1
|
Tang P, Huang B, Ou Q, Liu F, Lin L, Zheng Y, Xie H, Yang X, Zhang X, Kuang Z, Xie Y, Sun J, Lin B, Li J, Lin B. A mouse model of sepsis-associated DIC induced by Kappa-carrageenan and Lipopolysaccharides: Establishment and characteristics. J Adv Res 2025:S2090-1232(25)00189-4. [PMID: 40118341 DOI: 10.1016/j.jare.2025.03.029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2024] [Revised: 03/02/2025] [Accepted: 03/16/2025] [Indexed: 03/23/2025] Open
Abstract
BACKGROUND No animal models fully replicate the pathogenesis and clinical features of sepsis-associated disseminated intravascular coagulation (DIC), which hinders mechanistic understanding and treatment development. Kappa-carrageenan (KCG) and lipopolysaccharides (LPS) induce thrombosis and systemic inflammation in mice, respectively. The combination of LPS and KCG provides a promising method for establishing a mouse model of sepsis-associated DIC. OBJECTIVE This study aimed to establish a standardized mouse model of sepsis-associated DIC using KCG and LPS. METHODS Kunming (KM) mice were intraperitoneally injected with KCG (25-200 mg/kg) alone or in combination with LPS (50-1250 μg/kg) to determine optimal dose. The effects of ambient temperature, gender and mouse strains on the mouse model were evaluated. Time-dependent changes in the model were examined. RESULTS The combined injection of KCG (100 mg/kg) and LPS (50 μg/kg) effectively induced tail thrombosis and prolonged activated partial thromboplastin time. Mice housed at 16 ± 1℃ exhibited more severe thrombosis and hypocoagulability than those at 24 ± 1℃. Male and female mice exhibited similar responses. Time-course analysis revealed inflammation and blood hypocoagulability beginning from 1.5 to 24 h, with fibrinolysis inhibition occurring within 1 h. Tail thrombosis and auricle petechial developed at 3 and 6 h, respectively, and stabilized by 12 h. Thrombi in the tail, lung and liver along with organ dysfunction were obeserved at 12 h. KM and BALB/c mice exhibited longer tail thrombi than Institute of Cancer Research (ICR) mice. KM mice showed more severe blood hypocoagulability than ICR and BALB/c mice. CONCLUSIONS This study establishes a standardized mouse model of sepsis-associated DIC using KCG and LPS, which more accurately replicates the key clinical and pathological characteristics of sepsis-associated DIC compared to existing models. This model serves as a novelty and valuable tool for investigating the mechanisms of sepsis-associated DIC and therapeutic evaluation.
Collapse
Affiliation(s)
- Ping Tang
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510405, China; Guangdong Clinical Research Academy of Chinese Medicine, Guangzhou, Guangdong 510405, China
| | - Boning Huang
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510405, China; Guangdong Clinical Research Academy of Chinese Medicine, Guangzhou, Guangdong 510405, China
| | - Qianqing Ou
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510405, China; Guangdong Clinical Research Academy of Chinese Medicine, Guangzhou, Guangdong 510405, China
| | - Fangle Liu
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510405, China; Guangdong Clinical Research Academy of Chinese Medicine, Guangzhou, Guangdong 510405, China
| | - Liuqing Lin
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510405, China; Guangdong Clinical Research Academy of Chinese Medicine, Guangzhou, Guangdong 510405, China
| | - Yuying Zheng
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510405, China; Guangdong Clinical Research Academy of Chinese Medicine, Guangzhou, Guangdong 510405, China
| | - Huiyi Xie
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510405, China; Guangdong Clinical Research Academy of Chinese Medicine, Guangzhou, Guangdong 510405, China
| | - Xinrong Yang
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510405, China; Guangdong Clinical Research Academy of Chinese Medicine, Guangzhou, Guangdong 510405, China
| | - Xiubing Zhang
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510405, China; Guangdong Clinical Research Academy of Chinese Medicine, Guangzhou, Guangdong 510405, China
| | - Zhongsheng Kuang
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510405, China; Guangdong Clinical Research Academy of Chinese Medicine, Guangzhou, Guangdong 510405, China
| | - Yuhui Xie
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510405, China; Guangdong Clinical Research Academy of Chinese Medicine, Guangzhou, Guangdong 510405, China
| | - Jingjing Sun
- Greater Bay Area Institute of Precision Medicine (Guangzhou), Guangzhou, Guangdong 511462, China
| | - Bingqing Lin
- School of Mathematical Sciences, Shenzhen University, Shenzhen, Guangdong 518060, China.
| | - Jun Li
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510405, China; Guangdong Clinical Research Academy of Chinese Medicine, Guangzhou, Guangdong 510405, China.
| | - Baoqin Lin
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510405, China; Guangdong Clinical Research Academy of Chinese Medicine, Guangzhou, Guangdong 510405, China.
| |
Collapse
|
2
|
Wang S, Zhao K, Chen Z, Liu D, Tang S, Sun C, Chen H, Wang Y, Wu C. Halicin: A New Horizon in Antibacterial Therapy against Veterinary Pathogens. Antibiotics (Basel) 2024; 13:492. [PMID: 38927159 PMCID: PMC11200678 DOI: 10.3390/antibiotics13060492] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Revised: 05/22/2024] [Accepted: 05/23/2024] [Indexed: 06/28/2024] Open
Abstract
It is crucial to discover novel antimicrobial drugs to combat resistance. This study investigated the antibacterial properties of halicin (SU3327), an AI-identified anti-diabetic drug, against 13 kinds of common clinical pathogens of animal origin, including multidrug-resistant strains. Employing minimum inhibitory concentration (MIC) and minimum bactericidal concentration (MBC) assessments, halicin demonstrated a broad-spectrum antibacterial effect. Time-killing assays revealed its concentration-dependent bactericidal activity against Escherichia coli ATCC 25922 (E. coli ATCC 25922), Staphylococcus aureus ATCC 29213 (S. aureus ATCC 29213), and Actinobacillus pleuropneumoniae S6 (APP S6) after 4 h of treatment at concentrations above the MIC. Halicin exhibited longer post-antibiotic effects (PAEs) and sub-MIC effects (PA-SMEs) for E. coli 25922, S. aureus 29213, and APP S6 compared to ceftiofur and ciprofloxacin, the commonly used veterinary antimicrobial agents, indicating sustained antibacterial action. Additionally, the results of consecutive passaging experiments over 40 d at sub-inhibitory concentrations showed that bacteria exhibited difficulty in developing resistance to halicin. Toxicology studies confirmed that halicin exhibited low acute toxicity, being non-mutagenic, non-reproductive-toxic, and non-genotoxic. Blood biochemical results suggested that halicin has no significant impact on hematological parameters, liver function, and kidney function. Furthermore, halicin effectively treated respiratory A. pleuropneumoniae infections in murine models. These results underscore the potential of halicin as a new antibacterial agent with applications against clinically relevant pathogens in veterinary medicine.
Collapse
Affiliation(s)
- Shuge Wang
- National Key Laboratory of Veterinary Public Health and Safety, College of Veterinary Medicine, China Agricultural University, Beijing 100094, China; (S.W.); (K.Z.); (Z.C.); (D.L.); (S.T.); (C.S.)
| | - Ke Zhao
- National Key Laboratory of Veterinary Public Health and Safety, College of Veterinary Medicine, China Agricultural University, Beijing 100094, China; (S.W.); (K.Z.); (Z.C.); (D.L.); (S.T.); (C.S.)
| | - Ziqi Chen
- National Key Laboratory of Veterinary Public Health and Safety, College of Veterinary Medicine, China Agricultural University, Beijing 100094, China; (S.W.); (K.Z.); (Z.C.); (D.L.); (S.T.); (C.S.)
| | - Dejun Liu
- National Key Laboratory of Veterinary Public Health and Safety, College of Veterinary Medicine, China Agricultural University, Beijing 100094, China; (S.W.); (K.Z.); (Z.C.); (D.L.); (S.T.); (C.S.)
| | - Shusheng Tang
- National Key Laboratory of Veterinary Public Health and Safety, College of Veterinary Medicine, China Agricultural University, Beijing 100094, China; (S.W.); (K.Z.); (Z.C.); (D.L.); (S.T.); (C.S.)
| | - Chengtao Sun
- National Key Laboratory of Veterinary Public Health and Safety, College of Veterinary Medicine, China Agricultural University, Beijing 100094, China; (S.W.); (K.Z.); (Z.C.); (D.L.); (S.T.); (C.S.)
| | - Hongliang Chen
- School of Life Sciences, Xiamen University, Xiamen 361005, China;
- Xiamen Vangenes Biotechnology Co., Ltd., Xiamen 361006, China
| | - Yang Wang
- National Key Laboratory of Veterinary Public Health and Safety, College of Veterinary Medicine, China Agricultural University, Beijing 100094, China; (S.W.); (K.Z.); (Z.C.); (D.L.); (S.T.); (C.S.)
| | - Congming Wu
- National Key Laboratory of Veterinary Public Health and Safety, College of Veterinary Medicine, China Agricultural University, Beijing 100094, China; (S.W.); (K.Z.); (Z.C.); (D.L.); (S.T.); (C.S.)
| |
Collapse
|
3
|
Bui DT, Lee YS, Kuo TF, Chen ZW, Yang WC. Novel Experimental Mouse Model to Study the Pathogenesis and Therapy of Actinobacillus pleuropneumoniae Infection. Pathogens 2024; 13:412. [PMID: 38787263 PMCID: PMC11123673 DOI: 10.3390/pathogens13050412] [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: 04/18/2024] [Revised: 05/12/2024] [Accepted: 05/14/2024] [Indexed: 05/25/2024] Open
Abstract
Actinobacillus pleuropneumoniae (APP) is a major cause of lung infections in pigs. An experimental mouse has the edge over pigs pertaining to the ease of experimental operation, disease study and therapy, abundance of genetic resources, and cost. However, it is a challenge to introduce APP into a mouse lung due to the small respiratory tract of mice and bacterial host tropism. In this study, an effective airborne transmission of APP serovar 1 (APP1) was developed in mice for lung infection. Consequently, APP1 infected BALB/c mice and caused 60% death within three days of infection at the indicated condition. APP1 seemed to enter the lung and, in turn, spread to other organs of the mice over the first 5 days after infection. Accordingly, APP1 damaged the lung as evidenced by its morphological and histological examinations. Furthermore, ampicillin fully protected mice against APP1 as shown by their survival, clinical symptoms, body weight loss, APP1 count, and lung damages. Finally, the virulence of two extra APP strains, APP2 and APP5, in the model was compared based on the survival rate of mice. Collectively, this study successfully established a fast and reliable mouse model of APP which can benefit APP research and therapy. Such a model is a potentially useful model for airway bacterial infections.
Collapse
Affiliation(s)
- Duc-Thang Bui
- Agricultural Biotechnology Research Center, Academia Sinica, Taipei City 115, Taiwan; (D.-T.B.); (Y.-S.L.); (T.-F.K.)
- Institute of Biotechnology, National Taiwan University, Taipei City 106, Taiwan
| | - Yi-San Lee
- Agricultural Biotechnology Research Center, Academia Sinica, Taipei City 115, Taiwan; (D.-T.B.); (Y.-S.L.); (T.-F.K.)
- Institute of Biotechnology, National Taiwan University, Taipei City 106, Taiwan
| | - Tien-Fen Kuo
- Agricultural Biotechnology Research Center, Academia Sinica, Taipei City 115, Taiwan; (D.-T.B.); (Y.-S.L.); (T.-F.K.)
| | - Zeng-Weng Chen
- Animal Technology Research Center, Agricultural Technology Research Institute, Miaoli County 350, Taiwan;
| | - Wen-Chin Yang
- Agricultural Biotechnology Research Center, Academia Sinica, Taipei City 115, Taiwan; (D.-T.B.); (Y.-S.L.); (T.-F.K.)
- Institute of Biotechnology, National Taiwan University, Taipei City 106, Taiwan
- Department of Life Sciences, National Taiwan Ocean University, Keelung City 202, Taiwan
- Graduate Institute of Integrated Medicine, China Medical University, Taichung City 404, Taiwan
- Department of Life Sciences, National Chung-Hsing University, Taichung City 404, Taiwan
| |
Collapse
|
4
|
Wang J, Gan L, Li F, Li Q, Wu T, Wu Z, Chen P, Scicluna BP, Feng X, Gu J, Han W, Li N, Lei L. Tracheal epithelial cell-exosome-derived MiR-21-5p inhibits alveolar macrophage pyroptosis to resist pulmonary bacterial infection through PIK3CD-autophagy pathway. Life Sci 2024; 336:122340. [PMID: 38092143 DOI: 10.1016/j.lfs.2023.122340] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Revised: 06/06/2023] [Accepted: 12/06/2023] [Indexed: 12/24/2023]
Abstract
AIMS Structural cells play an important role in regulating immune cells during infection. Our aim was to determine whether structural porcine tracheal epithelial cells (PTECs) can regulate alveolar macrophages (AMs) to prevent bacterial pneumonia, explore the underlying mechanism(s) and therapeutic target. MATERIALS AND METHODS Actinobacillus pleuropneumoniae (APP) was used as the model strain for infection studies. Small RNA sequencing was used to identify differentially abundant exosome-derived miRNAs. The role of PTECs exosome-derived miR-21-5p in regulating AMs autophagy, pyroptosis, reactive oxygen species (ROS) was determined using RT-qPCR, western-blotting, flow cytometry, immunohistochemistry. Luciferase reporter assays were conducted to identify potential binding targets of miR-21-5p. The universality of miR-21-5p action on resistance to bacterial pulmonary infection was demonstrated using Klebsiella pneumoniae or Staphylococcus aureus in vitro and in vivo infection models. KEY FINDINGS MiR-21-5p was enriched in PETCs-derived exosomes, which protected AMs against pulmonary bacterial infection. Mechanistically, miR-21-5p targeted PIK3CD, to promote autophagy of AMs, which reduced the pyroptosis induced by APP infection via inhibiting the over-production of ROS, which in turn suppressed the over-expression of pro-inflammatory cytokines, and increased bacterial clearance. Importantly, the protective effect and mechanism of miR-21-5p were universal as they also occurred upon challenge with Klebsiella pneumoniae and Staphylococcus aureus. SIGNIFICANCE Our data reveals miR-21-5p can promote pulmonary resistance to bacterial infection by inhibiting pyroptosis of alveolar macrophages through the PIK3CD-autophagy-ROS pathway, suggesting PIK3CD may be a potential therapeutic target for bacterial pneumonia.
Collapse
Affiliation(s)
- Jun Wang
- State Key Laboratory for Zoonotic Diseases, College of Veterinary Medicine, Jilin University, Changchun, China
| | - Lin Gan
- State Key Laboratory for Zoonotic Diseases, College of Veterinary Medicine, Jilin University, Changchun, China
| | - Fengyang Li
- State Key Laboratory for Zoonotic Diseases, College of Veterinary Medicine, Jilin University, Changchun, China
| | - Qin Li
- State Key Laboratory for Zoonotic Diseases, College of Veterinary Medicine, Jilin University, Changchun, China
| | - Tong Wu
- State Key Laboratory for Zoonotic Diseases, College of Veterinary Medicine, Jilin University, Changchun, China
| | - Zengshuai Wu
- State Key Laboratory for Zoonotic Diseases, College of Veterinary Medicine, Jilin University, Changchun, China
| | - Peiru Chen
- State Key Laboratory for Zoonotic Diseases, College of Veterinary Medicine, Jilin University, Changchun, 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
| | - Xin Feng
- State Key Laboratory for Zoonotic Diseases, College of Veterinary Medicine, Jilin University, Changchun, China
| | - Jingmin Gu
- State Key Laboratory for Zoonotic Diseases, College of Veterinary Medicine, Jilin University, Changchun, China
| | - Wenyu Han
- State Key Laboratory for Zoonotic Diseases, College of Veterinary Medicine, Jilin University, Changchun, China
| | - Na Li
- State Key Laboratory for Zoonotic Diseases, College of Veterinary Medicine, Jilin University, Changchun, China.
| | - Liancheng Lei
- State Key Laboratory for Zoonotic Diseases, College of Veterinary Medicine, Jilin University, Changchun, China.
| |
Collapse
|
5
|
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
|
6
|
Yuan Y, An B, Xie S, Qu W, Hao H, Huang L, Luo W, Liang J, Peng D. The dose regimen formulation of doxycycline hydrochloride and florfenicol injection based on ex vivo pharmacokinetic-pharmacodynamic modeling against the Actinobacillus pleuropneumoniae in pigs. ANIMAL DISEASES 2023. [DOI: 10.1186/s44149-023-00066-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/26/2023] Open
Abstract
AbstractDoxycycline hydrochloride and florfenicol combination (DoxHcl&FF) is an effective treatment for respiratory diseases. In the study, our objective was to evaluate the activity of DoxHcl&FF against Actinobacillus pleuropneumoniae (APP) in porcine pulmonary epithelial lining fluid (PELF) and the optimal dosage scheme to avoid the development of resistance. The DoxHcl&FF was administered intramuscularly (IM) at 20 mg/kg, and the PELF was collected at different time points. The minimum inhibitory concentration (MIC) and time-mortality curves were also included in the study. Based on the sigmoid Emax equation and dose equations, the study integrated the in vivo pharmacokinetic data of infected pigs and ex vivo pharmacodynamic data to obtain the area under concentration time curve (AUC0-24h)/MIC values in PELF and achieve bacteriostatic activity, bactericidal activity and the virtual eradication of bacteria. The study showed that the combination of DoxHcl and FF caused no significant changes in PK parameters. The peak concentration (Cmax) of FF in healthy and diseased pigs was 8.87 ± 0.08 μg/mL and 8.67 ± 0.07 μg/mL, the AUC0-24h were 172.75 ± 2.52 h·μg/mL and 180.22 ± 3.13 h·μg/mL, the Cmax of DoxHcl was 7.91 ± 0.09 μg/mL and 7.99 ± 0.05 μg/mL, and the AUC0-24h was 129.96 ± 3.70 h·μg/mL and 169.82 ± 4.38 h·μg/mL. DoxHcl&FF showed strong concentration-dependent tendencies. The bacteriostatic, bactericidal, and elimination activity were calculated as 5.61, 18.83 and 32.68 h, and the doses were 1.37 (bacteriostatic), 4.59 (bactericidal) and 7.99 (elimination) mg/kg. These findings indicated that the calculated recommended dose could assist in achieving more precise administration, increasing the effectiveness of DoxHcl&FF treatment for APP infections.
Collapse
|
7
|
Jarosova R, Ondrackova P, Leva L, Nedbalcova K, Vicenova M, Masek J, Volf J, Gebauer J, Do T, Guran R, Sladek Z, Dominguez J, Faldyna M. Cytokine expression by CD163+ monocytes in healthy and Actinobacillus pleuropneumoniae-infected pigs. Res Vet Sci 2022; 152:1-9. [PMID: 35901636 DOI: 10.1016/j.rvsc.2022.07.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2021] [Revised: 06/30/2022] [Accepted: 07/18/2022] [Indexed: 11/29/2022]
Abstract
Distinct monocyte subpopulations have been previously described in healthy pigs and pigs experimentally infected with Actinobacillus pleuropneumoniae (APP). The CD163+ subpopulation of bone marrow (BM), peripheral blood (PB) and lung monocytes was found to play an important role in the inflammatory process. The inflammation is accompanied by elevation of inflammatory cytokines. The aim of the study was to evaluate the contribution of CD163+ monocytes and macrophages to cytokine production during APP-induced lung inflammation. Cytokine production was assessed by flow cytometry (FC) and quantitative PCR (qPCR) in CD163+ monocytes and by qPCR, immunohistochemistry/fluorescence in lungs and tracheobronchial lymph nodes (TBLN). Despite the systemic inflammatory response after APP infection, BM and PB CD163+ monocytes did not express elevated levels of a wide range of cytokines compared to control pigs. In contrast, significant amounts of IL-1β, IL-6, IL-8 and TNF-α were produced in lung lesions and IL-1β in the TBLN. At the protein level, TNF-α was expressed by both CD163+ monocytes and macrophages in lung lesions, whereas IL-1β, IL-6 and IL-8 expression was found only in CD163+ monocytes; no CD163+ macrophages were found to produce these cytokines. Furthermore, the quantification of CD163+ monocytes expressing the two cytokines IL-1β and IL-8 that were most elevated was performed. In lung lesions, 36.5% IL-1β positive CD163+ monocytes but only 18.3% IL-8 positive CD163+ monocytes were found. In conclusion, PB and BM CD163+ monocytes do not appear to contribute to the elevated cytokine levels in plasma. On the other hand, CD163+ monocytes contribute to inflammatory cytokine expression, especially IL-1β at the site of inflammation during the inflammatory process.
Collapse
Affiliation(s)
- Rea Jarosova
- Veterinary Research Institute, Brno, Czech Republic; Department of Morphology, Physiology and Animal Genetics, The Faculty of AgriSciences, Mendel University in Brno, Brno, Czech Republic.
| | | | - Lenka Leva
- Veterinary Research Institute, Brno, Czech Republic.
| | | | | | - Josef Masek
- Veterinary Research Institute, Brno, Czech Republic.
| | - Jiri Volf
- Veterinary Research Institute, Brno, Czech Republic.
| | - Jan Gebauer
- Veterinary Research Institute, Brno, Czech Republic.
| | - Tomas Do
- Department of Chemistry and Biochemistry, Faculty of AgriSciences, Mendel University in Brno, Brno, Czech Republic.
| | - Roman Guran
- Department of Chemistry and Biochemistry, Faculty of AgriSciences, Mendel University in Brno, Brno, Czech Republic; Central European Institute of Technology, Brno University of Technology, Brno, Czech Republic
| | - Zbysek Sladek
- Department of Morphology, Physiology and Animal Genetics, The Faculty of AgriSciences, Mendel University in Brno, Brno, Czech Republic.
| | - Javier Dominguez
- Departmento de Biotecnologia, Centro Nacional Instituto de Investigacion y Tecnologia Agraria y Alimentaria (CSIC-INIA), Madrid, Spain.
| | | |
Collapse
|
8
|
An Experimental Dermal Oedema Model for Apx Toxins of Actinobacillus pleuropneumoniae. J Comp Pathol 2022; 195:12-18. [DOI: 10.1016/j.jcpa.2022.04.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Revised: 03/18/2022] [Accepted: 04/27/2022] [Indexed: 11/21/2022]
|
9
|
Chen P, Bao C, Zhu R, Wang J, Zhu J, Li Z, Li F, Gu J, Feng X, Li N, Lei L. IL-5 enhances the resistance of Actinobacillus pleuropneumoniae infection in mice through maintaining appropriate levels of lung M2, PMN-II and highly effective neutrophil extracellular traps. Vet Microbiol 2022; 269:109438. [PMID: 35468400 DOI: 10.1016/j.vetmic.2022.109438] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Revised: 04/09/2022] [Accepted: 04/14/2022] [Indexed: 01/25/2023]
Abstract
Interleukin 5 (IL-5) regulates the maturation, activation, proliferation and function of immune cells, and plays an important role in the inflammatory response induced by an allergy. However, its anti-pathogen effect is poorly understood currently, especially on pneumonia. Here, this study was designed to elucidate the immunological role of IL-5 in the infection of mice with Actinobacillus pleuropneumoniae (APP). We established an acute lung infection model of APP in IL-5 knockout mice (IL-5-/-) and wild-type mice (WT) through nasal infusion or intraperitoneal injection, compared the survival rate, clinical symptoms, lung bacterial load, proportion of various immune cells, immune molecular expression, and neutrophil germicidal ability through flow cytometry, RT-qPCR, ELISA and immunofluorescence. Compared to WT mice, the IL-5-/- mice had a lower survival rate, more severe clinical symptoms, significantly increased bacterial load, and inflammatory cell infiltration in the lung after APP infection. In an uninfected state, IL-5 deficiency decreased the number of M1 interstitial macrophages and CD14- monocytes, while after infection, IL-5 deficiency significantly reduced the M2 alveolar macrophages, and increased PMN-II cells in the lung. Furthermore, the expression of IL-10, IL-4, IL-33, TNF-α, iNOS in the lung was lower in IL-5-/- mice under an uninfected condition, and the secretion of IL-18 was significantly increased after infection. In addition, IL-5 deficiency decreased bactericidal ability by inhibiting the formation of neutrophil extracellular traps (NETs). Collectively, these results provide evidence that IL-5 can enhance the resistance of APP infection, and its anti-infection mechanism, implying new targets and ideas for APP or similar respiratory agents' prevention and treatment.
Collapse
Affiliation(s)
- Peiru Chen
- Key Laboratory of Zoonosis, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, PR China
| | - Chuntong Bao
- Key Laboratory of Zoonosis, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, PR China
| | - Rining Zhu
- Key Laboratory of Zoonosis, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, PR China
| | - Jun Wang
- Key Laboratory of Zoonosis, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, PR China
| | - Junhui Zhu
- Key Laboratory of Zoonosis, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, PR China
| | - Ziheng Li
- Key Laboratory of Zoonosis, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, PR China
| | - Fengyang Li
- Key Laboratory of Zoonosis, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, PR China
| | - Jingmin Gu
- Key Laboratory of Zoonosis, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, PR China
| | - Xin Feng
- Key Laboratory of Zoonosis, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, PR China
| | - Na Li
- Key Laboratory of Zoonosis, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, PR China.
| | - Liancheng Lei
- Key Laboratory of Zoonosis, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, PR China; College of Animal Science, Yangtze University, Jingzhou, Hubei, 434023, PR China.
| |
Collapse
|
10
|
Chen X, Shao Z, Wu L, He B, Yang W, Chen J, Jin E, Huang Q, Lei L, Xu J, Li H, Zhang H, Wan Y, Liu W, Zhou R. Involvement of the Actinobacillus pleuropneumoniae ompW Gene in Confrontation of Environmental Pressure. Front Vet Sci 2022; 9:846322. [PMID: 35664844 PMCID: PMC9161549 DOI: 10.3389/fvets.2022.846322] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Accepted: 04/28/2022] [Indexed: 11/13/2022] Open
Abstract
Actinobacillus pleuropneumoniae causes porcine pleuropneumonia. The function of the outer membrane protein W gene (ompW) of A. pleuropneumoniae has not been evaluated. Thus a deletion mutant of ompW, ΔompW, was constructed to explore the effect of ompW gene deletion on bacterial growth, biofilm formation, bacterial morphology, oxidative tolerance, susceptibility to antibiotics, and the expression of ribosome synthesis and ABC transporter related genes. Results showed that the ompW gene deletion did not affect biofilm formation and the growth of A. pleuropneumoniae but did affect bacterial morphology during steady growth, oxidative tolerance, and bacterial susceptibility to polymyxin B, kanamycin, and penicillin. The ompW gene deletion also affected the expression of ribosome synthesis and ABC transporter related genes. These results suggested that ompW may regulate the biological phenotype of A. pleuropneumoniae.
Collapse
Affiliation(s)
- Xiabing Chen
- Institute of Animal Husbandry and Veterinary Science, Wuhan Academy of Agricultural Sciences, Wuhan, China
- *Correspondence: Xiabing Chen
| | - Zhiyong Shao
- Institute of Animal Husbandry and Veterinary Science, Wuhan Academy of Agricultural Sciences, Wuhan, China
| | - Lijun Wu
- Institute of Animal Husbandry and Veterinary Science, Wuhan Academy of Agricultural Sciences, Wuhan, China
| | - Bin He
- Institute of Animal Husbandry and Veterinary Science, Wuhan Academy of Agricultural Sciences, Wuhan, China
| | - Wenhai Yang
- Institute of Animal Husbandry and Veterinary Science, Wuhan Academy of Agricultural Sciences, Wuhan, China
| | - Jie Chen
- Institute of Animal Husbandry and Veterinary Science, Wuhan Academy of Agricultural Sciences, Wuhan, China
| | - Erguang Jin
- Institute of Animal Husbandry and Veterinary Science, Wuhan Academy of Agricultural Sciences, Wuhan, China
| | - Qi Huang
- State Key Laboratory of Agricultural Microbiology, College of Animal Science and Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
- Cooperative Innovation Center for Sustainable Pig Production, College of Animal Science and Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Liancheng Lei
- College of Veterinary Medicine and College of Animal Science, Jilin University, Changchun, China
| | - Jiajia Xu
- State Key Laboratory of Agricultural Microbiology, College of Animal Science and Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
- Cooperative Innovation Center for Sustainable Pig Production, College of Animal Science and Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Haotian Li
- State Key Laboratory of Agricultural Microbiology, College of Animal Science and Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
- Cooperative Innovation Center for Sustainable Pig Production, College of Animal Science and Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Hui Zhang
- State Key Laboratory of Agricultural Microbiology, College of Animal Science and Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
- Cooperative Innovation Center for Sustainable Pig Production, College of Animal Science and Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Yun Wan
- Wuhan Animal Disease Control Center, Wuhan, China
| | - Wu Liu
- Institute of Animal Husbandry and Veterinary Science, Wuhan Academy of Agricultural Sciences, Wuhan, China
- Wu Liu
| | - Rui Zhou
- State Key Laboratory of Agricultural Microbiology, College of Animal Science and Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
- Cooperative Innovation Center for Sustainable Pig Production, College of Animal Science and Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
- Rui Zhou
| |
Collapse
|
11
|
Bao C, Liu B, Zhu R, Xiao J, Li Z, Jiang H, Wang B, Langford PR, Fei R, Li N, Lei L. IFN-γ -/- Mice Resist Actinobacillus pleuropneumoniae Infection by Promoting Early Lung IL-18 Release and PMN-I Accumulation. Infect Immun 2021; 89:e00069-21. [PMID: 33685942 PMCID: PMC8316061 DOI: 10.1128/iai.00069-21] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Accepted: 03/01/2021] [Indexed: 12/15/2022] Open
Abstract
Porcine pleuropneumonia is a common infectious disease of pigs caused by Actinobacillus pleuropneumoniae Interferon gamma (IFN-γ) expression increases in the lung of pigs after A. pleuropneumoniae infection, but the role of IFN-γ during the infection is still obscure. In this study, an IFN-γ-/- mouse infection model was established, and bacterial load, levels of inflammatory cytokines, and types of neutrophils in the lungs were studied at different times post-A. pleuropneumoniae infection. We found that wild-type (WT) mice were more susceptible to A. pleuropneumoniae than IFN-γ-/- mice. At 6 h postinfection (hpi), the expression of interleukin 18 (IL-18) and IL-1β in the lungs of IFN-γ-/- mice was significantly increased compared to WT mice. The bacterial load and levels of inflammatory cytokines (IL-1β and IL-6) of IFN-γ-/- mice were significantly reduced at 12 hpi compared to WT mice. After an initial loss, the numbers of lung polymorphonuclear (PMN)-I cells dramatically increased in the lungs of IFN-γ-/- but not WT mice, whereas PMN-II cells continually decreased. Finally, in vivo administration of IL-18 significantly reduced clinical scores and bacterial load in the lungs of A. pleuropneumoniae-infected mice. This study identifies IFN-γ as a target for regulating the inflammatory response in the lung and provides a basis for understanding the course of clinical bacterial pneumonia and for the formulation of treatment protocols.
Collapse
Affiliation(s)
- Chuntong Bao
- College of Veterinary Medicine, Jilin University, Changchun, People's Republic of China
| | - Baijun Liu
- College of Veterinary Medicine, Jilin University, Changchun, People's Republic of China
| | - Rining Zhu
- College of Veterinary Medicine, Jilin University, Changchun, People's Republic of China
| | - Jiameng Xiao
- College of Veterinary Medicine, Jilin University, Changchun, People's Republic of China
| | - Ziheng Li
- College of Veterinary Medicine, Jilin University, Changchun, People's Republic of China
| | - Hexiang Jiang
- College of Veterinary Medicine, Jilin University, Changchun, People's Republic of China
| | - Beinan Wang
- Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, People's Republic of China
| | - P R Langford
- Section of Paediatric Infectious Diseases, Imperial College London, London, UK
| | - Rui Fei
- School of Basic Medicine, Jilin University, Changchun, People's Republic of China
| | - Na Li
- College of Veterinary Medicine, Jilin University, Changchun, People's Republic of China
| | - Liancheng Lei
- College of Veterinary Medicine, Jilin University, Changchun, People's Republic of China
- College of Animal Science, Yangtze University, Jingzhou, Hubei, People's Republic of China
| |
Collapse
|
12
|
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: 0.6] [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
|
13
|
Bao C, Jiang H, Zhu R, Liu B, Xiao J, Li Z, Chen P, Langford PR, Zhang F, Lei L. Differences in pig respiratory tract and peripheral blood immune responses to Actinobacillus pleuropneumoniae. Vet Microbiol 2020; 247:108755. [PMID: 32686648 DOI: 10.1016/j.vetmic.2020.108755] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2019] [Revised: 04/25/2020] [Accepted: 06/11/2020] [Indexed: 11/22/2022]
Abstract
Excessive cytokine production is an important component of the acute respiratory distress syndrome and multiple organ failure. Pneumonia can lead to an overexpression of cytokines, although comparatively little is known about the relevance and differences in cytokines between blood and lung. In this study, piglets were experimentally infected intranasally with Actinobacillus pleuropneumoniae (APP), and transcriptomes of lung tissue and peripheral blood mononuclear cells determined. In addition, the levels of 30 cytokines in broncheoalveolar lavage fluid (BALF) and sera were determined by ELISA. Post infection, there was an early increase in lung monocytes, and a later rise in inflammatory cytokines in BALF. Blood lymphocytes increased early in infection and there was a rise in inflammatory cytokines in the peripheral blood of infected piglets. Genes involved in cytokine production, leukocyte migration and differentiation, lymphocyte activation, and cytokine-mediated signaling pathways in the transcriptomes of lung tissue were significantly down-regulated early in infection. At this early phase of APP infection (0-6 h), the cytokines IL-1β, MCP-1, and IL-5 in sera increased rapidly and significantly, while many cytokines in BALF decreased. At 48 h post-infection, cytokines in sera were no longer significantly increased, although some were up-regulated in BALF, and there was aggravated pathological damage in the lungs at this time. The data indicate there are substantial differences between immune cells and cytokines in the lung and peripheral blood of APP infected piglets at equivalent time points. The results increase our understanding of pig-APP host interactive biology, and will be important in formulating future therapeutic and preventative strategies to prevent disease caused by APP.
Collapse
Affiliation(s)
- Chuntong Bao
- College of Veterinary Medicine, Jilin University, Changchun, PR China
| | - Hexiang Jiang
- College of Veterinary Medicine, Jilin University, Changchun, PR China
| | - Rining Zhu
- College of Veterinary Medicine, Jilin University, Changchun, PR China
| | - Baijun Liu
- College of Veterinary Medicine, Jilin University, Changchun, PR China
| | - Jiameng Xiao
- College of Veterinary Medicine, Jilin University, Changchun, PR China
| | - Ziheng Li
- College of Veterinary Medicine, Jilin University, Changchun, PR China
| | - Peiru Chen
- College of Veterinary Medicine, Jilin University, Changchun, PR China
| | - Paul R Langford
- Section of Paediatric Infectious Disease, Imperial College London, London, UK
| | - Fuxian Zhang
- College of Animal Science, Yangtze University, Jingzhou, Hubei, 434023, PR China.
| | - Liancheng Lei
- College of Veterinary Medicine, Jilin University, Changchun, PR China; College of Animal Science, Yangtze University, Jingzhou, Hubei, 434023, PR China.
| |
Collapse
|
14
|
Zhu R, Bao C, Liu B, Xiao J, Sun C, Feng X, Langford PR, Li Y, Lei L. iTRAQ-based quantitative proteomic analysis of peripheral blood serum in piglets infected with Actinobacillus pleuropneumoniae. AMB Express 2020; 10:121. [PMID: 32632500 PMCID: PMC7338327 DOI: 10.1186/s13568-020-01057-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2020] [Accepted: 07/02/2020] [Indexed: 12/15/2022] Open
Abstract
Porcine pleuropneumonia caused by Actinobacillus pleuropneumoniae (APP) is a swine respiratory disease with an important impact around the world either as a single infection or part of the porcine respiratory disease complex. The data of interaction between hosts and pathogens has becoming more crucial for exploration of the mechanism. However, up to now, comparatively little information is available on the systemic and dynamic changes that occur in pig serum in response to APP infection. This study used iTRAQ to identify differentially expressed proteins (DEPs) in pig serum in response to APP infection. Compared with the APP un-infected group (S0),there were 137 up-regulated and 68 down-regulated proteins at 24 h (S24), and 81 up-regulated and 107 down-regulated proteins at 120 h (S120). At 24 h, the immune response was not significantly enriched, but cell adhesion, cytosol, Golgi apparatus, GTP and ATP binding and regulation of cell cycle were extremely active, implying host preparation of immune response starting. Subsequently, innate immune response, negative regulation of apoptotic process, immunological synapse, adaptive immune response, the regulation of inflammatory response, positive regulation of T cell proliferation were more enhanced at 120 h then that of 24 h, representing innate immunity transferring to the adaptive, while endocytosis, cell adhesion and platelet aggregation showed obvious decline. The pathways of T cell receptor signaling pathway, cytokine–cytokine receptor interaction, complement and coagulation cascades, leukocyte transendothelial migration were active remarkably during all infection period, and more pathways could connect to form innate immune defense networks. Surprisingly, the pathways like amoebiasis, rheumatoid arthritis and malaria had been found up-regulated. As a conclusion, APP could delay host inflammatory response to the infection at early stage, and induced innate immunity to convert from adhesion, interaction into complement activation, proteasome digestion, bacterial invasion at later stage. This would increase our understanding of the porcine distinct response to APP infection.
Collapse
|
15
|
Gao L, Zhang L, Xu H, Zhao F, Ke W, Chen J, Yang J, Qi C, Liu J. The Actinobacillus pleuropneumoniae sulfate-binding protein is required for the acquisition of sulfate and methionine, but is not essential for virulence. Vet Microbiol 2020; 245:108704. [DOI: 10.1016/j.vetmic.2020.108704] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2019] [Revised: 04/23/2020] [Accepted: 04/23/2020] [Indexed: 12/20/2022]
|
16
|
The antimicrobial peptide MPX kills Actinobacillus pleuropneumoniae and reduces its pathogenicity in mice. Vet Microbiol 2020; 243:108634. [PMID: 32273013 DOI: 10.1016/j.vetmic.2020.108634] [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: 08/13/2019] [Revised: 02/22/2020] [Accepted: 03/02/2020] [Indexed: 01/03/2023]
Abstract
Actinobacillus pleuropneumoniae is the causative agent of highly contagious and fatal respiratory infections, causing substantial economic losses to the global pig industry. Due to increased antibiotic resistance, there is an urgent need to find new antibiotic alternatives for treating A. pleuropneumoniae infections. MPX is obtained from wasp venom and has a killing effect on various bacteria. This study found that MPX had a good killing effect on A. pleuropneumoniae and that the minimum inhibitory concentration (MIC) was 16 μg/mL. The bacterial density of A. pleuropneumoniae decreased 1000 times after MPX (1 × MIC) treatment for 1 h, and the antibacterial activity was not affected by pH or temperature. Fluorescence microscopy showed that MPX (1 × MIC) destroyed the bacterial cell membrane after treatment for 0.5 h, increasing membrane permeability and releasing bacterial proteins and Ca2+, Na+ and other cations. In addition, MPX (1 × MIC) treatment significantly reduced the formation of bacterial biofilms. Quantitative RT-PCR results showed that MPX treatment significantly upregulated the expression of the PurC virulence gene and downregulated that of ApxI, ApxII, and Apa1. In addition, the Sap A gene was found to play an important role in the tolerance of A. pleuropneumoniae to antimicrobial peptides. Therapeutic evaluation in a murine model showed that MPX protects mice from a lethal dose of A. pleuropneumoniae and relieves lung inflammation. This study reports the use of MPX to treat A. pleuropneumonia infections, laying the foundation for the development of new drugs for bacterial infections.
Collapse
|
17
|
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: 0.6] [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
|