1
|
Frazzini S, Turin L, Vanosi G, Rossi L, Hejna M. Seaweed-derived mixed extracts exhibit immunomodulatory properties on porcine alveolar macrophages. Vet J 2025; 312:106358. [PMID: 40246016 DOI: 10.1016/j.tvjl.2025.106358] [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: 11/16/2024] [Revised: 04/10/2025] [Accepted: 04/12/2025] [Indexed: 04/19/2025]
Abstract
Antimicrobial resistance is a growing global concern, prompting for antibiotic alternatives in animal production. Seaweed, abundant in bioactive compounds with anti-inflammatory properties, offers a natural substitute to synthetic compounds. Considering this, the objective of the present study was to evaluate the anti-inflammatory bioactivity of three seaweeds 1:1 combination of Ascophyllum nodosum, Palmaria palmata, and Ulva lactuca. Initially, polyphenol, flavonoid, and total phlorotannin content of the three seaweed species were assessed through colorimetric assays. Subsequently, the anti-inflammatory bioactivity was first evaluated through an inhibition protein precipitation assay and then confirmed in vitro through gene expression assays in LPS-stimulated porcine alveolar macrophages (PAMs). The evaluation of the bioactive molecules revealed a high content of TPC (1487.67 ± 40.39 and 1763.57 ± 69.01 mg TAE/100 mg of sample, respectively), as well as of TFC (95.68 ± 3.62 and 126.09 ± 7.34 mg CE/100 mg of sample) and TPhC (0.167 ± 0.02 and 0.23 ± 0.01 mg PGE/100 mg) for AN and UL, respectively. The assay for inhibiting protein precipitation disclosed that the extracts combining two algae species (ANUL, ANPP, PPUL) were more effective than the effect exhibited by each single extract. The assessment of anti-inflammatory bioactivity revealed a significant down-regulation of IL-1β and TNF-α in the algae combination extracts. In contrast, TGF-β showed an increasing trend. These findings, along with confirmation of the high content of bioactive molecules, highlight the algae's anti-inflammatory potential, making them suitable as natural alternatives to antibiotics for disease prevention in the livestock sector. Therefore, future research should explore the specific bioactive compounds and validate their efficacy in vivo to confirm their potential use in animal production.
Collapse
Affiliation(s)
- Sara Frazzini
- Department of Veterinary Medicine and Animal Sciences - DIVAS, Università degli Studi di Milano, dell'Università 6, Lodi 26900, Italy
| | - Lauretta Turin
- Department of Veterinary Medicine and Animal Sciences - DIVAS, Università degli Studi di Milano, dell'Università 6, Lodi 26900, Italy
| | - Graziella Vanosi
- Department of Veterinary Medicine and Animal Sciences - DIVAS, Università degli Studi di Milano, dell'Università 6, Lodi 26900, Italy
| | - Luciana Rossi
- Department of Veterinary Medicine and Animal Sciences - DIVAS, Università degli Studi di Milano, dell'Università 6, Lodi 26900, Italy.
| | - Monika Hejna
- Department of Biotechnology and Nutrigenomics, Institute of Genetics and Animal Biotechnology of the Polish Academy of Sciences, Postępu 36A, Jastrzębiec 05-552, Poland
| |
Collapse
|
2
|
Yan K, He Q, Tang J, Peng W, Dou B, Chen H, Bei W. Actinobacillus pleuropneumoniae infection activates IL-1β expression in porcine alveolar macrophages via β-amyloid production. Microb Pathog 2025; 204:107559. [PMID: 40220800 DOI: 10.1016/j.micpath.2025.107559] [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: 12/21/2024] [Revised: 03/30/2025] [Accepted: 04/07/2025] [Indexed: 04/14/2025]
Abstract
Actinobacillus pleuropneumoniae (A. pleuropneumoniae), a porcine respiratory tract pathogen, causes porcine pleuropneumonia. Porcine alveolar macrophages (PAMs) play a crucial role during A. pleuropneumoniae infection. Amyloid precursor protein (APP) can be cleaved by β- and γ-secretase to produce β-amyloid (Aβ). APP and Aβ are associated with the inflammatory response. They activate microglia and astrocytes to secrete IL-1β, IL-6, and other cytokines. In this study, we found that during the interaction between A. pleuropneumoniae and PAMs, the two-component system CpxAR upregulates wecA expression, increasing lipopolysaccharide (LPS) production. LPS promotes APP production and cleavage to generate Aβ. The Aβ activates NF-κB, leading to increased IL-1β expression. We hypothesize that A. pleuropneumoniae infection of PAMs regulates APP production and cleavage to control Aβ levels. Different quantities of Aβ induce PAMs to produce varying amounts of cytokines, leading to different pathological processes in porcine pleuropneumonia.
Collapse
Affiliation(s)
- Kang Yan
- National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, China; The Cooperative Innovation Center for Sustainable Pig Production, Huazhong Agricultural University, Wuhan, Hubei, China
| | - Qiyun He
- National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, China; The Cooperative Innovation Center for Sustainable Pig Production, Huazhong Agricultural University, Wuhan, Hubei, China
| | - Jia Tang
- National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, China; The Cooperative Innovation Center for Sustainable Pig Production, Huazhong Agricultural University, Wuhan, Hubei, China
| | - Wei Peng
- National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, China; The Cooperative Innovation Center for Sustainable Pig Production, Huazhong Agricultural University, Wuhan, Hubei, China
| | - Beibei Dou
- National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, China
| | - Huanchun Chen
- National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, China; The Cooperative Innovation Center for Sustainable Pig Production, Huazhong Agricultural University, Wuhan, Hubei, China; Hubei Hongshan Laboratory, Wuhan, China
| | - Weicheng Bei
- National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, China; The Cooperative Innovation Center for Sustainable Pig Production, Huazhong Agricultural University, Wuhan, Hubei, China; Hubei Hongshan Laboratory, Wuhan, China.
| |
Collapse
|
3
|
Ma B, Wan Y, Zhuo R, Gong H, Gao Y, Luo R, Hu X, Hua K, Xiao Y, Jin H. Glaesserella parasuis induces tissue transglutaminase-mediated fibrinogen crosslinking through NF-κB activation. Vet Microbiol 2025; 302:110418. [PMID: 39919498 DOI: 10.1016/j.vetmic.2025.110418] [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: 09/03/2024] [Revised: 01/19/2025] [Accepted: 01/27/2025] [Indexed: 02/09/2025]
Abstract
Glaesserella parasuis (GPS) is the causative agent of Glässer's disease, leading to significant economic losses in the global swine industry. During post-mortem inspection, the main observation in pigs affected by Glässer's disease is the presence of serofibrinous or fibrinopurulent exudate on the mucosal surface. Nevertheless, the mechanism by which fibrinogen is converted into a fibrin clot during Glässer's disease is not fully understood. In this study, we discovered in the liver, lung, and kidney, that GPS infection upregulates the expression of tissue transglutaminase (tTG) and promotes the co-localization of tTG with fibrin. In porcine aortic endothelial cells, knockdown of tTG significantly reduced fibrinogen cross-linking and pro-inflammatory factor production after GPS infection. In addition, in investigating the mechanism of tTG upregulation by GPS infection, inhibitor assays revealed the involvement of the NF-κB signaling pathway in the upregulation of tTG expression during GPS infection. Further data from dual-luciferase assays and chromatin immunoprecipitation confirmed that phosphorylated p65 binding to the tTG promoter sequences increased tTG expression and the specific binding site was discovered at GACCTTCCCT (-1082 to -1072 bp), AGGGAAATTG (-807 to -797 bp), and TAAGTTCCCC (+22 to +32 bp). The above results indicate that GPS infection may promote the cross-linking of fibrinogen by tTG, thereby mediating exudative fibrinous inflammation, providing new insights into the pathogenesis of GPS infection, and suggesting potential molecular targets for therapeutic intervention.
Collapse
Affiliation(s)
- Bin Ma
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, Hubei 430070, China; College of Veterinery Medicine, Huazhong Agricultural University, Wuhan, Hubei 430070, China; Hubei Provincial Key Laboratory of Preventive Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei 430070, China
| | - Yanxi Wan
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, Hubei 430070, China; College of Veterinery Medicine, Huazhong Agricultural University, Wuhan, Hubei 430070, China; Hubei Provincial Key Laboratory of Preventive Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei 430070, China
| | - Ran Zhuo
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, Hubei 430070, China; College of Veterinery Medicine, Huazhong Agricultural University, Wuhan, Hubei 430070, China; Hubei Provincial Key Laboratory of Preventive Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei 430070, China
| | - Huimin Gong
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, Hubei 430070, China; College of Veterinery Medicine, Huazhong Agricultural University, Wuhan, Hubei 430070, China; Hubei Provincial Key Laboratory of Preventive Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei 430070, China
| | - Yuan Gao
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, Hubei 430070, China; College of Veterinery Medicine, Huazhong Agricultural University, Wuhan, Hubei 430070, China; Hubei Provincial Key Laboratory of Preventive Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei 430070, China
| | - Rui Luo
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, Hubei 430070, China; College of Veterinery Medicine, Huazhong Agricultural University, Wuhan, Hubei 430070, China; Hubei Provincial Key Laboratory of Preventive Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei 430070, China
| | - Xueying Hu
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, Hubei 430070, China; College of Veterinery Medicine, Huazhong Agricultural University, Wuhan, Hubei 430070, China; Hubei Provincial Key Laboratory of Preventive Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei 430070, China
| | - Kexin Hua
- Swine Genome and Breeding Team, Yazhouwan National Laboratory, Sanya, Hainan 572024, China
| | - Yuncai Xiao
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, Hubei 430070, China; College of Veterinery Medicine, Huazhong Agricultural University, Wuhan, Hubei 430070, China; Hubei Provincial Key Laboratory of Preventive Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei 430070, China
| | - Hui Jin
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, Hubei 430070, China; College of Veterinery Medicine, Huazhong Agricultural University, Wuhan, Hubei 430070, China; Hubei Provincial Key Laboratory of Preventive Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei 430070, China.
| |
Collapse
|
4
|
Huang J, Kang W, Yi D, Zhu S, Xiang Y, Liu C, Li H, Dai D, Su J, He J, Liang Z. Intranasal B5 promotes mucosal defence against Actinobacillus pleuropneumoniae via ameliorating early immunosuppression. Virulence 2024; 15:2316459. [PMID: 38378464 PMCID: PMC10880497 DOI: 10.1080/21505594.2024.2316459] [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: 08/10/2023] [Accepted: 02/04/2024] [Indexed: 02/22/2024] Open
Abstract
Actinobacillus pleuropneumoniae (APP) is an important pathogen of the porcine respiratory disease complex, which leads to huge economic losses worldwide. We previously demonstrated that Pichia pastoris-producing bovine neutrophil β-defensin-5 (B5) could resist the infection by the bovine intracellular pathogen Mycobacterium bovis. In this study, the roles of synthetic B5 in regulating mucosal innate immune response and protecting against extracellular APP infection were further investigated using a mouse model. Results showed that B5 promoted the production of tumour necrosis factor (TNF)-α, interleukin (IL)-1β, and interferon (IFN)-β in macrophages as well as dendritic cells (DC) and enhanced DC maturation in vitro. Importantly, intranasal B5 was safe and conferred effective protection against APP via reducing the bacterial load in lungs and alleviating pulmonary inflammatory damage. Furthermore, in the early stage of APP infection, we found that intranasal B5 up-regulated the secretion of TNF-α, IL-1β, IL-17, and IL-22; enhanced the rapid recruitment of macrophages, neutrophils, and DC; and facilitated the generation of group 3 innate lymphoid cells in lungs. In addition, B5 activated signalling pathways associated with cellular response to IFN-β and activation of innate immune response in APP-challenged lungs. Collectively, B5 via the intranasal route can effectively ameliorate the immune suppression caused by early APP infection and provide protection against APP. The immunization strategy may be applied to animals or human respiratory bacterial infectious diseases. Our findings highlight the potential importance of B5, enhancing mucosal defence against intracellular bacteria like APP which causes early-phase immune suppression.
Collapse
Affiliation(s)
- Jingsheng Huang
- College of Animal Science and Technology, Guangxi University, Nanning, China
| | - Weichao Kang
- College of Animal Science and Technology, Guangxi University, Nanning, China
| | - Dandan Yi
- College of Animal Science and Technology, Guangxi University, Nanning, China
| | - Shuxin Zhu
- College of Animal Science and Technology, Guangxi University, Nanning, China
| | - Yifei Xiang
- College of Animal Science and Technology, Guangxi University, Nanning, China
| | - Chengzhi Liu
- College of Animal Science and Technology, Guangxi University, Nanning, China
| | - Han Li
- College of Animal Science and Technology, Guangxi University, Nanning, China
| | - Dejia Dai
- College of Animal Science and Technology, Guangxi University, Nanning, China
| | - Jieyu Su
- College of Animal Science and Technology, Guangxi University, Nanning, China
| | - Jiakang He
- College of Animal Science and Technology, Guangxi University, Nanning, China
| | - Zhengmin Liang
- College of Animal Science and Technology, Guangxi University, Nanning, China
| |
Collapse
|
5
|
Pang Z, Chen S, Cui S, Zhai W, Huang Y, Gao X, Wang Y, Jiang F, Guo X, Hao Y, Li W, Wang L, Zhu H, Wu J, Jia H. Identification of Potential miRNA-mRNA Regulatory Network Associated with Regulating Immunity and Metabolism in Pigs Induced by ASFV Infection. Animals (Basel) 2023; 13:ani13071246. [PMID: 37048502 PMCID: PMC10093425 DOI: 10.3390/ani13071246] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Revised: 03/14/2023] [Accepted: 03/16/2023] [Indexed: 04/07/2023] Open
Abstract
African swine fever (ASF) is a devastating infectious disease in domestic pigs caused by African swine fever virus (ASFV) with a mortality rate of about 100%. However, the understanding of the interaction between ASFV and host is still not clear. In this study, the expression differences and functional analysis of microRNA (miRNA) in porcine peripheral blood lymphocytes of ASFV infected pigs and healthy pigs were compared based on Illumina high-throughput sequencing, then the GO and KEGG signal pathways were analyzed. The miRNA related to immunity and inflammation were screened, and the regulatory network of miRNA-mRNA was drawn. A total of 70 differentially expressed miRNAs were found (p ≤ 0.05). Of these, 45 were upregulated and 25 were downregulated in ASFV-infected pigs vs. healthy pigs. A total of 8179 mRNA genes targeted by these 70 differentially expressed miRNA were predicted, of which 1447 mRNA genes were targeted by ssc-miR-2320-5p. Five differentially expressed miRNA were validated by RT-qPCR, which were consistent with the RNA-Seq results. The GO analysis revealed that a total of 30 gene functions were significantly enriched, including 7 molecular functions (MF), 13 cellular components (CC), and 10 biological processes (BP). The KEGG enrichment analysis revealed that the differentially expressed genes were significantly enriched in pathways related to immunity, inflammation, and various metabolic processes, in which a total of two downregulated miRNAs after infection and eight upregulated miRNAs related to immunity and inflammation were screened in ASFV-infected pigs vs. healthy pigs. The network of miRNA-mRNA showed that the mRNA target genes were strongly regulated by ssc-miR-214, ssc-miR-199b-3p, and ssc-miR-199a-3p. The mRNA target genes were enriched into the MAPK signaling pathway, Toll-like receptor signaling pathway, TNF signaling pathway, and IL-17 signaling pathway by using a KEGG enrichment analysis. Therefore, ASFV could regulate immunity and metabolism-related pathways in infected pigs by inducing differential expression of miRNAs. These results provided a new basis for further elucidating the interactions between ASFV and the host as well as the immunity regulation mechanisms of ASFV, which will be conducive to better controlling ASF.
Collapse
Affiliation(s)
- Zhongbao Pang
- Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100081, China
| | - Shiyu Chen
- Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100081, China
| | - Shuai Cui
- College of Animal Medicine, Shandong Vocational Animal Science and Veterinary College, Weifang 261061, China
| | - Wenzhu Zhai
- Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100081, China
| | - Ying Huang
- Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100081, China
| | - Xintao Gao
- Biotechnology Research Institute, Chinese Academy of Agricultural Sciences, Beijing 100081, China
| | - Yang Wang
- Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100081, China
| | - Fei Jiang
- China Animal Disease Control Center, Beijing 100026, China
| | - Xiaoyu Guo
- Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100081, China
| | - Yuxin Hao
- China Animal Disease Control Center, Beijing 100026, China
| | - Wencai Li
- China Animal Disease Control Center, Beijing 100026, China
| | - Lei Wang
- China Animal Disease Control Center, Beijing 100026, China
| | - Hongfei Zhu
- Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100081, China
| | - Jiajun Wu
- China Animal Disease Control Center, Beijing 100026, China
| | - Hong Jia
- Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100081, China
| |
Collapse
|
6
|
Zhu J, Zhu R, Jiang H, Li Z, Jiang X, Li F, Zhang F, Feng X, Gu J, Li N, Lei L. Adh Promotes Actinobacillus pleuropneumoniae Survival in Porcine Alveolar Macrophages by Inhibiting CHAC2-Mediated Respiratory Burst and Inflammatory Cytokine Expression. Cells 2023; 12:696. [PMID: 36899832 PMCID: PMC10001268 DOI: 10.3390/cells12050696] [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: 12/28/2022] [Revised: 01/30/2023] [Accepted: 02/14/2023] [Indexed: 02/25/2023] Open
Abstract
Actinobacillus pleuropneumoniae (A. pleuropneumoniae) causes porcine pleuropneumonia that seriously endangers pig's health. Adh, located in the head region of trimeric autotransporter adhesion of A. pleuropneumoniae, affects bacterial adhesion and pathogenicity. However, how Adh mediates A. pleuropneumoniae immune invasion is still unclear. Here, we established the A. pleuropneumoniae strain L20 or L20 ΔAdh-infected porcine alveolar macrophages (PAM) model, and applied protein overexpression, RNA interference, qRT-PCR, Western blot and immunoflourescence techniques to dissect the effects of Adh on PAM during A. pleuropneumoniae infection. We found that Adh could increase the A. pleuropneumoniae adhesion and intracellular survival in PAM. Gene chip analysis of piglet lungs further showed that Adh significantly induced cation transport regulatory-like protein 2 (CHAC2) expression, whose overexpression suppressed the phagocytic capacity of PAM. Furthermore, CHAC2 overexpression dramatically increased glutathione (GSH) expression, decreased reactive oxygen species (ROS), and promoted A. pleuropneumoniae survival in PAM, while the knockdown of CHAC2 reversed these phenomena. Meanwhile, CHAC2 silence activated the NOD1/NF-κB pathway, resulting in an increase in IL-1β, IL-6, and TNF-α expression, whereas this effect was weakened by CHAC2 overexpression and addition of NOD1/NF-κB inhibitor ML130. Moreover, Adh enhanced the secretion of LPS of A. pleuropneumoniae, which regulated the expression of CHAC2 via TLR4. In conclusion, through a LPS-TLR4-CHAC2 pathway, Adh inhibits respiratory burst and inflammatory cytokines expression to promote A. pleuropneumoniae survival in PAM. This finding may provide a novel target for the prevention and treatment of A. pleuropneumoniae.
Collapse
Affiliation(s)
- Junhui Zhu
- College of Veterinary Medicine, Jilin University, Changchun 130062, China
| | - Rining Zhu
- College of Veterinary Medicine, Jilin University, Changchun 130062, China
| | - Hexiang Jiang
- College of Veterinary Medicine, Jilin University, Changchun 130062, China
| | - Ziheng Li
- College of Veterinary Medicine, Jilin University, Changchun 130062, China
| | - Xuan Jiang
- College of Veterinary Medicine, Jilin University, Changchun 130062, China
| | - Fengyang Li
- College of Veterinary Medicine, Jilin University, Changchun 130062, China
| | - Fuxian Zhang
- College of Animal Science, Yangtze University, Jingzhou 434025, China
| | - Xin Feng
- College of Veterinary Medicine, Jilin University, Changchun 130062, China
| | - Jingmin Gu
- College of Veterinary Medicine, Jilin University, Changchun 130062, China
| | - Na Li
- College of Veterinary Medicine, Jilin University, Changchun 130062, China
| | - Liancheng Lei
- College of Veterinary Medicine, Jilin University, Changchun 130062, China
- College of Animal Science, Yangtze University, Jingzhou 434025, China
| |
Collapse
|
7
|
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: 1.0] [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
|
8
|
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
|
9
|
Swain T, Chavez C, Myers MJ. Effects of swine microRNA mimics on lipopolysaccharide (LPS) induced inflammatory changes in 3D4/21 cells. Res Vet Sci 2022; 150:115-121. [PMID: 35816767 DOI: 10.1016/j.rvsc.2022.06.017] [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: 09/24/2021] [Revised: 05/07/2022] [Accepted: 06/28/2022] [Indexed: 11/27/2022]
Abstract
There have been limited studies focused on validation of swine microRNAs (miRNA) with mRNA targets. The objective of this study was to validate a defined set of targets using artificial miRNA mimics transfected into cell lines to confirm specific targets of endogenous miRNAs after administration of Escherichia coli lipopolysaccharide (LPS). Sixteen hours after mimic transfection of 3D4/21 cell lines, the cells were stimulated with 1 μg/ml LPS or phosphate-buffered saline (PBS). The cells were harvested and collected at 0, 1, 3, and 8 h post administration. The selected genes DAD1, IL8, and ESR, which are involved in known pathways of inflammation. and are predicted or validated human targets of either miR-146a, let-7a, or miR-22-3p. These were then evaluated by quantitative real-time-PCR (qRT-PCR) to verify microRNA-mRNA interaction in swine. Using the ROX reference dye, mRNA changes in expression were assessed using the comparative CT Method (ΔΔCT method) for normalization against the PBS control group. DAD1 and ESR1 were negatively regulated by miR-22-3p and miR-146a-5p, respectively in 3D4/21 cells after LPS stimulation. However, miR-146a-5p may play an indirect positive regulatory role of both DAD1 and IL8 mRNA expression. Furthermore, we found an inverse relationship between LPS stimulation compared with the let-7a-5p overexpression with DAD1. Our inflammation study provides new evidence on the roles and predicted targets of miR-146a, let-7a, and miR-22-3p in swine.
Collapse
Affiliation(s)
- Trevon Swain
- U.S. Food and Drug Administration Center for Veterinary Medicine, Laurel, MD 20708, United States of America
| | - Chris Chavez
- U.S. Food and Drug Administration Center for Veterinary Medicine, Laurel, MD 20708, United States of America
| | - Michael J Myers
- U.S. Food and Drug Administration Center for Veterinary Medicine, Laurel, MD 20708, United States of America.
| |
Collapse
|
10
|
Hua K, Wang M, Jin Y, Gao Y, Luo R, Bi D, Zhou R, Jin H. P38 MAPK pathway regulates the expression of resistin in porcine alveolar macrophages via Ets2 during Haemophilus parasuis stimulation. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2022; 128:104327. [PMID: 34863954 DOI: 10.1016/j.dci.2021.104327] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Revised: 11/30/2021] [Accepted: 11/30/2021] [Indexed: 06/13/2023]
Abstract
Haemophilus parasuis is a widespread bacterial pathogen causing acute systemic inflammation and leading to the sudden death of piglets. Resistin, a multifunctional peptide hormone previously demonstrated to influence the inflammation in porcine, was extremely increased in H. parasuis-infected tissues. However, the mechanism of resistin expression regulation in porcine, especially during pathogen infection, remains unclear. In the present study, we explored for the first time the transcription factor and signaling pathway mediating the expression of pig resistin during H. parasuis stimulation. We found that H. parasuis induced the expression of pig resistin in a time- and dose-dependent manner via the transcription factor Ets2 in porcine alveolar macrophages during H. parasuis stimulation. Moreover, the expression of Ets2 was mediated by the activation of the p38 MAPK pathway induced by H. parasuis, thus promoting resistin production. These results revealed a novel view of the molecular mechanism of pig resistin production during acute inflammation induced by pathogenic bacteria.
Collapse
Affiliation(s)
- Kexin Hua
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, China; College of Veterinary Medicine, Huazhong Agricultural University, China; Hubei Provincial Key Laboratory of Preventive Veterinary Medicine, Huazhong Agricultural University, China
| | - Mingyang Wang
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, China; College of Veterinary Medicine, Huazhong Agricultural University, China; Hubei Provincial Key Laboratory of Preventive Veterinary Medicine, Huazhong Agricultural University, China
| | - Yishun Jin
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, China; College of Veterinary Medicine, Huazhong Agricultural University, China; Hubei Provincial Key Laboratory of Preventive Veterinary Medicine, Huazhong Agricultural University, China
| | - Yuan Gao
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, China; College of Veterinary Medicine, Huazhong Agricultural University, China; Hubei Provincial Key Laboratory of Preventive Veterinary Medicine, Huazhong Agricultural University, China
| | - Rui Luo
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, China; College of Veterinary Medicine, Huazhong Agricultural University, China; Hubei Provincial Key Laboratory of Preventive Veterinary Medicine, Huazhong Agricultural University, China
| | - Dingren Bi
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, China; College of Veterinary Medicine, Huazhong Agricultural University, China; Hubei Provincial Key Laboratory of Preventive Veterinary Medicine, Huazhong Agricultural University, China
| | - Rui Zhou
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, China; College of Veterinary Medicine, Huazhong Agricultural University, China; Hubei Provincial Key Laboratory of Preventive Veterinary Medicine, Huazhong Agricultural University, China
| | - Hui Jin
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, China; College of Veterinary Medicine, Huazhong Agricultural University, China; Hubei Provincial Key Laboratory of Preventive Veterinary Medicine, Huazhong Agricultural University, China.
| |
Collapse
|
11
|
Application of the MISTEACHING(S) disease susceptibility framework to Actinobacillus pleuropneumoniae to identify research gaps: an exemplar of a veterinary pathogen. Anim Health Res Rev 2021; 22:120-135. [PMID: 34275511 DOI: 10.1017/s1466252321000074] [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] [Indexed: 12/13/2022]
Abstract
Historically, the MISTEACHING (microbiome, immunity, sex, temperature, environment, age, chance, history, inoculum, nutrition, genetics) framework to describe the outcome of host-pathogen interaction, has been applied to human pathogens. Here, we show, using Actinobacillus pleuropneumoniae as an exemplar, that the MISTEACHING framework can be applied to a strict veterinary pathogen, enabling the identification of major research gaps, the formulation of hypotheses whose study will lead to a greater understanding of pathogenic mechanisms, and/or improved prevention/therapeutic measures. We also suggest that the MISTEACHING framework should be extended with the inclusion of a 'strain' category, to become MISTEACHINGS. We conclude that the MISTEACHINGS framework can be applied to veterinary pathogens, whether they be bacteria, fungi, viruses, or parasites, and hope to stimulate others to use it to identify research gaps and to formulate hypotheses worthy of study with their own pathogens.
Collapse
|
12
|
Li Y, Yang Q, Cai D, Guo H, Fang J, Cui H, Gou L, Deng J, Wang Z, Zuo Z. Resistin, a Novel Host Defense Peptide of Innate Immunity. Front Immunol 2021; 12:699807. [PMID: 34220862 PMCID: PMC8253364 DOI: 10.3389/fimmu.2021.699807] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2021] [Accepted: 06/07/2021] [Indexed: 12/24/2022] Open
Abstract
Resistin, a cysteine-rich protein, expressed in adipocytes, was initially proposed as a link between obesity and diabetes in mice. In humans, resistin is considered to be a pro-inflammatory molecule expressed in immune cells, which plays a regulatory role in many chronic inflammatory diseases, metabolic diseases, infectious diseases, and cancers. However, increasing evidence shows that resistin functions as a host defense peptide of innate immunity, in terms of its wide-spectrum anti-microbial activity, modulation of immunity, and limitation of microbial product-induced inflammation. To date, the understanding of resistin participating in host defense mechanism is still limited. The review aims to summarize current knowledge about the biological properties, functions, and related mechanisms of resistin in host defense, which provides new insights into the pleiotropic biological function of resistin and yields promising strategies for developing new antimicrobial therapeutic agents.
Collapse
Affiliation(s)
- Yanran Li
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Qiyuan Yang
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Dongjie Cai
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Hongrui Guo
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Jing Fang
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Hengmin Cui
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Liping Gou
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Junliang Deng
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Zhisheng Wang
- Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, China
| | - Zhicai Zuo
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| |
Collapse
|
13
|
Liu JX, Chao XY, Chen P, Wang YD, Su TJ, Li M, Xu RY, Wu Q. Transcriptome Analysis of Selenium-Treated Porcine Alveolar Macrophages Against Lipopolysaccharide Infection. Front Genet 2021; 12:645401. [PMID: 33747052 PMCID: PMC7970123 DOI: 10.3389/fgene.2021.645401] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Accepted: 02/02/2021] [Indexed: 11/25/2022] Open
Affiliation(s)
- Jia-Xuan Liu
- Animal Science and Technology College, Beijing University of Agriculture, Beijing, China
| | - Xin-Yu Chao
- Animal Science and Technology College, Beijing University of Agriculture, Beijing, China
| | - Peng Chen
- Animal Science and Technology College, Beijing University of Agriculture, Beijing, China
| | - Yi-Ding Wang
- Animal Science and Technology College, Beijing University of Agriculture, Beijing, China
| | - Tong-Jian Su
- Animal Science and Technology College, Beijing University of Agriculture, Beijing, China
| | - Meng Li
- Animal Science and Technology College, Beijing University of Agriculture, Beijing, China
| | - Ru-Yu Xu
- Animal Science and Technology College, Beijing University of Agriculture, Beijing, China
| | - Qiong Wu
- Animal Science and Technology College, Beijing University of Agriculture, Beijing, China
| |
Collapse
|
14
|
Jia L, Liu K, Fei T, Liu Q, Zhao X, Hou L, Zhang W. Programmed cell death-1/programmed cell death-ligand 1 inhibitors exert antiapoptosis and antiinflammatory activity in lipopolysaccharide stimulated murine alveolar macrophages. Exp Ther Med 2021; 21:400. [PMID: 33680122 PMCID: PMC7918472 DOI: 10.3892/etm.2021.9831] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2020] [Accepted: 06/17/2020] [Indexed: 12/23/2022] Open
Abstract
Acute lung injury caused by sepsis remains one of the most difficult challenges faced by patients in intensive care units and is associated with a high mortality rate. The aim of the present study was to investigate whether programmed cell death (PD)-1/programmed cell death-ligand 1 (PD-L1) inhibitors reduce alveolar macrophage apoptosis, reduce inflammatory factor release and relieve inflammation. For this purpose, murine alveolar macrophages, MH-S, were cultured and divided into control, lipopolysaccharide (LPS) and LPS+BMS-1 (PD-1/PD-L1 inhibitors) groups. LPS (10 ng/ml) was added to the LPS and LPS+BMS-1 groups for 24 h and PD-1/PD-L1 inhibitor BMS-1 (1 µmol/l) was added to the LPS+BMS-1 group for 72 h. PD-1 mRNA expression was detected using reverse transcription-quantitative PCR and PD-1 protein expression was detected using western blotting in the control, LPS and LPS+BMS-1 groups of macrophages. MH-S apoptosis was detected using flow cytometry with Annexin V/PI staining. The levels of the inflammatory factors interleukin (IL)-1β, IL-6, tumor necrosis factor (TNF)-α and IL-10 were detected by ELISA. Murine alveolar macrophages expressed PD-1 at both the molecular and protein levels and PD-1 expression was increased in MH-S cells stimulated with LPS. Compared with the LPS group, the expression of PD-1 in the LPS+BMS-1 group was significantly decreased. Flow cytometry demonstrated that there was increased apoptosis of alveolar macrophages in the LPS group compared with the control group, whereas, alveolar macrophages notably decreased apoptosis in the LPS+BMS-1 group compared with the LPS group. There was no statistical difference between the control group and the LPS+BMS-1 group. IL-1β, IL-6, TNF-α and IL-10 were increased in the LPS group compared with the control group. The levels of IL-1β, IL-6 and TNF-α in the LPS+BMS-1 group were lower compared with those in the LPS group whereas IL-10 was further increased. In vitro, the PD-1/PD-L1 inhibitor, BMS-1, decreases alveolar macrophage apoptosis compared with the LPS group to maintain effective immune clearance and reduce inflammatory factor release. This decreased the inflammatory response and reduced acute lung injury caused by sepsis. Therefore, PD-1/PD-L1 inhibitors may be a potential therapeutic target for acute lung injury in patients with sepsis.
Collapse
Affiliation(s)
- Lingling Jia
- Department of Thoracic Surgery, Shanxi Medical University, Taiyuan, Shanxi 030001, P.R. China
| | - Kai Liu
- Department of Thoracic Surgery, Shanxi Medical University, Taiyuan, Shanxi 030001, P.R. China
| | - Teng Fei
- Department of Thoracic Surgery, Shanxi Medical University, Taiyuan, Shanxi 030001, P.R. China
| | - Qian Liu
- Department of Thoracic Surgery, Shanxi Medical University, Taiyuan, Shanxi 030001, P.R. China
| | - Xiwei Zhao
- Department of Thoracic Surgery, Shanxi Medical University, Taiyuan, Shanxi 030001, P.R. China
| | - Linyi Hou
- Department of Intensive Care Unit, Second Hospital of Shanxi Medical University, Taiyuan, Shanxi 030001, P.R. China
| | - Wenkai Zhang
- Department of Intensive Care Unit, Second Hospital of Shanxi Medical University, Taiyuan, Shanxi 030001, P.R. China
| |
Collapse
|
15
|
Nahar N, Turni C, Tram G, Blackall PJ, Atack JM. Actinobacillus pleuropneumoniae: The molecular determinants of virulence and pathogenesis. Adv Microb Physiol 2021; 78:179-216. [PMID: 34147185 DOI: 10.1016/bs.ampbs.2020.12.001] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Actinobacillus pleuropneumoniae, the causative agent of porcine pleuropneumonia, is responsible for high economic losses in swine herds across the globe. Pleuropneumonia is characterized by severe respiratory distress and high mortality. The knowledge about the interaction between bacterium and host within the porcine respiratory tract has improved significantly in recent years. A. pleuropneumoniae expresses multiple virulence factors, which are required for colonization, immune clearance, and tissue damage. Although vaccines are used to protect swine herds against A. pleuropneumoniae infection, they do not offer complete coverage, and often only protect against the serovar, or serovars, used to prepare the vaccine. This review will summarize the role of individual A. pleuropneumoniae virulence factors that are required during key stages of pathogenesis and disease progression, and highlight progress made toward developing effective and broadly protective vaccines against an organism of great importance to global agriculture and food production.
Collapse
Affiliation(s)
- Nusrat Nahar
- Institute for Glycomics, Griffith University, Gold Coast, QLD, Australia
| | - Conny Turni
- Queensland Alliance for Agriculture and Food Innovation, The University of Queensland, St. Lucia, QLD, Australia
| | - Greg Tram
- Institute for Glycomics, Griffith University, Gold Coast, QLD, Australia
| | - Patrick J Blackall
- Queensland Alliance for Agriculture and Food Innovation, The University of Queensland, St. Lucia, QLD, Australia.
| | - John M Atack
- Institute for Glycomics, Griffith University, Gold Coast, QLD, Australia.
| |
Collapse
|
16
|
Zhang H, He F, Li P, Hardwidge PR, Li N, Peng Y. The Role of Innate Immunity in Pulmonary Infections. BIOMED RESEARCH INTERNATIONAL 2021; 2021:6646071. [PMID: 33553427 PMCID: PMC7847335 DOI: 10.1155/2021/6646071] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/31/2020] [Revised: 12/26/2020] [Accepted: 01/08/2021] [Indexed: 02/07/2023]
Abstract
Innate immunity forms a protective line of defense in the early stages of pulmonary infection. The primary cellular players of the innate immunity against respiratory infections are alveolar macrophages (AMs), dendritic cells (DCs), neutrophils, natural killer (NK) cells, and innate lymphoid cells (ILCs). They recognize conserved structures of microorganisms through membrane-bound and intracellular receptors to initiate appropriate responses. In this review, we focus on the prominent roles of innate immune cells and summarize transmembrane and cytosolic pattern recognition receptor (PRR) signaling recognition mechanisms during pulmonary microbial infections. Understanding the mechanisms of PRR signal recognition during pulmonary pathogen infections will help us to understand pulmonary immunopathology and lay a foundation for the development of effective therapies to treat and/or prevent pulmonary infections.
Collapse
Affiliation(s)
- Huihui Zhang
- College of Animal Medicine, Southwest University, Chongqing, China
| | - Fang He
- College of Animal Medicine, Southwest University, Chongqing, China
| | - Pan Li
- College of Animal Medicine, Southwest University, Chongqing, China
| | | | - Nengzhang Li
- College of Animal Medicine, Southwest University, Chongqing, China
| | - Yuanyi Peng
- College of Animal Medicine, Southwest University, Chongqing, China
| |
Collapse
|
17
|
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: 0.2] [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
|
18
|
Weng J, Chen M, Lin Q, Chen J, Wang S, Fang D. Penehyclidine hydrochloride defends against LPS-induced ALI in rats by mitigating endoplasmic reticulum stress and promoting the Hes1/Notch1 pathway. Gene 2019; 721:144095. [PMID: 31476403 DOI: 10.1016/j.gene.2019.144095] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2019] [Revised: 08/29/2019] [Accepted: 08/29/2019] [Indexed: 12/12/2022]
Abstract
Penehyclidine hydrochloride (PHC) is a novel anticholinergic drug applied broadly in surgeries as a preanesthetic medication. A substantial amount of research indicates that PHC has lung defensive properties. Considering that endoplasmic reticulum (ER) stress exerts a crucial function in cell apoptosis associated with the lipopolysaccharides (LPS)-induced acute lung injury (ALI) model, we aimed to determine whether regulation of ER stress in the LPS-induced ALI model was associated with the lung defensive role of PHC. Adult male SD rats were administered LPS (5 mg/kg, intratracheally) followed by PHC (1.0 mg/kg, intravenously) for 24 h. The NR8383 alveolar macrophages were randomly separated into Sham, LPS (100 ng/mL), and PHC (1, 2.5, or 5 μg/mL) + LPS groups. PHC (1, 2.5, or 5 μg/mL) + LPS groups were treated with PHC alone for 1 h after LPS exposure. Posttreatment with PHC relieved LPS-induced pulmonary impairment and blocked LPS-mediated lung apoptosis, indicated by the downregulation of the lung apoptotic indicators malondialdehyde and superoxide dismutase in serum at 24 h after LPS-induced ALI. PHC (1-5 μg/mL) did not influence the activity of cultivated NR8383 alveolar macrophages in vitro. However, postconditioning with PHC dosage-dependently reduced LPS-mediated cell apoptosis. Additionally, many studies have indicated that PHC administration inhibits ER stress and initiates hairy and enhancer of split 1 (Hes1)/(Notch1) signaling by decreasing phosphorylated α subunit of eukaryotic initiation factor 2α (p-eIF2α)/eukaryotic translation initiation factor 2α (eIF2α) and Phospho-protein kinase R-like ER kinase (p-PERK)/ protein kinase R-like ER kinase (PERK) proportions; inhibiting C/EBP-homologous protein (CHOP), activating transcription factor 4 (ATF4), caspase-3, and Bcl2-associated x (Bax) activity; and enhancing notch1 intracellular domain (NICD), Notch1, B-cell lymphoma-2 (Bcl-2), and Hes1 activity in vivo and in vitro. In addition, the defensive functions of PHC on LPS-activated NR8383 alveolar macrophages were abrogated through the Notch1 pathway antagonist [(3,5-difluorophenacetyl)-1-alanyl] -phenylglycine-butyl ester (DAPT). In conclusion, PHC alleviates LPS-induced ALI by ameliorating ER stress-mediated apoptosis and promoting Hes1/Notch1 signaling in vivo and in vitro.
Collapse
Affiliation(s)
- Junting Weng
- Department of Critical Care Medicine, The Affiliated Hospital of Putian University, Putian 351100, China
| | - Min Chen
- Department of Critical Care Medicine, The Affiliated Hospital of Putian University, Putian 351100, China
| | - Qunying Lin
- Department of Respiratory and Critical Care, The Affiliated Hospital of Putian University, Putian 351100, China
| | - Jianfei Chen
- Department of Critical Care Medicine, The Affiliated Hospital of Putian University, Putian 351100, China
| | - ShanZuan Wang
- Department of Respiratory and Critical Care, The Affiliated Hospital of Putian University, Putian 351100, China
| | - Dexiang Fang
- Department of Critical Care Medicine, The Affiliated Hospital of Putian University, Putian 351100, China.
| |
Collapse
|
19
|
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: 1.2] [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
|
20
|
Zhang F, Zhao Q, Tian J, Chang YF, Wen X, Huang X, Wu R, Wen Y, Yan Q, Huang Y, Ma X, Han X, Miao C, Cao S. Effective Pro-Inflammatory Induced Activity of GALT, a Conserved Antigen in A. Pleuropneumoniae, Improves the Cytokines Secretion of Macrophage via p38, ERK1/2 and JNK MAPKs Signal Pathway. Front Cell Infect Microbiol 2018; 8:337. [PMID: 30319993 PMCID: PMC6167544 DOI: 10.3389/fcimb.2018.00337] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2018] [Accepted: 09/03/2018] [Indexed: 12/17/2022] Open
Abstract
GALT is a highly conserved antigen in gram-negative bacteria, and has been shown to play a crucial role in the pathogenesis of many zoonoses. Actinobacillus pleuropneumoniae (APP) is a widespread respiratory system pathogen belonging to the Pasteuriaceae family. The functional mechanisms of GALT in the process of infection remain unclear. The aim of this study is to analyze roles of GALT in the pathogenesis of APP infection. Recombinant GALT was expressed in E. coli, purified, and was used to treat a Raw 264.7 macrophage line. Stimulation of Raw 264.7 macrophages with recombinant GALT protein induced the expression of pro-inflammatory cytokines (TNF-α, IL-1β, and IL-6). Compared with negative control, GALT led to increased production of pro-inflammatory cytokines in treated cells. Furthermore, specific inhibitors of the extracellular signal-regulated P38 and JNK MAPKs pathways significantly decreased GALT-induced pro-inflammatory cytokine production, and a western blot assay showed that GALT stimulation induced the activation of the MAPKs pathway. This process included cell-signaling pathways like P38, ERK1/2 and JNK MAPKs, and NF-κB. Both TLR2 and TLR4 were receptors of GALT antigens, whereas they played negative and positive roles (respectively) in the process of induction and expression of pro-inflammatory cytokines. Taken together, our data indicate that GALT is a novel pro-inflammatory mediator and induces TLR2 and TLR4-dependent pro-inflammatory activity in Raw 264.7 macrophages through P38, ERK1/2, and JNK MAPKs pathways.
Collapse
Affiliation(s)
- Fei Zhang
- College of Veterinary Medicine, Research Center of Swine Disease, Sichuan Agricultural University, Chengdu, China.,Department of Population Medicine and Diagnostic Sciences, College of Veterinary Medicine, Cornell University, Ithaca, NY, United States
| | - Qin Zhao
- College of Veterinary Medicine, Research Center of Swine Disease, Sichuan Agricultural University, Chengdu, China.,National Teaching and Experimental Center of Animal, Sichuan Agricultural University, Chengdu, China
| | - Jin Tian
- College of Veterinary Medicine, Research Center of Swine Disease, Sichuan Agricultural University, Chengdu, China
| | - Yung-Fu Chang
- Department of Population Medicine and Diagnostic Sciences, College of Veterinary Medicine, Cornell University, Ithaca, NY, United States
| | - Xintian Wen
- College of Veterinary Medicine, Research Center of Swine Disease, Sichuan Agricultural University, Chengdu, China.,Sichuan Science-Observation Experimental Station of Veterinary Drugs and Veterinary Diagnostic Technology, Ministry of Agriculture, Chengdu, China
| | - Xiaobo Huang
- College of Veterinary Medicine, Research Center of Swine Disease, Sichuan Agricultural University, Chengdu, China.,Sichuan Science-Observation Experimental Station of Veterinary Drugs and Veterinary Diagnostic Technology, Ministry of Agriculture, Chengdu, China
| | - Rui Wu
- College of Veterinary Medicine, Research Center of Swine Disease, Sichuan Agricultural University, Chengdu, China.,Sichuan Science-Observation Experimental Station of Veterinary Drugs and Veterinary Diagnostic Technology, Ministry of Agriculture, Chengdu, China
| | - Yiping Wen
- College of Veterinary Medicine, Research Center of Swine Disease, Sichuan Agricultural University, Chengdu, China.,Sichuan Science-Observation Experimental Station of Veterinary Drugs and Veterinary Diagnostic Technology, Ministry of Agriculture, Chengdu, China
| | - Qigui Yan
- College of Veterinary Medicine, Research Center of Swine Disease, Sichuan Agricultural University, Chengdu, China.,National Teaching and Experimental Center of Animal, Sichuan Agricultural University, Chengdu, China
| | - Yong Huang
- College of Veterinary Medicine, Research Center of Swine Disease, Sichuan Agricultural University, Chengdu, China.,National Teaching and Experimental Center of Animal, Sichuan Agricultural University, Chengdu, China
| | - Xiaoping Ma
- College of Veterinary Medicine, Research Center of Swine Disease, Sichuan Agricultural University, Chengdu, China.,National Teaching and Experimental Center of Animal, Sichuan Agricultural University, Chengdu, China
| | - Xinfeng Han
- College of Veterinary Medicine, Research Center of Swine Disease, Sichuan Agricultural University, Chengdu, China.,National Teaching and Experimental Center of Animal, Sichuan Agricultural University, Chengdu, China
| | - Chang Miao
- College of Veterinary Medicine, Research Center of Swine Disease, Sichuan Agricultural University, Chengdu, China
| | - Sanjie Cao
- College of Veterinary Medicine, Research Center of Swine Disease, Sichuan Agricultural University, Chengdu, China.,National Teaching and Experimental Center of Animal, Sichuan Agricultural University, Chengdu, China.,Sichuan Science-Observation Experimental Station of Veterinary Drugs and Veterinary Diagnostic Technology, Ministry of Agriculture, Chengdu, China
| |
Collapse
|