1
|
Albert E, Kis IE, Kiss K, K-Jánosi K, de Oliveira Costa M, Tolnai G, Biksi I. Abortion and Lethal Septicaemia in Sows Caused by a Non-ST194 Streptococcus equi subsp. zooepidemicus. Transbound Emerg Dis 2024; 2024:4008946. [PMID: 40303154 PMCID: PMC12016921 DOI: 10.1155/2024/4008946] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Revised: 06/18/2024] [Accepted: 07/01/2024] [Indexed: 05/02/2025]
Abstract
Outbreaks of zoonotic Streptococcus equi subsp. zooepidemicus (SEZ) have caused severe epidemics in the pig sector since the 1970s in Southeastern Asia, China, and more recently North America. Cases of high mortality caused by peracute septicaemia were all attributed to strains of a highly virulent clonal lineage belonging to the sequence type (ST) 194. In Europe, only two outbreaks have been reported with similar features, caused by other sequence types. In August 2023, a febrile disease followed by abortion and subsequent death was observed among sows kept in a small-scale organic pig farm in West Hungary. Symptoms, pathological lesions, and microbiological findings were suggestive of septicaemia from bacterial origin caused by SEZ. According to the results of the routine laboratory testing, no other relevant infectious agents were involved. Whole-genome sequence analysis assigned the examined strains to ST138, unrelated to any of the European isolates. It also revealed a few common SEZ virulence genes, compared to the highly virulent ST194 strains. A sudden weather change and subsequent extremely high average daily temperature before the outbreak could be identified as the only predisposing factor. The immediate antibiotic treatment and applied biosecurity measures might have helped to restrict and terminate the outbreak. To our knowledge, this is the first report on abortion and lethal septicaemia in sows from Central and Eastern Europe. The results call attention to the potential of non-ST194 SEZ strains to cause outbreaks in pig farms.
Collapse
Affiliation(s)
- Ervin Albert
- Department of PathologyUniversity of Veterinary Medicine Budapest, Üllő, Hungary
- Institute of MetagenomicsUniversity of Debrecen, Debrecen, Hungary
| | - István Emil Kis
- Department of PathologyUniversity of Veterinary Medicine Budapest, Üllő, Hungary
| | | | - Katalin K-Jánosi
- Department of PathologyUniversity of Veterinary Medicine Budapest, Üllő, Hungary
| | - Matheus de Oliveira Costa
- Department of Large Animal Clinical SciencesWestern College of Veterinary MedicineUniversity of Saskatchewan, Saskatoon, Canada
- Department of Population HealthFaculty of Veterinary MedicineUtrecht University, Utrecht, Netherlands
| | | | - Imre Biksi
- Department of PathologyUniversity of Veterinary Medicine Budapest, Üllő, Hungary
| |
Collapse
|
2
|
Xie H, Zhang R, Guo R, Zhang Y, Zhang J, Li H, Fu Q, Wang X. Characterization of AI-2/LuxS quorum sensing system in biofilm formation, pathogenesis of Streptococcus equi subsp. zooepidemicus. Front Cell Infect Microbiol 2024; 14:1339131. [PMID: 38379770 PMCID: PMC10876813 DOI: 10.3389/fcimb.2024.1339131] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Accepted: 01/24/2024] [Indexed: 02/22/2024] Open
Abstract
Streptococcus equi subsp. zooepidemicus (SEZ) is an opportunistic pathogen of both humans and animals. Quorum sensing (QS) plays an important role in the regulation of bacterial group behaviors. The aim of this study was to characterize the LuxS in SEZ and evaluate its impact on biofilm formation, pathogenesis and gene expression. The wild-type SEZ and its LuxS mutant (ΔluxS) were examined for growth, biofilm formation, virulence factors, and transcriptomic profiles. Our results showed that LuxS deficiency did not affect SEZ hemolytic activity, adhesion or capsule production. For biofilm assay demonstrated that mutation in the luxS gene significantly enhances biofilm formation, produced a denser biofilm and attached to a glass surface. RAW264.7 cell infection indicated that ΔluxS promoted macrophage apoptosis and pro-inflammatory responses. In mice infection, there was no significant difference in mortality between SEZ and ΔluxS. However, the bacterial load in the spleen of mice infected with ΔluxS was significantly higher than in those infected with SEZ. And the pathological analysis further indicated that spleen damage was more severe in the ΔluxS group. Moreover, transcriptomics analysis revealed significant alterations in carbon metabolism, RNA binding and stress response genes in ΔluxS. In summary, this study provides the first evidence of AI-2/LuxS QS system in SEZ and reveals its regulatory effects on biofilm formation, pathogenicity and gene expression.
Collapse
Affiliation(s)
- Honglin Xie
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China
| | - Riteng Zhang
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China
| | - Ruhai Guo
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China
| | - Yining Zhang
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China
| | - Jingya Zhang
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China
| | - Hui Li
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China
| | - Qiang Fu
- School of Life Science and Engineering, Foshan University, Foshan, China
| | - Xinglong Wang
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China
| |
Collapse
|
3
|
Xu G, Guo Z, Liu Y, Yang Y, Lin Y, Li C, Huang Y, Fu Q. Gasdermin D protects against Streptococcus equi subsp. zooepidemicus infection through macrophage pyroptosis. Front Immunol 2022; 13:1005925. [PMID: 36311722 PMCID: PMC9614658 DOI: 10.3389/fimmu.2022.1005925] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Accepted: 09/28/2022] [Indexed: 11/13/2022] Open
Abstract
Streptococcus equi subsp. zooepidemicus (S. zooepidemicus, SEZ) is an essential zoonotic bacterial pathogen that can cause various inflammation, such as meningitis, endocarditis, and pneumonia. Gasdermin D (GSDMD) is involved in cytokine release and cell death, indicating an important role in controlling the microbial infection. This study investigated the protective role of GSDMD in mice infected with SEZ and examined the role of GSDMD in peritoneal macrophages in the infection. GSDMD-deficient mice were more susceptible to intraperitoneal infection with SEZ, and the white pulp structure of the spleen was seriously damaged in GSDMD-deficient mice. Although the increased proportion of macrophages did not depend on GSDMD in both spleen and peritoneal lavage fluid (PLF), deficiency of GSDMD caused the minor release of the pro-inflammatory cytokines interleukin-1β (IL-1β) and interleukin-18 (IL-18) during the infection in vivo. In vitro, SEZ infection induced more release of IL-1β, IL-18, and lactate dehydrogenase (LDH) in wild-type macrophages than in GSDMD-deficient macrophages. Finally, we demonstrated that pore formation and pyroptosis of macrophages depended on GSDMD. Our findings highlight the host defense mechanisms of GSDMD against SEZ infection, providing a potential therapeutic target in SEZ infection.
Collapse
Affiliation(s)
- Guobin Xu
- School of Life Science and Engineering, Foshan University, Foshan, China
| | - Zheng Guo
- School of Life Science and Engineering, Foshan University, Foshan, China
| | - Yuxuan Liu
- School of Life Science and Engineering, Foshan University, Foshan, China
| | - Yalin Yang
- School of Life Science and Engineering, Foshan University, Foshan, China
| | - Yongjin Lin
- School of Life Science and Engineering, Foshan University, Foshan, China
| | - Chunliu Li
- School of Life Science and Engineering, Foshan University, Foshan, China
| | - Yunfei Huang
- School of Life Science and Engineering, Foshan University, Foshan, China
- Foshan University Veterinary Teaching Hospital, Foshan University, Foshan, China
| | - Qiang Fu
- School of Life Science and Engineering, Foshan University, Foshan, China
- Foshan University Veterinary Teaching Hospital, Foshan University, Foshan, China
- *Correspondence: Qiang Fu,
| |
Collapse
|
4
|
Li G, Zong X, Cheng Y, Xu J, Deng J, Huang Y, Ma C, Fu Q. miR-223-3p contributes to suppressing NLRP3 inflammasome activation in Streptococcus equi ssp. zooepidemicus infection. Vet Microbiol 2022; 269:109430. [PMID: 35427992 DOI: 10.1016/j.vetmic.2022.109430] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 03/21/2022] [Accepted: 04/03/2022] [Indexed: 11/20/2022]
Abstract
Streptococcus equi subsp. zooepidemicus (SEZ) is an essential pathogen in a range of species, causing a worldwide variety of diseases, such as meningitis, endocarditis, and septicaemia. Studies have shown that microRNAs (miRNAs), which regulate target genes at the post-transcriptional level, play an important regulatory role in the organism. In this study, the infection of J774A.1 murine macrophages with SEZ up-regulated NLRP3 inflammasome and downstream pathways accompanied by miR-223-3p down-regulation. Through computational prediction and experimental confirmation, we have shown that miR-223-3p directly targets the NLRP3 mRNA. Consequently, overexpression of miR-223-3p suppressed NLRP3 inflammasome activation and downstream pathways in response to SEZ infection. The miR-223-3p inhibitor exhibited the opposite effect, causing hyperactivation of NLRP3 inflammation activation and downstream pathways. Additionally, we further demonstrated that miRNA-223-3p inhibited the secretion of IL-1β and IL-18 by regulating the NLRP3/caspase-1 pathway. Furthermore, intravenous administration of miR-223-3p significantly decreased inflammation in mice in response to SEZ. In conclusion, our results demonstrated that miR-223-3p contributes to suppressing the NLRP3 inflammasome activation in SEZ infection, contributing novel evidence to identify a therapeutic target for treating SEZ.
Collapse
Affiliation(s)
- Guochao Li
- School of Life Science and Engineering, Foshan University, Guangdong 528225, China
| | - Xueqing Zong
- School of Life Science and Engineering, Foshan University, Guangdong 528225, China
| | - Yun Cheng
- School of Life Science and Engineering, Foshan University, Guangdong 528225, China
| | - Jianqi Xu
- School of Life Science and Engineering, Foshan University, Guangdong 528225, China
| | - Jingfei Deng
- School of Life Science and Engineering, Foshan University, Guangdong 528225, China
| | - Yunfei Huang
- School of Life Science and Engineering, Foshan University, Guangdong 528225, China; Foshan University Veterinary Teaching Hospital, Foshan University, Guangdong 528225, China
| | - Chunquan Ma
- School of Life Science and Engineering, Foshan University, Guangdong 528225, China; Foshan University Veterinary Teaching Hospital, Foshan University, Guangdong 528225, China
| | - Qiang Fu
- School of Life Science and Engineering, Foshan University, Guangdong 528225, China; Foshan University Veterinary Teaching Hospital, Foshan University, Guangdong 528225, China.
| |
Collapse
|
5
|
Pei X, Liu J, Liu M, Zhou H, Wang X, Fan H. Quantitative proteomics revealed modulation of macrophages by MetQ gene of Streptococcus suis serotype 2. AMB Express 2020; 10:195. [PMID: 33125582 PMCID: PMC7599288 DOI: 10.1186/s13568-020-01131-2] [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: 09/20/2020] [Accepted: 10/18/2020] [Indexed: 11/10/2022] Open
Abstract
Streptococcus suis serotype 2 (SS2) is a serious zoonotic pathogen; it can lead to symptoms of streptococcal toxic shock syndrome (STSS) in humans and sepsis in pigs, and poses a great threat to public health. The SS2 MetQ gene deletion strain has attenuated antiphagocytosis, although the mechanism of antiphagocytosis and pathogenesis of MetQ in SS2 has remained unclear. In this study, stable isotope labeling by amino acids in cell culture (SILAC) based liquid chromatography–mass spectrometry (LC–MS) and subsequent bioinformatics analysis was used to determine differentially expressed proteins of RAW264.7 cells infected with △MetQ and ZY05719. Proteomic results were verified by quantitative real-time polymerase chain reaction (qRT-PCR) and Western blotting for selected proteins. Further research was focused mainly on immune system processes related to downregulated proteins, such as Src and Ccl9, and actin cytoskeleton and endocytosis related upregulated proteins, like Pstpip1 and Ppp1r9b. The proteomic results in this study shed light on the mechanism of antiphagocytosis and innate immunity of macrophages infected with △MetQ and ZY05719, which might provide novel targets to prevent or control the infection of SS2.
Collapse
|
6
|
Guo G, Wei D, Zhang Y, Wang K, Liu R, Wu Z, Kong L, Cheng L, Zhang W. M-like protein SrM is not crucial to the virulence of a novel isolate of Streptococcus equi subsp. ruminatorum from Macaca mulatta. Res Vet Sci 2020; 132:221-228. [PMID: 32615343 DOI: 10.1016/j.rvsc.2020.05.006] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2019] [Revised: 03/27/2020] [Accepted: 05/04/2020] [Indexed: 11/28/2022]
Abstract
In this study, a Streptococcus strainnamed FJ1804, was isolated from a blood sample collected from a dead Macaca mulatta in China and, was subsequently classified as Streptococcus equi subsp. ruminatorum (S.e. ruminatorum) through 16S rRNA gene sequence analysis. After whole genome sequencing and analysis, an M-like protein encoding gene that encodes an SrM protein that is homologous to the crucial S.e. zooepidemicus crucial virulence factor SzP, was identified in the genome of FJ1804. To determinethe function of SrM in this bacterium, a strain deleted of srm as well as a complement strain were constructed. The results of in vitro cell adherence, invasion and phagocytosis assays and in vivo animal challenge and histopathology showed that the anti-phagocytosis was decreased and the adherence rate was increased in the srm deletion strain, whereas the invasion rate, pathological features and LD50 values inboth zebrafish and BALB/c mice model showed no difference compared to that observed for the WT strain. To the best of our knowledge, this is first of an infection caused by S.e. ruminatorum, which is a newly identified zoonotic pathogen, in Macaca mulatta, and our data suggest that, compared with other S.e. zooepidemicus strains, the SzP homologous protein is not crucial to the virulence of this bacterium.
Collapse
Affiliation(s)
- Genglin Guo
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China; Key Lab of Animal Bacteriology, Ministry of Agriculture, Nanjing 210095, China; OIE Reference Lab for Swine Streptococcosis, Nanjing 210095, China.
| | - Dan Wei
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China; Key Lab of Animal Bacteriology, Ministry of Agriculture, Nanjing 210095, China; OIE Reference Lab for Swine Streptococcosis, Nanjing 210095, China.
| | - Yuhang Zhang
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China; Key Lab of Animal Bacteriology, Ministry of Agriculture, Nanjing 210095, China; OIE Reference Lab for Swine Streptococcosis, Nanjing 210095, China.
| | - Kaicheng Wang
- China Animal Health and Epidemiology Center, Qingdao, Shandong, China.
| | - Rongchang Liu
- Institute of Animal Husbandry and Veterinary Medicine, Fujian Academy of Agricultural Sciences, Fuzhou, Fujian 350013, China.
| | - Zhiqiang Wu
- City Park Management Office of Nanping City, Nanping, Fujian 353000, China.
| | - Lifang Kong
- City Park Management Office of Nanping City, Nanping, Fujian 353000, China.
| | - Longfei Cheng
- Institute of Animal Husbandry and Veterinary Medicine, Fujian Academy of Agricultural Sciences, Fuzhou, Fujian 350013, China.
| | - Wei Zhang
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China; Key Lab of Animal Bacteriology, Ministry of Agriculture, Nanjing 210095, China; OIE Reference Lab for Swine Streptococcosis, Nanjing 210095, China.
| |
Collapse
|
7
|
Ma Z, Peng J, Yu D, Park JS, Lin H, Xu B, Lu C, Fan H, Waldor MK. A streptococcal Fic domain-containing protein disrupts blood-brain barrier integrity by activating moesin in endothelial cells. PLoS Pathog 2019; 15:e1007737. [PMID: 31071198 PMCID: PMC6529018 DOI: 10.1371/journal.ppat.1007737] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2019] [Revised: 05/21/2019] [Accepted: 03/27/2019] [Indexed: 01/04/2023] Open
Abstract
Streptococcus equi subsp. zooepidemicus (SEZ) is a zoonotic pathogen capable of causing meningitis in humans. The mechanisms that enable pathogens to traverse the blood-brain barrier (BBB) are incompletely understood. Here, we investigated the role of a newly identified Fic domain-containing protein, BifA, in SEZ virulence. BifA was required for SEZ to cross the BBB and to cause meningitis in mice. BifA also enhanced SEZ translocation across human Brain Microvascular Endothelial Cell (hBMEC) monolayers. Purified BifA or its Fic domain-containing C-terminus alone were able to enter into hBMECs, leading to disruption of monolayer barrier integrity. A SILAC-based proteomic screen revealed that BifA binds moesin. BifA’s Fic domain was required for its binding to this regulator of host cell cytoskeletal processes. BifA treatment of hBMECs led to moesin phosphorylation and downstream RhoA activation. Inhibition of moesin activation or moesin depletion in hBMEC monolayers abrogated BifA-mediated increases in barrier permeability and SEZ’s capacity to translocate across monolayers. Thus, BifA activation of moesin appears to constitute a key mechanism by which SEZ disrupts endothelial monolayer integrity to penetrate the BBB. Streptococcus equi subsp. zooepidemicus (SEZ) is an important animal pathogen and can cause meningitis in humans. Little is known about how this Group C streptococcal species penetrates the blood-brain barrier (BBB). We identified bifA, a gene that is critical for SEZ to cause meningitis in mice and to penetrate a human brain endothelial monolayer in a tissue culture model. BifA’s Fic domain enables the protein to enter into endothelial monolayers and to bind to moesin, a cytoskeletal regulatory protein, leading to its activation. Preventing moesin activation abolished BifA-induced barrier leakiness and SEZ’s capacity to penetrate a monolayer barrier. Together, our findings suggest that SEZ meningitis depends on BifA, a Fic-domain protein that manipulates moesin-dependent signaling to modulate BBB permeability.
Collapse
Affiliation(s)
- Zhe Ma
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, Jiangsu, China
- Division of Infectious Diseases, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
- Department of Microbiology, Harvard Medical School, Boston, Massachusetts, United States of America
- Ministry of Agriculture Key Laboratory of Animal Bacteriology, Nanjing, Jiangsu, China
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, Jiangsu, China
| | - Jie Peng
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, Jiangsu, China
| | - Dandan Yu
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, Jiangsu, China
| | - Joseph S. Park
- Division of Infectious Diseases, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
- Department of Microbiology, Harvard Medical School, Boston, Massachusetts, United States of America
- Howard Hughes Medical Institute, Boston, Massachusetts, United States of America
| | - Huixing Lin
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, Jiangsu, China
- Ministry of Agriculture Key Laboratory of Animal Bacteriology, Nanjing, Jiangsu, China
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, Jiangsu, China
| | - Bin Xu
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, Jiangsu, China
| | - Chengping Lu
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, Jiangsu, China
| | - Hongjie Fan
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, Jiangsu, China
- Ministry of Agriculture Key Laboratory of Animal Bacteriology, Nanjing, Jiangsu, China
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, Jiangsu, China
- * E-mail: (HF); (MKW)
| | - Matthew K. Waldor
- Division of Infectious Diseases, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
- Department of Microbiology, Harvard Medical School, Boston, Massachusetts, United States of America
- Howard Hughes Medical Institute, Boston, Massachusetts, United States of America
- * E-mail: (HF); (MKW)
| |
Collapse
|
8
|
Xu B, Zhang P, Zhou H, Sun Y, Tang J, Fan H. Identification of novel genes associated with anti-phagocytic functions in Streptococcus equi subsp. zooepidemicus. Vet Microbiol 2019; 233:28-38. [PMID: 31176409 DOI: 10.1016/j.vetmic.2019.04.023] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2018] [Revised: 04/12/2019] [Accepted: 04/16/2019] [Indexed: 01/13/2023]
Abstract
The anti-phagocytic abilities of bacteria often affect bacterial pathogenicity. Here, random mutant library of Streptococcus equi subsp. zooepidemicus (SEZ) was constructed using transposon mutagenesis. After careful screening, 30 transposon mutants with different transposon insertion sites were identified by conducting quantitative phagocytosis and insertion-site confirmation assays, whose anti-phagocytic abilities were significantly reduced relative to the wild-type strain. Insertion sites of 19 strains were monocistronic, including genes coding membrane proteins, transporters, and enzymes with unknown pathological function, such as sadM, adhP, purD, guaA, alpha-galactosidase coding gene, ABC transporter permease coding gene, metallo-beta-lactamase coding gene, and three secreted enzyme coding genes spuZ, slaB, and endoS, as well as known virulence factor coding genes, such as hasA and szM. The insertion sites of another 11 strains were polycistronic. We focused on four monocistronic-mutant strains: MhtpZ, MspuZ, MslaB, and MendoS. The anti-phagocytic abilities of not only the mutants that were precoincubated with the recombinant proteins, but also the complement strains were significantly more pronounced than those of all four corresponding mutants. The polyclonal antiserum against SlaB or EndoS also significantly decreased the anti-phagocytic capacity of wild-type SEZ. All four mutants exhibited significantly decreased viability in whole blood and reduced lethality in mice relative to the wild-type strain. Thus, we identified a variety of new anti-phagocytic factors, particularly multiple SEZ secreted enzymes. These factors are instrumental in the phagocytic resistance of SEZ in the absence of opsonin. Our results provide a framework for further studies of SEZ pathogenesis and relevant vaccine development for novel potential targets.
Collapse
Affiliation(s)
- Bin Xu
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China; National Research Center of Veterinary Biologicals engineering and Technology, Institute of Veterinary Medicine, Jiangsu Academy of Agricultural Sciences, Nanjing, China
| | - Ping Zhang
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China; Poultry Institute, Chinese Academy of Agricultural Sciences, Yangzhou, China
| | - Hong Zhou
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Yu Sun
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Jinsheng Tang
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Hongjie Fan
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, China.
| |
Collapse
|