1
|
Fan Q, Wang H, Yuan S, Quan Y, Li R, Yi L, Jia A, Wang Y, Wang Y. Pyruvate formate lyase regulates fermentation metabolism and virulence of Streptococcus suis. Virulence 2025; 16:2467156. [PMID: 39977342 PMCID: PMC11845055 DOI: 10.1080/21505594.2025.2467156] [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: 09/17/2024] [Revised: 01/17/2025] [Accepted: 02/10/2025] [Indexed: 02/22/2025] Open
Abstract
Streptococcus suis, a zoonotic pathogen, is commonly found as a commensal bacterium in the respiratory tracts of pigs. Under specific conditions, it becomes invasive and enters the blood, causing severe systemic infections. For S. suis, effective acquisition of carbon sources in different host niches is necessary for its survival. However, as of now, our understanding of the metabolism of S. suis within the host is highly restricted. Pyruvate formate lyase (PFL) plays a crucial role in bacterial survival of in glucose-limited and hypoxic host tissues. Here, we investigated the physiological and metabolic functions of PFL PflB in S. suis and elucidated its pivotal role in regulating virulence within the mucosal and blood niches. We demonstrate that PflB is a key enzyme for S. suis to support mixed-acid fermentation under glucose-limited and hypoxic conditions. Additionally, PflB is involved in regulating S. suis morphology and stress tolerance, and its regulation of capsular polysaccharide content depends on dynamic carbon availability. We also found that PflB is associated with the capacity of S. suis to cause bacteremia and persist in the upper respiratory tract to induce persistent infection. Our results provide highly persuasive evidence for the relationship between metabolic regulation and the virulence of S. suis.
Collapse
Affiliation(s)
- Qingying Fan
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang, Henan, China
- College of Life Sciences, Northwest A&F University, Yangling, Shaanxi, China
- Henan Provincial Engineering Research Center for Detection and Prevention and Control of Emerging Infectious Diseases in Livestock and Poultry, Luoyang, Henan, China
| | - Haikun Wang
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang, Henan, China
- Henan Provincial Engineering Research Center for Detection and Prevention and Control of Emerging Infectious Diseases in Livestock and Poultry, Luoyang, Henan, China
| | - Shuo Yuan
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang, Henan, China
- Henan Provincial Engineering Research Center for Detection and Prevention and Control of Emerging Infectious Diseases in Livestock and Poultry, Luoyang, Henan, China
| | - Yingying Quan
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang, Henan, China
- Henan Provincial Engineering Research Center for Detection and Prevention and Control of Emerging Infectious Diseases in Livestock and Poultry, Luoyang, Henan, China
| | - Rishun Li
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang, Henan, China
- Henan Provincial Engineering Research Center for Detection and Prevention and Control of Emerging Infectious Diseases in Livestock and Poultry, Luoyang, Henan, China
| | - Li Yi
- Henan Provincial Engineering Research Center for Detection and Prevention and Control of Emerging Infectious Diseases in Livestock and Poultry, Luoyang, Henan, China
- College of Life Science, Luoyang Normal University, Luoyang, China
| | - Aiqing Jia
- Guangdong Haid Institute of Animal Husbandry and Veterinary, Guangzhou, P.R. China
| | - Yuxin Wang
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang, Henan, China
- Henan Provincial Engineering Research Center for Detection and Prevention and Control of Emerging Infectious Diseases in Livestock and Poultry, Luoyang, Henan, China
| | - Yang Wang
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang, Henan, China
- Henan Provincial Engineering Research Center for Detection and Prevention and Control of Emerging Infectious Diseases in Livestock and Poultry, Luoyang, Henan, China
| |
Collapse
|
2
|
Yuan S, Liu B, Quan Y, Gao S, Zuo J, Jin W, Shen Y, Li Y, Wang Y, Wang Y. Streptococcus suis regulates central carbon fluxes in response to environment to balance drug resistance and virulence. Microbiol Res 2025; 296:128157. [PMID: 40174362 DOI: 10.1016/j.micres.2025.128157] [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: 10/01/2024] [Revised: 02/28/2025] [Accepted: 03/25/2025] [Indexed: 04/04/2025]
Abstract
Streptococcus suis, a zoonotic pathogen, must adapt to the distinct nutritional environment of the host microhabitat during infection and the establishment of invasive disease, primarily by modulating its metabolic pathways. Metabolic plasticity endows S. suis with an enhanced capacity for environmental adaptation. Multidrug-resistant S. suis is increasingly prevalent due to the extensive use of antibiotics in swine production. In this study, an environment-dependent evolutionary model demonstrated that S. suis could modulate its metabolism in response to environmental changes, thereby altering its drug resistance and virulence. The central carbon flux regulated by pyruvate dehydrogenase (PDH) was identified as a pivotal node in balancing drug resistance and virulence in S. suis. Within the in vivo host environment, increased carbon flux through PDH enhances the production of capsular polysaccharide (CPS), thereby improving immune evasion. Conversely, in the antibiotic environment, reduced carbon flux through PDH downregulates the bacterial metabolic state, which diminishes the induction of toxic metabolites by antibiotics, thereby augmenting drug resistance. This concept provides a reasonable explanation for the puzzling phenomena observed with S. suis in clinical settings. For instance, antibiotic-resistant S. suis has a survival advantage in pig farms where antibiotics are frequently used but is less frequently associated with invasive infections. Furthermore, this study demonstrates that exogenous pyruvate can enhance the bactericidal effect of gentamicin against clinically multidrug-resistant S. suis, offering new insights and potential strategies for controlling clinical multidrug-resistant S. suis infections.
Collapse
Affiliation(s)
- Shuo Yuan
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang 471000, China; Henan Provincial Engineering Research Center for Detection and Prevention and Control of Emerging Infectious Diseases in Livestock and Poultry, Luoyang 471003, China.
| | - Baobao Liu
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang 471000, China; Henan Provincial Engineering Research Center for Detection and Prevention and Control of Emerging Infectious Diseases in Livestock and Poultry, Luoyang 471003, China.
| | - Yingying Quan
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang 471000, China; Henan Provincial Engineering Research Center for Detection and Prevention and Control of Emerging Infectious Diseases in Livestock and Poultry, Luoyang 471003, China.
| | - Shuji Gao
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang 471000, China; Henan Provincial Engineering Research Center for Detection and Prevention and Control of Emerging Infectious Diseases in Livestock and Poultry, Luoyang 471003, China.
| | - Jing Zuo
- Henan Provincial Engineering Research Center for Detection and Prevention and Control of Emerging Infectious Diseases in Livestock and Poultry, Luoyang 471003, China; College of Life Sciences, Sichuan University, Chengdu 610000, China.
| | - Wenjie Jin
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang 471000, China; Henan Provincial Engineering Research Center for Detection and Prevention and Control of Emerging Infectious Diseases in Livestock and Poultry, Luoyang 471003, China.
| | - Yamin Shen
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang 471000, China; Henan Provincial Engineering Research Center for Detection and Prevention and Control of Emerging Infectious Diseases in Livestock and Poultry, Luoyang 471003, China.
| | - Yue Li
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang 471000, China; Henan Provincial Engineering Research Center for Detection and Prevention and Control of Emerging Infectious Diseases in Livestock and Poultry, Luoyang 471003, China.
| | - Yuxin Wang
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang 471000, China; Henan Provincial Engineering Research Center for Detection and Prevention and Control of Emerging Infectious Diseases in Livestock and Poultry, Luoyang 471003, China.
| | - Yang Wang
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang 471000, China; Henan Provincial Engineering Research Center for Detection and Prevention and Control of Emerging Infectious Diseases in Livestock and Poultry, Luoyang 471003, China.
| |
Collapse
|
3
|
Luo W, Yi X, Zhang X, Yuan C, Wei W, Li X, Pu D, Yang J, Zheng H. Taxonomic reassessment of genomes from a divergent population of Streptococcus suis by average nucleotide identity analysis. INFECTION, GENETICS AND EVOLUTION : JOURNAL OF MOLECULAR EPIDEMIOLOGY AND EVOLUTIONARY GENETICS IN INFECTIOUS DISEASES 2025; 131:105753. [PMID: 40287079 DOI: 10.1016/j.meegid.2025.105753] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/18/2025] [Revised: 04/22/2025] [Accepted: 04/23/2025] [Indexed: 04/29/2025]
Abstract
Streptococcus spp., including the emerging zoonotic pathogen S. suis, represent a dominant bacterial population in the porcine nasopharynx. Species identification within the Streptococcus genus remains challenging and frequently ambiguous, resulting in indistinct species boundaries. By employing comparative genomic analyses, a previous study categorized S. suis into a central population and divergent lineages, based on the single nucleotide polymorphisms (SNPs) within core genes and the presence or absence of accessory genes, indicating evolutionary divergence. The divergent lineages were designated as the "out population" in this study for clarity. The 16S rRNA gene sequences of seven putative novel Streptococcus strains isolated from the throats of healthy pigs in China exhibited 100 % similarity to that of the 684_17B strain of S. suis, which clustered in the out population. This study established a threshold average nucleotide identity (ANI) value of 93.17 % for the identification of authentic S. suis. All the 645 genomes from the out population fell below this threshold, indicating that they did not belong to S. suis. Further taxonomic assessment of the 645 genomes from the out population revealed 18 clusters based on pairwise ANI comparisons, using a 92.33 % threshold, determined by pairwise ANI comparisons among the 2422 genomes from the central population of S. suis. These clusters were identified as 12 novel Streptococcus spp. (Streptococcus sp. nov-1-12) and six known Streptococcus spp. by ANI comparisons with type or reference genomes of 98 known Streptococcus spp. The study provides a useful framework for the identification of authentic S. suis and the determination of Streptococcus sp. nov.
Collapse
Affiliation(s)
- Wenbo Luo
- National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Xueli Yi
- Center for Medical Laboratory Science, Affiliated Hospital of Youjiang Medical University for Nationalities, Guangxi, 533000, China
| | - Xiyan Zhang
- National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | | | - Wenfei Wei
- Baise Center for Animal Disease Prevention and Control, Baise, China
| | - Xuezhen Li
- Baise Center for Animal Disease Prevention and Control, Baise, China
| | - Danna Pu
- Research Institute of Public Health, School of Medicine, Nankai University, Tianjin, China
| | - Jing Yang
- National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Han Zheng
- National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China; MOE Key Laboratory of Coal Environmental Pathogenicity and Prevention, School of Public Health, Shanxi Medical University, Taiyuan, China.
| |
Collapse
|
4
|
Yang Q, Li N, Zheng Y, Tian Y, Liang Q, Zhao M, Chu H, Gong Y, Wu T, Wei S, Wang H, Yan G, Li F, Lei L. Identification and characterization of ugpE associated with the full virulence of Streptococcus suis. Vet Res 2025; 56:82. [PMID: 40241177 PMCID: PMC12001685 DOI: 10.1186/s13567-025-01513-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Accepted: 01/28/2025] [Indexed: 04/18/2025] Open
Abstract
Streptococcus suis (S. suis) is an emerging zoonotic pathogen that threatens both animal and human health worldwide. UgpE is a protein subunit of the Ugp (uptake of glycerol phosphate) transporter system that is involved in glycerophospholipid synthesis in bacterial membranes. In this study, an ugpE deletion mutant was constructed and the effects of ugpE deletion on cell morphology, biofilm formation, and virulence were investigated. Deletion of ugpE slowed down bacterial growth and impaired cell chain formation and capsular synthesis by downregulating the mRNA levels of the capsular regulon genes cps-2B, cps-2C, and cps-2S. Deletion of ugpE also led to decreased tolerance to heat, oxidative, and acid-base stress. Crystal violet staining and scanning electron microscopy demonstrate that ugpE may negatively regulate biofilm formation in liquid culture and the rdar biofilm morphotype on agar plates. Moreover, ugpE deletion not only reduced hemolysin activity, survival in whole human blood, and anti-phagocytosis ability against porcine alveolar macrophages (PAM) but also enhanced bacterial adhesion and invasion of human cerebral microvascular endothelial cells (hCMEC/D3) by upregulating the expression of multiple genes associated with cell adhesion. In a mouse infection model, ugpE deletion significantly attenuated virulence and lowered the number of viable bacteria in the blood and major organs, as well as distribution of macrophages. In conclusion, this study identified that UgpE may play a pivotal role in the regulation of various properties including virulence and biofilm formation of S. suis.
Collapse
Affiliation(s)
- Qiulei Yang
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun, China
| | - Na Li
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun, China
| | - Yu Zheng
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun, China
| | - Yanyan Tian
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun, China
| | - Qiao Liang
- Department of First Hospital, Jilin University, Changchun, China
| | - Miaomiao Zhao
- College of Animal Science, Yangtze University, Jingzhou, China
| | - Hong Chu
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun, China
| | - Yan Gong
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun, China
| | - Tong Wu
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun, China
| | - Shaopeng Wei
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun, China
| | - He Wang
- Department of Rehabilitation, The Second Hospital of Jilin University, Changchun, China
| | - Guangmou Yan
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun, China
| | - Fengyang Li
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun, China.
| | - Liancheng Lei
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun, China.
- College of Animal Science, Yangtze University, Jingzhou, China.
| |
Collapse
|
5
|
Gao S, Mao C, Yuan S, Quan Y, Jin W, Shen Y, Zhang X, Wang Y, Yi L, Wang Y. AI-2 quorum sensing-induced galactose metabolism activation in Streptococcus suis enhances capsular polysaccharide-associated virulence. Vet Res 2024; 55:80. [PMID: 38886823 PMCID: PMC11184709 DOI: 10.1186/s13567-024-01335-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Accepted: 05/29/2024] [Indexed: 06/20/2024] Open
Abstract
Bacteria utilize intercellular communication to orchestrate essential cellular processes, adapt to environmental changes, develop antibiotic tolerance, and enhance virulence. This communication, known as quorum sensing (QS), is mediated by the exchange of small signalling molecules called autoinducers. AI-2 QS, regulated by the metabolic enzyme LuxS (S-ribosylhomocysteine lyase), acts as a universal intercellular communication mechanism across gram-positive and gram-negative bacteria and is crucial for diverse bacterial processes. In this study, we demonstrated that in Streptococcus suis (S. suis), a notable zoonotic pathogen, AI-2 QS enhances galactose utilization, upregulates the Leloir pathway for capsular polysaccharide (CPS) precursor production, and boosts CPS synthesis, leading to increased resistance to macrophage phagocytosis. Additionally, our molecular docking and dynamics simulations suggest that, similar to S. pneumoniae, FruA, a fructose-specific phosphoenolpyruvate phosphotransferase system prevalent in gram-positive pathogens, may also function as an AI-2 membrane surface receptor in S. suis. In conclusion, our study demonstrated the significance of AI-2 in the synthesis of galactose metabolism-dependent CPS in S. suis. Additionally, we conducted a preliminary analysis of the potential role of FruA as a membrane surface receptor for S. suis AI-2.
Collapse
Affiliation(s)
- Shuji Gao
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang, 471000, China
- College of Life Science, Luoyang Normal University, Luoyang, 471934, China
- Henan Provincial Engineering Research Center for Detection and Prevention and Control of Emerging Infectious Diseases in Livestock and Poultry, Luoyang, 471003, China
| | - Chenlong Mao
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang, 471000, China
- Henan Provincial Engineering Research Center for Detection and Prevention and Control of Emerging Infectious Diseases in Livestock and Poultry, Luoyang, 471003, China
| | - Shuo Yuan
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang, 471000, China
- Henan Provincial Engineering Research Center for Detection and Prevention and Control of Emerging Infectious Diseases in Livestock and Poultry, Luoyang, 471003, China
| | - Yingying Quan
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang, 471000, China
- Henan Provincial Engineering Research Center for Detection and Prevention and Control of Emerging Infectious Diseases in Livestock and Poultry, Luoyang, 471003, China
| | - Wenjie Jin
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang, 471000, China
- Henan Provincial Engineering Research Center for Detection and Prevention and Control of Emerging Infectious Diseases in Livestock and Poultry, Luoyang, 471003, China
| | - Yamin Shen
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang, 471000, China
- Henan Provincial Engineering Research Center for Detection and Prevention and Control of Emerging Infectious Diseases in Livestock and Poultry, Luoyang, 471003, China
| | - Xiaoling Zhang
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang, 471000, China
- Henan Provincial Engineering Research Center for Detection and Prevention and Control of Emerging Infectious Diseases in Livestock and Poultry, Luoyang, 471003, China
| | - Yuxin Wang
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang, 471000, China
- Henan Provincial Engineering Research Center for Detection and Prevention and Control of Emerging Infectious Diseases in Livestock and Poultry, Luoyang, 471003, China
| | - Li Yi
- College of Life Science, Luoyang Normal University, Luoyang, 471934, China.
- Henan Provincial Engineering Research Center for Detection and Prevention and Control of Emerging Infectious Diseases in Livestock and Poultry, Luoyang, 471003, China.
| | - Yang Wang
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang, 471000, China.
- Henan Provincial Engineering Research Center for Detection and Prevention and Control of Emerging Infectious Diseases in Livestock and Poultry, Luoyang, 471003, China.
| |
Collapse
|
6
|
Liu Y, Lu M, Sun Q, Guo Z, Lin Y, Li S, Huang Y, Li Y, Fu Q. Magnolol attenuates macrophage pyroptosis triggered by Streptococcus equi subsp. zooepidemicus. Int Immunopharmacol 2024; 131:111922. [PMID: 38522137 DOI: 10.1016/j.intimp.2024.111922] [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: 08/05/2023] [Revised: 12/11/2023] [Accepted: 03/21/2024] [Indexed: 03/26/2024]
Abstract
Streptococcus equi subsp. zooepidemicus (SEZ) is a zoonotic bacterial pathogen that causes life-threatening infections and various diseases such as meningitis, endocarditis and pneumonia. With the use of antibiotics being severely restricted in the international community, an alternative to antibiotics is urgently needed against bacterial. In the present study, the herbal extract magnolol protected mice against SEZ infection, reflected by increased survival rate and reduced bacterial burden. A pro-inflammatory form of cell death occurred in SEZ-infected macrophage. Magnolol downregulated the expression of pyroptosis-related proteins and reduced the formation of cell membrane pores in infected macrophages to suppress the development of subsequent inflammation. We further demonstrated that magnolol directly suppressed SEZ-induced macrophage pyroptosis, which partially protected macrophages from SEZ infection. Our study revealed that magnolol suppressed inflammation and protected mice against SEZ infection, providing a possible treatment for SEZ infection.
Collapse
Affiliation(s)
- Yuxuan Liu
- School of Life Science and Engineering, Foshan University, Foshan, China
| | - Meijun Lu
- School of Life Science and Engineering, Foshan University, Foshan, China
| | - Qian Sun
- School of Life Science and Engineering, Foshan University, Foshan, China
| | - Zheng Guo
- School of Life Science and Engineering, Foshan University, Foshan, China
| | - Yongjin Lin
- School of Life Science and Engineering, Foshan University, Foshan, China
| | - Shun Li
- School of Life Science and Engineering, Foshan University, Foshan, China; Foshan University Veterinary Teaching Hospital, 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
| | - Yajuan Li
- 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.
| |
Collapse
|
7
|
Gao S, Jin W, Quan Y, Li Y, Shen Y, Yuan S, Yi L, Wang Y, Wang Y. Bacterial capsules: Occurrence, mechanism, and function. NPJ Biofilms Microbiomes 2024; 10:21. [PMID: 38480745 PMCID: PMC10937973 DOI: 10.1038/s41522-024-00497-6] [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: 09/15/2023] [Accepted: 03/05/2024] [Indexed: 03/17/2024] Open
Abstract
In environments characterized by extended multi-stress conditions, pathogens develop a variety of immune escape mechanisms to enhance their ability to infect the host. The capsules, polymers that bacteria secrete near their cell wall, participates in numerous bacterial life processes and plays a crucial role in resisting host immune attacks and adapting to their niche. Here, we discuss the relationship between capsules and bacterial virulence, summarizing the molecular mechanisms of capsular regulation and pathogenesis to provide new insights into the research on the pathogenesis of pathogenic bacteria.
Collapse
Affiliation(s)
- Shuji Gao
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang, 471000, China
- Henan Provincial Engineering Research Center for Detection and Prevention and Control of Emerging Infectious Diseases in Livestock and Poultry, Luoyang, 471003, China
| | - Wenjie Jin
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang, 471000, China
- Henan Provincial Engineering Research Center for Detection and Prevention and Control of Emerging Infectious Diseases in Livestock and Poultry, Luoyang, 471003, China
| | - Yingying Quan
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang, 471000, China
- Henan Provincial Engineering Research Center for Detection and Prevention and Control of Emerging Infectious Diseases in Livestock and Poultry, Luoyang, 471003, China
| | - Yue Li
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang, 471000, China
- Henan Provincial Engineering Research Center for Detection and Prevention and Control of Emerging Infectious Diseases in Livestock and Poultry, Luoyang, 471003, China
| | - Yamin Shen
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang, 471000, China
- Henan Provincial Engineering Research Center for Detection and Prevention and Control of Emerging Infectious Diseases in Livestock and Poultry, Luoyang, 471003, China
| | - Shuo Yuan
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang, 471000, China
- Henan Provincial Engineering Research Center for Detection and Prevention and Control of Emerging Infectious Diseases in Livestock and Poultry, Luoyang, 471003, China
| | - Li Yi
- Henan Provincial Engineering Research Center for Detection and Prevention and Control of Emerging Infectious Diseases in Livestock and Poultry, Luoyang, 471003, China
- College of Life Science, Luoyang Normal University, Luoyang, 471934, China
| | - Yuxin Wang
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang, 471000, China.
- Henan Provincial Engineering Research Center for Detection and Prevention and Control of Emerging Infectious Diseases in Livestock and Poultry, Luoyang, 471003, China.
| | - Yang Wang
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang, 471000, China.
- Henan Provincial Engineering Research Center for Detection and Prevention and Control of Emerging Infectious Diseases in Livestock and Poultry, Luoyang, 471003, China.
| |
Collapse
|
8
|
Kobayashi K, Kubota H, Tohya M, Ushikubo M, Yamamoto M, Ariyoshi T, Uchitani Y, Mitobe M, Okuno R, Nakagawa I, Sekizaki T, Suzuki J, Sadamasu K. Characterization of pig tonsils as niches for the generation of Streptococcus suis diversity. Vet Res 2024; 55:17. [PMID: 38321502 PMCID: PMC10848530 DOI: 10.1186/s13567-024-01270-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Accepted: 01/12/2024] [Indexed: 02/08/2024] Open
Abstract
Streptococcus suis is a gram-positive bacterium that causes meningitis, septicemia, endocarditis, and other disorders in pigs and humans. We obtained 42 and 50 S. suis isolates from lesions of porcine endocarditis and palatine tonsils, respectively, of clinically healthy pigs in Japan; we then determined their sequence types (STs) by multilocus sequence typing (MLST), cps genotypes, serotypes, and presence of classical major virulence-associated marker genes (mrp, epf, and sly). The 42 isolates from endocarditis lesions were assigned to a limited number of STs and clonal complexes (CCs). On the other hand, the 50 isolates from tonsils were diverse in these traits and seemingly in the degree of virulence, suggesting that tonsils can accommodate a variety of S. suis isolates. The goeBURST full algorithm using tonsil isolates obtained in this study and those retrieved from the database showed that major CCs as well as many other clusters were composed of isolates originating from different countries, and some of the STs were very similar to each other despite the difference in country of origin. These findings indicate that S. suis with not only different but also similar mutations in the genome have survived in tonsils independently across different geographical locations. Therefore, unlike the lesions of endocarditis, the tonsils of pigs seemingly accommodate various S. suis lineages. The present study suggests that S. suis acquired its diversity by natural mutations during colonization and persistence in the tonsils of pigs.
Collapse
Affiliation(s)
- Kai Kobayashi
- Department of Microbiology, Tokyo Metropolitan Institute of Public Health, Hyakunincho 3-24-1, Shinjuku-ku, Tokyo, 169-0073, Japan.
| | - Hiroaki Kubota
- Department of Microbiology, Tokyo Metropolitan Institute of Public Health, Hyakunincho 3-24-1, Shinjuku-ku, Tokyo, 169-0073, Japan
| | - Mari Tohya
- Division of Biomedical Food Research, National Institute of Health Sciences, Tonomachi 3-25-26, Kawasaki-ku, Kawasaki-shi, Kanagawa, 210-9501, Japan
- Department of Microbiology and Department of Microbiome Research, Juntendo University School of Medicine, Hongo 2-1-1, Bunkyo-ku, Tokyo, 113-8421, Japan
| | - Megumi Ushikubo
- Shibaura Meat Sanitary Inspection Station, Tokyo Metropolitan Government, Konan 2-7-19, Minato-ku, Tokyo, 108-0075, Japan
| | - Miki Yamamoto
- Shibaura Meat Sanitary Inspection Station, Tokyo Metropolitan Government, Konan 2-7-19, Minato-ku, Tokyo, 108-0075, Japan
| | - Tsukasa Ariyoshi
- Department of Microbiology, Tokyo Metropolitan Institute of Public Health, Hyakunincho 3-24-1, Shinjuku-ku, Tokyo, 169-0073, Japan
| | - Yumi Uchitani
- Department of Microbiology, Tokyo Metropolitan Institute of Public Health, Hyakunincho 3-24-1, Shinjuku-ku, Tokyo, 169-0073, Japan
| | - Morika Mitobe
- Department of Microbiology, Tokyo Metropolitan Institute of Public Health, Hyakunincho 3-24-1, Shinjuku-ku, Tokyo, 169-0073, Japan
| | - Rumi Okuno
- Department of Microbiology, Tokyo Metropolitan Institute of Public Health, Hyakunincho 3-24-1, Shinjuku-ku, Tokyo, 169-0073, Japan
| | - Ichiro Nakagawa
- Department of Microbiology, Graduate School of Medicine, Kyoto University, Yoshida-Konoe-cho, Sakyo-ku, Kyoto, 606-8501, Japan
| | - Tsutomu Sekizaki
- Department of Microbiology, Graduate School of Medicine, Kyoto University, Yoshida-Konoe-cho, Sakyo-ku, Kyoto, 606-8501, Japan
- Research Center for Food Safety, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Yayoi 1-1-1, Bunkyo-ku, Tokyo, 113-8657, Japan
| | - Jun Suzuki
- Department of Microbiology, Tokyo Metropolitan Institute of Public Health, Hyakunincho 3-24-1, Shinjuku-ku, Tokyo, 169-0073, Japan
| | - Kenji Sadamasu
- Department of Microbiology, Tokyo Metropolitan Institute of Public Health, Hyakunincho 3-24-1, Shinjuku-ku, Tokyo, 169-0073, Japan
| |
Collapse
|
9
|
Gao S, Shen Y, Yuan S, Quan Y, Li X, Wang Y, Yi L, Wang Y. Methyl anthranilate deteriorates biofilm structure of Streptococcus suis and antagonizes the capsular polysaccharide defence effect. Int J Antimicrob Agents 2023; 62:106996. [PMID: 37788717 DOI: 10.1016/j.ijantimicag.2023.106996] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Revised: 08/28/2023] [Accepted: 09/27/2023] [Indexed: 10/05/2023]
Abstract
BACKGROUND Streptococcus suis is an important zoonotic pathogen that often causes biofilm-associated infection. Bacterial biofilm-dependent infection is associated with enhanced drug resistance, making it difficult to eradicate. Novel therapeutic approaches are required urgently to treat infections associated with S. suis biofilm. This study aimed to investigate the effects and mechanisms of methyl anthranilate (MA) on S. suis biofilm. METHODS The effect of MA on S. suis biofilm was determined using the crystal violet method, and the microstructure of the biofilm was observed by electron microscopy. The effects on capsular polysaccharides were determined using the phenol-sulphuric acid method and high-performance liquid chromatography. Adhesion and antiphagocytosis properties of S. suis were detected via cell assays. Molecular docking, molecular dynamics simulation and enzyme activity inhibition assays were used to further explore the effect of MA on AI-2 quorum sensing (QS) of S. suis. Finally, the therapeutic effect of MA was investigated using a mouse infection model. RESULTS MA destroyed the structure of S. suis biofilm, hindered biofilm formation, and reduced the synthesis of capsular polysaccharides significantly, which further weakened the adhesion and antiphagocytosis ability of S. suis. MA had a docking effect and binding site (SER76 and ASP197) similar to S-adenosylhomocysteine (SAH). Further analysis showed that MA competitively bound 5'-methyladenosine/S-adenosine homocysteine nucleosidase with SAH to interfere with AI-2 QS. In a mouse model, MA reduced the bacterial burden and inflammatory infiltrates effectively. CONCLUSION This study revealed the antibiofilm effects of MA, and highlighted its potential as a QS inhibitor against S. suis infection.
Collapse
Affiliation(s)
- Shuji Gao
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang, China; Henan Provincial Engineering Research Centre for Detection and Prevention and Control of Emerging Infectious Diseases in Livestock and Poultry, Luoyang, China
| | - Yamin Shen
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang, China; Henan Provincial Engineering Research Centre for Detection and Prevention and Control of Emerging Infectious Diseases in Livestock and Poultry, Luoyang, China
| | - Shuo Yuan
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang, China; Henan Provincial Engineering Research Centre for Detection and Prevention and Control of Emerging Infectious Diseases in Livestock and Poultry, Luoyang, China
| | - Yingying Quan
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang, China; Henan Provincial Engineering Research Centre for Detection and Prevention and Control of Emerging Infectious Diseases in Livestock and Poultry, Luoyang, China
| | - Xingping Li
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang, China; Henan Provincial Engineering Research Centre for Detection and Prevention and Control of Emerging Infectious Diseases in Livestock and Poultry, Luoyang, China
| | - Yuxin Wang
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang, China; Henan Provincial Engineering Research Centre for Detection and Prevention and Control of Emerging Infectious Diseases in Livestock and Poultry, Luoyang, China
| | - Li Yi
- Henan Provincial Engineering Research Centre for Detection and Prevention and Control of Emerging Infectious Diseases in Livestock and Poultry, Luoyang, China; College of Life Science, Luoyang Normal University, Luoyang, China.
| | - Yang Wang
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang, China; Henan Provincial Engineering Research Centre for Detection and Prevention and Control of Emerging Infectious Diseases in Livestock and Poultry, Luoyang, China.
| |
Collapse
|
10
|
Dudek B, Rybka J, Bugla-Płoskońska G, Korzeniowska-Kowal A, Futoma-Kołoch B, Pawlak A, Gamian A. Biological functions of sialic acid as a component of bacterial endotoxin. Front Microbiol 2022; 13:1028796. [PMID: 36338080 PMCID: PMC9631793 DOI: 10.3389/fmicb.2022.1028796] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Accepted: 10/05/2022] [Indexed: 11/28/2022] Open
Abstract
Lipopolysaccharide (endotoxin, LPS) is an important Gram-negative bacteria antigen. LPS of some bacteria contains sialic acid (Neu5Ac) as a component of O-antigen (O-Ag), in this review we present an overview of bacteria in which the presence of Neu5Ac has been confirmed in their outer envelope and the possible ways that bacteria can acquire Neu5Ac. We explain the role of Neu5Ac in bacterial pathogenesis, and also involvement of Neu5Ac in bacterial evading the host innate immunity response and molecular mimicry phenomenon. We also highlight the role of sialic acid in the mechanism of bacterial resistance to action of serum complement. Despite a number of studies on involvement of Neu5Ac in bacterial pathogenesis many aspects of this phenomenon are still not understood.
Collapse
Affiliation(s)
- Bartłomiej Dudek
- Department of Microbiology, University of Wrocław, Wrocław, Poland
- *Correspondence: Bartłomiej Dudek,
| | - Jacek Rybka
- Department of Immunology of Infectious Diseases, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Wrocław, Poland
| | | | - Agnieszka Korzeniowska-Kowal
- Department of Immunology of Infectious Diseases, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Wrocław, Poland
| | | | | | - Andrzej Gamian
- Department of Immunology of Infectious Diseases, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Wrocław, Poland
- Andrzej Gamian,
| |
Collapse
|
11
|
Liu Z, Xu Q, Liang P, Peng Z, Yao H, Zheng H, Wu Z. The characteristics of population structure and antimicrobial resistance of Streptococcus suis serotype 8, a non-negligible pathotype. Transbound Emerg Dis 2022; 69:e2495-e2505. [PMID: 35560732 DOI: 10.1111/tbed.14592] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Revised: 04/23/2022] [Accepted: 05/04/2022] [Indexed: 11/29/2022]
Abstract
Streptococcus suis, the leading causative agent of swine streptococcosis, is considered as a severe zoonotic and foodborne pathogen for humans. Characteristics of population structure and pathogenicity of S. suis vary significantly by serotypes. As one of the main pathogenic serotypes causing clinical disease in pigs, very little is known about the pathogenicity, population structure, and antimicrobial resistance of S. suis serotype 8 (SS8). In this study, the genome of 26 SS8 strains isolated from healthy and diseased pigs was sequenced. Together with 38 sequences from NCBI, we found that SS8 population was clustered into 12 sequence types (ST) and 4 minimum core genome (MCG) groups, linked to the geographical distribution. Noteworthily, 10 strains belonged to MCG group 1 which was defined to possess the capacity to cause global outbreaks in our previous study. We found that 75% (9/12) of representative SS8 strains were virulent in mice and zebrafish, including all ST1241 strains. No virulence indicators were identified from 67 putative virulence-associated genes mainly identified among pathogenic serotype 2 strains. Instead, we found that the genotype of some of these genes was correlated to their evolution. All 26 isolates were classified as multidrug-resistant strains by antimicrobial susceptibility testing. The high carrying rate of tetO and ermB, mainly disseminated by integrative mobilizable elements, contributed to the prevalent resistance phenotypes to macrolides, lincosamides and tetracyclines. These findings indicated that the pathogenic potential of SS8 cannot be ignored and provided valuable information for SS8 surveillance.
Collapse
Affiliation(s)
- Zhaoying Liu
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
- Key Lab of Animal Bacteriology, Ministry of Agriculture, Nanjing, China
- OIE Reference Lab for Swine Streptococcosis, Nanjing, China
| | - Qiuhua Xu
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
- Key Lab of Animal Bacteriology, Ministry of Agriculture, Nanjing, China
- OIE Reference Lab for Swine Streptococcosis, Nanjing, China
| | - Pujun Liang
- State Key Laboratory of Infectious Disease Prevention and Control, National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Changping, Beijing, China
| | - Zeren Peng
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
- Key Lab of Animal Bacteriology, Ministry of Agriculture, Nanjing, China
- OIE Reference Lab for Swine Streptococcosis, Nanjing, China
| | - Huochun Yao
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
- Key Lab of Animal Bacteriology, Ministry of Agriculture, Nanjing, China
- OIE Reference Lab for Swine Streptococcosis, Nanjing, China
| | - Han Zheng
- State Key Laboratory of Infectious Disease Prevention and Control, National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Changping, Beijing, China
| | - Zongfu Wu
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
- Key Lab of Animal Bacteriology, Ministry of Agriculture, Nanjing, China
- OIE Reference Lab for Swine Streptococcosis, Nanjing, China
| |
Collapse
|
12
|
Hem S, Jarocki VM, Baker DJ, Charles IG, Drigo B, Aucote S, Donner E, Burnard D, Bauer MJ, Harris PNA, Wyrsch ER, Djordjevic SP. Genomic analysis of Elizabethkingia species from aquatic environments: Evidence for potential clinical transmission. CURRENT RESEARCH IN MICROBIAL SCIENCES 2022; 3:100083. [PMID: 34988536 PMCID: PMC8703026 DOI: 10.1016/j.crmicr.2021.100083] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Revised: 11/18/2021] [Accepted: 11/22/2021] [Indexed: 12/12/2022] Open
Abstract
Identification of closely related (< 50 SNV) clinical and environmental aquatic Elizabethkingia anophelis isolates. Identification of a provisional novel species Elizabethkingia umaracha. Novel blaGOB and blaB carbapenemases and extended spectrum β-lactamase blaCME alleles identified in Elizabethkingia spp. Analysis of the global phylogeny and pangenome of Elizabethkingia spp. Identification of novel ICE elements carrying uncharacterised genetic cargo in 67 / 94 (71.3%) of the aquatic environments Elizabethkingia spp.
Elizabethkingia species are ubiquitous in aquatic environments, colonize water systems in healthcare settings and are emerging opportunistic pathogens with reports surfacing in 25 countries across six continents. Elizabethkingia infections are challenging to treat, and case fatality rates are high. Chromosomal blaB, blaGOB and blaCME genes encoding carbapenemases and cephalosporinases are unique to Elizabethkingia spp. and reports of concomitant resistance to aminoglycosides, fluoroquinolones and sulfamethoxazole-trimethoprim are known. Here, we characterized whole-genome sequences of 94 Elizabethkingia isolates carrying multiple wide-spectrum metallo-β-lactamase (blaBand blaGOB) and extended-spectrum serine‑β-lactamase (blaCME) genes from Australian aquatic environments and performed comparative phylogenomic analyses against national clinical and international strains. qPCR was performed to quantify the levels of Elizabethkingia species in the source environments. Antibiotic MIC testing revealed significant resistance to carbapenems and cephalosporins but susceptibility to fluoroquinolones, tetracyclines and trimethoprim-sulfamethoxazole. Phylogenetics show that three environmental E. anophelis isolates are closely related to E. anophelis from Australian clinical isolates (∼36 SNPs), and a new species, E. umeracha sp. novel, was discovered. Genomic signatures provide insight into potentially shared origins and a capacity to transfer mobile genetic elements with both national and international isolates.
Collapse
Affiliation(s)
- Sopheak Hem
- iThree Institute, University of Technology Sydney, Ultimo, NSW 2007, Australia.,Australian Centre for Genomic Epidemiological Microbiology, University of Technology Sydney, PO Box 123, Broadway, NSW 2007, Australia
| | - Veronica M Jarocki
- iThree Institute, University of Technology Sydney, Ultimo, NSW 2007, Australia.,Australian Centre for Genomic Epidemiological Microbiology, University of Technology Sydney, PO Box 123, Broadway, NSW 2007, Australia
| | - Dave J Baker
- Quadram Institute Bioscience, Norwich, United Kingdom
| | - Ian G Charles
- Quadram Institute Bioscience, Norwich, United Kingdom.,Norwich Medical School, Norwich Research Park, Colney Lane, Norwich NR4 7TJ, United Kingdom
| | - Barbara Drigo
- Future Industries Institute, University of South Australia, Adelaide, SA 5001, Australia
| | - Sarah Aucote
- Future Industries Institute, University of South Australia, Adelaide, SA 5001, Australia
| | - Erica Donner
- Future Industries Institute, University of South Australia, Adelaide, SA 5001, Australia
| | - Delaney Burnard
- University of Queensland Centre for Clinical Research, Royal Brisbane and Woman's Hospital, Building 71/918 Royal Brisbane and Women's Hospital Campus, Herston, QLD 4029, Australia
| | - Michelle J Bauer
- University of Queensland Centre for Clinical Research, Royal Brisbane and Woman's Hospital, Building 71/918 Royal Brisbane and Women's Hospital Campus, Herston, QLD 4029, Australia
| | - Patrick N A Harris
- University of Queensland Centre for Clinical Research, Royal Brisbane and Woman's Hospital, Building 71/918 Royal Brisbane and Women's Hospital Campus, Herston, QLD 4029, Australia
| | - Ethan R Wyrsch
- iThree Institute, University of Technology Sydney, Ultimo, NSW 2007, Australia.,Australian Centre for Genomic Epidemiological Microbiology, University of Technology Sydney, PO Box 123, Broadway, NSW 2007, Australia
| | - Steven P Djordjevic
- iThree Institute, University of Technology Sydney, Ultimo, NSW 2007, Australia.,Australian Centre for Genomic Epidemiological Microbiology, University of Technology Sydney, PO Box 123, Broadway, NSW 2007, Australia
| |
Collapse
|
13
|
Li Q, Fei X, Zhang Y, Guo G, Shi H, Zhang W. The biological role of MutT in the pathogenesis of the zoonotic pathogen Streptococcus suis serotype 2. Virulence 2021; 12:1538-1549. [PMID: 34077309 PMCID: PMC8183525 DOI: 10.1080/21505594.2021.1936770] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Revised: 04/27/2021] [Accepted: 05/26/2021] [Indexed: 11/08/2022] Open
Abstract
Streptococcus suis (S. suis) is an important rising pathogen that causes serious diseases in humans and pigs. Although some putative virulence factors of S. suis have been identified, its pathogenic mechanisms are largely unclear. Here, we identified a putative virulence-associated factor MutT, which is unique to S. suis serotype 2 (SS2) virulent strains. To investigate the biological roles of MutT in the SS2 virulent strain ZY05719, the mutT knockout mutant (ΔmutT) was generated and used to explore the phenotypic and virulent variations between the parental and ΔmutT strains. We found that the mutT mutation significantly inhibited cell growth ability, shortened the chain length, and displayed a high susceptibility to H2O2-induced oxidative stress. Moreover, this study revealed that MutT induced the adhesion and invasion of SS2 to host cells. Deletion of mutT increased microbial clearance in host tissues of the infected mice. Sequence alignment results suggested that mutT was encoded in a strain-specific manner, in which the detection was strongly linked to bacterial pathogenicity. In both zebrafish and mice infection models, the virulence of ΔmutT was largely reduced compared with that of ZY05719. Overall, this study provides compelling evidence that MutT is indispensable for the virulence of SS2 and highlights the biological role of MutT in bacteria pathogenesis during infection.
Collapse
Affiliation(s)
- Quan Li
- College of Veterinary Medicine, Yangzhou University, Yangzhou, China
- Jiangsu Co-innovation Center for the Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, China
- MOE Joint International Research Laboratory of Animal Health and Food Safety, Key Lab of Animal Bacteriology, Ministry of Agriculture, OIE Reference Lab for Swine Streptococcosis, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Xia Fei
- College of Veterinary Medicine, Yangzhou University, Yangzhou, China
- Jiangsu Co-innovation Center for the Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, China
| | - Yuhang Zhang
- MOE Joint International Research Laboratory of Animal Health and Food Safety, Key Lab of Animal Bacteriology, Ministry of Agriculture, OIE Reference Lab for Swine Streptococcosis, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Genglin Guo
- MOE Joint International Research Laboratory of Animal Health and Food Safety, Key Lab of Animal Bacteriology, Ministry of Agriculture, OIE Reference Lab for Swine Streptococcosis, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Huoying Shi
- College of Veterinary Medicine, Yangzhou University, Yangzhou, China
- Jiangsu Co-innovation Center for the Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, China
| | - Wei Zhang
- MOE Joint International Research Laboratory of Animal Health and Food Safety, Key Lab of Animal Bacteriology, Ministry of Agriculture, OIE Reference Lab for Swine Streptococcosis, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| |
Collapse
|
14
|
Roodsant TJ, Van Der Putten BCL, Tamminga SM, Schultsz C, Van Der Ark KCH. Identification of Streptococcus suis putative zoonotic virulence factors: A systematic review and genomic meta-analysis. Virulence 2021; 12:2787-2797. [PMID: 34666617 PMCID: PMC8632099 DOI: 10.1080/21505594.2021.1985760] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022] Open
Abstract
Streptococcus suis is an emerging zoonotic pathogen. Over 100 putative virulence factors have been described, but it is unclear to what extent these virulence factors could contribute to zoonotic potential of S. suis. We identified all S. suis virulence factors studied in experimental models of human origin in a systematic review and assessed their contribution to zoonotic potential in a subsequent genomic meta-analysis. PubMed and Scopus were searched for English-language articles that studied S. suis virulence published until 31 March 2021. Articles that analyzed a virulence factor by knockout mutation, purified protein, and/or recombinant protein in a model of human origin, were included. Data on virulence factor, strain characteristics, used human models and experimental outcomes were extracted. All publicly available S. suis genomes with available metadata on host, disease status and country of origin, were included in a genomic meta-analysis. We calculated the ratio of the prevalence of each virulence factor in human and pig isolates. We included 130 articles and 1703 S. suis genomes in the analysis. We identified 53 putative virulence factors that were encoded by genes which are part of the S. suis core genome and 26 factors that were at least twice as prevalent in human isolates as in pig isolates. Hhly3 and NisK/R were particularly enriched in human isolates, after stratification by genetic lineage and country of isolation. This systematic review and genomic meta-analysis have identified virulence factors that are likely to contribute to the zoonotic potential of S. suis.
Collapse
Affiliation(s)
- Thomas J Roodsant
- Amsterdam UMC, University of Amsterdam, Department of Global Health-Amsterdam, Institute for Global Health and Development, Amsterdam, Netherlands.,Department of Medical Microbiology, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| | - Boas C L Van Der Putten
- Amsterdam UMC, University of Amsterdam, Department of Global Health-Amsterdam, Institute for Global Health and Development, Amsterdam, Netherlands.,Department of Medical Microbiology, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| | - Sara M Tamminga
- Department of Medical Microbiology, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| | - Constance Schultsz
- Amsterdam UMC, University of Amsterdam, Department of Global Health-Amsterdam, Institute for Global Health and Development, Amsterdam, Netherlands.,Department of Medical Microbiology, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| | - Kees C H Van Der Ark
- Amsterdam UMC, University of Amsterdam, Department of Global Health-Amsterdam, Institute for Global Health and Development, Amsterdam, Netherlands.,Department of Medical Microbiology, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| |
Collapse
|
15
|
Zhu Y, Dong W, Ma J, Zhang Y, Zhong X, Pan Z, Liu G, Wu Z, Yao H. Comparative genetic analyses provide clues about capsule switching in Streptococcus suis 2 strains with different virulence levels and genetic backgrounds. Microbiol Res 2021; 250:126814. [PMID: 34256310 DOI: 10.1016/j.micres.2021.126814] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Revised: 06/22/2021] [Accepted: 07/04/2021] [Indexed: 12/19/2022]
Abstract
Streptococcus suis (S. suis) is a major bacterial pathogen in the swine industry and an emerging zoonotic agent. S. suis produces an important extracellular component, capsular polysaccharide (CPS), based on which dozens of serotypes have been identified. Through virulence genotyping, we revealed the relatedness between subpopulations of S. suis serotype 2 (SS2), S. suis serotype 3 (SS3) and S. suis serotype 7 (SS7) strains despite their serotype differences. Multilocus sequence typing (MLST) was used to characterize the whole S. suis population and revealed capsule switching between S. suis strains. Importantly, capsule switching occurred in the SS2, SS3 and SS7 strains belonging to CC28 and CC29, which are phylogenetically distinct from the main CC1 SS2 lineage. To further explore capsule switching in S. suis, comparative genomic analyses were performed using available complete S. suis genomes. Phylogenetic analyses suggested that the SS2 strains could be divided into two clades (1 and 2), and those classified into clade 2 colocalized with SS3 and SS7 strains, in accordance with the above virulence genotyping and MLST analyses. Clade 2 SS2 strains presented high genetic similarity to SS3 and SS7 and shared common competence and defensive elements with them but were significantly different from Clade 1 SS2 strains. Notably, although the cps loci shared by Clade 1 and 2 SS2 strains were almost identical, a specific region of the cps locus of strain NSUI002 (Clade 2 SS2) could be found in the SS3 cps locus but not in the Clade 1 SS2 strain. These data indicated that the SS2 strains in CC28 and CC29 might have acquired the cps locus through capsule switching, which could explain the distinct genetic lineages within the SS2 population.
Collapse
Affiliation(s)
- Yinchu Zhu
- Institute of Animal Husbandry and Veterinary Sciences, Zhejiang Academy of Agricultural Sciences, Hangzhou, 310021, China; 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
| | - Wenyang Dong
- 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; Beijing Advanced Innovation Center for Genomics (ICG) & Biomedical Pioneering Innovation Center (BIOPIC), Peking University, Beijing, 100871, China
| | - Jiale Ma
- 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
| | - Yue 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; College of Animal Science and Veterinary Medicine, Henan Agricultural University, Zhengzhou, 450002, China
| | - Xiaojun Zhong
- 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
| | - Zihao Pan
- 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
| | - Guangjin Liu
- 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
| | - Zongfu Wu
- 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
| | - Huochun Yao
- 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
|
16
|
Vieira AZ, Raittz RT, Faoro H. Origin and evolution of nonulosonic acid synthases and their relationship with bacterial pathogenicity revealed by a large-scale phylogenetic analysis. Microb Genom 2021; 7:000563. [PMID: 33848237 PMCID: PMC8208679 DOI: 10.1099/mgen.0.000563] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Accepted: 03/16/2021] [Indexed: 12/28/2022] Open
Abstract
Nonulosonic acids (NulOs) are a group of nine-carbon monosaccharides with different functions in nature. N-acetylneuraminic acid (Neu5Ac) is the most common NulO. It covers the membrane surface of all human cells and is a central molecule in the process of self-recognition via SIGLECS receptors. Some pathogenic bacteria escape the immune system by copying the sialylation of the host cell membrane. Neu5Ac production in these bacteria is catalysed by the enzyme NeuB. Some bacteria can also produce other NulOs named pseudaminic and legionaminic acids, through the NeuB homologues PseI and LegI, respectively. In Opisthokonta eukaryotes, the biosynthesis of Neu5Ac is catalysed by the enzyme NanS. In this study, we used publicly available data of sequences of NulOs synthases to investigate its distribution within the three domains of life and its relationship with pathogenic bacteria. We mined the KEGG database and found 425 NeuB sequences. Most NeuB sequences (58.74 %) from the KEGG orthology database were classified as from environmental bacteria; however, sequences from pathogenic bacteria showed higher conservation and prevalence of a specific domain named SAF. Using the HMM profile we identified 13 941 NulO synthase sequences in UniProt. Phylogenetic analysis of these sequences showed that the synthases were divided into three main groups that can be related to the lifestyle of these bacteria: (I) predominantly environmental, (II) intermediate and (III) predominantly pathogenic. NeuB was widely distributed in the groups. However, LegI and PseI were more concentrated in groups II and III, respectively. We also found that PseI appeared later in the evolutionary process, derived from NeuB. We use this same methodology to retrieve sialic acid synthase sequences from Archaea and Eukarya. A large-scale phylogenetic analysis showed that while the Archaea sequences are spread across the tree, the eukaryotic NanS sequences were grouped in a specific branch in group II. None of the bacterial NanS sequences grouped with the eukaryotic branch. The analysis of conserved residues showed that the synthases of Archaea and Eukarya present a mutation in one of the three catalytic residues, an E134D change, related to a Neisseria meningitidis reference sequence. We also found that the conservation profile is higher between NeuB of pathogenic bacteria and NanS of eukaryotes than between NeuB of environmental bacteria and NanS of eukaryotes. Our large-scale analysis brings new perspectives on the evolution of NulOs synthases, suggesting their presence in the last common universal ancestor.
Collapse
Affiliation(s)
- Alexandre Zanatta Vieira
- Laboratory for Applied Science and Technology in Health, Carlos Chagas Institute, Fiocruz-PR, Algacyr Munhoz Mader street, 3775, Curitiba, Paraná, Brazil
- Graduation Program on Bioinformatics – Universidade Federal do Paraná, Alcides Viera Arcoverde street 1225, Curitiba, Paraná, Brazil
| | - Roberto Tadeu Raittz
- Graduation Program on Bioinformatics – Universidade Federal do Paraná, Alcides Viera Arcoverde street 1225, Curitiba, Paraná, Brazil
| | - Helisson Faoro
- Laboratory for Applied Science and Technology in Health, Carlos Chagas Institute, Fiocruz-PR, Algacyr Munhoz Mader street, 3775, Curitiba, Paraná, Brazil
- Graduation Program on Bioinformatics – Universidade Federal do Paraná, Alcides Viera Arcoverde street 1225, Curitiba, Paraná, Brazil
| |
Collapse
|
17
|
Vezina B, Al-Harbi H, Ramay HR, Soust M, Moore RJ, Olchowy TWJ, Alawneh JI. Sequence characterisation and novel insights into bovine mastitis-associated Streptococcus uberis in dairy herds. Sci Rep 2021; 11:3046. [PMID: 33542314 PMCID: PMC7862697 DOI: 10.1038/s41598-021-82357-3] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Accepted: 01/20/2021] [Indexed: 12/16/2022] Open
Abstract
Streptococcus uberis is one of the most frequent mastitis-causing pathogens isolated from dairy cows. Further understanding of S. uberis genetics may help elucidate the disease pathogenesis. We compared the genomes of S. uberis isolates cultured from dairy cows located in distinctly different geographic regions of Australia. All isolates had novel multi locus sequence types (MLST) indicating a highly diverse population of S. uberis. Global clonal complexes (GCC) were more conserved. GCC ST86 and GCC ST143 represented 30% of the total isolates (n = 27) and were clustered within different geographic regions. Core genome phylogeny revealed low phylogenetic clustering by region, isolation source, and MLST. Identification of putative sortase (srtA) substrates and generation of a custom putative virulence factor database revealed genes which may explain the affinity of S. uberis for mammary tissue, evasion of antimicrobial efforts and disease pathogenesis. Of 27 isolates, four contained antibiotic resistance genes including an antimicrobial resistance cluster containing mel/mef(A), mrsE, vatD, lnuD, and transposon-mediated lnuC was also identified. These are novel genes for S. uberis, which suggests interspecies lateral gene transfer. The presence of resistance genes across the two geographic regions tested within one country supports the need for a careful, tailored, implementation and monitoring of antimicrobial stewardship.
Collapse
Affiliation(s)
- Ben Vezina
- Good Clinical Practice Research Group (GCPRG), The University of Queensland, School of Veterinary Science, Gatton, QLD, 4343, Australia.,Centre for Cell Factories and Biopolymers, Griffith Institute for Drug Discovery, Griffith University, Nathan, Australia
| | - Hulayyil Al-Harbi
- The University of Queensland, School of Veterinary Science, Gatton, QLD, 4343, Australia
| | - Hena R Ramay
- International Microbiome Centre, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Martin Soust
- Terragen Biotech Pty Ltd., Coolum Beach, QLD, 4573, Australia
| | - Robert J Moore
- School of Science, RMIT University, Bundoora, Melbourne, 3083, Australia
| | - Timothy W J Olchowy
- Good Clinical Practice Research Group (GCPRG), The University of Queensland, School of Veterinary Science, Gatton, QLD, 4343, Australia.,Faculty of Veterinary Medicine, University of Calgary, Calgary, AB, T3R 1J3, Canada
| | - John I Alawneh
- Good Clinical Practice Research Group (GCPRG), The University of Queensland, School of Veterinary Science, Gatton, QLD, 4343, Australia. .,The University of Queensland, School of Veterinary Science, Gatton, QLD, 4343, Australia.
| |
Collapse
|
18
|
Tram G, Jennings MP, Blackall PJ, Atack JM. Streptococcus suis pathogenesis-A diverse array of virulence factors for a zoonotic lifestyle. Adv Microb Physiol 2021; 78:217-257. [PMID: 34147186 DOI: 10.1016/bs.ampbs.2020.12.002] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Streptococcus suis is a major cause of respiratory tract and invasive infections in pigs and is responsible for a substantial disease burden in the pig industry. S. suis is also a significant cause of bacterial meningitis in humans, particularly in South East Asia. S. suis expresses a wide array of virulence factors, and although many are described as being required for disease, no single factor has been demonstrated to be absolutely required. The lack of uniform distribution of known virulence factors among individual strains and lack of evidence that any particular virulence factor is essential for disease makes the development of vaccines and treatments challenging. Here we review the current understanding of S. suis virulence factors and their role in the pathogenesis of this important zoonotic pathogen.
Collapse
Affiliation(s)
- Greg Tram
- Institute for Glycomics, Griffith University, Gold Coast, QLD, Australia
| | - Michael P Jennings
- 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
|
19
|
Segura M, Aragon V, Brockmeier SL, Gebhart C, de Greeff A, Kerdsin A, O’Dea MA, Okura M, Saléry M, Schultsz C, Valentin-Weigand P, Weinert LA, Wells JM, Gottschalk M. Update on Streptococcus suis Research and Prevention in the Era of Antimicrobial Restriction: 4th International Workshop on S. suis. Pathogens 2020; 9:pathogens9050374. [PMID: 32422856 PMCID: PMC7281350 DOI: 10.3390/pathogens9050374] [Citation(s) in RCA: 92] [Impact Index Per Article: 18.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2020] [Revised: 05/07/2020] [Accepted: 05/08/2020] [Indexed: 12/16/2022] Open
Abstract
Streptococcus suis is a swine pathogen and a zoonotic agent afflicting people in close contact with infected pigs or pork meat. Sporadic cases of human infections have been reported worldwide. In addition, S. suis outbreaks emerged in Asia, making this bacterium a primary health concern in this part of the globe. In pigs, S. suis disease results in decreased performance and increased mortality, which have a significant economic impact on swine production worldwide. Facing the new regulations in preventive use of antimicrobials in livestock and lack of effective vaccines, control of S. suis infections is worrisome. Increasing and sharing of knowledge on this pathogen is of utmost importance. As such, the pathogenesis and epidemiology of the infection, antimicrobial resistance, progress on diagnosis, prevention, and control were among the topics discussed during the 4th International Workshop on Streptococcus suis (held in Montreal, Canada, June 2019). This review gathers together recent findings on this important pathogen from lectures performed by lead researchers from several countries including Australia, Canada, France, Germany, Japan, Spain, Thailand, The Netherlands, UK, and USA. Finally, policies and recommendations for the manufacture, quality control, and use of inactivated autogenous vaccines are addressed to advance this important field in veterinary medicine.
Collapse
Affiliation(s)
- Mariela Segura
- Research Group on Infectious Diseases in Production Animals and Swine and Poultry Infectious Diseases Research Centre, Faculty of Veterinary Medicine, University of Montreal, St-Hyacinthe, QC J2S 2M2, Canada
- Correspondence: (M.S.); (M.G.); Tel.: +1-450-773-8521 (ext. 0080) (M.S.); +1-450-773-8521 (ext. 8374) (M.G.)
| | - Virginia Aragon
- IRTA, Centre de Recerca en Sanitat Animal (CReSA, IRTA-UAB), Campus de la Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain;
| | | | - Connie Gebhart
- College of Veterinary Medicine, University of Minnesota, St. Paul, MN 55108, USA;
| | - Astrid de Greeff
- Wageningen Bioveterinary Research, 8221 RA Lelystad, The Netherlands;
| | - Anusak Kerdsin
- Faculty of Public Health, Kasetsart University Chalermphrakiat Sakon Nakhon Province Campus, Sakon Nakhon 47000, Thailand;
| | - Mark A O’Dea
- Antimicrobial Resistance and Infectious Disease Laboratory, School of Veterinary and Life Sciences, Murdoch University, Perth, Western Australia 6150, Australia;
| | - Masatoshi Okura
- Division of Bacterial and Parasitic Diseases, National Institute of Animal Health, National Agriculture and Food Research Organization, Tsukuba, Ibaraki 305-0856, Japan;
| | - Mariette Saléry
- French Agency for Veterinary Medicinal Products-French Agency for food, Environmental and Occupational Health Safety (Anses-ANMV), 35302 Fougères, France;
| | - Constance Schultsz
- Department of Global Health-Amsterdam Institute for Global Health and Development and Department of Medical Microbiology, Amsterdam University Medical Centers, University of Amsterdam, 1105 BP Amsterdam, The Netherlands;
| | | | - Lucy A. Weinert
- Department of Veterinary Medicine, University of Cambridge, Cambridge CB3 0ES, UK;
| | - Jerry M. Wells
- Host-Microbe Interactomics Group, Department Animal Sciences, Wageningen University and Research, 6709 PG Wageningen, The Netherlands;
- Department of Veterinary Medicine, University of Cambridge, Cambridge CB3 0ES, UK
| | - Marcelo Gottschalk
- Research Group on Infectious Diseases in Production Animals and Swine and Poultry Infectious Diseases Research Centre, Faculty of Veterinary Medicine, University of Montreal, St-Hyacinthe, QC J2S 2M2, Canada
- Correspondence: (M.S.); (M.G.); Tel.: +1-450-773-8521 (ext. 0080) (M.S.); +1-450-773-8521 (ext. 8374) (M.G.)
| |
Collapse
|
20
|
Arenas J, Bossers-de Vries R, Harders-Westerveen J, Buys H, Ruuls-van Stalle LMF, Stockhofe-Zurwieden N, Zaccaria E, Tommassen J, Wells JM, Smith HE, de Greeff A. In vivo transcriptomes of Streptococcus suis reveal genes required for niche-specific adaptation and pathogenesis. Virulence 2020; 10:334-351. [PMID: 30957693 PMCID: PMC6527017 DOI: 10.1080/21505594.2019.1599669] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Streptococcus suis is a Gram-positive bacterium and a zoonotic pathogen residing in the nasopharynx or the gastrointestinal tract of pigs with a potential of causing life-threatening invasive disease. It is endemic in the porcine production industry worldwide, and it is also an emerging human pathogen. After invasion, the pathogen adapts to cause bacteremia and disseminates to different organs including the brain. To gain insights in this process, we infected piglets with a highly virulent strain of S. suis, and bacterial transcriptomes were obtained from blood and different organs (brain, joints, and heart) when animals had severe clinical symptoms of infection. Microarrays were used to determine the genome-wide transcriptional profile at different infection sites and during growth in standard growth medium in vitro. We observed differential expression of around 30% of the Open Reading Frames (ORFs) and infection-site specific patterns of gene expression. Genes with major changes in expression were involved in transcriptional regulation, metabolism, nutrient acquisition, stress defenses, and virulence, amongst others, and results were confirmed for a subset of selected genes using RT-qPCR. Mutants were generated in two selected genes, and the encoded proteins, i.e., NADH oxidase and MetQ, were shown to be important virulence factors in coinfection experiments and in vitro assays. The knowledge derived from this study regarding S. suis gene expression in vivo and identification of virulence factors is important for the development of novel diagnostic and therapeutic strategies to control S. suis disease.
Collapse
Affiliation(s)
- Jesús Arenas
- a Department of Infection Biology , Wageningen BioVeterinary Research (WBVR) , Lelystad , The Netherlands
| | - Ruth Bossers-de Vries
- a Department of Infection Biology , Wageningen BioVeterinary Research (WBVR) , Lelystad , The Netherlands
| | - José Harders-Westerveen
- a Department of Infection Biology , Wageningen BioVeterinary Research (WBVR) , Lelystad , The Netherlands
| | - Herma Buys
- a Department of Infection Biology , Wageningen BioVeterinary Research (WBVR) , Lelystad , The Netherlands
| | | | | | - Edoardo Zaccaria
- b Host Microbe Interactions , Wageningen UR , Wageningen , The Netherlands
| | - Jan Tommassen
- c Department of Molecular Microbiology and Institute of Biomembranes , Utrecht University , Utrecht , The Netherlands
| | - Jerry M Wells
- b Host Microbe Interactions , Wageningen UR , Wageningen , The Netherlands
| | - Hilde E Smith
- a Department of Infection Biology , Wageningen BioVeterinary Research (WBVR) , Lelystad , The Netherlands
| | - Astrid de Greeff
- a Department of Infection Biology , Wageningen BioVeterinary Research (WBVR) , Lelystad , The Netherlands
| |
Collapse
|
21
|
Auger JP, Payen S, Roy D, Dumesnil A, Segura M, Gottschalk M. Interactions of Streptococcus suis serotype 9 with host cells and role of the capsular polysaccharide: Comparison with serotypes 2 and 14. PLoS One 2019; 14:e0223864. [PMID: 31600314 PMCID: PMC6786723 DOI: 10.1371/journal.pone.0223864] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2019] [Accepted: 09/30/2019] [Indexed: 12/21/2022] Open
Abstract
Streptococcus suis is an important porcine bacterial pathogen and a zoonotic agent responsible for sudden death, septic shock and meningitis, of which serotype 2 is the most widespread, with serotype 14 also causing infections in humans in South-East Asia. Knowledge of its pathogenesis and virulence are almost exclusively based on these two serotypes. Though serotype 9 is responsible for the greatest number of porcine cases in Spain, the Netherlands and Germany, very little information is currently available regarding this serotype. Of the different virulence factors, the capsular polysaccharide (CPS) is required for S. suis virulence as it promotes resistance to phagocytosis and killing and masks surface components responsible for host cell activation. However, these roles have been described for serotypes 2 and 14, whose CPSs are structurally and compositionally similar, both containing sialic acid. Consequently, we evaluated herein the interactions of serotype 9 with host cells and the role of its CPS, which greatly differs from those of serotypes 2 and 14. Results demonstrated that serotype 9 adhesion to but not invasion of respiratory epithelial cells was greater than that of serotypes 2 and 14. Furthermore serotype 9 was more internalized by macrophages but equally resistant to whole blood killing. Though recognition of serotypes 2, 9 and 14 by DCs required MyD88-dependent signaling, in vitro pro-inflammatory mediator production induced by serotype 9 was much lower. In vivo, however, serotype 9 causes an exacerbated inflammatory response, which combined with persistent bacterial presence, is probably responsible for host death during the systemic infection. Though presence of the serotype 9 CPS masks surface components less efficiently than those of serotypes 2 and 14, the serotype 9 CPS remains critical for virulence as it is required for survival in blood and development of clinical disease, and this regardless of its unique composition and structure.
Collapse
Affiliation(s)
- Jean-Philippe Auger
- Research Group on Infectious Diseases in Production Animals (GREMIP) and Swine and Poultry Infectious Diseases Research Center (CRIPA), Department of Pathology and Microbiology, Faculty of Veterinary Medicine, Université de Montréal, Saint-Hyacinthe, Quebec, Canada
| | - Servane Payen
- Research Group on Infectious Diseases in Production Animals (GREMIP) and Swine and Poultry Infectious Diseases Research Center (CRIPA), Department of Pathology and Microbiology, Faculty of Veterinary Medicine, Université de Montréal, Saint-Hyacinthe, Quebec, Canada
| | - David Roy
- Research Group on Infectious Diseases in Production Animals (GREMIP) and Swine and Poultry Infectious Diseases Research Center (CRIPA), Department of Pathology and Microbiology, Faculty of Veterinary Medicine, Université de Montréal, Saint-Hyacinthe, Quebec, Canada
| | - Audrey Dumesnil
- Research Group on Infectious Diseases in Production Animals (GREMIP) and Swine and Poultry Infectious Diseases Research Center (CRIPA), Department of Pathology and Microbiology, Faculty of Veterinary Medicine, Université de Montréal, Saint-Hyacinthe, Quebec, Canada
| | - Mariela Segura
- Research Group on Infectious Diseases in Production Animals (GREMIP) and Swine and Poultry Infectious Diseases Research Center (CRIPA), Department of Pathology and Microbiology, Faculty of Veterinary Medicine, Université de Montréal, Saint-Hyacinthe, Quebec, Canada
| | - Marcelo Gottschalk
- Research Group on Infectious Diseases in Production Animals (GREMIP) and Swine and Poultry Infectious Diseases Research Center (CRIPA), Department of Pathology and Microbiology, Faculty of Veterinary Medicine, Université de Montréal, Saint-Hyacinthe, Quebec, Canada
| |
Collapse
|
22
|
Faoro H, Oliveira WK, Weiss VA, Tadra-Sfeir MZ, Cardoso RL, Balsanelli E, Brusamarello-Santos LCC, Camilios-Neto D, Cruz LM, Raittz RT, Marques ACQ, LiPuma J, Fadel-Picheth CMT, Souza EM, Pedrosa FO. Genome comparison between clinical and environmental strains of Herbaspirillum seropedicae reveals a potential new emerging bacterium adapted to human hosts. BMC Genomics 2019; 20:630. [PMID: 31375067 PMCID: PMC6679464 DOI: 10.1186/s12864-019-5982-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2019] [Accepted: 07/17/2019] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Herbaspirillum seropedicae is an environmental β-proteobacterium that is capable of promoting the growth of economically relevant plants through biological nitrogen fixation and phytohormone production. However, strains of H. seropedicae have been isolated from immunocompromised patients and associated with human infections and deaths. In this work, we sequenced the genomes of two clinical strains of H. seropedicae, AU14040 and AU13965, and compared them with the genomes of strains described as having an environmental origin. RESULTS Both genomes were closed, indicating a single circular chromosome; however, strain AU13965 also carried a plasmid of 42,977 bp, the first described in the genus Herbaspirillum. Genome comparison revealed that the clinical strains lost the gene sets related to biological nitrogen fixation (nif) and the type 3 secretion system (T3SS), which has been described to be essential for interactions with plants. Comparison of the pan-genomes of clinical and environmental strains revealed different sets of accessorial genes. However, antimicrobial resistance genes were found in the same proportion in all analyzed genomes. The clinical strains also acquired new genes and genomic islands that may be related to host interactions. Among the acquired islands was a cluster of genes related to lipopolysaccharide (LPS) biosynthesis. Although highly conserved in environmental strains, the LPS biosynthesis genes in the two clinical strains presented unique and non-orthologous genes within the genus Herbaspirillum. Furthermore, the AU14040 strain cluster contained the neuABC genes, which are responsible for sialic acid (Neu5Ac) biosynthesis, indicating that this bacterium could add it to its lipopolysaccharide. The Neu5Ac-linked LPS could increase the bacterial resilience in the host aiding in the evasion of the immune system. CONCLUSIONS Our findings suggest that the lifestyle transition from environment to opportunist led to the loss and acquisition of specific genes allowing adaptations to colonize and survive in new hosts. It is possible that these substitutions may be the starting point for interactions with new hosts.
Collapse
Affiliation(s)
- Helisson Faoro
- Department of Biochemistry and Molecular Biology, Universidade Federal do Paraná, Coronel Francisco H. dos Santos street, Curitiba, Paraná 81531-980 Brazil
- Graduate Program on Bioinformatics, Universidade Federal do Paraná, Alcides Viera Arcoverde street 1225, Curitiba, Paraná 81520-260 Brazil
- Laboratory of Gene Expression Regulation, Instituto Carlos Chagas, FIOCRUZ, Algacyr Munhoz Mader street, 3775, Curitiba, Paraná 81350-010 Brazil
| | - Willian K. Oliveira
- Graduate Program on Bioinformatics, Universidade Federal do Paraná, Alcides Viera Arcoverde street 1225, Curitiba, Paraná 81520-260 Brazil
- Laboratory of Gene Expression Regulation, Instituto Carlos Chagas, FIOCRUZ, Algacyr Munhoz Mader street, 3775, Curitiba, Paraná 81350-010 Brazil
| | - Vinicius A. Weiss
- Department of Biochemistry and Molecular Biology, Universidade Federal do Paraná, Coronel Francisco H. dos Santos street, Curitiba, Paraná 81531-980 Brazil
- Graduate Program on Bioinformatics, Universidade Federal do Paraná, Alcides Viera Arcoverde street 1225, Curitiba, Paraná 81520-260 Brazil
| | - Michelle Z. Tadra-Sfeir
- Department of Biochemistry and Molecular Biology, Universidade Federal do Paraná, Coronel Francisco H. dos Santos street, Curitiba, Paraná 81531-980 Brazil
| | - Rodrigo L. Cardoso
- Department of Biochemistry and Molecular Biology, Universidade Federal do Paraná, Coronel Francisco H. dos Santos street, Curitiba, Paraná 81531-980 Brazil
| | - Eduardo Balsanelli
- Department of Biochemistry and Molecular Biology, Universidade Federal do Paraná, Coronel Francisco H. dos Santos street, Curitiba, Paraná 81531-980 Brazil
| | - Liziane C. C. Brusamarello-Santos
- Department of Biochemistry and Molecular Biology, Universidade Federal do Paraná, Coronel Francisco H. dos Santos street, Curitiba, Paraná 81531-980 Brazil
| | - Doumit Camilios-Neto
- Department of Biochemistry and Molecular Biology, Universidade Federal do Paraná, Coronel Francisco H. dos Santos street, Curitiba, Paraná 81531-980 Brazil
- Department of Biochemistry and Biothecnology, Universidade Estadual de Londrina, Celso Garcia Cid street, Londrina, Paraná 86057-970 Brazil
| | - Leonardo M. Cruz
- Department of Biochemistry and Molecular Biology, Universidade Federal do Paraná, Coronel Francisco H. dos Santos street, Curitiba, Paraná 81531-980 Brazil
| | - Roberto T. Raittz
- Graduate Program on Bioinformatics, Universidade Federal do Paraná, Alcides Viera Arcoverde street 1225, Curitiba, Paraná 81520-260 Brazil
| | - Ana C. Q. Marques
- Department of Clinical Analyses, Universidade Federal do Paraná, Av. Lothário Meissner 632, Curitiba, Paraná 80210-170 Brazil
| | - John LiPuma
- Department of Pediatrics, University of Michigan, 1500 E. Medical Center Dr, Ann Arbor, MI 48109 USA
| | - Cyntia M. T. Fadel-Picheth
- Department of Clinical Analyses, Universidade Federal do Paraná, Av. Lothário Meissner 632, Curitiba, Paraná 80210-170 Brazil
| | - Emanuel M. Souza
- Department of Biochemistry and Molecular Biology, Universidade Federal do Paraná, Coronel Francisco H. dos Santos street, Curitiba, Paraná 81531-980 Brazil
| | - Fabio O. Pedrosa
- Department of Biochemistry and Molecular Biology, Universidade Federal do Paraná, Coronel Francisco H. dos Santos street, Curitiba, Paraná 81531-980 Brazil
| |
Collapse
|
23
|
PrsA contributes to Streptococcus suis serotype 2 pathogenicity by modulating secretion of selected virulence factors. Vet Microbiol 2019; 236:108375. [PMID: 31500724 DOI: 10.1016/j.vetmic.2019.07.027] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2019] [Revised: 07/22/2019] [Accepted: 07/25/2019] [Indexed: 12/11/2022]
Abstract
Streptococcus suis serotype 2 (S. suis 2) is a major zoonotic pathogen. Parvulin-type peptidyl-prolyl isomerase (PrsA) in S. suis 2 is found surface-associated, pro-inflammatory and cytotoxic. To further explore the roles of PrsA in S. suis 2 infection, we constructed a prsA deletion mutant (ΔprsA) and a complemented strain (CΔprsA). The ΔprsA mutant showed increased length of bacterial chains and decreased growth. Deletion of prsA increased bacterial adhesion to host epithelial cells but with weakened invasion. The ΔprsA mutant had reduced survival in RAW264.7 macrophages and pig whole blood, and significantly attenuated in virulence to mice. All these phenotypes of the mutant could be reversed largely to the levels of its parental strain by gene complementation. Western blotting revealed that suilysin was markedly reduced both in surface-associated (SAP) and secreted fractions (SecP) of ΔprsA, which might be responsible for reduced hemolytic activity. Glyceraldehyde-3-phosphate dehydrogenase (GAPDH) and enolase were significantly increased in both SAP and SecP fractions as a result of prsA deletion. Increased adhesion of the ΔprsA mutant to bEND.3 cells was prevented using polyclonal antibodies against GAPDH and enolase. Overall, we propose that S. suis 2 deploys PrsA to control translocation of important virulence factors, thereby favoring its survival in the host with enhanced pathogenicity by compromising its interactions with the host cells. Further investigation is required to find out how PrsA modulates protein translocation to benefit S. suis infection and if there are other S. suis 2 substrates of potential virulence regulated by PrsA.
Collapse
|
24
|
Lin L, Xu L, Lv W, Han L, Xiang Y, Fu L, Jin M, Zhou R, Chen H, Zhang A. An NLRP3 inflammasome-triggered cytokine storm contributes to Streptococcal toxic shock-like syndrome (STSLS). PLoS Pathog 2019; 15:e1007795. [PMID: 31170267 PMCID: PMC6553798 DOI: 10.1371/journal.ppat.1007795] [Citation(s) in RCA: 95] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2019] [Accepted: 04/28/2019] [Indexed: 02/01/2023] Open
Abstract
Infection with the Streptococcus suis (S. suis) epidemic strain can cause Streptococcal toxic shock-like syndrome (STSLS), which is characterized by a cytokine storm, dysfunction of multiple organs and a high incidence of mortality despite adequate treatment. Despite some progress concerning the contribution of the inflammatory response to STSLS, the precise mechanism underlying STSLS development remains elusive. Here, we use a murine model to demonstrate that caspase-1 activity is critical for STSLS development. Furthermore, we show that inflammasome activation by S. suis is mainly dependent on NLRP3 but not on NLRP1, AIM2 or NLRC4. The important role of NLRP3 activation in STSLS is further confirmed in vivo with the NLRP3 inhibitor MCC950 and nlrp3-knockout mice. By comparison of WT strain with isogenic strains with mutation of various virulence genes for inflammasome activation, Suilysin is essential for inflammasome activation, which is dependent on the membrane perforation activity to cause cytosolic K+ efflux. Moreover, the mutant strain msly (P353L) expressing mutagenic SLY without hemolytic activity was unable to activate the inflammasome and does not cause STSLS. In summary, we demonstrate that the high membrane perforation activity of the epidemic strain induces a high level of NLRP3 inflammasome activation, which is essential for the development of the cytokine storm and multi-organ dysfunction in STSLS and suggests NLRP3 inflammasome as an attractive target for the treatment of STSLS.
Collapse
Affiliation(s)
- Lan Lin
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, China
- Key Laboratory of Preventive Veterinary Medicine in Hubei Province, The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, Hubei, China
| | - Lei Xu
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, China
- Key Laboratory of Preventive Veterinary Medicine in Hubei Province, The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, Hubei, China
| | - Weihua Lv
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, China
- Key Laboratory of Preventive Veterinary Medicine in Hubei Province, The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, Hubei, China
| | - Li Han
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, China
| | - Yaozu Xiang
- Shanghai East Hospital, School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Lei Fu
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, China
- Key Laboratory of Preventive Veterinary Medicine in Hubei Province, The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, Hubei, China
| | - Meilin Jin
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, China
- Key Laboratory of Preventive Veterinary Medicine in Hubei Province, The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, Hubei, China
- Key Laboratory of Development of Veterinary Diagnostic Products, Ministry of Agriculture of the People’s Republic of China, Wuhan, Hubei, China
| | - Rui Zhou
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, China
- Key Laboratory of Preventive Veterinary Medicine in Hubei Province, The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, Hubei, China
- International Research Center for Animal Disease, Ministry of Science and Technology of the People’s Republic of China, Wuhan, Hubei, China
| | - Huanchun Chen
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, China
- Key Laboratory of Preventive Veterinary Medicine in Hubei Province, The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, Hubei, China
- Key Laboratory of Development of Veterinary Diagnostic Products, Ministry of Agriculture of the People’s Republic of China, Wuhan, Hubei, China
| | - Anding Zhang
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, China
- Key Laboratory of Preventive Veterinary Medicine in Hubei Province, The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, Hubei, China
- Key Laboratory of Development of Veterinary Diagnostic Products, Ministry of Agriculture of the People’s Republic of China, Wuhan, Hubei, China
| |
Collapse
|
25
|
Xia X, Qin W, Zhu H, Wang X, Jiang J, Hu J. How Streptococcus suis serotype 2 attempts to avoid attack by host immune defenses. JOURNAL OF MICROBIOLOGY, IMMUNOLOGY, AND INFECTION = WEI MIAN YU GAN RAN ZA ZHI 2019; 52:516-525. [PMID: 30954397 DOI: 10.1016/j.jmii.2019.03.003] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/15/2018] [Revised: 02/20/2019] [Accepted: 03/07/2019] [Indexed: 01/08/2023]
Abstract
Streptococcus suis (S. suis) type 2 (SS2) is an important zoonotic pathogen that causes swine streptococcosis, a widespread infectious disease that occurs in pig production areas worldwide and causes serious economic losses in the pork industry. Hosts recognize pathogen-associated molecular patterns (PAMPs) through pattern recognition receptors (PRRs) to activate both innate and acquired immune responses. However, S. suis has evolved multiple mechanisms to escape host defenses. Pathogenic proteins, such as enolase, double-component regulatory systems, factor H-combining proteins and other pathogenic and virulence factors, contribute to immune escape by evading host phagocytosis, reactive oxygen species (ROS), complement-mediated immune destruction, etc. SS2 can prevent neutrophil extracellular trap (NET) formation to avoid being trapped by porcine neutrophils and disintegrate host immunoglobulins via IgA1 hydrolases and IgM proteases. Currently, the pathogenesis of arthritis and meningitis caused by SS2 infection remains unclear, and further studies are necessary to elucidate it. Understanding immune evasion mechanisms after SS2 infection is important for developing high-efficiency vaccines and targeted drugs.
Collapse
Affiliation(s)
- Xiaojing Xia
- College of Animal Science and Veterinary Medicine, Henan Institute of Science and Technology, Xinxiang, China; Postdoctoral Research Base, Henan Institute of Science and Technology, Xinxiang, China; Postdoctoral Research Station, Henan Agriculture University, Zhengzhou, China
| | - Wanhai Qin
- Amsterdam UMC, University of Amsterdam, Center for Experimental and Molecular Medicine, Amsterdam Infection & Immunity, Meibergdreef 9, 1105AZ Amsterdam, Netherlands
| | - Huili Zhu
- College of Animal Science and Veterinary Medicine, Henan Institute of Science and Technology, Xinxiang, China
| | - Xin Wang
- College of Agriculture and Forestry Science, Linyi University, Linyi, China
| | - Jinqing Jiang
- College of Animal Science and Veterinary Medicine, Henan Institute of Science and Technology, Xinxiang, China.
| | - Jianhe Hu
- College of Animal Science and Veterinary Medicine, Henan Institute of Science and Technology, Xinxiang, China; Postdoctoral Research Base, Henan Institute of Science and Technology, Xinxiang, China.
| |
Collapse
|
26
|
Faulds-Pain A, Shaw HA, Terra VS, Kellner S, Brockmeier SL, Wren BW. The Streptococcos suis sortases SrtB and SrtF are essential for disease in pigs. MICROBIOLOGY-SGM 2018; 165:163-173. [PMID: 30543506 DOI: 10.1099/mic.0.000752] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
The porcine pathogen Streptococcus suis colonizes the upper respiratory tracts of pigs, potentially causing septicaemia, meningitis and death, thus placing a severe burden on the agricultural industry worldwide. It is also a zoonotic pathogen that is known to cause systemic infections and meningitis in humans. Understanding how S. suis colonizes and interacts with its hosts is relevant for future strategies of drug and vaccine development. As with other Gram-positive bacteria, S. suis utilizes enzymes known as sortases to attach specific proteins bearing cell wall sorting signals to its surface, where they can play a role in host-pathogen interactions. The surface proteins of bacteria are often important in adhesion to and invasion of host cells. In this study, markerless in-frame deletion mutants of the housekeeping sortase srtA and the two pilus-associated sortases, srtB and srtF, were generated and their importance in S. suis infections was investigated. We found that all three of these sortases are essential to disease in pigs, concluding that their cognate-sorted proteins may also be useful in protecting pigs against infection.
Collapse
Affiliation(s)
- Alexandra Faulds-Pain
- 1Department of Pathogen Molecular Biology, London School of Hygiene and Tropical Medicine, Keppel Street, London, WC1E 7HT, UK
| | - Helen Alexandra Shaw
- 1Department of Pathogen Molecular Biology, London School of Hygiene and Tropical Medicine, Keppel Street, London, WC1E 7HT, UK.,‡Present address: National Institute for Biological Standards and Control (NIBSC), South Mimms, Potters Bar, EN6 3QG, UK
| | - Vanessa Sofia Terra
- 1Department of Pathogen Molecular Biology, London School of Hygiene and Tropical Medicine, Keppel Street, London, WC1E 7HT, UK
| | - Steven Kellner
- 2USDA, ARS, National Animal Disease Center, 1920 Dayton Avenue, Ames, Iowa 50010, USA
| | - Susan L Brockmeier
- 2USDA, ARS, National Animal Disease Center, 1920 Dayton Avenue, Ames, Iowa 50010, USA
| | - Brendan W Wren
- 1Department of Pathogen Molecular Biology, London School of Hygiene and Tropical Medicine, Keppel Street, London, WC1E 7HT, UK
| |
Collapse
|
27
|
Auger JP, Dolbec D, Roy D, Segura M, Gottschalk M. Role of the Streptococcus suis serotype 2 capsular polysaccharide in the interactions with dendritic cells is strain-dependent but remains critical for virulence. PLoS One 2018; 13:e0200453. [PMID: 30001363 PMCID: PMC6042740 DOI: 10.1371/journal.pone.0200453] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2018] [Accepted: 06/26/2018] [Indexed: 11/18/2022] Open
Abstract
Streptococcus suis serotype 2 is an important porcine bacterial pathogen and zoonotic agent responsible for sudden death, septic shock, and meningitis. However, serotype 2 strains are heterogeneous, composed of a multitude of sequence types (STs) whose distribution greatly varies worldwide. Of the virulence factors presently described for S. suis, the capsular polysaccharide (CPS) is a critical factor implicated in a multitude of functions, including in impairment of phagocytosis and innate immune cell activation by masking underlying bacterial components. However, these roles have been described using Eurasian ST1 and ST7 strains, which greatly differ from North American ST25 strains. Consequently, the capacity of the CPS to mask surface antigens and putative virulence factors in non-Eurasian strains remains unknown. Herein, the role of the S. suis serotype 2 CPS of a prototype intermediate virulent North American ST25 strain, in comparison with that of a virulent European ST1 strain, with regards to interactions with dendritic cells, as well as virulence during the systemic phase of infection, was evaluated. Results demonstrated that the CPS remains critical for virulence and development of clinical disease regardless of strain background, due to its requirement for survival in blood. However, its role in the interactions with dendritic cells is strain-dependent. Consequently, certain key characteristics associated with the CPS are not necessarily applicable to all S. suis serotype 2 strains. This indicates that though certain factors may be important for S. suis serotype 2 virulence, strain background could be as determining, reiterating the need in using strains from varying backgrounds in order to better characterize the S. suis pathogenesis.
Collapse
Affiliation(s)
- Jean-Philippe Auger
- Research Group on Infectious Diseases in Production Animals (GREMIP) and Swine and Poultry Infectious Diseases Research Center (CRIPA), Department of Pathology and Microbiology, Faculty of Veterinary Medicine, University of Montreal, Saint-Hyacinthe, Quebec, Canada
| | - Dominic Dolbec
- Research Group on Infectious Diseases in Production Animals (GREMIP) and Swine and Poultry Infectious Diseases Research Center (CRIPA), Department of Pathology and Microbiology, Faculty of Veterinary Medicine, University of Montreal, Saint-Hyacinthe, Quebec, Canada
| | - David Roy
- Research Group on Infectious Diseases in Production Animals (GREMIP) and Swine and Poultry Infectious Diseases Research Center (CRIPA), Department of Pathology and Microbiology, Faculty of Veterinary Medicine, University of Montreal, Saint-Hyacinthe, Quebec, Canada
| | - Mariela Segura
- Research Group on Infectious Diseases in Production Animals (GREMIP) and Swine and Poultry Infectious Diseases Research Center (CRIPA), Department of Pathology and Microbiology, Faculty of Veterinary Medicine, University of Montreal, Saint-Hyacinthe, Quebec, Canada
| | - Marcelo Gottschalk
- Research Group on Infectious Diseases in Production Animals (GREMIP) and Swine and Poultry Infectious Diseases Research Center (CRIPA), Department of Pathology and Microbiology, Faculty of Veterinary Medicine, University of Montreal, Saint-Hyacinthe, Quebec, Canada
| |
Collapse
|
28
|
Rapid and Visual Detection of Coxiella burnetii Using Recombinase Polymerase Amplification Combined with Lateral Flow Strips. BIOMED RESEARCH INTERNATIONAL 2018; 2018:6417354. [PMID: 29850545 PMCID: PMC5925019 DOI: 10.1155/2018/6417354] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/20/2017] [Accepted: 02/27/2018] [Indexed: 12/25/2022]
Abstract
Coxiella burnetii, a global-distributed biological warfare agent, is the causative agent of Q fever. Correct diagnosis of Q fever is challenging and developing a fast, simple, and reliable detection method is necessary. In this study, recombinase polymerase amplification (RPA) assay combined with lateral flow (LF) test was developed targeting 23S rRNA gene of C. burnetii Xinqiao strain. Primers and probe were designed and synthesized, with one set with high amplification efficiency screened for establishment of the method. Reaction conditions were optimized. Sensitivity, specificity, and accuracy were evaluated. The established RPA-LF reaction could be completed in 30 minutes by combining RPA at 37°C with LF at room temperature, with visually judged results. The method showed good specificity without recognizing other bacteria evaluated. It detected positive plasmid and genomic DNA at levels of 10 copies/reaction and 7 copies/reaction, respectively, levels comparable to that of real-time quantitative PCR (RT-qPCR) targeting 23S rRNA gene established previously. Both RPA-LF and RT-qPCR were used to detect C. burnetii-infected mouse samples and the results were fully consistent. The method showed superior detection performance and will provide technical support against C. burnetii in resources-limited areas.
Collapse
|
29
|
Li Q, Fu Y, He Y, Zhang Y, Qian Y, Yu Y, Yao H, Lu C, Zhang W. Fibronectin-/fibrinogen-binding protein (FBPS) is not a critical virulence factor for the Streptococcus suis serotype 2 strain ZY05719. Vet Microbiol 2017; 208:38-46. [DOI: 10.1016/j.vetmic.2017.07.010] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2017] [Revised: 07/09/2017] [Accepted: 07/09/2017] [Indexed: 01/06/2023]
|
30
|
Abstract
The capsular polysaccharide (CPS) is the major virulence factor of the emerging zoonotic pathogen Streptococcus suis. CPS differences are also the basis for serological differentiation of the species into 29 serotypes. Serotypes 2 and 1/2, which possess identical gene content in their cps loci, express CPSs that differ only by substitution of galactose (Gal) by N-acetylgalactosamine (GalNAc) in the CPS side chain. The same sugar substitution differentiates the CPS of serotypes 14 and 1, whose cps loci are also identical in gene content. Here, using mutagenesis, CPS structural analysis, and protein structure modeling, we report that a single amino acid polymorphism in the glycosyltransferase CpsK defines the enzyme substrate predilection for Gal or GalNAc and therefore determines CPS composition, structure, and strain serotype. We also show that the different CPS structures have similar antiphagocytic properties and that serotype switching has limited impact on the virulence of S. suis.
Collapse
|
31
|
Li Q, Fu Y, Ma C, He Y, Yu Y, Du D, Yao H, Lu C, Zhang W. The non-conserved region of MRP is involved in the virulence of Streptococcus suis serotype 2. Virulence 2017; 8:1274-1289. [PMID: 28362221 DOI: 10.1080/21505594.2017.1313373] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022] Open
Abstract
Muramidase-released protein (MRP) of Streptococcus suis serotype 2 (SS2) is an important epidemic virulence marker with an unclear role in bacterial infection. To investigate the biologic functions of MRP, 3 mutants named Δmrp, Δmrp domain 1 (Δmrp-d1), and Δmrp domain 2 (Δmrp-d2) were constructed to assess the phenotypic changes between the parental strain and the mutant strains. The results indicated that MRP domain 1 (MRP-D1, the non-conserved region of MRP from a virulent strain, a.a. 242-596) played a critical role in adherence of SS2 to host cells, compared with MRP domain 1* (MRP-D1*, the non-conserved region of MRP from a low virulent strain, a.a. 239-598) or MRP domain 2 (MRP-D2, the conserved region of MRP, a.a. 848-1222). We found that MRP-D1 but not MRP-D2, could bind specifically to fibronectin (FN), factor H (FH), fibrinogen (FG), and immunoglobulin G (IgG). Additionally, we confirmed that mrp-d1 mutation significantly inhibited bacteremia and brain invasion in a mouse infection model. The mrp-d1 mutation also attenuated the intracellular survival of SS2 in RAW246.7 macrophages, shortened the growth ability in pig blood and decreased the virulence of SS2 in BALB/c mice. Furthermore, antiserum against MRP-D1 was found to dramatically impede SS2 survival in pig blood. Finally, immunization with recombinant MRP-D1 efficiently enhanced murine viability after SS2 challenge, indicating its potential use in vaccination strategies. Collectively, these results indicated that MRP-D1 is involved in SS2 virulence and eloquently demonstrate the function of MRP in pathogenesis of infection.
Collapse
Affiliation(s)
- Quan Li
- a Key Lab of Animal Bacteriology, OIE Reference Lab for Swine Streptococcosis, College of Veterinary Medicine, Ministry of Agriculture , Nanjing Agricultural University , Nanjing , China
| | - Yang Fu
- a Key Lab of Animal Bacteriology, OIE Reference Lab for Swine Streptococcosis, College of Veterinary Medicine, Ministry of Agriculture , Nanjing Agricultural University , Nanjing , China
| | - Caifeng Ma
- a Key Lab of Animal Bacteriology, OIE Reference Lab for Swine Streptococcosis, College of Veterinary Medicine, Ministry of Agriculture , Nanjing Agricultural University , Nanjing , China
| | - Yanan He
- a Key Lab of Animal Bacteriology, OIE Reference Lab for Swine Streptococcosis, College of Veterinary Medicine, Ministry of Agriculture , Nanjing Agricultural University , Nanjing , China
| | - Yanfei Yu
- a Key Lab of Animal Bacteriology, OIE Reference Lab for Swine Streptococcosis, College of Veterinary Medicine, Ministry of Agriculture , Nanjing Agricultural University , Nanjing , China
| | - Dechao Du
- a Key Lab of Animal Bacteriology, OIE Reference Lab for Swine Streptococcosis, College of Veterinary Medicine, Ministry of Agriculture , Nanjing Agricultural University , Nanjing , China
| | - Huochun Yao
- a Key Lab of Animal Bacteriology, OIE Reference Lab for Swine Streptococcosis, College of Veterinary Medicine, Ministry of Agriculture , Nanjing Agricultural University , Nanjing , China
| | - Chengping Lu
- a Key Lab of Animal Bacteriology, OIE Reference Lab for Swine Streptococcosis, College of Veterinary Medicine, Ministry of Agriculture , Nanjing Agricultural University , Nanjing , China
| | - Wei Zhang
- a Key Lab of Animal Bacteriology, OIE Reference Lab for Swine Streptococcosis, College of Veterinary Medicine, Ministry of Agriculture , Nanjing Agricultural University , Nanjing , China
| |
Collapse
|
32
|
Li M, Cai RJ, Li CL, Song S, Li Y, Jiang ZY, Yang DX. Deletion of ssnA Attenuates the Pathogenicity of Streptococcus suis and Confers Protection against Serovar 2 Strain Challenge. PLoS One 2017; 12:e0169791. [PMID: 28081204 PMCID: PMC5232344 DOI: 10.1371/journal.pone.0169791] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2016] [Accepted: 12/21/2016] [Indexed: 11/19/2022] Open
Abstract
Streptococcus suis serotype 2 (SS2) is a major porcine and human pathogen which causes arthritis, meningitis, and septicemia. Streptococcus suis nuclease A (SsnA) is a recently discovered deoxyribonuclease (DNase), which has been demonstrated to contribute to escape killing in neutrophil extracellular traps (NETs). To further determine the effects of ssnA on virulence, the ssnA deletion mutant (ΔssnA) and its complemented strain (C-ΔssnA) were constructed. The ability of ΔssnA mutant to interact with human laryngeal epithelial cell (Hep-2) was evaluated and it exhibited dramatically decreased ability to adhere to and invade Hep-2 cells. This mutation was found to exhibit significant attenuation of virulence when evaluated in CD1 mice, suggesting ssnA plays a critical role in the pathogenesis of SS2. Finally, we found that immunization with the ΔssnA mutant triggered both antibody responses and cell-mediated immunity, and conferred 80% protection against virulent SS2 challenge in mice. Taken together, our results suggest that ΔssnA represents an attractive candidate for designing an attenuated live vaccine against SS2.
Collapse
Affiliation(s)
- Miao Li
- Institute of Animal Health, Guangdong Academy of Agricultural Sciences, Guangzhou, China
- Guangdong Open Laboratory of Veterinary Public Health, Guangzhou, China
- Guangdong Provincial Key Laboratory of Livestock Disease Prevention, Guangzhou, China
| | - Ru-Jian Cai
- Institute of Animal Health, Guangdong Academy of Agricultural Sciences, Guangzhou, China
- Guangdong Open Laboratory of Veterinary Public Health, Guangzhou, China
- Guangdong Provincial Key Laboratory of Livestock Disease Prevention, Guangzhou, China
- * E-mail: (CLL); (RJC)
| | - Chun-Ling Li
- Institute of Animal Health, Guangdong Academy of Agricultural Sciences, Guangzhou, China
- Guangdong Open Laboratory of Veterinary Public Health, Guangzhou, China
- Guangdong Provincial Key Laboratory of Livestock Disease Prevention, Guangzhou, China
- * E-mail: (CLL); (RJC)
| | - Shuai Song
- Institute of Animal Health, Guangdong Academy of Agricultural Sciences, Guangzhou, China
- Guangdong Open Laboratory of Veterinary Public Health, Guangzhou, China
- Guangdong Provincial Key Laboratory of Livestock Disease Prevention, Guangzhou, China
| | - Yan Li
- Institute of Animal Health, Guangdong Academy of Agricultural Sciences, Guangzhou, China
- Guangdong Open Laboratory of Veterinary Public Health, Guangzhou, China
- Guangdong Provincial Key Laboratory of Livestock Disease Prevention, Guangzhou, China
| | - Zhi-Yong Jiang
- Institute of Animal Health, Guangdong Academy of Agricultural Sciences, Guangzhou, China
- Guangdong Open Laboratory of Veterinary Public Health, Guangzhou, China
- Guangdong Provincial Key Laboratory of Livestock Disease Prevention, Guangzhou, China
| | - Dong-Xia Yang
- Institute of Animal Health, Guangdong Academy of Agricultural Sciences, Guangzhou, China
- Guangdong Open Laboratory of Veterinary Public Health, Guangzhou, China
- Guangdong Provincial Key Laboratory of Livestock Disease Prevention, Guangzhou, China
| |
Collapse
|
33
|
Jiang X, Yang Y, Zhou J, Zhu L, Gu Y, Zhang X, Li X, Fang W. Roles of the Putative Type IV-like Secretion System Key Component VirD4 and PrsA in Pathogenesis of Streptococcus suis Type 2. Front Cell Infect Microbiol 2016; 6:172. [PMID: 27995095 PMCID: PMC5133265 DOI: 10.3389/fcimb.2016.00172] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2016] [Accepted: 11/17/2016] [Indexed: 12/19/2022] Open
Abstract
Streptococcus suis type 2 (SS2) is a zoonotic pathogen causing septic infection, meningitis and pneumonia in pigs and humans. SS2 may cause streptococcal toxic shock syndrome (STSS) probably due to excessive release of inflammatory cytokines. A previous study indicated that the virD4 gene in the putative type IV-like secretion system (T4SS) within the 89K pathogenicity island specific for recent epidemic strains contributed to the development of STSS. However, the functional basis of VirD4 in STSS remains unclear. Here we show that deletion of virD4 led to reduced virulence as shown by about 65% higher LD50, lower bacterial load in liver and brain, and lower level of expression of inflammatory cytokines in mice and cell lines than its parent strain. The ΔVirD4 mutant was more easily phagocytosed, suggesting its role as an anti-phagocytic factor. Oxidative stress that mimic bacterial exposure to respiratory burst of phagocytes upregulated expression of virD4. Proteomic analysis identified 10 secreted proteins of significant differences between the parent and mutant strains under oxidative stress, including PrsA, a peptidyl-prolyl isomerase. The SS2 PrsA expressed in E. coli caused a dose-dependent cell death and increased expression of proinflammatory IL-1β, IL-6 and TNF-α in murine macrophage cells. Our data provide novel insights into the contribution of the VirD4 factor to STSS pathogenesis, possibly via its anti-phagocytic activity, upregulation of its expression upon oxidative stress and its involvement in increased secretion of PrsA as a cell death inducer and proinflammatory effector.
Collapse
Affiliation(s)
- Xiaowu Jiang
- Zhejiang University Institute of Preventive Veterinary Medicine, and Zhejiang Provincial Key Laboratory of Preventive Veterinary Medicine Zhejiang, China
| | - Yunkai Yang
- Zhejiang University Institute of Preventive Veterinary Medicine, and Zhejiang Provincial Key Laboratory of Preventive Veterinary Medicine Zhejiang, China
| | - Jingjing Zhou
- Zhejiang University Institute of Preventive Veterinary Medicine, and Zhejiang Provincial Key Laboratory of Preventive Veterinary Medicine Zhejiang, China
| | - Lexin Zhu
- Zhejiang University Institute of Preventive Veterinary Medicine, and Zhejiang Provincial Key Laboratory of Preventive Veterinary Medicine Zhejiang, China
| | - Yuanxing Gu
- Zhejiang University Institute of Preventive Veterinary Medicine, and Zhejiang Provincial Key Laboratory of Preventive Veterinary Medicine Zhejiang, China
| | - Xiaoyan Zhang
- Zhejiang University Institute of Preventive Veterinary Medicine, and Zhejiang Provincial Key Laboratory of Preventive Veterinary Medicine Zhejiang, China
| | - Xiaoliang Li
- Zhejiang University Institute of Preventive Veterinary Medicine, and Zhejiang Provincial Key Laboratory of Preventive Veterinary Medicine Zhejiang, China
| | - Weihuan Fang
- Zhejiang University Institute of Preventive Veterinary Medicine, and Zhejiang Provincial Key Laboratory of Preventive Veterinary Medicine Zhejiang, China
| |
Collapse
|
34
|
Abstract
A 2-year longitudinal microbiome study of 22 patients who underwent colectomy with an ileal pouch anal anastomosis detected significant increases in distinct populations of Bacteroides during 9 of 11 patient visits that coincided with inflammation (pouchitis). Oligotyping and metagenomic short-read annotation identified Bacteroides populations that occurred in early samples, bloomed during inflammation, and reappeared after antibiotic treatment. Targeted cultivation of Bacteroides isolates from the same individual at multiple time points and from several patients detected subtle genomic changes, including the identification of rapidly evolving genomic elements that differentiate isogenic strains of Bacteroides fragilis from the mucosa versus lumen. Each patient harbored Bacteroides spp. that are closely related to commonly occurring clinical isolates, including Bacteroides ovatus, B. thetaiotaomicron, B. vulgatus, and B. fragilis, which contained unique loci in different patients for synthesis of capsular polysaccharides. The presence of unique Bacteroides capsular polysaccharide loci within different hosts and between the lumen and mucosa may represent adaptations to stimulate, suppress, and evade host-specific immune responses at different microsites of the ileal pouch. This longitudinal study provides an opportunity to describe shifts in the microbiomes of individual patients who suffer from ulcerative colitis (UC) prior to and following inflammation. Pouchitis serves as a model for UC with a predictable incidence of disease onset and enables prospective longitudinal investigations of UC etiology prior to inflammation. Because of insufficient criteria for predicting which patients will develop UC or pouchitis, the interpretation of cross-sectional study designs suffers from lack of information about the microbiome structure and host gene expression patterns that directly correlate with the onset of disease. Our unique longitudinal study design allows each patient to serve as their own control, providing information about the state of the microbiome and host prior to and during the course of disease. Of significance to the broader community, this study identifies microbial strains that may have genetic elements that trigger the onset of disease in susceptible hosts.
Collapse
|
35
|
Ang MY, Dutta A, Wee WY, Dymock D, Paterson IC, Choo SW. Comparative Genome Analysis of Fusobacterium nucleatum. Genome Biol Evol 2016; 8:2928-2938. [PMID: 27540086 PMCID: PMC5630926 DOI: 10.1093/gbe/evw199] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Fusobacterium nucleatum is considered to be a key oral bacterium in recruiting periodontal pathogens into subgingival dental plaque. Currently F. nucleatum can be subdivided into five subspecies. Our previous genome analysis of F. nucleatum W1481 (referred to hereafter as W1481), isolated from an 8-mm periodontal pocket in a patient with chronic periodontitis, suggested the possibility of a new subspecies. To further investigate the biology and relationships of this possible subspecies with other known subspecies, we performed comparative analysis between W1481 and 35 genome sequences represented by the five known Fusobacterium subspecies. Our analyses suggest that W1481 is most likely a new F. nucleatum subspecies, supported by evidence from phylogenetic analyses and maximal unique match indices (MUMi). Interestingly, we found a horizontally transferred W1481-specific genomic island harboring the tripartite ATP-independent (TRAP)-like transporter genes, suggesting this bacterium might have a high-affinity transport system for the C4-dicarboxylates malate, succinate, and fumarate. Moreover, we found virulence genes in the W1481 genome that may provide a strong defense mechanism which might enable it to colonize and survive within the host by evading immune surveillance. This comparative study provides better understanding of F. nucleatum and the basis for future functional work on this important pathogen.
Collapse
Affiliation(s)
- Mia Yang Ang
- Genome Informatics Research Laboratory, Centre for Research in Biotechnology for Agriculture (CEBAR), High Impact Research Building, University of Malaya, Kuala Lumpur, Malaysia Department of Oral and Craniofacial Sciences, Faculty of Dentistry, University of Malaya, Kuala Lumpur, Malaysia
| | - Avirup Dutta
- Genome Informatics Research Laboratory, Centre for Research in Biotechnology for Agriculture (CEBAR), High Impact Research Building, University of Malaya, Kuala Lumpur, Malaysia
| | - Wei Yee Wee
- Genome Informatics Research Laboratory, Centre for Research in Biotechnology for Agriculture (CEBAR), High Impact Research Building, University of Malaya, Kuala Lumpur, Malaysia Department of Oral and Craniofacial Sciences, Faculty of Dentistry, University of Malaya, Kuala Lumpur, Malaysia
| | - David Dymock
- School of Oral & Dental Sciences, University of Bristol, Bristol, United Kingdom
| | - Ian C Paterson
- Department of Oral and Craniofacial Sciences, Faculty of Dentistry, University of Malaya, Kuala Lumpur, Malaysia Oral Cancer Research and Coordinating Centre (OCRCC), Faculty of Dentistry, University of Malaya, Kuala Lumpur, Malaysia
| | - Siew Woh Choo
- Genome Informatics Research Laboratory, Centre for Research in Biotechnology for Agriculture (CEBAR), High Impact Research Building, University of Malaya, Kuala Lumpur, Malaysia Department of Oral and Craniofacial Sciences, Faculty of Dentistry, University of Malaya, Kuala Lumpur, Malaysia Genome Solutions Sdn Bhd, Suite 8, Innovation Incubator UM, Level 5, Research Management & Innovation Complex, University of Malaya, Kuala Lumpur, Malaysia
| |
Collapse
|
36
|
Segura M, Calzas C, Grenier D, Gottschalk M. Initial steps of the pathogenesis of the infection caused by Streptococcus suis: fighting against nonspecific defenses. FEBS Lett 2016; 590:3772-3799. [PMID: 27539145 DOI: 10.1002/1873-3468.12364] [Citation(s) in RCA: 96] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2016] [Revised: 08/11/2016] [Accepted: 08/16/2016] [Indexed: 12/16/2022]
Abstract
Interactions between a bacterial pathogen and its potentially susceptible host are initiated with the colonization step. During respiratory/oral infection, the pathogens must compete with the normal microflora, resist defense mechanisms of the local mucosal immunity, and finally reach, adhere, and breach the mucosal epithelial cell barrier in order to induce invasive disease. This is the case during infection by the swine and zoonotic pathogen Streptococcus suis, which is able to counteract mucosal barriers to induce severe meningitis and sepsis in swine and in humans. The initial steps of the pathogenesis of S. suis infection has been a neglected area of research, overshadowed by studies on the systemic and central nervous phases of the disease. In this Review article, we provide for the first time, an exclusive focus on S. suis colonization and the potential mechanisms involved in S. suis establishment at the mucosa, as well as the mechanisms regulating mucosal barrier breakdown. The role of mucosal immunity is also addressed. Finally, we demystify the extensive list of putative adhesins and virulence factors reported to be involved in the initial steps of pathogenesis by S. suis.
Collapse
Affiliation(s)
- Mariela Segura
- Laboratory of Immunology, Faculty of Veterinary Medicine, University of Montreal, Saint-Hyacinthe, Quebec, Canada.,Swine and Poultry Infectious Diseases Research Centre (CRIPA), Faculty of Veterinary Medicine, University of Montreal, Saint-Hyacinthe, QC, Canada
| | - Cynthia Calzas
- Laboratory of Immunology, Faculty of Veterinary Medicine, University of Montreal, Saint-Hyacinthe, Quebec, Canada.,Swine and Poultry Infectious Diseases Research Centre (CRIPA), Faculty of Veterinary Medicine, University of Montreal, Saint-Hyacinthe, QC, Canada.,Laboratory of Streptococcus suis, Faculty of Veterinary Medicine, University of Montreal, Saint-Hyacinthe, Quebec, Canada
| | - Daniel Grenier
- Swine and Poultry Infectious Diseases Research Centre (CRIPA), Faculty of Veterinary Medicine, University of Montreal, Saint-Hyacinthe, QC, Canada.,Groupe de Recherche en Écologie Buccale (GREB), Faculté de Médecine Dentaire, Université Laval, Quebec City, Quebec, Canada
| | - Marcelo Gottschalk
- Swine and Poultry Infectious Diseases Research Centre (CRIPA), Faculty of Veterinary Medicine, University of Montreal, Saint-Hyacinthe, QC, Canada.,Laboratory of Streptococcus suis, Faculty of Veterinary Medicine, University of Montreal, Saint-Hyacinthe, Quebec, Canada
| |
Collapse
|
37
|
Auger JP, Fittipaldi N, Benoit-Biancamano MO, Segura M, Gottschalk M. Virulence Studies of Different Sequence Types and Geographical Origins of Streptococcus suis Serotype 2 in a Mouse Model of Infection. Pathogens 2016; 5:pathogens5030048. [PMID: 27409640 PMCID: PMC5039428 DOI: 10.3390/pathogens5030048] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2016] [Revised: 06/30/2016] [Accepted: 07/05/2016] [Indexed: 01/22/2023] Open
Abstract
Multilocus sequence typing previously identified three predominant sequence types (STs) of Streptococcus suis serotype 2: ST1 strains predominate in Eurasia while North American (NA) strains are generally ST25 and ST28. However, ST25/ST28 and ST1 strains have also been isolated in Asia and NA, respectively. Using a well-standardized mouse model of infection, the virulence of strains belonging to different STs and different geographical origins was evaluated. Results demonstrated that although a certain tendency may be observed, S. suis serotype 2 virulence is difficult to predict based on ST and geographical origin alone; strains belonging to the same ST presented important differences of virulence and did not always correlate with origin. The only exception appears to be NA ST28 strains, which were generally less virulent in both systemic and central nervous system (CNS) infection models. Persistent and high levels of bacteremia accompanied by elevated CNS inflammation are required to cause meningitis. Although widely used, in vitro tests such as phagocytosis and killing assays require further standardization in order to be used as predictive tests for evaluating virulence of strains. The use of strains other than archetypal strains has increased our knowledge and understanding of the S. suis serotype 2 population dynamics.
Collapse
Affiliation(s)
- Jean-Philippe Auger
- Swine and Poultry Infectious Diseases Research Center (CRIPA), Department of Pathology and Microbiology, Faculty of Veterinary Medicine, University of Montreal, 3200 Sicotte St., Saint-Hyacinthe, QC J2S 2M2, Canada.
| | - Nahuel Fittipaldi
- Public Health Ontario and Department of Laboratory Medicine and Pathobiology, Faculty of Medicine, University of Toronto, 661 University Avenue, Toronto, ON M5G 1M1, Canada.
| | - Marie-Odile Benoit-Biancamano
- Swine and Poultry Infectious Diseases Research Center (CRIPA), Department of Pathology and Microbiology, Faculty of Veterinary Medicine, University of Montreal, 3200 Sicotte St., Saint-Hyacinthe, QC J2S 2M2, Canada.
| | - Mariela Segura
- Swine and Poultry Infectious Diseases Research Center (CRIPA), Department of Pathology and Microbiology, Faculty of Veterinary Medicine, University of Montreal, 3200 Sicotte St., Saint-Hyacinthe, QC J2S 2M2, Canada.
| | - Marcelo Gottschalk
- Swine and Poultry Infectious Diseases Research Center (CRIPA), Department of Pathology and Microbiology, Faculty of Veterinary Medicine, University of Montreal, 3200 Sicotte St., Saint-Hyacinthe, QC J2S 2M2, Canada.
| |
Collapse
|
38
|
Li M, Shao ZQ, Guo Y, Wang L, Hou T, Hu D, Zheng F, Tang J, Wang C, Feng Y, Gao J, Pan X. The type II histidine triad protein HtpsC is a novel adhesion with the involvement of Streptococcus suis virulence. Virulence 2016; 6:631-41. [PMID: 26151575 PMCID: PMC4720241 DOI: 10.1080/21505594.2015.1056971] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Streptococcal histidine triad proteins HTPs are widely distributed within the Streptococcus genus. Based on the phylogenetic relationship and domain composition, HTPs are classified into type I and type II subfamilies. Previous studies revealed that several pathogenic streptococci contain more than one htp gene. We found that the highly virulent strain of Streptococcus suis 2 (S. suis 2), 05ZYH33 encodes 3 HTPs, designated HtpsA (previously described as HtpS), HtpsB, and HtpsC. Among them, HtpsC is the only member that contains leucine-rich repeat (LRR) domains at the C-terminal. In this study, we demonstrated that the recombinant HtpsC could bind to 2 different components of human ECM complex laminin and fibronectin in vitro, suggesting that it is a novel adhesin of S. suis 2. Having constructed an htpsC mutant, we evaluated its role in the pathogenesis of the highly virulent S. suis 2 strain 05ZYH33. Our data showed that inactivation of htpsC significantly affected adherence of S. suis 2 to Hep-2 cells and shortened the survival of the bacteria in whole blood. Furthermore, deletion of htpsC significantly attenuated the virulence of S. suis 2 in mice. These results demonstrated that htpsC was involved in the pathogenesis of the highly virulent S. suis 2 strain 05ZYH33. In line with the observation, immunization with HtpsC significantly prolonged mice's survival after S. suis 05ZYH33 challenge, indicating its potential use in the vaccine development against S. suis.
Collapse
Affiliation(s)
- Min Li
- a School of Laboratory Medicine and Life Science ; Wenzhou Medical University ; Wenzhou , China.,b Department of Epidemiology ; Research Institute for Medicine of Nanjing Command ; Nanjing , China
| | - Zhu-Qing Shao
- b Department of Epidemiology ; Research Institute for Medicine of Nanjing Command ; Nanjing , China.,c State Key Laboratory of Pharmaceutical Biotechnology ; School of Life Sciences ; Nanjing University ; Nanjing , China
| | - Yuqing Guo
- a School of Laboratory Medicine and Life Science ; Wenzhou Medical University ; Wenzhou , China.,b Department of Epidemiology ; Research Institute for Medicine of Nanjing Command ; Nanjing , China
| | - Ling Wang
- b Department of Epidemiology ; Research Institute for Medicine of Nanjing Command ; Nanjing , China.,d School of Life Sciences ; Nanjing Normal University ; Nanjing , China
| | - Tianqing Hou
- b Department of Epidemiology ; Research Institute for Medicine of Nanjing Command ; Nanjing , China
| | - Dan Hu
- b Department of Epidemiology ; Research Institute for Medicine of Nanjing Command ; Nanjing , China
| | - Feng Zheng
- b Department of Epidemiology ; Research Institute for Medicine of Nanjing Command ; Nanjing , China
| | - Jiaqi Tang
- b Department of Epidemiology ; Research Institute for Medicine of Nanjing Command ; Nanjing , China.,e Institute of Laboratory Medicine ; Jinling Hospital ; Nanjing , China
| | - Changjun Wang
- b Department of Epidemiology ; Research Institute for Medicine of Nanjing Command ; Nanjing , China
| | - Youjun Feng
- f Center for Infection & Immunity ; Department of Medical Microbiology & Parasitology ; Zhejiang University School of Medicine ; Hangzhou, Zhejiang , China
| | - Jimin Gao
- a School of Laboratory Medicine and Life Science ; Wenzhou Medical University ; Wenzhou , China
| | - Xiuzhen Pan
- a School of Laboratory Medicine and Life Science ; Wenzhou Medical University ; Wenzhou , China.,b Department of Epidemiology ; Research Institute for Medicine of Nanjing Command ; Nanjing , China
| |
Collapse
|
39
|
Efficient suilysin-mediated invasion and apoptosis in porcine respiratory epithelial cells after streptococcal infection under air-liquid interface conditions. Sci Rep 2016; 6:26748. [PMID: 27229328 PMCID: PMC4882623 DOI: 10.1038/srep26748] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2015] [Accepted: 05/05/2016] [Indexed: 12/25/2022] Open
Abstract
Streptococci may colonize the epithelium in the airways and other entry sites. While local infection often remains asymptomatic, severe or even fatal diseases occur when streptococci become invasive and spread to different sites in the infected host. We have established porcine respiratory air-liquid interface cultures (ALI) from the porcine lung to analyze the interaction of streptococci with their primary target cells. As representative of the streptococcal family we chose Streptococcus suis (S. suis) that is not only a major swine respiratory pathogen but can also infect humans. Suilysin, a cholesterol-dependent cytolysin (CDC), is an important virulence factor. By comparing a S. suis wt strain with a suilysin-deficient mutant, we demonstrate that suilysin contributes to (i) adherence to airway cells (ii) loss of ciliated cells (iii) apoptosis, and (iv) invasion. Furthermore, we show that cytolytic activity of suilysin is crucial for these effects. A striking result of our analysis was the high efficiency of S. suis-induced apoptosis and invasion upon infection under ALI conditions. These properties have been reported to be less efficient when analyzed with immortalized cells. We hypothesize that soluble effectors such as suilysin are present at higher concentrations in cells kept at ALI conditions and thus more effective. These results should be relevant also for infection of the respiratory tract by other respiratory pathogens.
Collapse
|
40
|
Functional definition of BirA suggests a biotin utilization pathway in the zoonotic pathogen Streptococcus suis. Sci Rep 2016; 6:26479. [PMID: 27217336 PMCID: PMC4877710 DOI: 10.1038/srep26479] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2016] [Accepted: 05/04/2016] [Indexed: 11/30/2022] Open
Abstract
Biotin protein ligase is universal in three domains of life. The paradigm version of BPL is the Escherichia coli BirA that is also a repressor for the biotin biosynthesis pathway. Streptococcus suis, a leading bacterial agent for swine diseases, seems to be an increasingly-important opportunistic human pathogen. Unlike the scenario in E. coli, S. suis lacks the de novo biotin biosynthesis pathway. In contrast, it retains a bioY, a biotin transporter-encoding gene, indicating an alternative survival strategy for S. suis to scavenge biotin from its inhabiting niche. Here we report functional definition of S. suis birA homologue. The in vivo functions of the birA paralogue with only 23.6% identity to the counterpart of E. coli, was judged by its ability to complement the conditional lethal mutants of E. coli birA. The recombinant BirA protein of S. suis was overexpressed in E. coli, purified to homogeneity and verified with MS. Both cellulose TLC and MALDI-TOFF-MS assays demonstrated that the S. suis BirA protein catalyzed the biotinylation reaction of its acceptor biotin carboxyl carrier protein. EMSA assays confirmed binding of the bioY gene to the S. suis BirA. The data defined the first example of the bifunctional BirA ligase/repressor in Streptococcus.
Collapse
|
41
|
Gao T, Tan M, Liu W, Zhang C, Zhang T, Zheng L, Zhu J, Li L, Zhou R. GidA, a tRNA Modification Enzyme, Contributes to the Growth, and Virulence of Streptococcus suis Serotype 2. Front Cell Infect Microbiol 2016; 6:44. [PMID: 27148493 PMCID: PMC4835480 DOI: 10.3389/fcimb.2016.00044] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2016] [Accepted: 03/27/2016] [Indexed: 11/16/2022] Open
Abstract
Glucose-inhibited division protein (GidA), is a tRNA modification enzyme functioning together with MnmE in the addition of a carboxymethylaminomethyl group to position 5 of the anticodon wobble uridine of tRNA. Here, we report a GidA homolog from a Chinese isolate SC-19 of the zoonotic Streptococcus suis serotype 2 (SS2). gidA disruption led to a defective growth, increased capsule thickness, and reduced hemolytic activity. Moreover, the gidA deletion mutant (ΔgidA) displayed reduced mortality and bacterial loads in mice, reduced ability of adhesion to and invasion in epithelial cells, and increased sensitivity to phagocytosis. The iTRAQ analysis identified 372 differentially expressed (182 up- and 190 down-regulated) proteins in ΔgidA and SC-19. Numerous DNA replication, cell division, and virulence associated proteins were downregulated, whereas many capsule synthesis enzymes were upregulated by gidA disruption. This is consistent with the phenotypes of the mutant. Thus, GidA is a translational regulator that plays an important role in the growth, cell division, capsule biosynthesis, and virulence of SS2. Our findings provide new insight into the regulatory function of GidA in bacterial pathogens.
Collapse
Affiliation(s)
- Ting Gao
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural UniversityWuhan, China; Veterinary Medicine Laboratory, Institute of Animal Husbandry and Veterinary Science, Hubei Academy of Agricultural ScienceWuhan, China; Wuhan Chopper Biology Co., Ltd.Wuhan, China
| | - Meifang Tan
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University Wuhan, China
| | - Wanquan Liu
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University Wuhan, China
| | - Chunyan Zhang
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University Wuhan, China
| | - Tengfei Zhang
- Veterinary Medicine Laboratory, Institute of Animal Husbandry and Veterinary Science, Hubei Academy of Agricultural Science Wuhan, China
| | - Linlin Zheng
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University Wuhan, China
| | - Jiawen Zhu
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University Wuhan, China
| | - Lu Li
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural UniversityWuhan, China; Cooperative Innovation Center of Sustainable Pig ProductionWuhan, China
| | - Rui Zhou
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural UniversityWuhan, China; Cooperative Innovation Center of Sustainable Pig ProductionWuhan, China
| |
Collapse
|
42
|
Haas B, Grenier D. Impact of Sub-Inhibitory Concentrations of Amoxicillin on Streptococcus suis Capsule Gene Expression and Inflammatory Potential. Pathogens 2016; 5:pathogens5020037. [PMID: 27104570 PMCID: PMC4931388 DOI: 10.3390/pathogens5020037] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2016] [Revised: 04/11/2016] [Accepted: 04/13/2016] [Indexed: 11/16/2022] Open
Abstract
Streptococcus suis is an important swine pathogen and emerging zoonotic agent worldwide causing meningitis, endocarditis, arthritis and septicemia. Among the 29 serotypes identified to date, serotype 2 is mostly isolated from diseased pigs. Although several virulence mechanisms have been characterized in S. suis, the pathogenesis of S. suis infections remains only partially understood. This study focuses on the response of S. suis P1/7 to sub-inhibitory concentrations of amoxicillin. First, capsule expression was monitored by qRT-PCR when S. suis was cultivated in the presence of amoxicillin. Then, the pro-inflammatory potential of S. suis P1/7 culture supernatants or whole cells conditioned with amoxicillin was evaluated by monitoring the activation of the NF-κB pathway in monocytes and quantifying pro-inflammatory cytokines secreted by macrophages. It was found that amoxicillin decreased capsule expression in S. suis. Moreover, conditioning the bacterium with sub-inhibitory concentrations of amoxicillin caused an increased activation of the NF-κB pathway in monocytes following exposure to bacterial culture supernatants and to a lesser extent to whole bacterial cells. This was associated with an increased secretion of pro-inflammatory cytokines (CXCL8, IL-6, IL-1β) by macrophages. This study identified a new mechanism by which S. suis may increase its inflammatory potential in the presence of sub-inhibitory concentrations of amoxicillin, a cell wall-active antibiotic, thus challenging its use for preventive treatments or as growth factor.
Collapse
Affiliation(s)
- Bruno Haas
- Groupe de Recherche en Écologie Buccale (GREB), Faculté de Médecine Dentaire, Université Laval, Quebec City, QC G1V 0A6, Canada.
| | - Daniel Grenier
- Groupe de Recherche en Écologie Buccale (GREB), Faculté de Médecine Dentaire, Université Laval, Quebec City, QC G1V 0A6, Canada.
- Centre de Recherche en Infectiologie Porcine et Avicole (CRIPA), Fonds de Recherche du Québec-Nature et Technologies (FRQNT), Saint-Hyacinthe, QC J2S 2M2, Canada.
| |
Collapse
|
43
|
Feng L, Zhu J, Chang H, Gao X, Gao C, Wei X, Yuan F, Bei W. The CodY regulator is essential for virulence in Streptococcus suis serotype 2. Sci Rep 2016; 6:21241. [PMID: 26883762 PMCID: PMC4756307 DOI: 10.1038/srep21241] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2015] [Accepted: 01/20/2016] [Indexed: 01/10/2023] Open
Abstract
The main role of CodY, a global regulatory protein in most low G + C gram-positive bacteria, is in transcriptional repression. To study the functions of CodY in Streptococcus suis serotype 2 (S. suis 2), a mutant codY clone named ∆codY was constructed to explore the phenotypic variation between ∆codY and the wild-type strain. The result showed that the codY mutation significantly inhibited cell growth, adherence and invasion ability of S. suis 2 to HEp-2 cells. The codY mutation led to decreased binding of the pathogen to the host cells, easier clearance by RAW264.7 macrophages and decreased growth ability in fresh blood of Cavia porcellus. The codY mutation also attenuated the virulence of S. suis 2 in BALB/c mice. Morphological analysis revealed that the codY mutation decreased the thickness of the capsule of S. suis 2 and changed the surface structures analylized by SDS-PAGE. Finally, the codY mutation altered the expressions of many virulence related genes, including sialic acid synthesis genes, leading to a decreased sialic acid content in capsule. Overall, mutation of codY modulated bacterial virulence by affecting the growth and colonization of S. suis 2, and at least via regulating sialic acid synthesis and capsule thickness.
Collapse
Affiliation(s)
- Liping Feng
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China.,Shanghai Laboratory Animal Research Center, Shanghai 201203, China
| | - Jiawen Zhu
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China
| | - Haitao Chang
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China
| | - Xiaoping Gao
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China
| | - Cheng Gao
- Shanghai Laboratory Animal Research Center, Shanghai 201203, China
| | - Xiaofeng Wei
- Shanghai Laboratory Animal Research Center, Shanghai 201203, China
| | - Fangyan Yuan
- Hubei key laboratory of Animal Embryo and Molecular Breeding, Institute of Animal Husbandry and Veterinary Sciences, Hubei Academy of Agricultural Sciences, Wuhan 430070, China
| | - Weicheng Bei
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China.,Cooperative Innovation Center of Sustainable Pig Production, Wuhan 430070, China
| |
Collapse
|
44
|
Zhang Y, Ding D, Liu M, Yang X, Zong B, Wang X, Chen H, Bei W, Tan C. Effect of the glycosyltransferases on the capsular polysaccharide synthesis of Streptococcus suis serotype 2. Microbiol Res 2016; 185:45-54. [PMID: 26946377 DOI: 10.1016/j.micres.2016.02.002] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2015] [Revised: 01/04/2016] [Accepted: 02/03/2016] [Indexed: 02/03/2023]
Abstract
Streptococcus suis serotype 2 (S. suis 2) is a serious zoonotic pathogen causing septicemia and meningitis in piglets and humans. The capsular polysaccharide (CPS) is an essential virulence factor for S. suis 2 to infect the host. The synthesis of CPS repeating units involves multiple glycosyltransferases. In this study, four genes (cps2E, cps2G, cps2J and cps2L) encoding different glycosyltransferases involved in CPS synthesis were researched in S. suis 2. Four deletion mutants (Δcps2E, Δcps2G, Δcps2J and Δcps2L) with their CPS incomplete and their sialic acid content significantly decreased were constructed in S. suis 2 SC19. All these four mutant strains showed enhanced adhesion to Hep-2 cells and increased sensitivity to phagocytosis. Flow cytometric analysis also revealed that these four mutants were more susceptible to the attack by the complement system. In a mouse model of infection, the mutant strains were rapidly cleared by the immune system, compared with the wild-type strain. In summary, this study characterized four genes (cps2E, cps2G, cps2J and cps2L) involved in CPS synthesis of S. suis 2 SC19 and it revealed that these genes were all crucial for SC19 to invade and survive in the host.
Collapse
Affiliation(s)
- Yanyan Zhang
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, Hubei, China; The Cooperative Innovation Center for Sustainable Pig Production, Huazhong Agricultural University, Wuhan 430070, Hubei, China; Key Laboratory of Development of Veterinary Diagnostic Products of Ministry of Agriculture, Huazhong Agricultural University, Wuhan 430070, Hubei, China
| | - Dandan Ding
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, Hubei, China; The Cooperative Innovation Center for Sustainable Pig Production, Huazhong Agricultural University, Wuhan 430070, Hubei, China; Key Laboratory of Development of Veterinary Diagnostic Products of Ministry of Agriculture, Huazhong Agricultural University, Wuhan 430070, Hubei, China
| | - Manli Liu
- Center of Bio-Pesticide Engineering Research, Hubei Academy of Agricultural Science, Wuhan 430064, Hubei, China
| | - Xiaopei Yang
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, Hubei, China; The Cooperative Innovation Center for Sustainable Pig Production, Huazhong Agricultural University, Wuhan 430070, Hubei, China; Key Laboratory of Development of Veterinary Diagnostic Products of Ministry of Agriculture, Huazhong Agricultural University, Wuhan 430070, Hubei, China
| | - Bingbing Zong
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, Hubei, China; The Cooperative Innovation Center for Sustainable Pig Production, Huazhong Agricultural University, Wuhan 430070, Hubei, China; Key Laboratory of Development of Veterinary Diagnostic Products of Ministry of Agriculture, Huazhong Agricultural University, Wuhan 430070, Hubei, China
| | - Xiangru Wang
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, Hubei, China; The Cooperative Innovation Center for Sustainable Pig Production, Huazhong Agricultural University, Wuhan 430070, Hubei, China; Key Laboratory of Development of Veterinary Diagnostic Products of Ministry of Agriculture, Huazhong Agricultural University, Wuhan 430070, Hubei, China
| | - Huanchun Chen
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, Hubei, China; The Cooperative Innovation Center for Sustainable Pig Production, Huazhong Agricultural University, Wuhan 430070, Hubei, China; Key Laboratory of Development of Veterinary Diagnostic Products of Ministry of Agriculture, Huazhong Agricultural University, Wuhan 430070, Hubei, China
| | - Weicheng Bei
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, Hubei, China; The Cooperative Innovation Center for Sustainable Pig Production, Huazhong Agricultural University, Wuhan 430070, Hubei, China; Key Laboratory of Development of Veterinary Diagnostic Products of Ministry of Agriculture, Huazhong Agricultural University, Wuhan 430070, Hubei, China
| | - Chen Tan
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, Hubei, China; The Cooperative Innovation Center for Sustainable Pig Production, Huazhong Agricultural University, Wuhan 430070, Hubei, China; Key Laboratory of Development of Veterinary Diagnostic Products of Ministry of Agriculture, Huazhong Agricultural University, Wuhan 430070, Hubei, China.
| |
Collapse
|
45
|
Zhang H, Ravcheev DA, Hu D, Zhang F, Gong X, Hao L, Cao M, Rodionov DA, Wang C, Feng Y. Two novel regulators of N-acetyl-galactosamine utilization pathway and distinct roles in bacterial infections. Microbiologyopen 2015; 4:983-1000. [PMID: 26540018 PMCID: PMC4694137 DOI: 10.1002/mbo3.307] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2015] [Revised: 09/21/2015] [Accepted: 09/28/2015] [Indexed: 12/19/2022] Open
Abstract
Bacterial pathogens can exploit metabolic pathways to facilitate their successful infection cycles, but little is known about roles of d‐galactosamine (GalN)/N‐acetyl‐d‐galactosamine (GalNAc) catabolism pathway in bacterial pathogenesis. Here, we report the genomic reconstruction of GalN/GalNAc utilization pathway in Streptococci and the diversified aga regulons. We delineated two new paralogous AgaR regulators for the GalN/GalNAc catabolism pathway. The electrophoretic mobility shift assays experiment demonstrated that AgaR2 (AgaR1) binds the predicted palindromes, and the combined in vivo data from reverse transcription quantitative polymerase chain reaction and RNA‐seq suggested that AgaR2 (not AgaR1) can effectively repress the transcription of the target genes. Removal of agaR2 (not agaR1) from Streptococcus suis 05ZYH33 augments significantly the abilities of both adherence to Hep‐2 cells and anti‐phagocytosis against RAW264.7 macrophage. As anticipated, the dysfunction in AgaR2‐mediated regulation of S. suis impairs its pathogenicity in experimental models of both mice and piglets. Our finding discovered two novel regulators specific for GalN/GalNAc catabolism and assigned them distinct roles into bacterial infections. To the best of our knowledge, it might represent a first paradigm that links the GalN/GalNAc catabolism pathway to bacterial pathogenesis.
Collapse
Affiliation(s)
- Huimin Zhang
- Department of Medical Microbiology and Parasitology, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310058, China
| | - Dmitry A Ravcheev
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Esch-sur-Alzette, L-4360, Luxembourg
| | - Dan Hu
- Department of Epidemiology, Research Institute for Medicine of Nanjing Command, Nanjing, 210002, China
| | - Fengyu Zhang
- Department of Epidemiology, Research Institute for Medicine of Nanjing Command, Nanjing, 210002, China
| | - Xiufang Gong
- Department of Epidemiology, Research Institute for Medicine of Nanjing Command, Nanjing, 210002, China
| | - Lina Hao
- Department of Epidemiology, Research Institute for Medicine of Nanjing Command, Nanjing, 210002, China
| | - Min Cao
- Department of Epidemiology, Research Institute for Medicine of Nanjing Command, Nanjing, 210002, China
| | - Dmitry A Rodionov
- A.A. Kharkevich Institute for Information Transmission Problems, Russian Academy of Sciences, Moscow, 127994, Russia
| | - Changjun Wang
- Department of Epidemiology, Research Institute for Medicine of Nanjing Command, Nanjing, 210002, China
| | - Youjun Feng
- Department of Medical Microbiology and Parasitology, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310058, China
| |
Collapse
|
46
|
Schieck E, Lartigue C, Frey J, Vozza N, Hegermann J, Miller RA, Valguarnera E, Muriuki C, Meens J, Nene V, Naessens J, Weber J, Lowary TL, Vashee S, Feldman MF, Jores J. Galactofuranose in Mycoplasma mycoides is important for membrane integrity and conceals adhesins but does not contribute to serum resistance. Mol Microbiol 2015; 99:55-70. [PMID: 26354009 DOI: 10.1111/mmi.13213] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/07/2015] [Indexed: 12/20/2022]
Abstract
Mycoplasma mycoides subsp. capri (Mmc) and subsp. mycoides (Mmm) are important ruminant pathogens worldwide causing diseases such as pleuropneumonia, mastitis and septicaemia. They express galactofuranose residues on their surface, but their role in pathogenesis has not yet been determined. The M. mycoides genomes contain up to several copies of the glf gene, which encodes an enzyme catalysing the last step in the synthesis of galactofuranose. We generated a deletion of the glf gene in a strain of Mmc using genome transplantation and tandem repeat endonuclease coupled cleavage (TREC) with yeast as an intermediary host for the genome editing. As expected, the resulting YCp1.1-Δglf strain did not produce the galactofuranose-containing glycans as shown by immunoblots and immuno-electronmicroscopy employing a galactofuranose specific monoclonal antibody. The mutant lacking galactofuranose exhibited a decreased growth rate and a significantly enhanced adhesion to small ruminant cells. The mutant was also 'leaking' as revealed by a β-galactosidase-based assay employing a membrane impermeable substrate. These findings indicate that galactofuranose-containing polysaccharides conceal adhesins and are important for membrane integrity. Unexpectedly, the mutant strain showed increased serum resistance.
Collapse
Affiliation(s)
- Elise Schieck
- International Livestock Research Institute, Old Naivasha Road, P.O. Box 30709, 00100, Nairobi, Kenya
| | - Carole Lartigue
- UMR 1332 Biologie du Fruit et Pathologie, The French National Institute for Agricultural Research, INRA-Université Bordeaux, Segalen, 71, avenue Edouard Bourlaux, CS20032, F-33882, Villenave D'Ornon CEDEX, Bordeaux, France.,UMR 1332 de Biologie du Fruit et Pathologie, Université Bordeaux, F-33140, Villenave d'Ornon, Bordeaux, France
| | - Joachim Frey
- Institute of Veterinary Bacteriology, University of Bern, CH-3001, Bern, Switzerland
| | - Nicolas Vozza
- Department of Biological Sciences, University of Alberta, Edmonton, Alberta, T6G 2E9, Canada
| | - Jan Hegermann
- Institute of Functional and Applied Anatomy, Hannover Medical School, Hannover, Germany.,Research in Endstage and Obstructive Lung Disease Hannover (BREATH), Member of the German Center for Lung Research (DZL), Hannover, Germany.,REBIRTH Cluster of Excellence, Hannover, Germany
| | - Rachel A Miller
- Department of Food Science, Cornell University, Ithaca, NY, USA
| | - Ezequiel Valguarnera
- Department of Molecular Microbiology, Washington University School of Medicine St Louis, 660 South Euclid Avenue, St Louis, MO 63110, USA
| | - Cecilia Muriuki
- International Livestock Research Institute, Old Naivasha Road, P.O. Box 30709, 00100, Nairobi, Kenya
| | - Jochen Meens
- Institute for Microbiology, Department of Infectious Diseases, University of Veterinary Medicine Hannover, Hannover, Germany
| | - Vish Nene
- International Livestock Research Institute, Old Naivasha Road, P.O. Box 30709, 00100, Nairobi, Kenya
| | - Jan Naessens
- International Livestock Research Institute, Old Naivasha Road, P.O. Box 30709, 00100, Nairobi, Kenya
| | - Johann Weber
- Center for Integrative Genomics, Lausanne Genomic Technologies Facility,University of Lausanne, Lausanne, Switzerland
| | - Todd L Lowary
- Department of Chemistry, Alberta Glycomics Centre, University of Alberta, Edmonton, Alberta, T6G 2E9, Canada
| | - Sanjay Vashee
- J. Craig Venter Institute, 9704 Medical Center Drive, MD 20850, Rockville, USA
| | - Mario F Feldman
- Department of Biological Sciences, University of Alberta, Edmonton, Alberta, T6G 2E9, Canada.,Department of Molecular Microbiology, Washington University School of Medicine St Louis, 660 South Euclid Avenue, St Louis, MO 63110, USA
| | - Joerg Jores
- International Livestock Research Institute, Old Naivasha Road, P.O. Box 30709, 00100, Nairobi, Kenya.,Institute of Veterinary Bacteriology, University of Bern, CH-3001, Bern, Switzerland
| |
Collapse
|
47
|
Abstract
Acquisition and metabolism of carbohydrates are essential for host colonization and pathogenesis of bacterial pathogens. Different bacteria can uptake different lines of carbohydrates via ABC transporters, in which ATPase subunits energize the transport though ATP hydrolysis. Some ABC transporters possess their own ATPases, while some share a common ATPase. Here we identified MsmK, an ATPase from Streptococcus suis, an emerging zoonotic bacterium causing dead infections in pigs and humans. Genetic and biochemistry studies revealed that the MsmK was responsible for the utilization of raffinose, melibiose, maltotetraose, glycogen and maltotriose. In infected mice, the msmK-deletion mutant showed significant defects of survival and colonization when compared with its parental and complementary strains. Taken together, MsmK is an ATPase that contributes to multiple carbohydrates utilization and host colonization of S. suis. This study gives new insight into our understanding of the carbohydrates utilization and its relationship to the pathogenesis of this zoonotic pathogen.
Collapse
|
48
|
Host-like carbohydrates promote bloodstream survival of Vibrio vulnificus in vivo. Infect Immun 2015; 83:3126-36. [PMID: 26015477 PMCID: PMC4496609 DOI: 10.1128/iai.00345-15] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2015] [Accepted: 05/13/2015] [Indexed: 01/15/2023] Open
Abstract
Sialic acids are found on all vertebrate cell surfaces and are part of a larger class of molecules known as nonulosonic acids. Many bacterial pathogens synthesize related nine-carbon backbone sugars; however, the role(s) of these non-sialic acid molecules in host-pathogen interactions is poorly understood. Vibrio vulnificus is the leading cause of seafood-related death in the United States due to its ability to quickly access the host bloodstream, which it can accomplish through gastrointestinal or wound infection. However, little is known about how this organism persists systemically. Here we demonstrate that sialic acid-like molecules are present on the lipopolysaccharide of V. vulnificus, are required for full motility and biofilm formation, and also contribute to the organism's natural resistance to polymyxin B. Further experiments in a murine model of intravenous V. vulnificus infection demonstrated that expression of nonulosonic acids had a striking benefit for bacterial survival during bloodstream infection and dissemination to other tissues in vivo. In fact, levels of bacterial persistence in the blood corresponded to the overall levels of these molecules expressed by V. vulnificus isolates. Taken together, these results suggest that molecules similar to sialic acids evolved to facilitate the aquatic lifestyle of V. vulnificus but that their emergence also resulted in a gain of function with life-threatening potential in the human host.
Collapse
|
49
|
Roy D, Auger JP, Segura M, Fittipaldi N, Takamatsu D, Okura M, Gottschalk M. Role of the capsular polysaccharide as a virulence factor for Streptococcus suis serotype 14. CANADIAN JOURNAL OF VETERINARY RESEARCH = REVUE CANADIENNE DE RECHERCHE VETERINAIRE 2015; 79:141-146. [PMID: 25852230 PMCID: PMC4365706] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Subscribe] [Scholar Register] [Received: 07/11/2014] [Accepted: 08/10/2014] [Indexed: 06/04/2023]
Abstract
Streptococcus suis is an important swine pathogen and a zoonotic agent causing meningitis and septicemia. Although serotype 2 is the most virulent type, serotype 14 is emerging, and understanding of its pathogenesis is limited. To study the role of the capsular polysaccharide (CPS) of serotype 14 as a virulence factor, we constructed knockout mutants devoid of either cps14B, a highly conserved regulatory gene, or neu14C, a gene coding for uridine diphospho-N-acetylglucosamine 2-epimerase, which is involved in sialic acid synthesis. The mutants showed total loss of the CPS with coagglutination assays and electron microscopy. Phagocytosis assays showed high susceptibility of mutant Δcps14B. An in vivo murine model was used to demonstrate attenuated virulence of this non-encapsulated mutant. Despite the difference in the CPS composition of different serotypes, this study has demonstrated for the first time that the CPS of a serotype other than 2 is also an important antiphagocytic factor and a critical virulence factor.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Marcelo Gottschalk
- Address all correspondence to Dr. Marcelo Gottschalk; telephone: (450) 773-8521, ext. 8374; fax: (450) 778-8108; e-mail:
| |
Collapse
|
50
|
Wu Z, Wu C, Shao J, Zhu Z, Wang W, Zhang W, Tang M, Pei N, Fan H, Li J, Yao H, Gu H, Xu X, Lu C. The Streptococcus suis transcriptional landscape reveals adaptation mechanisms in pig blood and cerebrospinal fluid. RNA (NEW YORK, N.Y.) 2014; 20:882-898. [PMID: 24759092 PMCID: PMC4024642 DOI: 10.1261/rna.041822.113] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/04/2013] [Accepted: 03/11/2014] [Indexed: 06/03/2023]
Abstract
Streptococcus suis (SS) is an important pathogen of pigs, and it is also recognized as a zoonotic agent for humans. SS infection may result in septicemia or meningitis in the host. However, little is known about genes that contribute to the virulence process and survival within host blood or cerebrospinal fluid (CSF). Small RNAs (sRNA) have emerged as key regulators of virulence in several bacteria, but they have not been investigated in SS. Here, using a differential RNA-sequencing approach and RNAs from SS strain P1/7 grown in rich medium, pig blood, or CSF, we present the SS genome-wide map of 793 transcriptional start sites and 370 operons. In addition to identifying 29 sRNAs, we show that five sRNA deletion mutants attenuate SS virulence in a zebrafish infection model. Homology searches revealed that 10 sRNAs were predicted to be present in other pathogenic Streptococcus species. Compared with wild-type strain P1/7, sRNAs rss03, rss05, and rss06 deletion mutants were significantly more sensitive to killing by pig blood. It is possible that rss06 contributes to SS virulence by indirectly activating expression of SSU0308, a virulence gene encoding a zinc-binding lipoprotein. In blood, genes involved in the synthesis of capsular polysaccharide (CPS) and subversion of host defenses were up-regulated. In contrast, in CSF, genes for CPS synthesis were down-regulated. Our study is the first analysis of SS sRNAs involved in virulence and has both improved our understanding of SS pathogenesis and increased the number of sRNAs known to play definitive roles in bacterial virulence.
Collapse
Affiliation(s)
- Zongfu Wu
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
- Key Lab of Animal Bacteriology, Ministry of Agriculture, Nanjing 210095, China
- OIE Reference Laboratory for Swine Streptococcosis, Nanjing 210095, China
| | | | - Jing Shao
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
- Key Lab of Animal Bacteriology, Ministry of Agriculture, Nanjing 210095, China
- OIE Reference Laboratory for Swine Streptococcosis, Nanjing 210095, China
| | | | - Weixue Wang
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
- Key Lab of Animal Bacteriology, Ministry of Agriculture, Nanjing 210095, China
- OIE Reference Laboratory for Swine Streptococcosis, Nanjing 210095, China
| | | | - Min Tang
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
- Key Lab of Animal Bacteriology, Ministry of Agriculture, Nanjing 210095, China
- OIE Reference Laboratory for Swine Streptococcosis, Nanjing 210095, China
| | - Na Pei
- BGI-Shenzhen, Shenzhen 518083, China
| | - Hongjie Fan
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
- Key Lab of Animal Bacteriology, Ministry of Agriculture, Nanjing 210095, China
- OIE Reference Laboratory for Swine Streptococcosis, Nanjing 210095, China
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou 225009, China
| | | | - Huochun Yao
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
- Key Lab of Animal Bacteriology, Ministry of Agriculture, Nanjing 210095, China
- OIE Reference Laboratory for Swine Streptococcosis, Nanjing 210095, China
| | - Hongwei Gu
- Jiangsu Engineering Research Center for microRNA Biology and Biotechnology, State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing 210093, China
| | - Xun Xu
- BGI-Shenzhen, Shenzhen 518083, China
| | - Chengping Lu
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
- Key Lab of Animal Bacteriology, Ministry of Agriculture, Nanjing 210095, China
- OIE Reference Laboratory for Swine Streptococcosis, Nanjing 210095, China
| |
Collapse
|