Department of Pharmacy, University of Oslo, Sem Sælandsvei 3, 0316 Oslo, Norway; (A.W.); (H.C.W.-L.)
| | - Anders Miki Bojesen
- Department of Veterinary and Animal Sciences, University of Copenhagen, Stigbøjlen 4, 1870 Frederiksberg C, Copenhagen, Denmark; (F.A.); (A.O.)
| |
Collapse
|
16
|
Detection of naturally aerosolized Actinobacillus pleuropneumoniae on pig farms by cyclonic air sampling and qPCR. Vet Microbiol 2020; 250:108856. [PMID: 33007607 DOI: 10.1016/j.vetmic.2020.108856] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2019] [Accepted: 09/04/2020] [Indexed: 11/20/2022]
Abstract
Respiratory infections caused by Actinobacillus pleuropneumoniae have a large impact on commercial pig farms globally. As current vaccines have limited efficacy, animal care and air hygiene are critical for disease control. Here we used a Coriolis μ cyclonic air sampler and an A. pleuropneumoniae-specific apxIV gene qPCR assay to detect the organism. Air samples were collected into a liquid medium by the Coriolis μ sampler for A. pleuropneumoniae detection by plate culture and qPCR assay. The method was validated by comparing the Coriolis μ sampler and a plate impactor (Millipore Air-T) in a specially designed aerosolization chamber. Two commercial farms, housing pigs between 3 and 21 weeks of age, were tested. On one farm, A. pleuropneumoniae was detected in low numbers (1000 organisms/m3 air) by qPCR, but not by culture, from sheds containing 8, 12, 16, and 18 weeks-old pigs. To our knowledge this is the first successful detection of naturally aerosolised A. pleuropneumoniae in commercial farms with the Coriolis μ air sampler, potentially allowing the identification of sub-clinically infected populations of pigs in the field.
Collapse
|
17
|
Dao HT, Truong QL, Do VT, Hahn TW. Construction and immunization with double mutant Δ apxIBD Δ pnp forms of Actinobacillus pleuropneumoniae serotypes 1 and 5. J Vet Sci 2020; 21:e20. [PMID: 32233129 PMCID: PMC7113565 DOI: 10.4142/jvs.2020.21.e20] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2019] [Revised: 12/06/2019] [Accepted: 12/06/2019] [Indexed: 12/14/2022] Open
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.
Collapse
Affiliation(s)
- Hoai Thu Dao
- College of Veterinary Medicine and Institute of Veterinary Science, Kangwon National University, Chuncheon 24341, Korea
| | - Quang Lam Truong
- College of Veterinary Medicine and Institute of Veterinary Science, Kangwon National University, Chuncheon 24341, Korea.,Key Laboratory of Veterinary Medicine, Faculty of Veterinary Medicine, Vietnam National University of Agriculture, Hanoi 100000, Vietnam
| | - Van Tan Do
- College of Veterinary Medicine and Institute of Veterinary Science, Kangwon National University, Chuncheon 24341, Korea
| | - Tae Wook Hahn
- College of Veterinary Medicine and Institute of Veterinary Science, Kangwon National University, Chuncheon 24341, Korea.
| |
Collapse
|
18
|
Dao HT, Do VT, Truong QL, Hahn TW. Enhancement of Apx Toxin Production in Actinobacillus pleuropneumoniae Serotypes 1, 2, and 5 by Optimizing Culture Condition. J Microbiol Biotechnol 2020; 30:1037-1043. [PMID: 32238774 PMCID: PMC9745662 DOI: 10.4014/jmb.1912.12042] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2019] [Accepted: 03/24/2020] [Indexed: 12/15/2022]
Abstract
Actinobacillus pleuropneumoniae (APP) is a causative agent of porcine pleuropneumonia. Therefore, the development of an effective vaccine for APP is necessary. Here, we optimized the culture medium and conditions to enhance the production yields of Apx toxins in APP serotype 1, 2, and 5 cultures. The use of Mycoplasma Broth Base (PPLO) medium improved both the quantity and quality of the harvested Apx toxins compared with Columbia Broth medium. Calcium chloride (CaCl2) was first demonstrated as a stimulation factor for the production of Apx toxins in APP serotype 2 cultures. Cultivation of APP serotype 2 in PPLO medium supplemented with 10 μg/ml of nicotinamide adenine dinucleotide (NAD) and 20 mM CaCl2 yielded the highest levels of Apx toxins. These findings suggest that the optimization of the culture medium and conditions increases the concentration of Apx toxins in the supernatants of APP serotype 1, 2, and 5 cultures and may be applied for the development of vaccines against APP infection.
Collapse
Affiliation(s)
- Hoai Thu Dao
- College of Veterinary Medicine and Institute of Veterinary Science, Kangwon National University, Chuncheon 24341, Republic of Korea
| | - Van Tan Do
- College of Veterinary Medicine and Institute of Veterinary Science, Kangwon National University, Chuncheon 24341, Republic of Korea
| | - Quang Lam Truong
- Key Laboratory of Veterinary Biotechnology, Faculty of Veterinary Medicine, Vietnam National University of Agriculture, Hanoi, Vietnam
| | - Tae-Wook Hahn
- College of Veterinary Medicine and Institute of Veterinary Science, Kangwon National University, Chuncheon 24341, Republic of Korea,Innovac Co., Kangwon National University, 1 Kangwondaehak-gil, Chuncheon 24341, Republic of Korea,Corresponding author Phone: +82-33-2508671 Fax: +82-33-2595625 E-mail:
| |
Collapse
|
19
|
Xiao J, Liu J, Bao C, Zhu R, Gu J, Sun C, Feng X, Du C, Han W, Li Y, Lei L. Recombinant tandem epitope vaccination provides cross protection against Actinobacillus pleuropneumoniae challenge in mice. AMB Express 2020; 10:123. [PMID: 32642871 PMCID: PMC7341470 DOI: 10.1186/s13568-020-01051-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Accepted: 06/15/2020] [Indexed: 11/10/2022] Open
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.
Collapse
|
20
|
Budde I, Litschko C, Führing JI, Gerardy-Schahn R, Schubert M, Fiebig T. An enzyme-based protocol for cell-free synthesis of nature-identical capsular oligosaccharides from Actinobacillus pleuropneumoniae serotype 1. J Biol Chem 2020; 295:5771-5784. [PMID: 32152227 PMCID: PMC7186170 DOI: 10.1074/jbc.ra120.012961] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Revised: 03/09/2020] [Indexed: 11/06/2022] Open
Abstract
Actinobacillus pleuropneumoniae (App) is the etiological agent of acute porcine pneumonia and responsible for severe economic losses worldwide. The capsule polymer of App serotype 1 (App1) consists of [4)-GlcNAc-β(1,6)-Gal-α-1-(PO4-] repeating units that are O-acetylated at O-6 of the GlcNAc. It is a major virulence factor and was used in previous studies in the successful generation of an experimental glycoconjugate vaccine. However, the application of glycoconjugate vaccines in the animal health sector is limited, presumably because of the high costs associated with harvesting the polymer from pathogen culture. Consequently, here we exploited the capsule polymerase Cps1B of App1 as an in vitro synthesis tool and an alternative for capsule polymer provision. Cps1B consists of two catalytic domains, as well as a domain rich in tetratricopeptide repeats (TPRs). We compared the elongation mechanism of Cps1B with that of a ΔTPR truncation (Cps1B-ΔTPR). Interestingly, the product profiles displayed by Cps1B suggested processive elongation of the nascent polymer, whereas Cps1B-ΔTPR appeared to work in a more distributive manner. The dispersity of the synthesized products could be reduced by generating single-action transferases and immobilizing them on individual columns, separating the two catalytic activities. Furthermore, we identified the O-acetyltransferase Cps1D of App1 and used it to modify the polymers produced by Cps1B. Two-dimensional NMR analyses of the products revealed O-acetylation levels identical to those of polymer harvested from App1 culture supernatants. In conclusion, we have established a protocol for the pathogen-free in vitro synthesis of tailored, nature-identical App1 capsule polymers.
Collapse
Affiliation(s)
- Insa Budde
- Institute of Clinical Biochemistry, Hannover Medical School, Carl-Neuberg Strasse 1, 30625 Hannover, Germany
| | - Christa Litschko
- Institute of Clinical Biochemistry, Hannover Medical School, Carl-Neuberg Strasse 1, 30625 Hannover, Germany
| | - Jana I Führing
- Institute of Clinical Biochemistry, Hannover Medical School, Carl-Neuberg Strasse 1, 30625 Hannover, Germany; Fraunhofer International Consortium for Anti-Infective Research (iCAIR), 30625 Hannover, Germany
| | - Rita Gerardy-Schahn
- Institute of Clinical Biochemistry, Hannover Medical School, Carl-Neuberg Strasse 1, 30625 Hannover, Germany; Fraunhofer International Consortium for Anti-Infective Research (iCAIR), 30625 Hannover, Germany
| | - Mario Schubert
- Department of Biosciences, University of Salzburg, 5020 Salzburg, Austria
| | - Timm Fiebig
- Institute of Clinical Biochemistry, Hannover Medical School, Carl-Neuberg Strasse 1, 30625 Hannover, Germany.
| |
Collapse
|
21
|
Oba P, Wieland B, Mwiine FN, Erume J, Gertzell E, Jacobson M, Dione MM. Status and gaps of research on respiratory disease pathogens of swine in Africa. Porcine Health Manag 2020; 6:5. [PMID: 32257367 PMCID: PMC7066813 DOI: 10.1186/s40813-020-0144-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2019] [Accepted: 01/22/2020] [Indexed: 11/10/2022] Open
Abstract
Over the last two decades, the pig population in Africa has grown rapidly, reflecting the increased adoption of pig production as an important economic activity. Of all species, pigs are likely to constitute a greater share of the growth in the livestock subsector. However, constraints such as respiratory infectious diseases cause significant economic losses to the pig industry worldwide. Compared to industrialized countries, the occurrence and impacts of respiratory diseases on pig production in Africa is under-documented. Hence, knowledge on prevalence and incidence of economically important swine respiratory pathogens in pigs in Africa is necessary to guide interventions for prevention and control. The purpose of this review was to document the current status of research on five important respiratory pathogens of swine in Africa to inform future research and interventions. The pathogens included were porcine reproductive and respiratory syndrome virus (PPRSv), porcine circovirus 2 (PCV2), Mycoplasma hyopneumoniae (M. hyopneumoniae), Actinobacillus pleuropneumoniae (APP) and swine influenza A viruses (IAV). For this review, published articles were obtained using Harzing's Publish or Perish software tool from GoogleScholar. Articles were also sourced from PubMed, ScienceDirect, FAO and OIE websites. The terms used for the search were Africa, swine or porcine, respiratory pathogens, M. hyopneumoniae, APP, PCV2, PPRSv, IAV, prevention and control. In all, 146 articles found were considered relevant, and upon further screening, only 85 articles were retained for the review. The search was limited to studies published from 2000 to 2019. Of all the studies that documented occurrence of the five respiratory pathogens, most were on IAV (48.4%, n = 15), followed by PCV2 (25.8%, n = 8), PPRSv (19.4%, n = 6), while only one study (3.2%, n = 1) reported APP and M. hyopneumoniae. This review highlights knowledge and information gaps on epidemiologic aspects as well as economic impacts of the various pathogens reported in swine in Africa, which calls for further studies.
Collapse
Affiliation(s)
- P Oba
- International Livestock Research Institute, P. O. Box 24384, Kampala, Uganda.,2College of Veterinary Medicine, Animal Resources and Biosecurity, Makerere University, P. O. Box 7062, Kampala, Uganda.,National Agricultural Research Organization, Abi Zonal Agricultural Research and Development Institute (Abi ZARDI), P. O. Box 219, Arua, Uganda
| | - B Wieland
- 4International Livestock Research Institute, P.O. Box 5689, Addis Ababa, Ethiopia
| | - F N Mwiine
- 2College of Veterinary Medicine, Animal Resources and Biosecurity, Makerere University, P. O. Box 7062, Kampala, Uganda
| | - J Erume
- 2College of Veterinary Medicine, Animal Resources and Biosecurity, Makerere University, P. O. Box 7062, Kampala, Uganda
| | - E Gertzell
- 5Department of Clinical Sciences, Swedish University of Agricultural Sciences (SLU), Box 7054, 750 07 Uppsala, Sweden
| | - M Jacobson
- 5Department of Clinical Sciences, Swedish University of Agricultural Sciences (SLU), Box 7054, 750 07 Uppsala, Sweden
| | - M M Dione
- International Livestock Research Institute, P. O. Box 24384, Kampala, Uganda
| |
Collapse
|
22
|
Park BS, Lee N. A bivalent fusion vaccine composed of recombinant Apx proteins shows strong protection against Actinobacillus pleuroneumoniae serovar 1 and 2 in a mouse model. Pathog Dis 2020; 77:5426212. [PMID: 30939190 DOI: 10.1093/femspd/ftz020] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2018] [Accepted: 03/31/2019] [Indexed: 11/12/2022] Open
Abstract
Actinobacillus pleuropneumonia (APP) causes porcine pleuropneumoniae, resulting in severe economic losses in the swine industry. Since there are diverse serotypes of APP, it is necessary for vaccines to induce cross-protection. In this report, we developed a bivalent fusion vaccine, the L vaccine composed of ApxIA and ApxIIA fragments. According to the experimental results of the L vaccine, recombinant protein specific-IgG antibody level increased significantly as well as Apx toxin specific-IgG antibody, suggesting toxin-neutralizing effect. Also, the production of both IgG1 and IgG2a indicates this fusion vaccine induces Th1 and Th2 immune reactions. In addition, lymphocytes were proliferated and immune related-cytokines of TNF-α, IL-12, IFN-γ and IL-5 were detected in the serum after the vaccination. The L vaccine showed a perfect cross-protection against APP serovar 1 and 2 that each secrete different Apx exotoxins. These findings reveal that the fusion L vaccine induces specific humoral and cellular immunity, leading to a perfect cross-protection against A. pleuropneumoniae infections in a murine model.
Collapse
Affiliation(s)
- Byung-Sun Park
- Technology Institute, KBNP, Inc., Anyang, Gyeonggi, South Korea
| | - Nakhyung Lee
- Technology Institute, KBNP, Inc., Anyang, Gyeonggi, South Korea
| |
Collapse
|
23
|
Cao Y, Gao L, Zhang L, Zhou L, Yang J, Deng L, Zhao J, Qi C, Liu J. Genome-wide screening of lipoproteins in Actinobacillus pleuropneumoniae identifies three antigens that confer protection against virulent challenge. Sci Rep 2020; 10:2343. [PMID: 32047221 PMCID: PMC7012816 DOI: 10.1038/s41598-020-58968-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2019] [Accepted: 01/23/2020] [Indexed: 11/24/2022] Open
Abstract
Actinobacillus pleuropneumoniae is an important veterinary pathogen that causes porcine pleuropneumonia. Lipoproteins of bacterial pathogens play pleiotropic roles in the infection process. In addition, many bacterial lipoproteins are antigenic and immunoprotective. Therefore, characterization of lipoproteins is a promising strategy for identification of novel vaccine candidates or diagnostic markers. We cloned 58 lipoproteins from A. pleuropneumoniae JL03 (serovar 3) and expressed them in Escherichia coli. Five proteins with strong positive signals in western blotting analysis were used to immunize mice. These proteins elicited significant antibody responses, and three of them (APJL_0922, APJL_1380 and APJL_1976) generated efficient immunoprotection in mice against lethal heterologous challenge with A. pleuropneumoniae 4074 (serovar 1), both in the active and passive immunization assays. Then immunogenicity of these three lipoproteins (APJL_0922, APJL_1380 and APJL_1976) were further tested in pigs. Results showed that these proteins elicited considerable humoral immune responses and effective protective immunity against virulent A. pleuropneumoniae challenge. Our findings suggest that these three novel lipoproteins could be potential subunit vaccine candidates.
Collapse
Affiliation(s)
- Yurou Cao
- Hubei Key Laboratory of Genetic Regulation and Integrative Biology, College of Life Sciences, Central China Normal University, Wuhan, Hubei, 430079, China
| | - Lulu Gao
- Hubei Key Laboratory of Genetic Regulation and Integrative Biology, College of Life Sciences, Central China Normal University, Wuhan, Hubei, 430079, China
| | - Li Zhang
- Hubei Key Laboratory of Genetic Regulation and Integrative Biology, College of Life Sciences, Central China Normal University, Wuhan, Hubei, 430079, China
| | - Lixiang Zhou
- Hubei Key Laboratory of Genetic Regulation and Integrative Biology, College of Life Sciences, Central China Normal University, Wuhan, Hubei, 430079, China
| | - Jihong Yang
- Hubei Key Laboratory of Genetic Regulation and Integrative Biology, College of Life Sciences, Central China Normal University, Wuhan, Hubei, 430079, China
| | - Lingfu Deng
- Hubei Key Laboratory of Genetic Regulation and Integrative Biology, College of Life Sciences, Central China Normal University, Wuhan, Hubei, 430079, China
| | - Jin Zhao
- Hubei Key Laboratory of Genetic Regulation and Integrative Biology, College of Life Sciences, Central China Normal University, Wuhan, Hubei, 430079, China
| | - Chao Qi
- Hubei Key Laboratory of Genetic Regulation and Integrative Biology, College of Life Sciences, Central China Normal University, Wuhan, Hubei, 430079, China.
| | - Jinlin Liu
- Hubei Key Laboratory of Genetic Regulation and Integrative Biology, College of Life Sciences, Central China Normal University, Wuhan, Hubei, 430079, China.
| |
Collapse
|
24
|
Antenucci F, Magnowska Z, Nimtz M, Roesch C, Jänsch L, Bojesen AM. Immunoproteomic characterization of outer membrane vesicles from hyper-vesiculating Actinobacillus pleuropneumoniae. Vet Microbiol 2019; 235:188-194. [PMID: 31383301 DOI: 10.1016/j.vetmic.2019.07.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2019] [Revised: 07/02/2019] [Accepted: 07/02/2019] [Indexed: 11/16/2022]
Abstract
Outer membrane vesicles (OMVs) are produced and secreted virtually by every known Gram-negative bacterium. Despite their non-live nature, they share antigenic characteristics with the bacteria they originate from. This, together with their relative ease of purification, casts the OMVs as a very promising and flexible tool in both human and veterinary vaccinology. The aim of the current work was to get an insight into the antigenic pattern of OMVs from the pig pathogen Actinobacillus pleuropneumoniae in the context of vaccine development. Accordingly, we designed a protocol combining 2D Western Blotting and mass spectrometric identification to robustly characterize the antigenic protein pattern of the vesicles. Our analysis revealed that A. pleuropneumoniae OMVs carry several immunoreactive virulence factors. Some of these proteins, LpoA, OsmY and MIDG2331_02184, have never previously been documented as antigenic in A. pleuropneumoniae or other pathogenic bacteria. Additionally, we showed that despite their relative abundance, proteins such as FrpB and DegQ do not contribute to the antigenic profile of A. pleuropneumoniae OMVs.
Collapse
Affiliation(s)
- Fabio Antenucci
- Department of Veterinary and Animal Sciences, Stigbøjlen 4, 1870 Frb. C., 1-20, Building: 301, University of Copenhagen, Copenhagen, Denmark.
| | - Zofia Magnowska
- Department of Veterinary and Animal Sciences, Stigbøjlen 4, 1870 Frb. C., 1-20, Building: 301, University of Copenhagen, Copenhagen, Denmark.
| | - Manfred Nimtz
- Helmholtz Centre for Infection Research, Inhoffenstraße 7, 38124, Braunschweig, Germany.
| | - Camille Roesch
- Izon Science Ltd, Batiment Laennec, 60 Avenue Rockefeller, 69008, Lyon, France.
| | - Lothar Jänsch
- Helmholtz Centre for Infection Research, Inhoffenstraße 7, 38124, Braunschweig, Germany.
| | - Anders Miki Bojesen
- Department of Veterinary and Animal Sciences, Stigbøjlen 4, 1870 Frb. C., 1-20, Building: 301, University of Copenhagen, Copenhagen, Denmark.
| |
Collapse
|
25
|
Bercier P, Gottschalk M, Grenier D. Effects of Actinobacillus pleuropneumoniae on barrier function and inflammatory response of pig tracheal epithelial cells. Pathog Dis 2019; 77:5159464. [PMID: 30395241 DOI: 10.1093/femspd/fty079] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2018] [Accepted: 11/01/2018] [Indexed: 12/11/2022] Open
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.
Collapse
Affiliation(s)
- Philippe Bercier
- Groupe de Recherche en Écologie Buccale (GREB), Faculté de médecine dentaire, Université Laval, Quebec City, Quebec, GIV 0A6, Canada
| | - Marcelo Gottschalk
- Groupe de Recherche sur les Maladies Infectieuses en Production Animale (GREMIP), Faculté de médecine vétérinaire, Université de Montréal, Saint-Hyacinthe, Quebec, J2S 2M2, Canada.,Centre de Recherche en Infectiologie Porcine et Avicole (CRIPA), Fonds de Recherche du Québec - Nature et Technologies (FRQNT), Saint-Hyacinthe, Quebec, J2S 2M2, Canada
| | - Daniel Grenier
- Groupe de Recherche en Écologie Buccale (GREB), Faculté de médecine dentaire, Université Laval, Quebec City, Quebec, GIV 0A6, Canada.,Centre de Recherche en Infectiologie Porcine et Avicole (CRIPA), Fonds de Recherche du Québec - Nature et Technologies (FRQNT), Saint-Hyacinthe, Quebec, J2S 2M2, Canada
| |
Collapse
|
26
|
Jung M, Won H, Shin MK, Oh MW, Shim S, Yoon I, Yoo HS. Development of Actinobacillus pleuropneumoniae ApxI, ApxII, and ApxIII-specific ELISA methods for evaluation of vaccine efficiency. J Vet Sci 2019; 20:e2. [PMID: 30944525 PMCID: PMC6441810 DOI: 10.4142/jvs.2019.20.e2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2018] [Revised: 11/03/2018] [Accepted: 12/16/2018] [Indexed: 11/20/2022] Open
Abstract
Among various vaccines against Actinobacillus pleuropneumoniae, subunit vaccines using recombinant proteins of ApxI, ApxII, and ApxIII as vaccine antigens have shown good efficacy in terms of safety and protection. Therefore, subunit vaccines are being applied worldwide and the development of new subunit vaccines is actively being conducted. To evaluate the efficacy of the subunit vaccines, it is important to measure immune responses to each Apx toxin separately. However, the cross-reactivity of antibodies makes it difficult to measure specific immune reactivity to each toxin. In the present study, specific antigen regions among the toxins were identified and cloned to solve this problem. The antigenicity of each recombinant protein was demonstrated by Western blot. Using the recombinant proteins, we developed enzyme-linked immunosorbent assay (ELISA) methods that can detect specific immune responses to each Apx toxin in laboratory guinea pigs. We suggest that the ELISA method developed in this study can be an important tool in the evaluation of vaccine efficiency and vaccine development.
Collapse
Affiliation(s)
- Myunghwan Jung
- Department of Microbiology, Research Institute of Life Sciences, Gyeongsang National University School of Medicine, Jinju 52727, Korea
| | - Hokeun Won
- Department of Infectious Diseases, College of Veterinary Medicine, Seoul National University, Seoul 08826, Korea.,Choong Ang Vaccine Laboratories Co., Ltd., Daejeon 34055, Korea
| | - Min-Kyoung Shin
- Department of Microbiology, Research Institute of Life Sciences, Gyeongsang National University School of Medicine, Jinju 52727, Korea
| | - Myung Whan Oh
- Department of Infectious Diseases, College of Veterinary Medicine, Seoul National University, Seoul 08826, Korea
| | - Soojin Shim
- Department of Infectious Diseases, College of Veterinary Medicine, Seoul National University, Seoul 08826, Korea
| | - Injoong Yoon
- Choong Ang Vaccine Laboratories Co., Ltd., Daejeon 34055, Korea
| | - Han Sang Yoo
- Department of Infectious Diseases, College of Veterinary Medicine, Seoul National University, Seoul 08826, Korea
| |
Collapse
|
27
|
Passmore IJ, Andrejeva A, Wren BW, Cuccui J. Cytoplasmic glycoengineering of Apx toxin fragments in the development of Actinobacillus pleuropneumoniae glycoconjugate vaccines. BMC Vet Res 2019; 15:6. [PMID: 30606265 PMCID: PMC6318927 DOI: 10.1186/s12917-018-1751-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2018] [Accepted: 12/17/2018] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Actinobacillus pleuropneumoniae is the causative agent of porcine pleuropneumonia and represents a major burden to the livestock industry. Virulence can largely be attributed to the secretion of a series of haemolytic toxins, which are highly immunogenic. A. pleuropneumoniae also encodes a cytoplasmic N-glycosylation system, which involves the modification of high molecular weight adhesins with glucose residues. Central to this process is the soluble N-glycosyl transferase, ngt, which is encoded in an operon with a subsequent glycosyl transferase, agt. Plasmid-borne recombinant expression of these genes in E. coli results in the production of a glucose polymer on peptides containing the appropriate acceptor sequon, NX(S/T). However to date, there is little evidence to suggest that such a glucose polymer is formed on its target peptides in A. pleuropneumoniae. Both the toxins and glycosylation system represent potential targets for the basis of a vaccine against A. pleuropneumoniae infection. RESULTS In this study, we developed cytoplasmic glycoengineering to construct glycoconjugate vaccine candidates composed of soluble toxin fragments modified by glucose. We transferred ngt and agt to the chromosome of Escherichia coli in order to generate a native-like operon for glycoengineering. A single chromosomal copy of ngt and agt resulted in the glucosylation of toxin fragments by a short glycan, rather than a polymer. CONCLUSIONS A vaccine candidate that combines toxin fragment with a conserved glycan offers a novel approach to generating epitopes important for both colonisation and disease progression.
Collapse
Affiliation(s)
- Ian J Passmore
- Department of Pathogen Molecular Biology, London School of Hygiene and Tropical Medicine, Keppel Street, London, WC1E 7HT, UK
| | - Anna Andrejeva
- Cambridge Centre for Proteomics, Department of Biochemistry, University of Cambridge, Tennis Court Road, Cambridge, CB2 1QR, UK
| | - Brendan W Wren
- Department of Pathogen Molecular Biology, London School of Hygiene and Tropical Medicine, Keppel Street, London, WC1E 7HT, UK
| | - Jon Cuccui
- Department of Pathogen Molecular Biology, London School of Hygiene and Tropical Medicine, Keppel Street, London, WC1E 7HT, UK.
| |
Collapse
|
28
|
Chen Y, Yang F, Yang J, Hou Y, He L, Hu H, Lv F. Aluminum (oxy) Hydroxide Nanorods Activate an Early Immune Response in Pseudomonas aeruginosa Vaccine. ACS APPLIED MATERIALS & INTERFACES 2018; 10:43533-43542. [PMID: 30480997 DOI: 10.1021/acsami.8b18164] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
Bacterial vaccines have been widely used to prevent infectious diseases, especially in veterinary medicine. Although there are many reports on bacterin adjuvants, only a few contain innovations in bacterin adjuvants. Taking this into consideration, in this study we designed and synthesized a new aluminum (oxy) hydroxide (AlOOH) nanorod (Al-NR) with a diameter of 200 ± 80 nm and a length of 1.1 ± 0.6 μm. Using whole- Pseudomonas aeruginosa PAO1 as antigens, we showed that the bacterial antigens of P. aeruginosa PAO1 adsorbed on the Al-NRs induced a quick and stronger antigen-specific antibody response than those of the other control groups, especially in the early stage of immunization. Furthermore, the level of antigen-specific IgG was approximately 4-fold higher than that of the no adjuvant group and 2.5-fold higher than those of other adjuvant groups in the first week after the initial immunization. The potent adjuvant activity of the Al-NRs was attributed to the rapid presentation of antigen adsorbed on them by APCs. Additionally, Al-NRs induced a milder local inflammation than the other adjuvants. In short, we confirmed that Al-NRs, enhancing both humoral and cellular immune responses, are a potentially promising vaccine adjuvant delivery system for inhibiting the whole- Pseudomonas aeruginosa infection.
Collapse
Affiliation(s)
- Yingli Chen
- College of Bioengineering, "111 Project" Laboratory of Biomechanics and Tissue Repair Engineering, Key Laboratory of Biorheological Science and Technology , Chongqing University , Chongqing 400030 , P.R. China
| | - Feng Yang
- College of Bioengineering, "111 Project" Laboratory of Biomechanics and Tissue Repair Engineering, Key Laboratory of Biorheological Science and Technology , Chongqing University , Chongqing 400030 , P.R. China
| | - Jun Yang
- College of Bioengineering, "111 Project" Laboratory of Biomechanics and Tissue Repair Engineering, Key Laboratory of Biorheological Science and Technology , Chongqing University , Chongqing 400030 , P.R. China
| | - Yali Hou
- College of Bioengineering, "111 Project" Laboratory of Biomechanics and Tissue Repair Engineering, Key Laboratory of Biorheological Science and Technology , Chongqing University , Chongqing 400030 , P.R. China
| | - Leilei He
- College of Bioengineering, "111 Project" Laboratory of Biomechanics and Tissue Repair Engineering, Key Laboratory of Biorheological Science and Technology , Chongqing University , Chongqing 400030 , P.R. China
| | - Houxiang Hu
- Department of Cardiology , Affiliated Hospital of North Sichuan Medical College , Nanchong 637000 , Sichuan , P.R. China
| | - Fenglin Lv
- College of Bioengineering, "111 Project" Laboratory of Biomechanics and Tissue Repair Engineering, Key Laboratory of Biorheological Science and Technology , Chongqing University , Chongqing 400030 , P.R. China
| |
Collapse
|
29
|
Shinkai H, Terada K, Toki D, Tohno M, Uenishi H. Q969R polymorphism in NLRP3 is associated with immune responses to vaccination against bacterial infections in pigs. Anim Sci J 2018; 89:1043-1050. [DOI: 10.1111/asj.13020] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2017] [Accepted: 01/11/2018] [Indexed: 11/28/2022]
Affiliation(s)
- Hiroki Shinkai
- Animal Bioregulation Unit; Division of Animal Sciences; Institute of Agrobiological Sciences; National Agriculture and Food Research Organization (NARO); Tsukuba Ibaraki Japan
| | - Kei Terada
- Shizuoka Swine and Poultry Experiment Center; Kikugawa Shizuoka Japan
| | - Daisuke Toki
- Animal Research Division; Institute of Japan Association for Techno-innovation in Agriculture, Forestry and Fisheries (JATAFF); Tsukuba Ibaraki Japan
| | - Masanori Tohno
- Feed Preparation Unit; Division of Animal Feeding and Management Research; Institute of Livestock and Grassland Science; National Agriculture and Food Research Organization (NARO); Nasushiobara Tochigi Japan
| | - Hirohide Uenishi
- Animal Bioregulation Unit; Division of Animal Sciences; Institute of Agrobiological Sciences; National Agriculture and Food Research Organization (NARO); Tsukuba Ibaraki Japan
| |
Collapse
|
30
|
Loera-Muro A, Angulo C. New trends in innovative vaccine development against Actinobacillus pleuropneumoniae. Vet Microbiol 2018; 217:66-75. [DOI: 10.1016/j.vetmic.2018.02.028] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2017] [Revised: 02/27/2018] [Accepted: 02/27/2018] [Indexed: 01/08/2023]
|
31
|
Yuan F, Liu J, You W, Bei W, Wang C, Zhao J, Tian Y, Liu S. Generation, safety and immunogenicity of an Actinobacillus pleuropneumoniae quintuple deletion mutant SLW07 (ΔapxICΔapxIICΔorf1ΔcpxARΔarcA). Vaccine 2018; 36:1830-1836. [PMID: 29486946 DOI: 10.1016/j.vaccine.2018.02.083] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2017] [Revised: 02/05/2018] [Accepted: 02/19/2018] [Indexed: 10/18/2022]
Abstract
We inactivated a virulence determinant, ArcA, in an Actinobacillus pleuropneumoniae quadruple deletion mutant SLW06 (ΔapxICΔapxIICΔorf1ΔcpxAR, serovar 1), and a quintuple deletion mutant SLW07 was generated. SLW07 showed decreased adherence to and invasion of host cells, compared to its parent strain SLW06. SLW07 was more sensitive in RAW264.7 macrophage-mediated phagocytosis and clearance. SLW07 was less virulent in mice. An immunization assay indicated that both SLW07 and SLW06 preferentially stimulated T helper cell type 2 response in mice. Live vaccines induced the production of interleukin-6 and tumor necrosis factor-α by splenic lymphocytes. Furthermore, the protective immunity of SLW07 was not affected after ArcA mutation. Immunization with SLW07 could provide a complete protection following virulent A. pleuropneumoniae challenge in mice. Our results suggest that SLW07 is a promising live vaccine candidate, which is further attenuated from and shares similar protective efficacy with its quadruple deletion parent SLW06.
Collapse
Affiliation(s)
- Fangyan Yuan
- Institute of Animal Husbandry and Veterinary Sciences, Hubei Academy of Agricultural Sciences, Key Laboratory of Prevention and Control Agents for Animal Bacteriosis (Ministry of Agriculture), Hubei Key Laboratory of Animal Embryo and Molecular Breeding, Wuhan, Hubei 430064, China
| | - Jinlin Liu
- College of Life Sciences, Central China Normal University, Wuhan, Hubei 430079, China
| | - Wujin You
- State Key Laboratory of Agricultural Microbiology, The Cooperative Innovation Center for Sustainable Pig Production, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei 430070, China
| | - Weicheng Bei
- State Key Laboratory of Agricultural Microbiology, The Cooperative Innovation Center for Sustainable Pig Production, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei 430070, China
| | - Chunlai Wang
- State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin 150001, China
| | - Jin Zhao
- College of Life Sciences, Central China Normal University, Wuhan, Hubei 430079, China
| | - Yiongxiang Tian
- Institute of Animal Husbandry and Veterinary Sciences, Hubei Academy of Agricultural Sciences, Key Laboratory of Prevention and Control Agents for Animal Bacteriosis (Ministry of Agriculture), Hubei Key Laboratory of Animal Embryo and Molecular Breeding, Wuhan, Hubei 430064, China.
| | - Siguo Liu
- State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin 150001, China.
| |
Collapse
|
32
|
Wu HC, Yeh PH, Hsueh KJ, Yang WJ, Chu CY. Recombinant ApxIV protein enhances protective efficacy against Actinobacillus pleuropneumoniae in mice and pigs. J Appl Microbiol 2018; 124:1366-1376. [PMID: 29431246 DOI: 10.1111/jam.13726] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2017] [Revised: 01/23/2018] [Accepted: 01/31/2018] [Indexed: 11/28/2022]
Abstract
AIMS Available bacterins, commercial or autogenous, for Actinobacillus pleuropneumoniae disease control have, thus far, shown debatable protective efficacy and only in homologous challenges. Our study sought to determine whether the addition of reombinant protein ApxIV to the multicomponent vaccine could enhance protection against homologous and heterologous challenge of A. pleuropneumoniae. METHODS AND RESULTS The virulence of ApxI, ApxII, ApxIV and OMP were cloned and expressed using a prokaryotic system; these recombinant proteins were combined with inactivated A. pleuropneumoniae serovar 1 to formulate different multicomponent vaccines. Immune response and protective efficacy of the vaccines were evaluated in mice and pigs. A protection rate of 67% was observed against heterologous challenge in mice vaccinated with the rApxIV formulation. Piglets vaccinated with vaccine containing ApxIV produced significantly higher antibody titre and provided complete protection and reduced gross lesions by 67% when compared with the nonimmunized group after homologous challenge. Additionally, flow cytometry analysis showed significant cellular immune response. CONCLUSIONS The results of our vaccination experiments revealed that a combination of inactivated bacteria and the recombinant antigens rApxI, rApxII, rApxIV and rOMP can provide effective protection against heterologous A. pleuropneumoniae challenge. SIGNIFICANCE AND IMPACT OF THE STUDY The addition of ApxIV to the multicomponent vaccine could enhance homologous and heterologous protection in mice and pigs, respectively, against challenge by A. pleuropneumoniae.
Collapse
Affiliation(s)
- H-C Wu
- Graduate Institute of Animal Vaccine Technology, College of Veterinary Medicine, National Pingtung University of Science and Technology, Pingtung, Taiwan
| | - P-H Yeh
- Graduate Institute of Animal Vaccine Technology, College of Veterinary Medicine, National Pingtung University of Science and Technology, Pingtung, Taiwan
| | - K-J Hsueh
- Graduate Institute of Animal Vaccine Technology, College of Veterinary Medicine, National Pingtung University of Science and Technology, Pingtung, Taiwan.,Department of Veterinary Medicine, National Pingtung University of Science and Technology, Pingtung, Taiwan
| | - W-J Yang
- Institute of Biotechnology, National University of Kaohsiung, Kaohsiung, Taiwan
| | - C-Y Chu
- Graduate Institute of Animal Vaccine Technology, College of Veterinary Medicine, National Pingtung University of Science and Technology, Pingtung, Taiwan
| |
Collapse
|
33
|
Antenucci F, Fougeroux C, Deeney A, Ørskov C, Rycroft A, Holst PJ, Bojesen AM. In vivo testing of novel vaccine prototypes against Actinobacillus pleuropneumoniae. Vet Res 2018; 49:4. [PMID: 29316978 PMCID: PMC5761136 DOI: 10.1186/s13567-017-0502-x] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2017] [Accepted: 12/18/2017] [Indexed: 02/05/2023] Open
Abstract
Actinobacillus pleuropneumoniae (A. pleuropneumoniae) is a Gram-negative bacterium that represents the main cause of porcine pleuropneumonia in pigs, causing significant economic losses to the livestock industry worldwide. A. pleuropneumoniae, as the majority of Gram-negative bacteria, excrete vesicles from its outer membrane (OM), accordingly defined as outer membrane vesicles (OMVs). Thanks to their antigenic similarity to the OM, OMVs have emerged as a promising tool in vaccinology. In this study we describe the in vivo testing of several vaccine prototypes for the prevention of infection by all known A. pleuropneumoniae serotypes. Previously identified vaccine candidates, the recombinant proteins ApfA and VacJ, administered individually or in various combinations with the OMVs, were employed as vaccination strategies. Our data show that the addition of the OMVs in the vaccine formulations significantly increased the specific IgG titer against both ApfA and VacJ in the immunized animals, confirming the previously postulated potential of the OMVs as adjuvant. Unfortunately, the antibody response raised did not translate into an effective protection against A. pleuropneumoniae infection, as none of the immunized groups following challenge showed a significantly lower degree of lesions than the controls. Interestingly, quite the opposite was true, as the animals with the highest IgG titers were also the ones bearing the most extensive lesions in their lungs. These results shed new light on A. pleuropneumoniae pathogenicity, suggesting that antibody-mediated cytotoxicity from the host immune response may play a central role in the development of the lesions typically associated with A. pleuropneumoniae infections.
Collapse
Affiliation(s)
- Fabio Antenucci
- Department of Veterinary and Animal Sciences, University of Copenhagen, Stigbøjlen 4, 1870 Frb. C., 1-20, Building: 301, Copenhagen, Denmark
| | - Cyrielle Fougeroux
- Department of International Health, Immunology and Microbiology ISIM, University of Copenhagen, Øster Farigmagsgade 5, Bldg 22/23, 1014 København K, Copenhagen, Denmark
| | - Alannah Deeney
- Department of Pathology and Pathogen Biology, Royal Veterinary College, Hawkshead Lane, North Mymms, Hertfordshire, AL9 7TA, UK
| | - Cathrine Ørskov
- Department of Biomedical Sciences, University of Copenhagen, Blegdamsvej 3, 2200 København N, 12.3, Building: 32, Copenhagen, Denmark
| | - Andrew Rycroft
- Department of Pathology and Pathogen Biology, Royal Veterinary College, Hawkshead Lane, North Mymms, Hertfordshire, AL9 7TA, UK
| | - Peter Johannes Holst
- Department of International Health, Immunology and Microbiology ISIM, University of Copenhagen, Øster Farigmagsgade 5, Bldg 22/23, 1014 København K, Copenhagen, Denmark
| | - Anders Miki Bojesen
- Department of Veterinary and Animal Sciences, University of Copenhagen, Stigbøjlen 4, 1870 Frb. C., 1-20, Building: 301, Copenhagen, Denmark.
| |
Collapse
|
34
|
Antenucci F, Fougeroux C, Bossé JT, Magnowska Z, Roesch C, Langford P, Holst PJ, Bojesen AM. Identification and characterization of serovar-independent immunogens in Actinobacillus pleuropneumoniae. Vet Res 2017; 48:74. [PMID: 29122004 PMCID: PMC5679336 DOI: 10.1186/s13567-017-0479-5] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2017] [Accepted: 10/20/2017] [Indexed: 11/17/2022] Open
Abstract
Despite numerous actions to prevent disease, Actinobacillus pleuropneumoniae (A. pleuropneumoniae) remains a major cause of porcine pleuropneumonia, resulting in economic losses to the swine industry worldwide. In this paper, we describe the utilization of a reverse vaccinology approach for the selection and in vitro testing of serovar-independent A. pleuropneumoniae immunogens. Potential immunogens were identified in the complete genomes of three A. pleuropneumoniae strains belonging to different serovars using the following parameters: predicted outer-membrane subcellular localization; ≤ 1 trans-membrane helices; presence of a signal peptide in the protein sequence; presence in all known A. pleuropneumoniae genomes; homology with other well characterized factors with relevant data regarding immunogenicity/protective potential. Using this approach, we selected the proteins ApfA and VacJ to be expressed and further characterized, both in silico and in vitro. Additionally, we analysed outer membrane vesicles (OMVs) of A. pleuropneumoniae MIDG2331 as potential immunogens, and compared deletions in degS and nlpI for increasing yields of OMVs compared to the parental strain. Our results indicated that ApfA and VacJ are highly conserved proteins, naturally expressed during infection by all A. pleuropneumoniae serovars tested. Furthermore, OMVs, ApfA and VacJ were shown to possess a high immunogenic potential in vitro. These findings favour the immunogen selection protocol used, and suggest that OMVs, along with ApfA and VacJ, could represent effective immunogens for the prevention of A. pleuropneumoniae infections in a serovar-independent manner. This hypothesis is nonetheless predictive in nature, and in vivo testing in a relevant animal model will be necessary to verify its validity.
Collapse
Affiliation(s)
- Fabio Antenucci
- Department of Veterinary and Animal Sciences, University of Copenhagen, Stigbøjlen 4, 1870 Frb. C., 1-20, Building: 301, Copenhagen, Denmark
| | - Cyrielle Fougeroux
- Department of International Health, Immunology and Microbiology ISIM, University of Copenhagen, Øster Farigmagsgade 5, Bldg 22/23, København K, 1014, Copenhagen, Denmark
| | - Janine T Bossé
- Department of Medicine, St Mary's Campus, Imperial College London, 236 Wright Fleming Wing, London, UK
| | - Zofia Magnowska
- Department of Veterinary and Animal Sciences, University of Copenhagen, Stigbøjlen 4, 1870 Frb. C., 1-20, Building: 301, Copenhagen, Denmark
| | - Camille Roesch
- Izon Science Ltd, Bâtiment Laennec, 60 Avenue Rockefeller, 69008, Lyon, France
| | - Paul Langford
- Department of Medicine, St Mary's Campus, Imperial College London, 236 Wright Fleming Wing, London, UK
| | - Peter Johannes Holst
- Department of International Health, Immunology and Microbiology ISIM, University of Copenhagen, Øster Farigmagsgade 5, Bldg 22/23, København K, 1014, Copenhagen, Denmark
| | - Anders Miki Bojesen
- Department of Veterinary and Animal Sciences, University of Copenhagen, Stigbøjlen 4, 1870 Frb. C., 1-20, Building: 301, Copenhagen, Denmark.
| |
Collapse
|
35
|
Sassu EL, Bossé JT, Tobias TJ, Gottschalk M, Langford PR, Hennig-Pauka I. Update on Actinobacillus pleuropneumoniae-knowledge, gaps and challenges. Transbound Emerg Dis 2017; 65 Suppl 1:72-90. [PMID: 29083117 DOI: 10.1111/tbed.12739] [Citation(s) in RCA: 113] [Impact Index Per Article: 16.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2017] [Indexed: 12/15/2022]
Abstract
Porcine pleuropneumonia, caused by the bacterial porcine respiratory tract pathogen Actinobacillus pleuropneumoniae, leads to high economic losses in affected swine herds in most countries of the world. Pigs affected by peracute and acute disease suffer from severe respiratory distress with high lethality. The agent was first described in 1957 and, since then, knowledge about the pathogen itself, and its interactions with the host, has increased continuously. This is, in part, due to the fact that experimental infections can be studied in the natural host. However, the fact that most commercial pigs are colonized by this pathogen has hampered the applicability of knowledge gained under experimental conditions. In addition, several factors are involved in development of disease, and these have often been studied individually. In a DISCONTOOLS initiative, members from science, industry and clinics exchanged their expertise and empirical observations and identified the major gaps in knowledge. This review sums up published results and expert opinions, within the fields of pathogenesis, epidemiology, transmission, immune response to infection, as well as the main means of prevention, detection and control. The gaps that still remain to be filled are highlighted, and present as well as future challenges in the control of this disease are addressed.
Collapse
Affiliation(s)
- E L Sassu
- Department of Pathobiology, Institute of Immunology, University of Veterinary Medicine, Vienna, Austria
| | - J T Bossé
- Section of Paediatrics, Department of Medicine, Imperial College London, London, UK
| | - T J Tobias
- Department of Farm Animal Health, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
| | - M Gottschalk
- Department of Pathology and Microbiology, Faculty of Veterinary Medicine, University of Montreal, Saint-Hyacinthe, QC, Canada
| | - P R Langford
- Section of Paediatrics, Department of Medicine, Imperial College London, London, UK
| | - I Hennig-Pauka
- Field Station for Epidemiology, University of Veterinary Medicine Hannover, Foundation, Bakum, Germany
| |
Collapse
|
36
|
An anti-Propionibacterium acnes antibody shows heterologous resistance to an Actinobacillus pleuropneumoniae infection independent of neutrophils in mice. Immunol Res 2017; 65:1124-1129. [PMID: 28929313 DOI: 10.1007/s12026-017-8954-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
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.
Collapse
|
37
|
Li P, Xu Z, Sun X, Yin Y, Fan Y, Zhao J, Mao X, Huang J, Yang F, Zhu L. Transcript profiling of the immunological interactions between Actinobacillus pleuropneumoniae serotype 7 and the host by dual RNA-seq. BMC Microbiol 2017; 17:193. [PMID: 28899359 PMCID: PMC5596872 DOI: 10.1186/s12866-017-1105-4] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2017] [Accepted: 09/05/2017] [Indexed: 02/08/2023] Open
Abstract
Background The complexity of the pathogenic mechanism underlying the host immune response to Actinobacillus pleuropneumonia (App) makes the use of preventive measures difficult, and a more global view of the host-pathogen interactions and new insights into this process are urgently needed to reveal the pathogenic and immune mechanisms underlying App infection. Here, we infected specific pathogen-free Mus musculus with App serotype 7 by intranasal inoculation to construct an acute hemorrhagic pneumonia infection model and isolated the infected lungs for analysis of the interactions by dual RNA-seq. Results Four cDNA libraries were constructed, and 2428 differentially expressed genes (DEGs) of the host and 333 DEGs of App were detected. The host DEGs were mainly enriched in inflammatory signaling pathways, such as the TLR, NLR, RLR, BCR and TCR signaling pathways, resulting in large-scale cytokine up-regulation and thereby yielding a cytokine cascade for anti-infection and lung damage. The majority of the up-regulated cytokines are involved in the IL-23/IL-17 cytokine-regulated network, which is crucial for host defense against bacterial infection. The DEGs of App were mainly related to the transport and metabolism of energy and materials. Most of these genes are metabolic genes involved in anaerobic metabolism and important for challenging the host and adapting to the anaerobic stress conditions observed in acute hemorrhagic pneumonia. Some of these genes, such as adhE, dmsA, and aspA, might be potential virulence genes. In addition, the up-regulation of genes associated with peptidoglycan and urease synthesis and the restriction of major virulence genes might be immune evasion strategies of App. The regulation of metabolic genes and major virulence genes indicate that the dominant antigens might differ during the infection process and that vaccines based on these antigens might allow establishment of a precise and targeted immune response during the early phase of infection. Conclusion Through an analysis of transcriptional data by dual RNA-seq, our study presents a novel global view of the interactions of App with its host and provides a basis for further study. Electronic supplementary material The online version of this article (10.1186/s12866-017-1105-4) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Ping Li
- College of Veterinary Medicine, Sichuan Agricultural University, Huimin Road 211, Weenjiang District, Chengdu, Sichuan, China.,Key Laboratory of Animal Diseases and Human Health of Sichuan Province, Chengdu, Sichuan, China
| | - Zhiwen Xu
- College of Veterinary Medicine, Sichuan Agricultural University, Huimin Road 211, Weenjiang District, Chengdu, Sichuan, China.,Key Laboratory of Animal Diseases and Human Health of Sichuan Province, Chengdu, Sichuan, China
| | - Xiangang Sun
- College of Veterinary Medicine, Sichuan Agricultural University, Huimin Road 211, Weenjiang District, Chengdu, Sichuan, China
| | - Yue Yin
- College of Veterinary Medicine, Sichuan Agricultural University, Huimin Road 211, Weenjiang District, Chengdu, Sichuan, China.,Key Laboratory of Animal Diseases and Human Health of Sichuan Province, Chengdu, Sichuan, China
| | - Yi Fan
- College of Veterinary Medicine, Sichuan Agricultural University, Huimin Road 211, Weenjiang District, Chengdu, Sichuan, China.,Key Laboratory of Animal Diseases and Human Health of Sichuan Province, Chengdu, Sichuan, China
| | - Jun Zhao
- College of Veterinary Medicine, Sichuan Agricultural University, Huimin Road 211, Weenjiang District, Chengdu, Sichuan, China.,Key Laboratory of Animal Diseases and Human Health of Sichuan Province, Chengdu, Sichuan, China
| | - Xiyu Mao
- College of Veterinary Medicine, Sichuan Agricultural University, Huimin Road 211, Weenjiang District, Chengdu, Sichuan, China.,Key Laboratory of Animal Diseases and Human Health of Sichuan Province, Chengdu, Sichuan, China
| | - Jianbo Huang
- College of Veterinary Medicine, Sichuan Agricultural University, Huimin Road 211, Weenjiang District, Chengdu, Sichuan, China.,Key Laboratory of Animal Diseases and Human Health of Sichuan Province, Chengdu, Sichuan, China
| | - Fan Yang
- College of Veterinary Medicine, Sichuan Agricultural University, Huimin Road 211, Weenjiang District, Chengdu, Sichuan, China.,Key Laboratory of Animal Diseases and Human Health of Sichuan Province, Chengdu, Sichuan, China
| | - Ling Zhu
- College of Veterinary Medicine, Sichuan Agricultural University, Huimin Road 211, Weenjiang District, Chengdu, Sichuan, China. .,Key Laboratory of Animal Diseases and Human Health of Sichuan Province, Chengdu, Sichuan, China.
| |
Collapse
|
38
|
To H, Nagai S, Iwata A, Koyama T, Oshima A, Tsutsumi N. Genetic and antigenic characteristics of ApxIIA and ApxIIIA from Actinobacillus pleuropneumoniae serovars 2, 3, 4, 6, 8 and 15. Microbiol Immunol 2017; 60:447-58. [PMID: 27211905 DOI: 10.1111/1348-0421.12388] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2015] [Revised: 04/21/2016] [Accepted: 05/16/2016] [Indexed: 11/30/2022]
Abstract
Apx toxins produced by Actinobacillus pleuropneumoniae are essential components of new generation vaccines. In this study, apxIIA and apxIIIA genes of serovars 2, 3, 4, 6, 8 and 15 were cloned and sequenced. Amino acid sequences of ApxIIA proteins of serovars 2, 3, 4, 6, 8 and 15 were almost identical to those of serovars 1, 5, 7, 9 and 11-13. Immunoblot analysis showed that rApxIIA from serovars 2 and 15 reacts strongly with sera from animals infected with various serovars. Sequence analysis revealed that ApxIIIA proteins has two variants, one in strains of serovar 2 and the other in strains of serovars 3, 4, 6, 8 and 15. A mouse cross-protection study showed that mice actively immunized with rApxIIIA/2 or rApxIIIA/15 are protected against challenge with A. pleuropneumoniae strains of serovars 3, 4, 6, 8, 15, and 2 expressing ApxIII/15 and ApxIII/2, respectively. Similarly, mice passively immunized with rabbit anti-rApxIIIA/2 or anti-rApxIIIA/15 sera were found to be protected against challenge with strains of serovars 2 and 15. Our study revealed antigenic and sequence similarities within ApxIIA and ApxIIIA proteins, which may help in the development of effective vaccines against disease caused by A. pleuropneumoniae.
Collapse
Affiliation(s)
- Ho To
- Nippon Institute for Biological Science, 9-2221-1 Shinmachi, Ome, Tokyo 198-0024, Japan
| | - Shinya Nagai
- Nippon Institute for Biological Science, 9-2221-1 Shinmachi, Ome, Tokyo 198-0024, Japan
| | - Akira Iwata
- Nippon Institute for Biological Science, 9-2221-1 Shinmachi, Ome, Tokyo 198-0024, Japan
| | - Tomohiro Koyama
- Nippon Institute for Biological Science, 9-2221-1 Shinmachi, Ome, Tokyo 198-0024, Japan
| | - Atsushi Oshima
- Nippon Institute for Biological Science, 9-2221-1 Shinmachi, Ome, Tokyo 198-0024, Japan
| | - Nobuyuki Tsutsumi
- Nippon Institute for Biological Science, 9-2221-1 Shinmachi, Ome, Tokyo 198-0024, Japan
| |
Collapse
|
39
|
Xie F, Li G, Zhou L, Zhang Y, Cui N, Liu S, Wang C. Attenuated Actinobacillus pleuropneumoniae double-deletion mutant S-8∆clpP/apxIIC confers protection against homologous or heterologous strain challenge. BMC Vet Res 2017; 13:14. [PMID: 28061786 PMCID: PMC5219649 DOI: 10.1186/s12917-016-0928-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2016] [Accepted: 12/15/2016] [Indexed: 11/18/2022] Open
Abstract
Background Actinobacillus pleuropneumoniae is the etiological agent of porcine pleuropneumonia, which leads to large economic losses to the swine industry worldwide. In this study, S-8△clpP△apxIIC, a double-deletion mutant of A. pleuropneumoniae was constructed, and its safety and protective efficacy were evaluated in pigs. Results The S-8△clpP△apxIIC mutant exhibited attenuated virulence in a murine (BALB/c) model, and caused no detrimental effects on pigs even at a dose of up to 1.0 × 109 CFU. Furthermore, the S-8△clpP△apxIIC mutant was able to induce a strong immune response in pigs, which included high levels of IgG1 and IgG2, stimulated gamma interferon (IFN-γ), interleukin 12 (IL-12), and interleukin 4 (IL-4) production, and conferred effective protection against the lethal challenge with A. pleuropneumoniae serovars 7 or 5a. The pigs in the S-8△clpP△apxIIC immunized groups have no lesions and reduced bacterial loads in the lung tissue after challenge. Conclusions The data obtained in this study suggest that the S-8△clpP△apxIIC mutant can serve as a highly immunogenic and potential live attenuated vaccine candidate against A. pleuropneumoniae infection.
Collapse
Affiliation(s)
- Fang Xie
- State Key Laboratory of Veterinary Biotechnology, Division of Bacterial Diseases, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, Heilongjiang, People's Republic of China
| | - Gang Li
- State Key Laboratory of Veterinary Biotechnology, Division of Bacterial Diseases, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, Heilongjiang, People's Republic of China
| | - Long Zhou
- State Key Laboratory of Veterinary Biotechnology, Division of Bacterial Diseases, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, Heilongjiang, People's Republic of China
| | - Yanhe Zhang
- State Key Laboratory of Veterinary Biotechnology, Division of Bacterial Diseases, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, Heilongjiang, People's Republic of China
| | - Ning Cui
- State Key Laboratory of Veterinary Biotechnology, Division of Bacterial Diseases, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, Heilongjiang, People's Republic of China
| | - Siguo Liu
- State Key Laboratory of Veterinary Biotechnology, Division of Bacterial Diseases, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, Heilongjiang, People's Republic of China
| | - Chunlai Wang
- State Key Laboratory of Veterinary Biotechnology, Division of Bacterial Diseases, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, Heilongjiang, People's Republic of China.
| |
Collapse
|
40
|
Qin W, Wang L, Zhai R, Ma Q, Liu J, Bao C, Sun D, Zhang H, Sun C, Feng X, Gu J, Du C, Han W, Langford PR, Lei L. Apa2H1, the first head domain of Apa2 trimeric autotransporter adhesin, activates mouse bone marrow-derived dendritic cells and immunization with Apa2H1 protects against Actinobacillus pleuropneumoniae infection. Mol Immunol 2016; 81:108-117. [PMID: 27940254 DOI: 10.1016/j.molimm.2016.12.004] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2016] [Revised: 11/29/2016] [Accepted: 12/01/2016] [Indexed: 12/23/2022]
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.
Collapse
Affiliation(s)
- Wanhai Qin
- College of Veterinary Medicine, Jilin University, Changchun, PR China
| | - Lei Wang
- College of Animal Science, Henan Institute of Science and Technology, Xinxiang, PR China
| | - Ruidong Zhai
- College of Veterinary Medicine, Jilin University, Changchun, PR China
| | - Qiuyue Ma
- College of Veterinary Medicine, Jilin University, Changchun, PR China
| | - Jianfang Liu
- College of Veterinary Medicine, Jilin University, Changchun, PR China
| | - Chuntong Bao
- College of Veterinary Medicine, Jilin University, Changchun, PR China
| | - Diangang Sun
- College of Veterinary Medicine, Jilin University, Changchun, PR China
| | - Hu Zhang
- School of Public Health, Jilin University, Changchun, PR China
| | - Changjiang Sun
- College of Veterinary Medicine, Jilin University, Changchun, PR China
| | - Xin Feng
- College of Veterinary Medicine, Jilin University, Changchun, PR China
| | - Jingmin Gu
- College of Veterinary Medicine, Jilin University, Changchun, PR China
| | - Chongtao Du
- College of Veterinary Medicine, Jilin University, Changchun, PR China
| | - Wenyu Han
- College of Veterinary Medicine, Jilin University, Changchun, PR China
| | - P R Langford
- Section of Paediatrics, Imperial College London, London, UK
| | - Liancheng Lei
- College of Veterinary Medicine, Jilin University, Changchun, PR China.
| |
Collapse
|
41
|
Production and immunogenicity of Actinobacillus pleuropneumoniae ApxIIA protein in transgenic rice callus. Protein Expr Purif 2016; 132:116-123. [PMID: 27215671 DOI: 10.1016/j.pep.2016.05.010] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2016] [Revised: 05/16/2016] [Accepted: 05/19/2016] [Indexed: 01/02/2023]
Abstract
Actinobacillus pleuropneumoniae is a major etiological agent that is responsible for swine pleuropneumonia, a highly contagious respiratory infection that causes severe economic losses in the swine production industry. ApxIIA is one of the virulence factors in A. pleuropneumoniae and has been considered as a candidate for developing a vaccine against the bacterial infection. A gene encoding an ApxIIA fragment (amino acids 439-801) was modified based on a plant-optimized codon and constructed into a plant expression vector under the control of a promoter and the 3' UTR of the rice amylase 3D gene. The plant expression vector was introduced into rice embryogenic callus (Oryza sativa L. cv. Dongjin) via particle bombardment-mediated transformation. The integration and transcription of the ApxIIA439-801 gene were confirmed by using genomic DNA PCR amplification and Northern blot analysis, respectively. The synthesis of ApxIIA439-801 antigen protein in transgenic rice callus was confirmed by western blot analysis. The concentration of antigen protein in lyophilized samples of transgenic rice callus was 250 μg/g. Immunizing mice with protein extracts from transgenic plants intranasally elicited secretory IgA. These results demonstrate the feasibility of using a transgenic plant to elicit immune responses against A. pleuropneumoniae.
Collapse
|
42
|
Xie F, Li G, Zhang Y, Zhou L, Liu S, Liu S, Wang C. The Lon protease homologue LonA, not LonC, contributes to the stress tolerance and biofilm formation of Actinobacillus pleuropneumoniae. Microb Pathog 2016; 93:38-43. [PMID: 26796296 DOI: 10.1016/j.micpath.2016.01.009] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2015] [Revised: 12/10/2015] [Accepted: 01/11/2016] [Indexed: 01/09/2023]
Abstract
Lon proteases are a family of ATP-dependent proteases that are involved in the degradation of abnormal proteins in bacteria exposed to adverse environmental stress. An analysis of the genome sequence of Actinobacillus pleuropneumoniae revealed the unusual presence of two putative ATP-dependent Lon homologues, LonA and LonC. Sequence comparisons indicated that LonA has the classical domain organization of the LonA subfamily, which includes the N-terminal domain, central ATPase (AAA) domain, and C-terminal proteolytic (P) domain. LonC belongs to the recently classified LonC subfamily, which includes Lon proteases that contain neither the N-terminal domain of LonA nor the transmembrane region that is present only in LonB subfamily members. To investigate the roles of LonA and LonC in A. pleuropneumoniae, mutants with deletions in the lonA and lonC genes were constructed. The impaired growth of the △lonA mutant exposed to low and high temperatures and osmotic and oxidative stress conditions indicates that the LonA protease is required for the stress tolerance of A. pleuropneumoniae. Furthermore, the △lonA mutant exhibited significantly reduced biofilm formation compared to the wild-type strain. However, no significant differences in stress responses or biofilm formation were observed between the △lonC mutant and the wild-type strain. The △lonA mutant exhibited reduced colonization ability and attenuated virulence of A. pleuropneumoniae in the BALB/c mouse model compared to the wild-type strain. Disruption of lonC gene did not significantly influence the colonization and virulence of A. pleuropneumoniae. The data presented in this study illustrate that the LonA protease, but not the LonC protease, is required for the stress tolerance, biofilm formation and pathogenicity of A. pleuropneumoniae.
Collapse
Affiliation(s)
- Fang Xie
- State Key Laboratory of Veterinary Biotechnology, Division of Bacterial Diseases, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin 15001, People's Republic of China
| | - Gang Li
- State Key Laboratory of Veterinary Biotechnology, Division of Bacterial Diseases, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin 15001, People's Republic of China
| | - Yanhe Zhang
- State Key Laboratory of Veterinary Biotechnology, Division of Bacterial Diseases, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin 15001, People's Republic of China
| | - Long Zhou
- State Key Laboratory of Veterinary Biotechnology, Division of Bacterial Diseases, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin 15001, People's Republic of China
| | - Shuanghong Liu
- State Key Laboratory of Veterinary Biotechnology, Division of Bacterial Diseases, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin 15001, People's Republic of China
| | - Siguo Liu
- State Key Laboratory of Veterinary Biotechnology, Division of Bacterial Diseases, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin 15001, People's Republic of China
| | - Chunlai Wang
- State Key Laboratory of Veterinary Biotechnology, Division of Bacterial Diseases, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin 15001, People's Republic of China.
| |
Collapse
|
43
|
Hathroubi S, Fontaine-Gosselin SÈ, Tremblay YDN, Labrie J, Jacques M. Sub-inhibitory concentrations of penicillin G induce biofilm formation by field isolates of Actinobacillus pleuropneumoniae. Vet Microbiol 2015; 179:277-86. [PMID: 26130517 DOI: 10.1016/j.vetmic.2015.06.011] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2014] [Revised: 06/05/2015] [Accepted: 06/17/2015] [Indexed: 11/28/2022]
Abstract
Actinobacillus pleuropneumoniae is a Gram-negative bacterium and causative agent of porcine pleuropneumonia. This is a highly contagious disease that causes important economic losses to the swine industry worldwide. Penicillins are extensively used in swine production and these antibiotics are associated with high systemic clearance and low oral bioavailability. This may expose A. pleuropneumoniae to sub-inhibitory concentrations of penicillin G when the antibiotic is administered orally. Our goal was to evaluate the effect of sub-minimum inhibitory concentration (MIC) of penicillin G on the biofilm formation of A. pleuropneumoniae. Biofilm production of 13 field isolates from serotypes 1, 5a, 7 and 15 was tested in the presence of sub-MIC of penicillin G using a polystyrene microtiter plate assay. Using microscopy techniques and enzymatic digestion, biofilm architecture and composition were also characterized after exposure to sub-MIC of penicillin G. Sub-MIC of penicillin G significantly induced biofilm formation of nine isolates. The penicillin G-induced biofilms contained more poly-N-acetyl-D-glucosamine (PGA), extracellular DNA and proteins when compared to control biofilms grown without penicillin G. Additionally, penicillin G-induced biofilms were sensitive to DNase which was not observed with the untreated controls. Furthermore, sub-MIC of penicillin G up-regulated the expression of pgaA, which encodes a protein involved in PGA synthesis, and the genes encoding the envelope-stress sensing two-component regulatory system CpxRA. In conclusion, sub-MICs of penicillin G significantly induce biofilm formation and this is likely the result of a cell envelope stress sensed by the CpxRA system resulting in an increased production of PGA and other matrix components.
Collapse
Affiliation(s)
- S Hathroubi
- Groupe de recherche sur les maladies infectieuses du porc, Faculté de médecine vétérinaire, Université de Montréal, C. P. 5000, St-Hyacinthe, Québec J2S 7C6, Canada
| | - S-È Fontaine-Gosselin
- Groupe de recherche sur les maladies infectieuses du porc, Faculté de médecine vétérinaire, Université de Montréal, C. P. 5000, St-Hyacinthe, Québec J2S 7C6, Canada
| | - Y D N Tremblay
- Groupe de recherche sur les maladies infectieuses du porc, Faculté de médecine vétérinaire, Université de Montréal, C. P. 5000, St-Hyacinthe, Québec J2S 7C6, Canada
| | - J Labrie
- Groupe de recherche sur les maladies infectieuses du porc, Faculté de médecine vétérinaire, Université de Montréal, C. P. 5000, St-Hyacinthe, Québec J2S 7C6, Canada
| | - M Jacques
- Groupe de recherche sur les maladies infectieuses du porc, Faculté de médecine vétérinaire, Université de Montréal, C. P. 5000, St-Hyacinthe, Québec J2S 7C6, Canada.
| |
Collapse
|
44
|
Overexpression of Porcine Beta-Defensin 2 Enhances Resistance to Actinobacillus pleuropneumoniae Infection in Pigs. Infect Immun 2015; 83:2836-43. [PMID: 25916992 DOI: 10.1128/iai.03101-14] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2014] [Accepted: 04/24/2015] [Indexed: 01/01/2023] Open
Abstract
To reduce the need for antibiotics in animal production, alternative approaches are needed to control infection. We hypothesized that overexpression of native defensin genes will provide food animals with enhanced resistance to bacterial infections. In this study, recombinant porcine beta-defensin 2 (PBD-2) was overexpressed in stably transfected PK-15 porcine kidney cells. PBD-2 antibacterial activities against Actinobacillus pleuropneumoniae, an important respiratory pathogen causing porcine contagious pleuropneumonia, were evaluated on agar plates. Transgenic pigs constitutively overexpressing PBD-2 were produced by a somatic cell cloning method, and their resistance to bacterial infection was evaluated by direct or cohabitation infection with A. pleuropneumoniae. Recombinant PBD-2 peptide that was overexpressed in the PK-15 cells showed antibacterial activity against A. pleuropneumoniae. PBD-2 was overexpressed in the heart, liver, spleen, lungs, kidneys, and jejunum of the transgenic pigs, which showed significantly lower bacterial loads in the lungs and reduced lung lesions after direct or cohabitation infection with A. pleuropneumoniae. The results demonstrate that transgenic overexpression of PBD-2 in pigs confers enhanced resistance against A. pleuropneumoniae infection.
Collapse
|
45
|
Park J, Seo KW, Kim SH, Lee HY, Kim B, Lim CW, Kim JH, Yoo HS, Jang YS. Nasal immunization with M cell-targeting ligand-conjugated ApxIIA toxin fragment induces protective immunity against Actinobacillus pleuropneumoniae infection in a murine model. Vet Microbiol 2015; 177:142-53. [PMID: 25818577 DOI: 10.1016/j.vetmic.2015.03.005] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2014] [Revised: 03/04/2015] [Accepted: 03/06/2015] [Indexed: 01/23/2023]
Abstract
Actinobacillus pleuropneumoniae is the causative agent of porcine pleuropneumonia and severe economic loss in the swine industry has been caused by the infection. Therefore, the development of an effective vaccine against the bacteria is necessary. ApxII toxin, among several virulence factors expressed by the bacteria, is considered to be a promising vaccine candidate because ApxII toxin not only accompanies cytotoxic and hemolytic activities, but is also expressed in all 15 serotypes of bacteria except serotypes 10 and 14. In this study, we identified the peptide ligand capable of targeting the ligand-conjugated ApxIIA #5 fragment antigen to nasopharynx-associated lymphoid tissue. It was found that nasal immunization with ligand-conjugated ApxIIA #5 induced efficient mucosal and systemic immune responses measured at the levels of antigen-specific antibodies, cytokine-secreting cells after antigen exposure, and antigen-specific lymphocyte proliferation. More importantly, the nasal immunization induced protective immunity against nasal challenge infection of the bacteria, which was confirmed by histopathological studies and bacterial clearance after challenge infection. Collectively, we confirmed that the ligand capable of targeting the ligand-conjugated antigen to nasopharynx-associated lymphoid tissue can be used as an effective nasal vaccine adjuvant to induce protective immunity against A. pleuropneumoniae infection.
Collapse
Affiliation(s)
- Jisang Park
- Department of Molecular Biology and the Institute for Molecular Biology and Genetics, Chonbuk National University, Jeonju 561-756, Republic of Korea; Department of Bioactive Material Sciences and Research Center of Bioactive Materials, Chonbuk National University, Jeonju 561-756, Republic of Korea
| | - Ki-Weon Seo
- Department of Molecular Biology and the Institute for Molecular Biology and Genetics, Chonbuk National University, Jeonju 561-756, Republic of Korea
| | - Sae-Hae Kim
- Department of Molecular Biology and the Institute for Molecular Biology and Genetics, Chonbuk National University, Jeonju 561-756, Republic of Korea; Department of Bioactive Material Sciences and Research Center of Bioactive Materials, Chonbuk National University, Jeonju 561-756, Republic of Korea
| | - Ha-Yan Lee
- Department of Molecular Biology and the Institute for Molecular Biology and Genetics, Chonbuk National University, Jeonju 561-756, Republic of Korea; Department of Bioactive Material Sciences and Research Center of Bioactive Materials, Chonbuk National University, Jeonju 561-756, Republic of Korea
| | - Bumseok Kim
- Department of Pathology, College of Veterinary Medicine, Chonbuk National University, Jeonju 561-756, Republic of Korea
| | - Chae Woong Lim
- Department of Pathology, College of Veterinary Medicine, Chonbuk National University, Jeonju 561-756, Republic of Korea
| | - Jin-Hee Kim
- Jeonbuk Provincial Office, National Agricultural Products Quality Management Service, Jeonju 561-202, Republic of Korea
| | - Han Sang Yoo
- Department of Infectious Diseases, College of Veterinary Medicine, Seoul National University, Seoul 151-742, Republic of Korea
| | - Yong-Suk Jang
- Department of Molecular Biology and the Institute for Molecular Biology and Genetics, Chonbuk National University, Jeonju 561-756, Republic of Korea; Department of Bioactive Material Sciences and Research Center of Bioactive Materials, Chonbuk National University, Jeonju 561-756, Republic of Korea.
| |
Collapse
|
46
|
Ma Q, Sun C, Yang F, Wang L, Qin W, Xia X, Feng X, Du C, Gu J, Han W, Lei L. Macrophages largely contribute to heterologous anti-Propionibacterium acnes antibody-mediated protection from Actinobacillus pleuropneumoniae infection in mice. Microbiol Immunol 2015; 59:166-73. [PMID: 25644652 DOI: 10.1111/1348-0421.12240] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2014] [Revised: 01/05/2015] [Accepted: 01/26/2015] [Indexed: 11/29/2022]
Abstract
Actinobacillus pleuropneumoniae is the causative agent of acute and chronic pleuropneumonia. Propionibacterium acnes is a facultative anaerobic gram-positive corynebacterium. We have previously found that anti-P. acnes antibodies can prevent A. pleuropneumoniae infections in mice. To investigate the role of macrophages in this process, affinity-purified anti-P. acnes IgG and anti-A. pleuropneumoniae IgG were used in opsonophagocytosis assays. Additionally, the efficacy of passive immunization with P. acnes serum against A. pleuropneumoniae was tested in macrophage-depleted mice. It was found that anti-P. acnes IgG had an effect similar to that of anti-A. pleuropneumoniae IgG (P > 0.05), which significantly promotes phagocytosis of A. pleuropneumoniae by macrophages (P < 0.01). It was also demonstrated that, after passive immunization with anti-P. acnes serum, macrophage-replete mice had the highest survival rate (90%), whereas the survival rate of macrophage-depleted mice was only 40% (P < 0.05). However, macrophage-depleted mice that had been passively immunized with naïve serum had the lowest survival rate (20%), this rate being lower than that of macrophage-replete mice that had been passively immunized with naïve serum. Overall, anti-P. acnes antibodies did not prevent A. pleuropneumoniae infection under conditions of macrophage depletion (P > 0.05). Furthermore, in mice that had been passively immunized with anti-P. acnes serum, macrophage depletion resulted in a greater A. pleuropneumoniae burden and more severe pathological features of pneumonia in lung tissues than occurred in macrophage-replete mice. It was concluded that macrophages are essential for the process by which anti-P. acnes antibody prevents A. pleuropneumoniae infection in mice.
Collapse
Affiliation(s)
- Qiuyue Ma
- College of Veterinary Medicine, Jilin University, Changchun, China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Lee KE, Choi HW, Kim HH, Song JY, Yang DK. Prevalence and Characterization ofActinobacillus pleuropneumoniaeIsolated from Korean Pigs. ACTA ACUST UNITED AC 2015. [DOI: 10.4167/jbv.2015.45.1.19] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Affiliation(s)
- Ki-Eun Lee
- Animal and Plant Quarantine Agency, MAFRA, Anyang, Korea
| | | | - Ha-Hyun Kim
- Animal and Plant Quarantine Agency, MAFRA, Anyang, Korea
| | - Jae-Young Song
- Animal and Plant Quarantine Agency, MAFRA, Anyang, Korea
| | - Dong-Kun Yang
- Animal and Plant Quarantine Agency, MAFRA, Anyang, Korea
| |
Collapse
|
48
|
Abstract
Swine are used in biomedical research as models for biomedical research and for teaching. This chapter covers normative biology and behavior along with common and emerging swine diseases. Xenotransplantation is discussed along with similarities and differences of swine immunology.
Collapse
Affiliation(s)
- Kristi L. Helke
- Departments of Comparative Medicine and Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, SC, USA
| | | | - Raimon Duran-Struuck
- Columbia Center of Translational Immunology, Department of Surgery; Institute of Comparative Medicine; Columbia University Medical Center, New York, NY, USA
| | - M. Michael Swindle
- Medical University of South Carolina, Department of Comparative Medicine and Department of Surgery, Charleston, SC, USA
| |
Collapse
|
49
|
The generation of successive unmarked mutations and chromosomal insertion of heterologous genes in Actinobacillus pleuropneumoniae using natural transformation. PLoS One 2014; 9:e111252. [PMID: 25409017 PMCID: PMC4237320 DOI: 10.1371/journal.pone.0111252] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2014] [Accepted: 09/26/2014] [Indexed: 12/24/2022] Open
Abstract
We have developed a simple method of generating scarless, unmarked mutations in Actinobacillus pleuropneumoniae by exploiting the ability of this bacterium to undergo natural transformation, and with no need to introduce plasmids encoding recombinases or resolvases. This method involves two successive rounds of natural transformation using linear DNA: the first introduces a cassette carrying cat (which allows selection by chloramphenicol) and sacB (which allows counter-selection using sucrose) flanked by sequences to either side of the target gene; the second transformation utilises the flanking sequences ligated directly to each other in order to remove the cat-sacB cassette. In order to ensure efficient uptake of the target DNA during transformation, A. pleuropneumoniae uptake sequences are added into the constructs used in both rounds of transformation. This method can be used to generate multiple successive deletions and can also be used to introduce targeted point mutations or insertions of heterologous genes into the A. pleuropneumoniae chromosome for development of live attenuated vaccine strains. So far, we have applied this method to highly transformable isolates of serovars 8 (MIDG2331), which is the most prevalent in the UK, and 15 (HS143). By screening clinical isolates of other serovars, it should be possible to identify other amenable strains.
Collapse
|
50
|
Yoo AN, Cha SB, Shin MK, Won HK, Kim EH, Choi HW, Yoo HS. Serotypes and antimicrobial resistance patterns of the recent Korean Actinobacillus pleuropneumoniae
isolates. Vet Rec 2014; 174:223. [DOI: 10.1136/vr.101863] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Affiliation(s)
- A. N. Yoo
- Department of Infectious Diseases; College of Veterinary Medicine and Brain Korea 21 Program for Veterinary Science; Seoul National University; Seoul 151-742 Korea
| | - S. B. Cha
- Department of Infectious Diseases; College of Veterinary Medicine and Brain Korea 21 Program for Veterinary Science; Seoul National University; Seoul 151-742 Korea
| | - M. K. Shin
- Department of Infectious Diseases; College of Veterinary Medicine and Brain Korea 21 Program for Veterinary Science; Seoul National University; Seoul 151-742 Korea
| | - H. K. Won
- Department of Infectious Diseases; College of Veterinary Medicine and Brain Korea 21 Program for Veterinary Science; Seoul National University; Seoul 151-742 Korea
- ChoongAng Vaccine Laboratory Co. Ltd.; Daejeon 305-348 Korea
| | - E. H. Kim
- ChoongAng Vaccine Laboratory Co. Ltd.; Daejeon 305-348 Korea
| | - H. W. Choi
- ChoongAng Vaccine Laboratory Co. Ltd.; Daejeon 305-348 Korea
| | - H. S. Yoo
- Department of Infectious Diseases; College of Veterinary Medicine and Brain Korea 21 Program for Veterinary Science; Seoul National University; Seoul 151-742 Korea
| |
Collapse
|