1
|
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
|
2
|
Boueroy P, Brizuela J, Roodsant TJ, Wongsurawat T, Jenjaroenpun P, Chopjitt P, Hatrongjit R, Phetburom N, Chareonsudjai S, Boonmars T, Schultsz C, Kerdsin A. Genomic analysis and virulence of human Streptococcus suis serotype 14. Eur J Clin Microbiol Infect Dis 2025; 44:639-651. [PMID: 39731619 DOI: 10.1007/s10096-024-05029-w] [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: 12/10/2024] [Accepted: 12/19/2024] [Indexed: 12/30/2024]
Abstract
PURPOSE Streptococcus suis serotype 14 is the second most prevalent serotype being highly prevalent in Southeast Asia. This study aimed to characterize genetic background, population structure, virulent genes, antimicrobial-resistant genes, and virulence of human S. suis serotype 14. METHODS Genomes of 11 S. suis serotype 14 were sequenced by short- and long-read sequencing platforms. The genomes were analyzed for genetic relationship, virulence-associated genes, and antimicrobial-resistant genes. Antimicrobial susceptibility was conducted and the virulence was tested based on cell assay. RESULTS All isolates belonged to clonal complex (CC) 1, with nine sequence type (ST) 105 isolates and each isolate of ST1 and ST237. They were susceptible to penicillin, whereas tetracycline and macrolide were resistance due to tetO and ermB. Genomic analysis revealed that the serotype 14-ST105 isolates were closely related to zoonotic serotype 14-ST105 isolates from Vietnam and the serotype 1-ST105 Thai strain. The serotype 14-ST1 isolate was closely related to pig-diseased serotype 1-ST1 isolates from UK and USA, whereas the serotype 14-ST237 isolate was related to serotype 1-ST237 strains recovered from healthy pig from Thailand. Of 150 virulence-associated genes, 13 were absent from the serotype 14 isolates, including atl1, atlAss, hhly3, nisK, nisR, pnuC, salK, salR, sp1, srtG, virB4, virD4, and zmp. The virulence of strain 32481, a representative S. suis serotype 14-ST105 isolate showed reduced adhesion and invasion of two epithelial cell lines (A549 and HeLa) when compared to the serotype 2-ST1 strain P1/7, whereas apoptosis was similar. CONCLUSION This study highlighted the pathogenic potential of virulent serotype 14-ST105 strains and the need for increased monitoring of S. suis serotypes other than for serotype 2.
Collapse
Affiliation(s)
- Parichart Boueroy
- Faculty of Public Health, Kasetsart University Chalermphrakiat Sakon Nakhon Province Campus, Sakon Nakhon, Thailand
| | - Jaime Brizuela
- Amsterdam UMC Location University of Amsterdam, Department of Global Health, Amsterdam Institute for Global Health and Development, Paasheuvelweg 25, Amsterdam, The Netherlands
- Amsterdam UMC location University of Amsterdam, Department of Medical Microbiology and Infection Prevention, Meibergdreef 9, Amsterdam, The Netherlands
| | - Thomas J Roodsant
- Amsterdam UMC Location University of Amsterdam, Department of Global Health, Amsterdam Institute for Global Health and Development, Paasheuvelweg 25, Amsterdam, The Netherlands
- Amsterdam UMC location University of Amsterdam, Department of Medical Microbiology and Infection Prevention, Meibergdreef 9, Amsterdam, The Netherlands
| | - Thidathip Wongsurawat
- Division of Bioinformatics and Data Management for Research, Department of Research and Development, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Piroon Jenjaroenpun
- Division of Bioinformatics and Data Management for Research, Department of Research and Development, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Peechanika Chopjitt
- Faculty of Public Health, Kasetsart University Chalermphrakiat Sakon Nakhon Province Campus, Sakon Nakhon, Thailand
| | - Rujirat Hatrongjit
- Department of General Sciences, Faculty of Science and Engineering, Kasetsart University Chalermphrakiat Sakon Nakhon Province Campus, Sakon Nakhon, Thailand
| | - Nattamol Phetburom
- Faculty of Public Health, Kasetsart University Chalermphrakiat Sakon Nakhon Province Campus, Sakon Nakhon, Thailand
| | - Sorujsiri Chareonsudjai
- Department of Microbiology, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
- Research and Diagnostic Center for Emerging Infectious Diseases (RCEID), Khon Kaen University, Khon Kaen, Thailand
| | - Thidarut Boonmars
- Cholangiocarcinoma Research Institute, Khon Kaen University, Khon Kaen, 40002, Thailand
- Department of Parasitology, Faculty of Medicine, Khon Kaen University, Khon Kaen, 40002, Thailand
| | - Constance Schultsz
- Amsterdam UMC Location University of Amsterdam, Department of Global Health, Amsterdam Institute for Global Health and Development, Paasheuvelweg 25, Amsterdam, The Netherlands
- Amsterdam UMC location University of Amsterdam, Department of Medical Microbiology and Infection Prevention, Meibergdreef 9, Amsterdam, The Netherlands
| | - Anusak Kerdsin
- Faculty of Public Health, Kasetsart University Chalermphrakiat Sakon Nakhon Province Campus, Sakon Nakhon, Thailand.
| |
Collapse
|
3
|
Zhao T, Pellegrini L, van der Hee B, Boekhorst J, Fernandes A, Brugman S, van Baarlen P, Wells JM. Choroid plexus organoids reveal mechanisms of Streptococcus suis translocation at the blood-cerebrospinal fluid barrier. Fluids Barriers CNS 2025; 22:14. [PMID: 39930492 PMCID: PMC11812244 DOI: 10.1186/s12987-025-00627-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2024] [Accepted: 01/27/2025] [Indexed: 02/13/2025] Open
Abstract
Streptococcus suis is a globally emerging zoonotic pathogen that can cause invasive disease commonly associated with meningitis in pigs and humans. To cause meningitis, S. suis must invade the central nervous system (CNS) by crossing the neurovascular unit, also known as the blood-brain barrier (BBB), or vascularized choroid plexus (ChP) epithelium known as the blood-cerebrospinal fluid barrier (BCSFB). Recently developed ChP organoids have been shown to accurately replicate the cytoarchitecture and physiological functions of the ChP epithelium in vivo. Here, we used human induced pluripotent stem cells (iPSC)-derived ChP organoids as an in vitro model to investigate S. suis interaction and infection at the BCSFB. Our study revealed that S. suis is capable of translocating across the epithelium of ChP organoids without causing significant cell death or compromising the barrier integrity. Plasminogen (Plg) binding to S. suis in the presence of tissue plasminogen activator (tPA), which converts immobilized Plg to plasmin (Pln), significantly increased the basolateral to apical translocation across ChP organoids into the CSF-like fluid in the lumen. S. suis was able to replicate at the same rate in CSF and laboratory S. suis culture medium but reached a lower final density. The analysis of transcriptomes in ChP organoids after S. suis infection indicated inflammatory responses, while the addition of Plg further suggested extracellular matrix (ECM) remodeling. To our knowledge, this is the first study using ChP organoids to investigate bacterial infection of the BCSFB. Our findings highlight the potential of ChP organoids as a valuable tool for studying the mechanisms of bacterial interaction and infection of the human ChP in vitro.
Collapse
Affiliation(s)
- Tiantong Zhao
- Host-Microbe Interactomics, Department Animal Science, Wageningen University & Research, De Elst 1, Wageningen, 6708 WD, The Netherlands
| | - Laura Pellegrini
- Centre for Developmental Neurobiology, King's College London, Guys Campus, New Hunt's House, London, UK
- MRC Laboratory of Molecular Biology, Cambridge Biomedical Campus, Cambridge, UK
| | - Bart van der Hee
- Host-Microbe Interactomics, Department Animal Science, Wageningen University & Research, De Elst 1, Wageningen, 6708 WD, The Netherlands
| | - Jos Boekhorst
- Host-Microbe Interactomics, Department Animal Science, Wageningen University & Research, De Elst 1, Wageningen, 6708 WD, The Netherlands
| | - Aline Fernandes
- Host-Microbe Interactomics, Department Animal Science, Wageningen University & Research, De Elst 1, Wageningen, 6708 WD, The Netherlands
| | - Sylvia Brugman
- Host-Microbe Interactomics, Department Animal Science, Wageningen University & Research, De Elst 1, Wageningen, 6708 WD, The Netherlands
| | - Peter van Baarlen
- Host-Microbe Interactomics, Department Animal Science, Wageningen University & Research, De Elst 1, Wageningen, 6708 WD, The Netherlands
| | - Jerry M Wells
- Host-Microbe Interactomics, Department Animal Science, Wageningen University & Research, De Elst 1, Wageningen, 6708 WD, The Netherlands.
| |
Collapse
|
4
|
Fu Y, Jie J, Lei L, Liu M, Wang J, Lei L, Liu H. Exploring the destructive synergy between IL-33 and Suilysin hemolysis on blood-brain barrier stability. Microbiol Spectr 2024; 12:e0061224. [PMID: 38980021 PMCID: PMC11302228 DOI: 10.1128/spectrum.00612-24] [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: 03/09/2024] [Accepted: 06/09/2024] [Indexed: 07/10/2024] Open
Abstract
Streptococcus suis type 2 (SS2) is a zoonotic pathogen capable of eliciting meningitis, presenting significant challenges to both the swine industry and public health. Suilysin (Sly), one of SS2 most potent virulence determinants, releases a surfeit of inflammatory agents following red blood cell lysis. Notably, while current research on Sly role in SS2-induced meningitis predominantly centers on its interaction with the blood-brain barrier (BBB), the repercussions of Sly hemolytic products on BBB function have largely been sidestepped. In this vein, our study delves into the ramifications of Sly-induced hemolysis on BBB integrity. We discern that Sly hemolytic derivatives exacerbate the permeability of Sly-induced in vitro BBB models. Within these Sly hemolytic products, Interleukin-33 (IL-33) disrupts the expression and distribution of Claudin-5 in brain microvascular endothelial cells, facilitating the release of Interleukin-6 (IL-6) and Interleukin-8 (IL-8), thereby amplifying BBB permeability. Preliminary mechanistic insights suggest that IL-33-driven expression of IL-6 and IL-8 is orchestrated by the p38-mitogen-activated protein kinase signaling, whereas matrix metalloproteinase 9 mediates IL-33-induced suppression of Claudin-5. To validate these in vitro findings, an SS2-infected mouse model was established, and upon intravenous administration of growth stimulation expressed gene 2 (ST2) antibodies, in vivo results further underscored the pivotal role of the IL-33/ST2 axis during SS2 cerebral invasion. In summation, this study pioneerly illuminates the involvement of Sly hemolytic products in SS2-mediated BBB compromise and spotlights the instrumental role and primary mechanism of IL-33 therein. These insights enrich our comprehension of SS2 meningitis pathogenesis, laying pivotal groundwork for therapeutic advancements against SS2-induced meningitis.IMPORTANCEThe treatment of meningitis caused by Streptococcus suis type 2 (SS2) has always been a clinical challenge. Elucidating the molecular mechanisms by which SS2 breaches the blood-brain barrier (BBB) is crucial for the development of meningitis therapeutics. Suilysin (Sly) is one of the most important virulence factors of SS2, which can quickly lyse red blood cells and release large amounts of damage-associated molecular patterns, such as hemoglobin, IL-33, cyclophilin A, and so on. However, the impact of these hemolytic products on the function of BBB is unknown and ignored. This study is the first to investigate the effect of Sly hemolytic products on BBB function. The data are crucial for the study of the pathogenesis of SS2 meningitis and can provide an important reference for the development of meningitis therapeutics.
Collapse
Affiliation(s)
- Yang Fu
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, College of Veterinary Medicine; Department of Respiratory Medicine, Center for Pathogen Biology and Infectious Diseases, Key Laboratory of Organ Regeneration and Transplantation of the Ministry of Education, The First Hospital of Jilin University, Jilin University, Changchun, China
- Key Laboratory of the Provincial Education, Department of Heilongjiang for Common Animal Disease Prevention and Treatment, College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| | - Jing Jie
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, College of Veterinary Medicine; Department of Respiratory Medicine, Center for Pathogen Biology and Infectious Diseases, Key Laboratory of Organ Regeneration and Transplantation of the Ministry of Education, The First Hospital of Jilin University, Jilin University, Changchun, China
| | - Liang Lei
- Department of Orthopaedics, Heilongjiang Provincial Hospital, Harbin, Heilongjiang, China
| | - Mengmeng Liu
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, College of Veterinary Medicine; Department of Respiratory Medicine, Center for Pathogen Biology and Infectious Diseases, Key Laboratory of Organ Regeneration and Transplantation of the Ministry of Education, The First Hospital of Jilin University, Jilin University, Changchun, China
| | - Junjie Wang
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, College of Veterinary Medicine; Department of Respiratory Medicine, Center for Pathogen Biology and Infectious Diseases, Key Laboratory of Organ Regeneration and Transplantation of the Ministry of Education, The First Hospital of Jilin University, 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, College of Veterinary Medicine; Department of Respiratory Medicine, Center for Pathogen Biology and Infectious Diseases, Key Laboratory of Organ Regeneration and Transplantation of the Ministry of Education, The First Hospital of Jilin University, Jilin University, Changchun, China
| | - Hongtao Liu
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, College of Veterinary Medicine; Department of Respiratory Medicine, Center for Pathogen Biology and Infectious Diseases, Key Laboratory of Organ Regeneration and Transplantation of the Ministry of Education, The First Hospital of Jilin University, Jilin University, Changchun, China
| |
Collapse
|
5
|
Payen S, Roy D, Okura M, Segura M, Gottschalk M. Study of the Role of Lipoprotein Maturation Enzymes in the Pathogenesis of the Infection Caused by the Streptococcus suis Serotype 2 Sequence Type 25 North American Prototype Strain. Pathogens 2023; 12:1325. [PMID: 38003790 PMCID: PMC10675726 DOI: 10.3390/pathogens12111325] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Revised: 11/01/2023] [Accepted: 11/02/2023] [Indexed: 11/26/2023] Open
Abstract
Streptococcus suis serotype 2 is an important swine bacterial pathogen causing sudden death, septic shock, and meningitis. However, serotype 2 strains are phenotypically and genotypically heterogeneous and composed of a multitude of sequence types (STs) whose distributions greatly vary worldwide. It has been previously shown that the lipoprotein (LPP) maturation enzymes diacylglyceryl transferase (Lgt) and signal peptidase (Lsp) significantly modulate the inflammatory host response and play a differential role in virulence depending on the genetic background of the strain. Differently from Eurasian ST1/ST7 strains, the capsular polysaccharide of a North American S. suis serotype 2 ST25 representative strain only partially masks sub-capsular domains and bacterial wall components. Thus, our hypothesis is that since LPPs would be more surface exposed in ST25 strains than in their ST1 or ST7 counterparts, the maturation enzymes would play a more important role in the pathogenesis of the infection caused by the North American strain. Using isogenic Δlgt and Δlsp mutants derived from the wild-type ST25 strain, our studies suggest that these enzymes do not seem to play a role in the interaction between S. suis and epithelial and endothelial cells, regardless of the genetics background of the strain used. However, a role in the formation of biofilms (also independently of the STs) has been demonstrated. Moreover, the involvement of LPP dendritic cell activation in vitro seems to be somehow more pronounced with the ST25 strain. Finally, the Lgt enzyme seems to play a more important role in the virulence of the ST25 strain. Although some differences between STs could be observed, our original hypothesis that LPPs would be significantly more important in ST25 strains due to a better bacterial surface exposition could not be confirmed.
Collapse
Affiliation(s)
- Servane Payen
- Groupe de Recherche sur les Maladies Infectieuses en Production Animale (GREMIP) and Swine and Poultry Infectious Diseases Research Center (CRIPA), Department of Pathology and Microbiology, Faculty of Veterinary Medicine, University of Montreal, Saint-Hyacinthe, QC J2S 2M2, Canada; (S.P.); (M.S.)
| | - David Roy
- Centre de Recherche du Centre Hospitalier Universitaire de Sherbrooke, Sherbrooke, QC J1H 5N4, Canada;
| | - Masatoshi Okura
- Division of Transboundary Animal Disease Research, National Institute of Animal Health, National Agriculture and Food Research Organization, Kagoshima 891-0105, Japan;
| | - Mariela Segura
- Groupe de Recherche sur les Maladies Infectieuses en Production Animale (GREMIP) and Swine and Poultry Infectious Diseases Research Center (CRIPA), Department of Pathology and Microbiology, Faculty of Veterinary Medicine, University of Montreal, Saint-Hyacinthe, QC J2S 2M2, Canada; (S.P.); (M.S.)
| | - Marcelo Gottschalk
- Groupe de Recherche sur les Maladies Infectieuses en Production Animale (GREMIP) and Swine and Poultry Infectious Diseases Research Center (CRIPA), Department of Pathology and Microbiology, Faculty of Veterinary Medicine, University of Montreal, Saint-Hyacinthe, QC J2S 2M2, Canada; (S.P.); (M.S.)
| |
Collapse
|
6
|
Pramitasuri TI, Susilawathi NM, Tarini NMA, Sudewi AAR, Evans MC. Cholesterol dependent cytolysins and the brain: Revealing a potential therapeutic avenue for bacterial meningitis. AIMS Microbiol 2023; 9:647-667. [PMID: 38173970 PMCID: PMC10758573 DOI: 10.3934/microbiol.2023033] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Revised: 07/24/2023] [Accepted: 08/14/2023] [Indexed: 01/05/2024] Open
Abstract
Bacterial meningitis is a catastrophic nervous system disorder with high mortality and wide range of morbidities. Some of the meningitis-causing bacteria occupy cholesterol dependent cytolysins (CDCs) to increase their pathogenicity and arrange immune-evasion strategy. Studies have observed that the relationship between CDCs and pathogenicity in these meningitides is complex and involves interactions between CDC, blood-brain barrier (BBB), glial cells and neurons. In BBB, these CDCs acts on capillary endothelium, tight junction (TJ) proteins and neurovascular unit (NVU). CDCs also observed to elicit intriguing effects on brain inflammation which involves microglia and astrocyte activations, along with neuronal damage as the end-point of pathological pathways in bacterial meningitis. As some studies mentioned potential advantage of CDC-targeted therapeutic mechanisms to combat CNS infections, it might be a fruitful avenue to deepen our understanding of CDC as a candidate for adjuvant therapy to combat bacterial meningitis.
Collapse
Affiliation(s)
- Tjokorda Istri Pramitasuri
- Doctoral Program in Medical Science, Faculty of Medicine, Universitas Udayana, Bali, Indonesia
- Postgraduate Research Student, Faculty of Medicine, Imperial College London, United Kingdom
| | - Ni Made Susilawathi
- Department of Neurology, Faculty of Medicine, Universitas Udayana, Bali, Indonesia
| | - Ni Made Adi Tarini
- Department of Microbiology, Faculty of Medicine, Universitas Udayana-Rumah Sakit Umum Pusat Prof Dr dr IGNG Ngoerah, Bali, Indonesia
| | - AA Raka Sudewi
- Department of Neurology, Faculty of Medicine, Universitas Udayana, Bali, Indonesia
| | - Matthew C Evans
- Pain Research, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, United Kingdom
- Department of Brain Sciences, Care Research and Technology Centre, UK Dementia Research Institute, London, United Kingdom
| |
Collapse
|
7
|
Yang R, Wang J, Wang F, Zhang H, Tan C, Chen H, Wang X. Blood-Brain Barrier Integrity Damage in Bacterial Meningitis: The Underlying Link, Mechanisms, and Therapeutic Targets. Int J Mol Sci 2023; 24:ijms24032852. [PMID: 36769171 PMCID: PMC9918147 DOI: 10.3390/ijms24032852] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Revised: 01/26/2023] [Accepted: 01/30/2023] [Indexed: 02/05/2023] Open
Abstract
Despite advances in supportive care and antimicrobial treatment, bacterial meningitis remains the most serious infection of the central nervous system (CNS) that poses a serious risk to life. This clinical dilemma is largely due to our insufficient knowledge of the pathology behind this disease. By controlling the entry of molecules into the CNS microenvironment, the blood-brain barrier (BBB), a highly selective cellular monolayer that is specific to the CNS's microvasculature, regulates communication between the CNS and the rest of the body. A defining feature of the pathogenesis of bacterial meningitis is the increase in BBB permeability. So far, several contributing factors for BBB disruption have been reported, including direct cellular damage brought on by bacterial virulence factors, as well as host-specific proteins or inflammatory pathways being activated. Recent studies have demonstrated that targeting pathological factors contributing to enhanced BBB permeability is an effective therapeutic complement to antimicrobial therapy for treating bacterial meningitis. Hence, understanding how these meningitis-causing pathogens affect the BBB permeability will provide novel perspectives for investigating bacterial meningitis's pathogenesis, prevention, and therapies. Here, we summarized the recent research progress on meningitis-causing pathogens disrupting the barrier function of BBB. This review provides handy information on BBB disruption by meningitis-causing pathogens, and helps design future research as well as develop potential combination therapies.
Collapse
Affiliation(s)
- Ruicheng Yang
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China
- Key Laboratory of Preventive Veterinary Medicine in Hubei Province, The Cooperative Innovation Center for Sustainable Pig Production, Wuhan 430070, China
| | - Jundan Wang
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China
- Key Laboratory of Preventive Veterinary Medicine in Hubei Province, The Cooperative Innovation Center for Sustainable Pig Production, Wuhan 430070, China
| | - Fen Wang
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China
- Key Laboratory of Preventive Veterinary Medicine in Hubei Province, The Cooperative Innovation Center for Sustainable Pig Production, Wuhan 430070, China
| | - Huipeng Zhang
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China
- Key Laboratory of Preventive Veterinary Medicine in Hubei Province, The Cooperative Innovation Center for Sustainable Pig Production, Wuhan 430070, China
| | - Chen Tan
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China
- Key Laboratory of Preventive Veterinary Medicine in Hubei Province, The Cooperative Innovation Center for Sustainable Pig Production, Wuhan 430070, China
- Key Laboratory of Development of Veterinary Diagnostic Products, Ministry of Agriculture of the People’s Republic of China, Wuhan 430070, China
- International Research Center for Animal Disease, Ministry of Science and Technology of the People’s Republic of China, Wuhan 430070, China
| | - Huanchun Chen
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China
- Key Laboratory of Preventive Veterinary Medicine in Hubei Province, The Cooperative Innovation Center for Sustainable Pig Production, Wuhan 430070, China
- Key Laboratory of Development of Veterinary Diagnostic Products, Ministry of Agriculture of the People’s Republic of China, Wuhan 430070, China
- International Research Center for Animal Disease, Ministry of Science and Technology of the People’s Republic of China, Wuhan 430070, China
| | - Xiangru Wang
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China
- Key Laboratory of Preventive Veterinary Medicine in Hubei Province, The Cooperative Innovation Center for Sustainable Pig Production, Wuhan 430070, China
- Key Laboratory of Development of Veterinary Diagnostic Products, Ministry of Agriculture of the People’s Republic of China, Wuhan 430070, China
- International Research Center for Animal Disease, Ministry of Science and Technology of the People’s Republic of China, Wuhan 430070, China
- Correspondence:
| |
Collapse
|
8
|
Li S, Wang C, Tang YD, Qin L, Chen T, Wang S, Bai Y, Cai X, Wang S. Interaction between Porcine Alveolar Macrophage-Tang Cells and Streptococcus suis Strains of Different Virulence: Phagocytosis and Apoptosis. Microorganisms 2023; 11:microorganisms11010160. [PMID: 36677452 PMCID: PMC9863715 DOI: 10.3390/microorganisms11010160] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2022] [Revised: 12/21/2022] [Accepted: 01/06/2023] [Indexed: 01/11/2023] Open
Abstract
Streptococcus suis is an important swine bacterial pathogen that activates macrophages to secrete inflammatory cytokines. Primary porcine alveolar macrophages (PAMs) are inconvenient to obtain, but it is unknown whether immortalized PAM-Tang cells can replace them as a better cell model for the study of the interaction between S. suis and macrophages. In this study, the phagocytic integrity, polarization, and pro-inflammatory cytokine secretion of PAM-Tang cells were confirmed by live-cell imaging, electron microscopy, confocal microscopy, and ELISA. Interestingly, the S. suis serotype 9 avirulent strain W7119 induced higher levels of adhesion and pro-inflammatory cytokines in PAM-Tang cells than the S. suis serotype 2 virulent strain 700794. Prolonged incubation with S. suis caused more cytotoxic cell damage, and the virulent strain induced higher levels of cytotoxicity to PAM-Tang cells. The virulent strain also induced higher levels of apoptosis in PAM-Tang cells, as shown by terminal deoxynucleotidyl transferase (TdT)-mediated dUTP-biotin nick end labeling (TUNEL) assay. In addition, it is the first report of virulent and avirulent S. suis inducing PAM-Tang polarization towards pro-inflammatory M1 macrophages and p53- and caspase-dependent apoptosis in PAMs. Taken together, this study contributes to a better understand of interactions between macrophages and S. suis isolates of different virulence, and confirms that PAM-Tang cells provide a long-term, renewable resource for investigating macrophage infections with bacteria.
Collapse
Affiliation(s)
- Siqi Li
- National Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin 150001, China
| | - Chunsheng Wang
- College of Life Science, Northeast Forestry University, Harbin 150040, China
| | - Yan-Dong Tang
- National Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin 150001, China
| | - Lei Qin
- National Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin 150001, China
| | - Tianfeng Chen
- National Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin 150001, China
| | - Shanghui Wang
- National Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin 150001, China
| | - Yuanzhe Bai
- National Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin 150001, China
| | - Xuehui Cai
- National Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin 150001, China
- Correspondence: (X.C.); (S.W.)
| | - Shujie Wang
- National Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin 150001, China
- Heilongjiang Provincial Key Laboratory of Veterinary Immunology, Harbin 150069, China
- Correspondence: (X.C.); (S.W.)
| |
Collapse
|
9
|
Payen S, Rrodriguez JA, Segura M, Gottschalk M. Laminin-binding protein of Streptococcus suis serotype 2 influences zinc acquisition and cytokine responses. Vet Res 2023; 54:1. [PMID: 36604750 PMCID: PMC9817373 DOI: 10.1186/s13567-022-01128-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Accepted: 11/08/2022] [Indexed: 01/07/2023] Open
Abstract
Streptococcus suis serotype 2 is an important bacterial pathogen of swine, responsible for substantial economic losses to the swine industry worldwide. The knowledge on the pathogenesis of the infection caused by S. suis is still poorly known. It has been previously described that S. suis possesses at least one lipoprotein with double laminin and zinc (Zn)-binding properties, which was described in the literature as either laminin-binding protein (Lmb, as in the current study), lipoprotein 103, CDS 0330 or AdcAII. In the present study, the role of the Lmb in the pathogenesis of the infection caused by S. suis serotype 2 was dissected. Using isogenic mutants, results showed that Lmb does not play an important role in the laminin-binding activity of S. suis, even when clearly exposed at the bacterial surface. In addition, the presence of this lipoprotein does not influence bacterial adhesion to and invasion of porcine respiratory epithelial and brain endothelial cells and it does not increase the susceptibility of S. suis to phagocytosis. On the other hand, the Lmb was shown to play an important role as cytokine activator when tested in vitro with dendritic cells. Finally, this lipoprotein plays a critical role in Zn acquisition from the host environment allowing bacteria to grow in vivo. The significant lower virulence of the Lmb defective mutant may be related to a combination of a lower bacterial survival due to the incapacity to acquire Zn from their surrounding milieu and a reduced cytokine activation.
Collapse
Affiliation(s)
- Servane Payen
- grid.14848.310000 0001 2292 3357Research 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, QC J2S 2M2 Canada
| | - Jesús Aranda Rrodriguez
- grid.7080.f0000 0001 2296 0625Department de Genètica I Microbiologia, Universitat Autónoma de Barcelona (UAB), Bellaterra (Cerdanyola del Vallès), 08193 Barcelona, Spain
| | - Mariela Segura
- grid.14848.310000 0001 2292 3357Research 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, QC J2S 2M2 Canada
| | - Marcelo Gottschalk
- grid.14848.310000 0001 2292 3357Research 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, QC J2S 2M2 Canada
| |
Collapse
|
10
|
Wu T, Jia L, Lei S, Jiang H, Liu J, Li N, Langford PR, Liu H, Lei L. Host HSPD1 Translocation from Mitochondria to the Cytoplasm Induced by Streptococcus suis Serovar 2 Enolase Mediates Apoptosis and Loss of Blood-Brain Barrier Integrity. Cells 2022; 11:2071. [PMID: 35805155 PMCID: PMC9265368 DOI: 10.3390/cells11132071] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Revised: 06/25/2022] [Accepted: 06/27/2022] [Indexed: 02/01/2023] Open
Abstract
Streptococcus suis serovar 2 (S. suis serovar 2) is a zoonotic pathogen that causes meningitis in pigs and humans, and is a serious threat to the swine industry and public health. Understanding the mechanism(s) by which S. suis serovar 2 penetrates the blood-brain barrier (BBB) is crucial to elucidating the pathogenesis of meningitis. In a previous study, we found that expression of the virulence factor enolase (Eno) by S. suis serovar 2 promotes the expression of host heat shock protein family D member 1 (HSPD1) in brain tissue, which leads to the apoptosis of porcine brain microvascular endothelial cells (PBMECs) and increased BBB permeability, which in turn promotes bacterial translocation across the BBB. However, the mechanism by which HSPD1 mediates Eno-induced apoptosis remains unclear. In this study, we demonstrate that Eno promotes the translocation of HSPD1 from mitochondria to the cytoplasm, where HSPD1 binds to β-actin (ACTB), the translocated HSPD1, and its interaction with ACTB led to adverse changes in cell morphology and promoted the expression of apoptosis-related proteins, second mitochondria-derived activator of caspases (Smac), and cleaved caspase-3; inhibited the expression of X-linked inhibitor of apoptosis protein (XIAP); and finally promoted cell apoptosis. These results further elucidate the role of HSPD1 in the process of Eno-induced apoptosis and increased BBB permeability, increasing our understanding of the pathogenic mechanisms of meningitis, and providing a framework for novel therapeutic strategies.
Collapse
Affiliation(s)
- Tong Wu
- State Key Laboratory for Zoonotic Diseases/Key Laboratory of Zoonosis, Ministry of Education, Institute of Zoonosis, College of Veterinary Medicine, Jilin University, Changchun 130062, China; (T.W.); (L.J.); (H.J.); (J.L.); (N.L.)
| | - Li Jia
- State Key Laboratory for Zoonotic Diseases/Key Laboratory of Zoonosis, Ministry of Education, Institute of Zoonosis, College of Veterinary Medicine, Jilin University, Changchun 130062, China; (T.W.); (L.J.); (H.J.); (J.L.); (N.L.)
| | - Siyu Lei
- School of Basic Medicine, Jilin University, Changchun 130021, China;
| | - Hexiang Jiang
- State Key Laboratory for Zoonotic Diseases/Key Laboratory of Zoonosis, Ministry of Education, Institute of Zoonosis, College of Veterinary Medicine, Jilin University, Changchun 130062, China; (T.W.); (L.J.); (H.J.); (J.L.); (N.L.)
| | - Jianan Liu
- State Key Laboratory for Zoonotic Diseases/Key Laboratory of Zoonosis, Ministry of Education, Institute of Zoonosis, College of Veterinary Medicine, Jilin University, Changchun 130062, China; (T.W.); (L.J.); (H.J.); (J.L.); (N.L.)
| | - Na Li
- State Key Laboratory for Zoonotic Diseases/Key Laboratory of Zoonosis, Ministry of Education, Institute of Zoonosis, College of Veterinary Medicine, Jilin University, Changchun 130062, China; (T.W.); (L.J.); (H.J.); (J.L.); (N.L.)
| | - Paul R. Langford
- Section of Paediatric Infectious Disease, Imperial College London, London W2 1NY, UK;
| | - Hongtao Liu
- State Key Laboratory for Zoonotic Diseases/Key Laboratory of Zoonosis, Ministry of Education, Institute of Zoonosis, College of Veterinary Medicine, Jilin University, Changchun 130062, China; (T.W.); (L.J.); (H.J.); (J.L.); (N.L.)
| | - Liancheng Lei
- State Key Laboratory for Zoonotic Diseases/Key Laboratory of Zoonosis, Ministry of Education, Institute of Zoonosis, College of Veterinary Medicine, Jilin University, Changchun 130062, China; (T.W.); (L.J.); (H.J.); (J.L.); (N.L.)
- Department of Veterinary Medicine, College of Animal Science, Yangtze University, Jingzhou 434023, China
| |
Collapse
|
11
|
Absence of Streptococcus pneumoniae Capsule Increases Bacterial Binding, Persistence, and Inflammation in Corneal Infection. Microorganisms 2022; 10:microorganisms10040710. [PMID: 35456761 PMCID: PMC9025271 DOI: 10.3390/microorganisms10040710] [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: 02/12/2022] [Revised: 03/07/2022] [Accepted: 03/22/2022] [Indexed: 11/17/2022] Open
Abstract
The role of the pneumococcal polysaccharide capsule is largely unclear for Streptococcus pneumoniae keratitis, an ocular inflammatory disease that develops as a result of bacterial infection of the cornea. In this study, capsule-deficient strains were compared to isogenic parent strains in their ability to adhere to human corneal epithelial cells. One isogenic pair was further used in topical ocular infection of mice to assess the contribution of the capsule to keratitis. The results showed that non-encapsulated pneumococci were significantly more adherent to cells, persisted in significantly higher numbers on mouse corneas in vivo, and caused significant increases in murine ocular IL9, IL10, IL12-p70, MIG, and MIP-1-gamma compared to encapsulated S. pneumoniae. These findings indicate that the bacterial capsule impedes virulence and the absence of capsule impacts inflammation following corneal infection.
Collapse
|
12
|
Hammerschmitt ME, Schwertz CI, Lopes BC, Pereira PR, Frandoloso R, Driemeier D. Clinical and pathological aspects of an outbreak of Streptococcus suis serotype 9 infection in pigs. PESQUISA VETERINÁRIA BRASILEIRA 2022. [DOI: 10.1590/1678-5150-pvb-7004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
ABSTRACT: Streptococcus suis is a Gram-positive pathogen that inhabits the upper respiratory tract and can cause severe systemic inflammatory disease in pigs, mainly during the nursery phase. Streptococcus suis is a reemergent pathogen, and outbreaks of its inducing disease represent significant economic losses for the pig industry worldwide. In this study, we described the clinical, pathological, and molecular aspects of an outbreak of S. suis infection with atypically high mortality. The outbreak occurred in nursery farms integrated into a cooperative in the state of Paraná, Brazil. Of the 30 nurseries, 10 were severely affected by the pathogen and had high economic losses. Clinical signs usually started approximately 10 days after weaning and were mainly characterized by acute nervous and locomotor disorders. The mortality of the affected batches usually ranged between 8% and 10%, but in some cases, it reached 18%. Nine piglets were submitted to post mortem examination. Macroscopically, the synovial joints were enlarged and contained fibrinous exudates. In the central nervous system, there was hyperemia of the leptomeningeal vessels associated with deposition of fibrin and purulent exudate in the leptomeninges. In three piglets, there was thickening of the choroid plexus associated with dilation of the lateral ventricles. Microscopic lesions were characterized mainly by fibrinosuppurative inflammation, which involved the synovial membranes, leptomeninges of the brain, and spinal cord. Furthermore, it also affects the choroid plexus, ependyma, nerve roots, and central canal of the spinal cord. S. suis was isolated from the cerebrospinal fluid, meningeal swabs, and/or synovial fluid of 8/9 piglets, and typified as serotype 9 by multiplex PCR.
Collapse
|
13
|
Bai Q, Ma J, Zhang Z, Zhong X, Pan Z, Zhu Y, Zhang Y, Wu Z, Liu G, Yao H. YSIRK-G/S-directed translocation is required for Streptococcus suis to deliver diverse cell wall anchoring effectors contributing to bacterial pathogenicity. Virulence 2021; 11:1539-1556. [PMID: 33138686 PMCID: PMC7644249 DOI: 10.1080/21505594.2020.1838740] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
The Streptococcus suis serotype 2 (SS2) is a significant zoonotic pathogen that is responsible for various swine diseases, even causing cytokine storms of Streptococcal toxic shock-like syndromes amongst human. Cell wall anchoring proteins with a C-terminal LPxTG are considered to play vital roles during SS2 infection; however, their exporting mechanism across cytoplasmic membranes has remained vague. This study found that YSIRK-G/S was involved in the exportation of LPxTG-anchoring virulence factors MRP and SspA in virulent SS2 strain ZY05719. The whole-genome analysis indicated that diverse LPxTG proteins fused with an N-terminal YSIRK-G/S motif are encoded in strain ZY05719. Two novel LPxTG proteins SspB and YzpA were verified to be exported via a putative transport system that was dependent on the YSIRK-G/S directed translocation, and portrayed vital functions during the infection of SS2 strain ZY05719. Instead of exhibiting an inactivation of C5a peptidase in SspB, another LPxTG protein with an N-terminal YSIRK-G/S motif from Streptococcus agalactiae was depicted to cleave the C5a component of the host complement. The consequent domain-architecture retrieval determined more than 10,000 SspB/YzpA like proteins that are extensively distributed in the Gram-positive bacteria, and most of them harbor diverse glycosyl hydrolase or peptidase domains within their middle regions, thus presenting their capability to interact with host cells. The said findings provide compelling evidence that LPxTG proteins with an N-terminal YSIRK-G/S motif are polymorphic effectors secreted by Gram-positive bacteria, which can be further proposed to define as cell wall anchoring effectors in a new subset.
Collapse
Affiliation(s)
- Qiankun Bai
- 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.,Department of pathogenic diagnosis, OIE Reference Lab for Swine Streptococcosis , Nanjing, China
| | - Jiale Ma
- 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.,Department of pathogenic diagnosis, OIE Reference Lab for Swine Streptococcosis , Nanjing, China
| | - Ze Zhang
- 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.,Department of pathogenic diagnosis, OIE Reference Lab for Swine Streptococcosis , Nanjing, China
| | - Xiaojun Zhong
- 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.,Department of pathogenic diagnosis, OIE Reference Lab for Swine Streptococcosis , Nanjing, China
| | - Zihao Pan
- 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.,Department of pathogenic diagnosis, OIE Reference Lab for Swine Streptococcosis , Nanjing, China
| | - Yinchu Zhu
- 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.,Department of pathogenic diagnosis, OIE Reference Lab for Swine Streptococcosis , Nanjing, China
| | - Yue Zhang
- 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.,Department of pathogenic diagnosis, OIE Reference Lab for Swine Streptococcosis , Nanjing, 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.,Department of pathogenic diagnosis, OIE Reference Lab for Swine Streptococcosis , Nanjing, China
| | - Guangjin 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.,Department of pathogenic diagnosis, 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.,Department of pathogenic diagnosis, OIE Reference Lab for Swine Streptococcosis , Nanjing, China
| |
Collapse
|
14
|
Lu Y, Li S, Shen X, Zhao Y, Zhou D, Hu D, Cai X, Lu L, Xiong X, Li M, Cao M. The type II histidine triad protein HtpsC facilitates invasion of epithelial cells by highly virulent Streptococcus suis serotype 2. J Microbiol 2021; 59:949-957. [PMID: 34491523 DOI: 10.1007/s12275-021-1129-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Revised: 07/19/2021] [Accepted: 07/20/2021] [Indexed: 10/20/2022]
Abstract
Streptococcus suis serotype 2 (S. suis 2) is an important zoonotic pathogen that presents a significant threat both to pigs and to workers in the pork industry. The initial steps of S. suis 2 pathogenesis are unclear. In this study, we found that the type II histidine triad protein HtpsC from the highly virulent Chinese isolate 05ZYH33 is structurally similar to internalin A (InlA) from Listeria monocytogenes, which plays an important role in mediating listerial invasion of epithelial cells. To determine if HtpsC and InlA function similarly, an isogenic htpsC mutant (ΔhtpsC) was generated in S. suis by homologous recombination. The htpsC deletion strain exhibited a diminished ability to adhere to and invade epithelial cells from different sources. Double immunofluorescence microscopy also revealed reduced survival of the ΔhtpsC mutant after co-cultivation with epithelium. Adhesion to epithelium and invasion by the wild type strain was inhibited by a monoclonal antibody against E-cadherin. In contrast, the htpsC-deficient mutant was unaffected by the same treatment, suggesting that E-cadherin is the host-cell receptor that interacts with HtpsC and facilitates bacterial internalization. Based on these results, we propose that HtpsC is involved in the process by which S. suis 2 penetrates host epithelial cells, and that this protein is an important virulence factor associated with cell adhesion and invasion.
Collapse
Affiliation(s)
- Yunjun Lu
- College Food Science and Light Industry, Nanjing Tech University, Nanjing, 211816, Jiangsu, P. R. China
| | - Shu Li
- Department of Microbiology, College of Basic Medical Sciences, Army Medical University (Third Military Medical University), Key Laboratory of Microbial Engineering under the Educational Committee in Chongqing, Chongqing, 400038, P. R. China
| | - Xiaodong Shen
- Department of Microbiology, College of Basic Medical Sciences, Army Medical University (Third Military Medical University), Key Laboratory of Microbial Engineering under the Educational Committee in Chongqing, Chongqing, 400038, P. R. China
| | - Yan Zhao
- Department of Microbiology, College of Basic Medical Sciences, Army Medical University (Third Military Medical University), Key Laboratory of Microbial Engineering under the Educational Committee in Chongqing, Chongqing, 400038, P. R. China
| | - Dongming Zhou
- Center for Disease Control and Prevention for the Eastern Theater Command, Nanjing, 210002, Jiangsu, P. R. China
| | - Dan Hu
- Center for Disease Control and Prevention for the Eastern Theater Command, Nanjing, 210002, Jiangsu, P. R. China
| | - Xushen Cai
- College Food Science and Light Industry, Nanjing Tech University, Nanjing, 211816, Jiangsu, P. R. China
| | - Lixia Lu
- College Food Science and Light Industry, Nanjing Tech University, Nanjing, 211816, Jiangsu, P. R. China
| | - Xiaohui Xiong
- College Food Science and Light Industry, Nanjing Tech University, Nanjing, 211816, Jiangsu, P. R. China
| | - Ming Li
- Department of Microbiology, College of Basic Medical Sciences, Army Medical University (Third Military Medical University), Key Laboratory of Microbial Engineering under the Educational Committee in Chongqing, Chongqing, 400038, P. R. China.
| | - Min Cao
- College Food Science and Light Industry, Nanjing Tech University, Nanjing, 211816, Jiangsu, P. R. China.
| |
Collapse
|
15
|
Liu H, Lei S, Jia L, Xia X, Sun Y, Jiang H, Zhu R, Li S, Qu G, Gu J, Sun C, Feng X, Han W, Langford PR, Lei L. Streptococcus suis serotype 2 enolase interaction with host brain microvascular endothelial cells and RPSA-induced apoptosis lead to loss of BBB integrity. Vet Res 2021; 52:30. [PMID: 33618766 PMCID: PMC7898445 DOI: 10.1186/s13567-020-00887-6] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Accepted: 12/23/2020] [Indexed: 11/10/2022] Open
Abstract
Host proteins interacting with pathogens are receiving more attention as potential therapeutic targets in molecular medicine. Streptococcus suis serotype 2 (SS2) is an important cause of meningitis in both humans and pigs worldwide. SS2 Enolase (Eno) has previously been identified as a virulence factor with a role in altering blood brain barrier (BBB) integrity, but the host cell membrane receptor of Eno and The mechanism(s) involved are unclear. This study identified that SS2 Eno binds to 40S ribosomal protein SA (RPSA) on the surface of porcine brain microvascular endothelial cells leading to activation of intracellular p38/ERK-eIF4E signalling, which promotes intracellular expression of HSPD1 (heat-shock protein family D member 1), and initiation of host-cell apoptosis, and increased BBB permeability facilitating bacterial invasion. This study reveals novel functions for the host-interactional molecules RPSA and HSPD1 in BBB integrity, and provides insight for new therapeutic strategies in meningitis.
Collapse
Affiliation(s)
- Hongtao Liu
- Key Laboratory of Zoonosis, Ministry of Education, Institute of Zoonosis/College of Veterinary Medicine, Jilin University, Changchun, Jilin, 130062, People's Republic of China
| | - Siyu Lei
- School of Basic Medicine, Jilin University, Changchun, 130021, China
| | - Li Jia
- Key Laboratory of Zoonosis, Ministry of Education, Institute of Zoonosis/College of Veterinary Medicine, Jilin University, Changchun, Jilin, 130062, People's Republic of China
| | - Xiaojing Xia
- Key Laboratory of Zoonosis, Ministry of Education, Institute of Zoonosis/College of Veterinary Medicine, Jilin University, Changchun, Jilin, 130062, People's Republic of China
| | - Yingying Sun
- Key Laboratory of Zoonosis, Ministry of Education, Institute of Zoonosis/College of Veterinary Medicine, Jilin University, Changchun, Jilin, 130062, People's Republic of China
| | - Hexiang Jiang
- Key Laboratory of Zoonosis, Ministry of Education, Institute of Zoonosis/College of Veterinary Medicine, Jilin University, Changchun, Jilin, 130062, People's Republic of China
| | - Rining Zhu
- Key Laboratory of Zoonosis, Ministry of Education, Institute of Zoonosis/College of Veterinary Medicine, Jilin University, Changchun, Jilin, 130062, People's Republic of China
| | - Shuguang Li
- Shandong Binzhou Animal Science and Veterinary Medicine Academy, Binzhou, Shandong, 256600, People's Republic of China
| | - Guanggang Qu
- Shandong Binzhou Animal Science and Veterinary Medicine Academy, Binzhou, Shandong, 256600, People's Republic of China
| | - Jingmin Gu
- Key Laboratory of Zoonosis, Ministry of Education, Institute of Zoonosis/College of Veterinary Medicine, Jilin University, Changchun, Jilin, 130062, People's Republic of China
| | - Changjiang Sun
- Key Laboratory of Zoonosis, Ministry of Education, Institute of Zoonosis/College of Veterinary Medicine, Jilin University, Changchun, Jilin, 130062, People's Republic of China
| | - Xin Feng
- Key Laboratory of Zoonosis, Ministry of Education, Institute of Zoonosis/College of Veterinary Medicine, Jilin University, Changchun, Jilin, 130062, People's Republic of China
| | - Wenyu Han
- Key Laboratory of Zoonosis, Ministry of Education, Institute of Zoonosis/College of Veterinary Medicine, Jilin University, Changchun, Jilin, 130062, People's Republic of China
| | - Paul R Langford
- Section of Paediatric Infectious Disease, Imperial College London, London, W2 1PG, UK
| | - Liancheng Lei
- Key Laboratory of Zoonosis, Ministry of Education, Institute of Zoonosis/College of Veterinary Medicine, Jilin University, Changchun, Jilin, 130062, People's Republic of China. .,College of Animal Science, Yangtze University, Jingzhou, Hubei, 434023, People's Republic of China.
| |
Collapse
|
16
|
Streptococcal Infections in Marine Mammals. Microorganisms 2021; 9:microorganisms9020350. [PMID: 33578962 PMCID: PMC7916692 DOI: 10.3390/microorganisms9020350] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Revised: 01/29/2021] [Accepted: 02/07/2021] [Indexed: 01/28/2023] Open
Abstract
Marine mammals are sentinels for the marine ecosystem and threatened by numerous factors including infectious diseases. One of the most frequently isolated bacteria are beta-hemolytic streptococci. However, knowledge on ecology and epidemiology of streptococcal species in marine mammals is very limited. This review summarizes published reports on streptococcal species, which have been detected in marine mammals. Furthermore, we discuss streptococcal transmission between and adaptation to their marine mammalian hosts. We conclude that streptococci colonize and/or infect marine mammals very frequently, but in many cases, streptococci isolated from marine mammals have not been further identified. How these bacteria disseminate and adapt to their specific niches can only be speculated due to the lack of respective research. Considering the relevance of pathogenic streptococci for marine mammals as part of the marine ecosystem, it seems that they have been neglected and should receive scientific interest in the future.
Collapse
|
17
|
Bünger M, Brunthaler R, Unterweger C, Loncaric I, Dippel M, Ruczizka U, Schwarz L, Griessler A, Voglmayr T, Verhovsek D, Ladinig A, Spergser J. Mycoplasma hyorhinis as a possible cause of fibrinopurulent meningitis in pigs? - a case series. Porcine Health Manag 2020; 6:38. [PMID: 33292668 PMCID: PMC7713030 DOI: 10.1186/s40813-020-00178-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Accepted: 11/25/2020] [Indexed: 12/18/2022] Open
Abstract
Background Mycoplasma hyorhinis is an invader of the upper respiratory tract in swine that is considered to have ubiquitous distribution. It is mainly known for causing polyserositis and polyarthritis in weaned piglets, even though the mechanisms of systemic spread are not fully understood. Mycoplasma hyorhinis has also been associated with other diseases in pigs such as pneumonia or otitis media, but so far has not been known to cause central nervous disorders. This case series reports the isolation of Mycoplasma hyorhinis from cerebrospinal fluid and/ or meningeal swabs from piglets originating from four different piglet producing farms in Austria. Case presentation On farm 1, coughing, stiff movement and central nervous signs occurred in nursery piglets. Mycoplasma hyorhinis was the only pathogen isolated from meningeal swabs from two piglets showing central nervous signs. Fibrinopurulent leptomeningitis was only observed in one piglet. Only one of two nursery piglets from farm 2 showed mild central nervous signs but no histologic lesions; Mycoplasma hyorhinis was isolated from cerebrospinal fluid of the piglet with neurologic signs. Mycoplasma hyorhinis was isolated from cerebrospinal fluid of all three investigated piglets from farm 3, all of which showed central nervous signs and purulent leptomeningitis. Further, Streptococcus suis was isolated from the cerebrospinal fluid of one piglet. Fibrinopurulent leptomeningitis was detected in two piglets from farm 4 that had died overnight without showing any clinical signs and Mycoplasma hyorhinis was isolated from meningeal swabs from both piglets. Conclusion While causality has yet to be proven by experimental infection and in situ detection of the pathogen in histologic sections, the findings of this study and the absence of other pathogens suggest Mycoplasma hyorhinis as a potential causative agent of meningitis in swine.
Collapse
Affiliation(s)
- Moritz Bünger
- University Clinic for Swine, Vetmeduni Vienna, Vienna, Austria
| | | | | | - Igor Loncaric
- Institute of Microbiology, Vetmeduni Vienna, Vienna, Austria
| | | | - Ursula Ruczizka
- University Clinic for Swine, Vetmeduni Vienna, Vienna, Austria
| | - Lukas Schwarz
- University Clinic for Swine, Vetmeduni Vienna, Vienna, Austria
| | | | - Thomas Voglmayr
- Traunkreis Vet Clinic, Ried im Traunkreis, Traunkreis, Austria
| | - Doris Verhovsek
- University Clinic for Swine, Vetmeduni Vienna, Vienna, Austria.,VetFarm Medau, Vetmeduni Vienna, Berndorf, Austria
| | - Andrea Ladinig
- University Clinic for Swine, Vetmeduni Vienna, Vienna, Austria.
| | | |
Collapse
|
18
|
Plasencia-Muñoz B, Avelar-González FJ, De la Garza M, Jacques M, Moreno-Flores A, Guerrero-Barrera AL. Actinobacillus pleuropneumoniae Interaction With Swine Endothelial Cells. Front Vet Sci 2020; 7:569370. [PMID: 33195549 PMCID: PMC7658479 DOI: 10.3389/fvets.2020.569370] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Accepted: 09/28/2020] [Indexed: 12/26/2022] Open
Abstract
Actinobacillus pleuropneumonia is a swine (host) specific respiratory pathogen and the etiological agent of swine pleuropneumonia which affects pigs of all ages, many being asymptomatic carriers. This pathogen has high morbidity and mortality rates which generates large economic losses for the pig industry. Actinobacillus pleuropneumoniae is a widely studied bacterium, however its pathogenesis is not yet fully understood. The prevalence of the 18 serotypes of A. pleuropneumoniae varies by geographic region, in North American area, more specifically in Mexico, serotypes 1, 3, 5b, and 7 show higher prevalence. Actinobacillus pleuropneumoniae is described as a strict extracellular pathogen with tropism for lower respiratory tract. However, this study depicts the ability of these serotypes to adhere to non-phagocytic cells, using an endothelial cell model, as well as their ability to internalize them, proposing it could be considered as an intracellular pathogen.
Collapse
Affiliation(s)
- Berenice Plasencia-Muñoz
- Laboratorio de Biología Celular y Tisular, Departamento de Morfología, Centro de Ciencias Básicas, Universidad Autónoma de Aguascalientes, Aguascalientes, Mexico
| | - Francisco J Avelar-González
- Laboratorio de Estudios Ambientales, Departamento de Fisiología y Farmacología, Centro de Ciencias Básicas, Universidad Autónoma de Aguascalientes, Aguascalientes, Mexico
| | - Mireya De la Garza
- Departamento de Biología Celular, Centro de Investigación y de Estudios Avanzados del IPN, Ciudad de México, Mexico
| | - Mario Jacques
- Groupe de Recherche sur les Maladies Infectieuses en Production Animale, Faculté de Médecine Vétérinaire, Université de Montréal, Saint-Hyacinthe, QC, Canada
| | - Adriana Moreno-Flores
- Laboratorio de Biología Celular y Tisular, Departamento de Morfología, Centro de Ciencias Básicas, Universidad Autónoma de Aguascalientes, Aguascalientes, Mexico
| | - Alma L Guerrero-Barrera
- Laboratorio de Biología Celular y Tisular, Departamento de Morfología, Centro de Ciencias Básicas, Universidad Autónoma de Aguascalientes, Aguascalientes, Mexico
| |
Collapse
|
19
|
Xu B, Liu R, Ding M, Zhang J, Sun H, Liu C, Lu F, Zhao S, Pan Q, Zhang X. Interaction of Mycoplasma synoviae with chicken synovial sheath cells contributes to macrophage recruitment and inflammation. Poult Sci 2020; 99:5366-5377. [PMID: 33142453 PMCID: PMC7647830 DOI: 10.1016/j.psj.2020.08.003] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2019] [Revised: 06/13/2020] [Accepted: 08/15/2020] [Indexed: 11/21/2022] Open
Abstract
Mycoplasma synoviae (MS) is an important avian pathogen causing considerable economic hardship in the poultry industry. A major inflammation caused by MS is synovitis that occurs in the synovial tendon sheath and joint synovium. However, the overall appearance of pathological changes in the tendon sheath and surrounding tissues caused by MS infection at the level of pathological tissue sections was poor. Studies on the role of MS and synovial sheath cells (SSCs) interaction in the development of synovitis have not been carried out. Through histopathological observation, our study found that a major MS-induced pathological change of the tendon sheath synovium was extensive scattered and focal inflammatory cell infiltration of the tendon sheath synovial layer. In vitro research experiments revealed that the CFU numbers of MS adherent and invading SSC, the levels of expression of various pattern recognition receptors, inflammatory cytokines, and chemokines coding genes, such as IL-1β, IL-6, IL-8, CCL-20, RANTES, MIP-1β, TLR7, and TLR15 in SSCs, and chemotaxis of macrophages were significantly increased when the multiplicity of infection (MOI) of MS to SSC were increased tenfold. The expression level of IL-12p40 in SSC was significantly higher when the MOIs of MS to SSC were increased by a factor of 100. The interaction between MS and SSC can activate macrophages, which was manifested by a significant increase in the expression of IL-1β, IL-6, IL-8, CCL-20, RANTES, MIP-1β, and CXCL-13. This study systematically demonstrated that the interaction of MS with chicken SSC contributes to the inflammatory response caused by the robust expression of related cytokines and macrophage chemotaxis. These findings are helpful in elucidating the molecular mechanism of MS-induced synovitis in chickens.
Collapse
Affiliation(s)
- Bin Xu
- Key Laboratory of Veterinary Biological Engineering and Technology of Ministry of Agriculture, Institute of Veterinary Medicine, Jiangsu Academy of Agricultural Sciences, Nanjing, China; National Center for Engineering Research of Veterinary Bio-Products, Jiangsu Academy of Agricultural Sciences, Nanjing, China
| | - Rui Liu
- Key Laboratory of Veterinary Biological Engineering and Technology of Ministry of Agriculture, Institute of Veterinary Medicine, Jiangsu Academy of Agricultural Sciences, Nanjing, China; National Center for Engineering Research of Veterinary Bio-Products, Jiangsu Academy of Agricultural Sciences, Nanjing, China
| | - Meijuan Ding
- Key Laboratory of Veterinary Biological Engineering and Technology of Ministry of Agriculture, Institute of Veterinary Medicine, Jiangsu Academy of Agricultural Sciences, Nanjing, China; National Center for Engineering Research of Veterinary Bio-Products, Jiangsu Academy of Agricultural Sciences, Nanjing, China
| | - Jingfeng Zhang
- Key Laboratory of Veterinary Biological Engineering and Technology of Ministry of Agriculture, Institute of Veterinary Medicine, Jiangsu Academy of Agricultural Sciences, Nanjing, China; National Center for Engineering Research of Veterinary Bio-Products, Jiangsu Academy of Agricultural Sciences, Nanjing, China
| | - Huawei Sun
- Key Laboratory of Veterinary Biological Engineering and Technology of Ministry of Agriculture, Institute of Veterinary Medicine, Jiangsu Academy of Agricultural Sciences, Nanjing, China; National Center for Engineering Research of Veterinary Bio-Products, Jiangsu Academy of Agricultural Sciences, Nanjing, China
| | - Chuanmin Liu
- Key Laboratory of Veterinary Biological Engineering and Technology of Ministry of Agriculture, Institute of Veterinary Medicine, Jiangsu Academy of Agricultural Sciences, Nanjing, China; National Center for Engineering Research of Veterinary Bio-Products, Jiangsu Academy of Agricultural Sciences, Nanjing, China
| | - Fengying Lu
- Key Laboratory of Veterinary Biological Engineering and Technology of Ministry of Agriculture, Institute of Veterinary Medicine, Jiangsu Academy of Agricultural Sciences, Nanjing, China; National Center for Engineering Research of Veterinary Bio-Products, Jiangsu Academy of Agricultural Sciences, Nanjing, China
| | - Sha Zhao
- Key Laboratory of Veterinary Biological Engineering and Technology of Ministry of Agriculture, Institute of Veterinary Medicine, Jiangsu Academy of Agricultural Sciences, Nanjing, China; National Center for Engineering Research of Veterinary Bio-Products, Jiangsu Academy of Agricultural Sciences, Nanjing, China
| | - Qunxing Pan
- Key Laboratory of Veterinary Biological Engineering and Technology of Ministry of Agriculture, Institute of Veterinary Medicine, Jiangsu Academy of Agricultural Sciences, Nanjing, China; National Center for Engineering Research of Veterinary Bio-Products, Jiangsu Academy of Agricultural Sciences, Nanjing, China
| | - Xiaofei Zhang
- Key Laboratory of Veterinary Biological Engineering and Technology of Ministry of Agriculture, Institute of Veterinary Medicine, Jiangsu Academy of Agricultural Sciences, Nanjing, China; National Center for Engineering Research of Veterinary Bio-Products, Jiangsu Academy of Agricultural Sciences, Nanjing, China.
| |
Collapse
|
20
|
Gajdács M, Németh A, Knausz M, Barrak I, Stájer A, Mestyán G, Melegh S, Nyul A, Tóth Á, Ágoston Z, Urbán E. Streptococcus suis: An Underestimated Emerging Pathogen in Hungary? Microorganisms 2020; 8:1292. [PMID: 32847011 PMCID: PMC7570012 DOI: 10.3390/microorganisms8091292] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Revised: 08/16/2020] [Accepted: 08/21/2020] [Indexed: 11/16/2022] Open
Abstract
Streptococcus suis (S. suis) is an emerging zoonotic pathogen, demonstrated as an etiological agent in human infections in increasing frequency, including diseases like purulent meningitis, sepsis, uveitis-endophtalmitis and arthritis. Due to the increased availability and utility of novel diagnostic technologies in clinical microbiology, more studies have been published on the epidemiology of S. suis, both in veterinary and human medicine; however, there are no comprehensive data available regarding human S. suis infections from East-Central European countries. As a part of our study, data were collected from the National Bacteriological Surveillance (NBS) system on patients who had at least one positive microbiological result for S. suis, corresponding to an 18-year study period (2002-2019). n = 74 S. suis strains were isolated from invasive human infections, corresponding to 34 patients. The number of affected patients was 1.89 ± 1.53/year (range: 0-5). Most isolates originated from blood culture (63.5%) and cerebrospinal fluid (18.9%) samples. Additionally, we present detailed documentation of three instructive cases from three regions of the country and with three distinctly different outcomes. Hungary has traditional agriculture, the significant portion of which includes the production and consumption of pork meat, with characteristic preparation and consumption customs and unfavorable epidemiological characteristics (alcohol consumption, prevalence of malignant diseases or diabetes), which have all been described as important predisposing factors for the development of serious infections. Clinicians and microbiologist need to be vigilant even in nonendemic areas, especially if the patients have a history of occupational hazards or having close contact with infected pigs.
Collapse
Affiliation(s)
- Márió Gajdács
- Department of Pharmacodynamics and Biopharmacy, Faculty of Pharmacy, University of Szeged, Eötvös utca 6., 6720 Szeged, Hungary
- Institute of Medical Microbiology, Faculty of Medicine, Semmelweis University, Nagyvárad tér 4., 1089 Budapest, Hungary
| | - Anita Németh
- Microbiology Laboratory, Petz Aladár County Teaching Hospital, Vasvári Pál utca 2–4., 9023 Győr, Hungary; (A.N.); (M.K.)
| | - Márta Knausz
- Microbiology Laboratory, Petz Aladár County Teaching Hospital, Vasvári Pál utca 2–4., 9023 Győr, Hungary; (A.N.); (M.K.)
| | - Ibrahim Barrak
- Department of Prosthodontics, Faculty of Dentistry, University of Szeged, Tiszta Lajos körút 62–64., 6720 Szeged, Hungary;
| | - Anette Stájer
- Department of Periodontology, Faculty of Dentistry, University of Szeged, Tiszta Lajos körút 62–64, 6720 Szeged, Hungary;
| | - Gyula Mestyán
- Department of Medical Microbiology and Immunology, Faculty of Medicine, University of Pécs, Szigeti út 12., 7624 Pécs, Hungary; (G.M.); (S.M.); (A.N.)
| | - Szilvia Melegh
- Department of Medical Microbiology and Immunology, Faculty of Medicine, University of Pécs, Szigeti út 12., 7624 Pécs, Hungary; (G.M.); (S.M.); (A.N.)
| | - Adrienn Nyul
- Department of Medical Microbiology and Immunology, Faculty of Medicine, University of Pécs, Szigeti út 12., 7624 Pécs, Hungary; (G.M.); (S.M.); (A.N.)
| | - Ákos Tóth
- Department of Bacteriology, Mycology and Parasitology, National Public Health Center, Albert Flórián út 2–6., 1097 Budapest, Hungary;
| | - Zsuzsanna Ágoston
- Department of Anaesthesiology and Intensive Therapy, Faculty of Medicine, University of Szeged, Semmelweis utca 6., 6725 Szeged, Hungary;
| | - Edit Urbán
- Institute of Translational Medicine, Faculty of Medicine, University of Pécs, Szigeti út 12., 7624 Pécs, Hungary;
| |
Collapse
|
21
|
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
|
22
|
Vötsch D, Willenborg M, Oelemann WM, Brogden G, Valentin-Weigand P. Membrane Binding, Cellular Cholesterol Content and Resealing Capacity Contribute to Epithelial Cell Damage Induced by Suilysin of Streptococcus suis. Pathogens 2019; 9:pathogens9010033. [PMID: 31905867 PMCID: PMC7168673 DOI: 10.3390/pathogens9010033] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2019] [Revised: 12/19/2019] [Accepted: 12/24/2019] [Indexed: 12/31/2022] Open
Abstract
Streptococcus (S.) suis is a major cause of economic losses in the pig industry worldwide and is an emerging zoonotic pathogen. One important virulence-associated factor is suilysin (SLY), a toxin that belongs to the family of cholesterol-dependent pore-forming cytolysins (CDC). However, the precise role of SLY in host–pathogen interactions is still unclear. Here, we investigated the susceptibility of different respiratory epithelial cells to SLY, including immortalized cell lines (HEp-2 and NPTr cells), which are frequently used in in vitro studies on S. suis virulence mechanisms, as well as primary porcine respiratory cells, which represent the first line of barrier during S. suis infections. SLY-induced cell damage was determined by measuring the release of lactate dehydrogenase after infection with a virulent S. suis serotype 2 strain, its isogenic SLY-deficient mutant strain, or treatment with the recombinant protein. HEp-2 cells were most susceptible, whereas primary epithelial cells were hardly affected by the toxin. This prompted us to study possible explanations for these differences. We first investigated the binding capacity of SLY using flow cytometry analysis. Since binding and pore-formation of CDC is dependent on the membrane composition, we also determined the cellular cholesterol content of the different cell types using TLC and HPLC. Finally, we examined the ability of those cells to reseal SLY-induced pores using flow cytometry analysis. Our results indicated that the amount of membrane-bound SLY, the cholesterol content of the cells, as well as their resealing capacity all affect the susceptibility of the different cells regarding the effects of SLY. These findings underline the differences of in vitro pathogenicity models and may further help to dissect the biological role of SLY during S. suis infections.
Collapse
Affiliation(s)
- Désirée Vötsch
- Institute for Microbiology, University of Veterinary Medicine Hannover, 30173 Hannover, Germany; (D.V.); (M.W.)
| | - Maren Willenborg
- Institute for Microbiology, University of Veterinary Medicine Hannover, 30173 Hannover, Germany; (D.V.); (M.W.)
| | - Walter M.R. Oelemann
- Institute for Microbiology, University of Veterinary Medicine Hannover, 30173 Hannover, Germany; (D.V.); (M.W.)
- Departamento de Imunologia, Instituto de Microbiologia Paulo Góes, Universidade Federal do Rio de Janeiro (UFRJ), 21941-901 Rio de Janeiro, Brazil
| | - Graham Brogden
- Department of Physiological Chemistry, University for Veterinary Medicine Hannover, 30559 Hannover, Germany;
| | - Peter Valentin-Weigand
- Institute for Microbiology, University of Veterinary Medicine Hannover, 30173 Hannover, Germany; (D.V.); (M.W.)
- Correspondence: ; Tel.: +49-(0)511-856-7362
| |
Collapse
|
23
|
Virulence Factors of Meningitis-Causing Bacteria: Enabling Brain Entry across the Blood-Brain Barrier. Int J Mol Sci 2019; 20:ijms20215393. [PMID: 31671896 PMCID: PMC6862235 DOI: 10.3390/ijms20215393] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2019] [Revised: 10/24/2019] [Accepted: 10/25/2019] [Indexed: 12/16/2022] Open
Abstract
Infections of the central nervous system (CNS) are still a major cause of morbidity and mortality worldwide. Traversal of the barriers protecting the brain by pathogens is a prerequisite for the development of meningitis. Bacteria have developed a variety of different strategies to cross these barriers and reach the CNS. To this end, they use a variety of different virulence factors that enable them to attach to and traverse these barriers. These virulence factors mediate adhesion to and invasion into host cells, intracellular survival, induction of host cell signaling and inflammatory response, and affect barrier function. While some of these mechanisms differ, others are shared by multiple pathogens. Further understanding of these processes, with special emphasis on the difference between the blood-brain barrier and the blood-cerebrospinal fluid barrier, as well as virulence factors used by the pathogens, is still needed.
Collapse
|
24
|
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
|
25
|
Zhang B, Ku X, Zhang X, Zhang Y, Chen G, Chen F, Zeng W, Li J, Zhu L, He Q. The AI-2/ luxS Quorum Sensing System Affects the Growth Characteristics, Biofilm Formation, and Virulence of Haemophilus parasuis. Front Cell Infect Microbiol 2019; 9:62. [PMID: 30941317 PMCID: PMC6434701 DOI: 10.3389/fcimb.2019.00062] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2018] [Accepted: 02/28/2019] [Indexed: 12/12/2022] Open
Abstract
Haemophilus parasuis (H. parasuis) is a kind of opportunistic pathogen of the upper respiratory tract of piglets. Under certain circumstances, virulent strains can breach the mucosal barrier and enter the bloodstream, causing severe Glässer's disease. Many virulence factors are found to be related to the pathogenicity of H. parasuis strain, but the pathogenic mechanism remains unclear. LuxS/AI-2, as a kind of very important quorum sensing system, affects the growth characteristics, biofilm formation, antibiotic production, virulence, and metabolism of different strains. In order to investigate the effect of luxS/AI-2 quorum sensing system on the virulence of H. parasuis, a deletion mutant strain (ΔluxS) and complemented strain (C-luxS) were constructed and characterized. The results showed that the luxS gene participated in regulating and controlling stress resistance, biofilm formation and virulence. Compared with wild-type strain, ΔluxS strain decreased the production of AI-2 molecules and the tolerance toward oxidative stress and heat shock, and it reduced the abilities of autoagglutination, hemagglutination, and adherence, whereas it increased the abilities to form biofilm in vitro. In vivo experiments showed that ΔluxS strain attenuated its virulence about 10-folds and significantly decreased its tissue burden of bacteria in mice, compared with the wild-type strain. Taken together, the luxS/AI-2 quorum sensing system in H. parasuis not only plays an important role in growth and biofilm formation, but also affects the pathogenicity of H. parasuis.
Collapse
Affiliation(s)
- Bingzhou Zhang
- State Key Laboratory of Agricultural Microbiology, Division of Animal Infectious Diseases, College of Animal Sciences and Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Xugang Ku
- State Key Laboratory of Agricultural Microbiology, Division of Animal Infectious Diseases, College of Animal Sciences and Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Xiaoqian Zhang
- State Key Laboratory of Agricultural Microbiology, Division of Animal Infectious Diseases, College of Animal Sciences and Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Yan Zhang
- College of Animal Sciences and Veterinary Medicine, Xinjiang Agricultural University, Urumqi, China
| | - Guo Chen
- State Key Laboratory of Agricultural Microbiology, Division of Animal Infectious Diseases, College of Animal Sciences and Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Fangzhou Chen
- State Key Laboratory of Agricultural Microbiology, Division of Animal Infectious Diseases, College of Animal Sciences and Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Wei Zeng
- State Key Laboratory of Agricultural Microbiology, Division of Animal Infectious Diseases, College of Animal Sciences and Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Jing Li
- State Key Laboratory of Agricultural Microbiology, Division of Animal Infectious Diseases, College of Animal Sciences and Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Ling Zhu
- State Key Laboratory of Agricultural Microbiology, Division of Animal Infectious Diseases, College of Animal Sciences and Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Qigai He
- State Key Laboratory of Agricultural Microbiology, Division of Animal Infectious Diseases, College of Animal Sciences and Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| |
Collapse
|
26
|
The Two-Component Signaling System VraSR ss Is Critical for Multidrug Resistance and Full Virulence in Streptococcus suis Serotype 2. Infect Immun 2018; 86:IAI.00096-18. [PMID: 29685990 DOI: 10.1128/iai.00096-18] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2018] [Accepted: 04/18/2018] [Indexed: 11/20/2022] Open
Abstract
Streptococcus suis has received increasing attention for its involvement in severe human infections worldwide as well as in multidrug resistance. Two-component signaling systems (TCSSs) play important roles in bacterial adaptation to various environmental stimuli. In this study, we identified a novel TCSS located in S. suis serotype 2 (SS2), designated VraSRSS, which is involved in bacterial pathogenicity and susceptibility to antimicrobials. Our data demonstrated that the yvqFSS gene, located upstream of vraSRSS , shared the same promoter with the TCSS genes, which was directly regulated by VraSRSS, as shown in electrophoretic mobility shift assays. Notably, YvqFSS and VraSRSS constitute a novel multidrug resistance module of SS2 that participates in resistance to certain groups of antimicrobials. Further analyses showed that VraSRSS inactivation significantly attenuated bacterial virulence in animal models, which, coupled with the significant activation of VraSRSS expression observed in host blood, strongly suggested that VraSRSS is an important regulator of SS2 pathogenicity. Indeed, RNA-sequencing analyses identified 106 genes that were differentially expressed between the wild-type and ΔvraSRSS strains, including genes involved in capsular polysaccharide (CPS) biosynthesis. Subsequent studies confirmed that VraSRSS indirectly regulated the transcription of CPS gene clusters and, thus, controlled the CPS thickness shown by transmission electron microscopy. Decreased CPS biosynthesis caused by vraSRSS deletion subsequently increased bacterial adhesion to epithelial cells and attenuated antiphagocytosis against macrophages, which partially clarified the pathogenic mechanism mediated by VraSRSS Taken together, our data suggest that the novel TCSS, VraSRSS, plays critical roles for multidrug resistance and full virulence in SS2.
Collapse
|
27
|
Limited Interactions between Streptococcus Suis and Haemophilus Parasuis in In Vitro Co-Infection Studies. Pathogens 2018; 7:pathogens7010007. [PMID: 29316613 PMCID: PMC5874733 DOI: 10.3390/pathogens7010007] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2017] [Revised: 01/01/2018] [Accepted: 01/04/2018] [Indexed: 01/14/2023] Open
Abstract
Streptococcus suis and Haemophilus parasuis are normal inhabitants of the porcine upper respiratory tract but are also among the most frequent causes of disease in weaned piglets worldwide, causing inflammatory diseases such as septicemia, meningitis and pneumonia. Using an in vitro model of infection with tracheal epithelial cells or primary alveolar macrophages (PAMs), it was possible to determine the interaction between S. suis serotype 2 and H. parasuis strains with different level of virulence. Within H. parasuis strains, the low-virulence F9 strain showed higher adhesion levels to respiratory epithelial cells and greater association levels to PAMs than the high-virulence Nagasaki strain. Accordingly, the low-virulence F9 strain induced, in general, higher levels of pro-inflammatory cytokines than the virulent Nagasaki strain from both cell types. In general, S. suis adhesion levels to respiratory epithelial cells were similar to H. parasuis Nagasaki strain. Yet, S. suis strains induced a significantly lower level of pro-inflammatory cytokine expression from epithelial cells and PAMs than those observed with both H. parasuis strains. Finally, this study has shown that, overall and under the conditions used in the present study, S. suis and H. parasuis have limited in vitro interactions between them and use probably different host receptors, regardless to their level of virulence.
Collapse
|
28
|
De la Rosa-Ramos MA, Muñoz-Solís K, Palma-Zepeda M, Gutierrez-Castillo AC, López Villegas EO, Guerra-Infante FM, Castro-Escarpulli G. Adherence of Ornithobacterium rhinotracheale to chicken embryo lung cells as a pathogenic mechanism. Avian Pathol 2017; 47:172-178. [PMID: 29016186 DOI: 10.1080/03079457.2017.1390208] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
Ornithobacterium rhinotracheale is a bacterium that causes respiratory disease in birds and it has been isolated in countries with a large poultry production, including Mexico. The pathogenicity mechanisms of this bacterium have not been completely elucidated yet. The capacity of the bacterium to adhere to epithelial cells of chicken in vitro has been evidenced, and since this bacterium has been isolated from the lungs and air sacs of several avian species, the aim of this study was to determine if this bacterium can adhere to chicken lung cells. We used five O. rhinotracheale reference serovars (A-E) that were in contact with primary lung cells cultured from a 19-day-old chicken embryo. O. rhinotracheale adherence was evaluated through optical and transmission electron microscopies. The results revealed that O. rhinotracheale is capable of adhering to chicken embryo lung cells within 3 h of incubation with a diffuse adherence pattern. The adherence percentages of the chicken embryo lung cells were 51-96% according to the serovar of the bacterium. Relative adherence was from 4 to 8 bacteria per cell. Transmission electron microscope data revealed intracellular bacteria inside a vacuole in less than 3 h of incubation.
Collapse
Affiliation(s)
- Miguel A De la Rosa-Ramos
- a Laboratorio de Microbiología Veterinaria, Departamento de Microbiología de la Escuela Nacional de Ciencias Biológicas (ENCB), Instituto Politécnico Nacional , Mexico City , Mexico.,b Laboratorio de Bacteriología Médica, Departamento de Microbiología de la Escuela Nacional de Ciencias Biológicas (ENCB), Instituto Politécnico Nacional , Mexico City , Mexico
| | - Karina Muñoz-Solís
- a Laboratorio de Microbiología Veterinaria, Departamento de Microbiología de la Escuela Nacional de Ciencias Biológicas (ENCB), Instituto Politécnico Nacional , Mexico City , Mexico
| | - Mario Palma-Zepeda
- c Laboratorio de Virología, Departamento de Microbiología e Inmunología, Facultad de Medicina Veterinaria y Zootecnia (FMVZ) , Universidad Nacional Autónoma de México (UNAM) , Mexico City , Mexico
| | - Adriana C Gutierrez-Castillo
- d Facultad de Medicina Veterinaria y Zootecnia (FMVZ) , Universidad Autónoma del Estado de México (UAEMex) , Toluca , Mexico
| | - Edgar O López Villegas
- e Central de Microscopia, Escuela Nacional de Ciencias Biológicas (ENCB) , Instituto Politécnico Nacional (IPN) , Mexico City , Mexico
| | - Fernando M Guerra-Infante
- a Laboratorio de Microbiología Veterinaria, Departamento de Microbiología de la Escuela Nacional de Ciencias Biológicas (ENCB), Instituto Politécnico Nacional , Mexico City , Mexico.,f Laboratorio de Bioinmunología Molecular del Instituto Nacional de Perinatología (INPerIER) , Mexico City , Mexico
| | - Graciela Castro-Escarpulli
- b Laboratorio de Bacteriología Médica, Departamento de Microbiología de la Escuela Nacional de Ciencias Biológicas (ENCB), Instituto Politécnico Nacional , Mexico City , Mexico
| |
Collapse
|
29
|
Auger JP, Gottschalk M. The Streptococcus suis factor H-binding protein: A key to unlocking the blood-brain barrier and access the central nervous system? Virulence 2017. [PMID: 28622084 DOI: 10.1080/21505594.2017.1342027] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Affiliation(s)
- Jean-Philippe Auger
- a Streptococcus suis Research Laboratory, Research Group on Infectious Diseases in Production Animals (GREMIP) & Swine and Poultry Infectious Diseases Research Center (CRIPA), Faculty of Veterinary Medicine , University of Montreal , St-Hyacinthe , QC , Canada
| | - Marcelo Gottschalk
- a Streptococcus suis Research Laboratory, Research Group on Infectious Diseases in Production Animals (GREMIP) & Swine and Poultry Infectious Diseases Research Center (CRIPA), Faculty of Veterinary Medicine , University of Montreal , St-Hyacinthe , QC , Canada
| |
Collapse
|
30
|
Kong D, Chen Z, Wang J, Lv Q, Jiang H, Zheng Y, Xu M, Zhou X, Hao H, Jiang Y. Interaction of factor H-binding protein of Streptococcus suis with globotriaosylceramide promotes the development of meningitis. Virulence 2017; 8:1290-1302. [PMID: 28402705 DOI: 10.1080/21505594.2017.1317426] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Streptococcus suis is an important emerging zoonotic agent that causes acute bacterial meningitis in humans with high mortality and morbidity. Our previous work showed that factor H-binding protein (Fhb) contributed to virulence of S. suis, but the role of Fhb in the development of S. suis meningitis remained unclear. In this study, we demonstrated for the first time that Fhb contributed to the traversal of S. suis across the human blood-brain barrier by allelic-exchange mutagenesis, complementation and specific antibody blocking studies. We also showed that globotriaosylceramide (Gb3), the receptor of Fhb, was involved in this process and affected S. suis infection-induced activation of myosin light chain 2 through Rho/ROCK signaling in hCMEC/D3 cells. Using a murine model of S. suis meningitis, we further demonstrated that Gb3-deficiency prevented the mice from developing severe brain inflammation or injury. Our results demonstrate that the Fhb-Gb3 interaction plays an important role in the development of S. suis meningitis and might be a potential therapeutic target against S. suis infection.
Collapse
Affiliation(s)
- Decong Kong
- a State Key Laboratory of Pathogen and Biosecurity , Institute of Microbiology and Epidemiology, Academy of Military Medical Sciences , Beijing , China
| | - Zhe Chen
- a State Key Laboratory of Pathogen and Biosecurity , Institute of Microbiology and Epidemiology, Academy of Military Medical Sciences , Beijing , China.,b College of Biological Science & Technology , Shenyang Agricultural University , Shenyang , China
| | - Junping Wang
- a State Key Laboratory of Pathogen and Biosecurity , Institute of Microbiology and Epidemiology, Academy of Military Medical Sciences , Beijing , China.,d Urumqi Ethnic Cadres' College , Urumqi , China
| | - Qingyu Lv
- a State Key Laboratory of Pathogen and Biosecurity , Institute of Microbiology and Epidemiology, Academy of Military Medical Sciences , Beijing , China
| | - Hua Jiang
- a State Key Laboratory of Pathogen and Biosecurity , Institute of Microbiology and Epidemiology, Academy of Military Medical Sciences , Beijing , China
| | - Yuling Zheng
- a State Key Laboratory of Pathogen and Biosecurity , Institute of Microbiology and Epidemiology, Academy of Military Medical Sciences , Beijing , China
| | - Maokai Xu
- a State Key Laboratory of Pathogen and Biosecurity , Institute of Microbiology and Epidemiology, Academy of Military Medical Sciences , Beijing , China
| | - Xuyu Zhou
- c CAS Key Laboratory of Pathogenic Microbiology and Immunology , Institute of Microbiology, Chinese Academy of Sciences , Beijing , China
| | - Huaijie Hao
- c CAS Key Laboratory of Pathogenic Microbiology and Immunology , Institute of Microbiology, Chinese Academy of Sciences , Beijing , China
| | - Yongqiang Jiang
- a State Key Laboratory of Pathogen and Biosecurity , Institute of Microbiology and Epidemiology, Academy of Military Medical Sciences , Beijing , China
| |
Collapse
|
31
|
Streptococcus suis small RNA rss04 contributes to the induction of meningitis by regulating capsule synthesis and by inducing biofilm formation in a mouse infection model. Vet Microbiol 2016; 199:111-119. [PMID: 28110777 DOI: 10.1016/j.vetmic.2016.12.034] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2016] [Revised: 12/26/2016] [Accepted: 12/27/2016] [Indexed: 12/30/2022]
Abstract
Streptococcus suis (SS) is an important pathogen for pigs, and it is also considered as a zoonotic agent for humans. Meningitis is one of the most common features of the infection caused by SS, but little is known about the mechanisms of SS meningitis. Recent studies have revealed that small RNAs (sRNAs) have emerged as key regulators of the virulence in several bacteria. In the previous study, we reported that SS sRNA rss04 was up-regulated in pig cerebrospinal fluid and contributes to SS virulence in a zebrafish infection model. Here, we show that rss04 facilitates SS invasion of mouse brain and lung in vivo. Label-free quantitation mass spectrometry analysis revealed that rss04 regulates transcriptional regulator CcpA and several virulence factors including LuxS. Transmission electron microscope and Dot-blot analyses indicated that rss04 represses capsular polysaccharide (CPS) production, which in turn facilitates SS adherence and invasion of mouse brain microvascular endothelial cells bEnd.3 in vitro and activates the mRNA expression of TLR2, CCL2, IL-6 and TNF-α in mouse brain in vivo at 12h post-infection. In addition, rss04 positively regulates SS biofilm formation. Survival analysis of infected mice showed that biofilm state in brain contributes to SS virulence by intracranial subarachnoidal route of infection. Together, our data reveal that SS sRNA rss04 contributes to the induction of meningitis by regulating the CPS synthesis and by inducing biofilm formation, thereby increasing the virulence in a mouse infection model. To our knowledge, rss04 represents the first bacterial sRNA that plays definitive roles in bacterial meningitis.
Collapse
|
32
|
Musyoki AM, Shi Z, Xuan C, Lu G, Qi J, Gao F, Zheng B, Zhang Q, Li Y, Haywood J, Liu C, Yan J, Shi Y, Gao GF. Structural and functional analysis of an anchorless fibronectin-binding protein FBPS from Gram-positive bacterium Streptococcus suis. Proc Natl Acad Sci U S A 2016; 113:13869-13874. [PMID: 27834729 PMCID: PMC5137682 DOI: 10.1073/pnas.1608406113] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
The anchorless fibronectin-binding proteins (FnBPs) are a group of important virulence factors for which the structures are not available and the functions are not well defined. In this study we performed comprehensive studies on a prototypic member of this group: the fibronectin-/fibrinogen-binding protein from Streptococcus suis (FBPS). The structures of the N- and C-terminal halves (FBPS-N and FBPS-C), which together cover the full-length protein in sequence, were solved at a resolution of 2.1 and 2.6 Å, respectively, and each was found to be composed of two domains with unique folds. Furthermore, we have elucidated the organization of these domains by small-angle X-ray scattering. We further showed that the fibronectin-binding site is located in FBPS-C and that FBPS promotes the adherence of S suis to host cells by attaching the bacteria via FBPS-N. Finally, we demonstrated that FBPS functions both as an adhesin, promoting S suis attachment to host cells, and as a bacterial factor, activating signaling pathways via β1 integrin receptors to induce chemokine production.
Collapse
Affiliation(s)
- Abednego Moki Musyoki
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, China
- University of the Chinese Academy of Sciences, Beijing 100049, China
| | - Zhongyu Shi
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, China
- University of the Chinese Academy of Sciences, Beijing 100049, China
| | - Chunling Xuan
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, China
- University of the Chinese Academy of Sciences, Beijing 100049, China
| | - Guangwen Lu
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, China
- West China Hospital Emergency Department (WCHED), State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Sichuan 610041, China
- Collaborative Innovation Center of Biotherapy, Chengdu, Sichuan 610041, China
| | - Jianxun Qi
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, China
| | - Feng Gao
- Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
| | - Beiwen Zheng
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, the First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310003, China
| | - Qiangmin Zhang
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, China
| | - Yan Li
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, China
| | - Joel Haywood
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, China
- University of the Chinese Academy of Sciences, Beijing 100049, China
| | - Cuihua Liu
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, China
| | - Jinghua Yan
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, China
| | - Yi Shi
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, China
- University of the Chinese Academy of Sciences, Beijing 100049, China
- Research Network of Immunity and Health, Beijing Institutes of Life Science, Chinese Academy of Sciences, Beijing 100101, China
| | - George F Gao
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, China;
- University of the Chinese Academy of Sciences, Beijing 100049, China
- Research Network of Immunity and Health, Beijing Institutes of Life Science, Chinese Academy of Sciences, Beijing 100101, China
- National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing 102206, China
| |
Collapse
|
33
|
Tenenbaum T, Asmat TM, Seitz M, Schroten H, Schwerk C. Biological activities of suilysin: role in Streptococcus suis pathogenesis. Future Microbiol 2016; 11:941-54. [PMID: 27357518 DOI: 10.2217/fmb-2016-0028] [Citation(s) in RCA: 56] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Streptococcus suis is an important swine and zoonotic pathogen equipped with several virulence factors. The pore-forming toxins are the most abundant bacterial toxins and classified as critical virulence (associated) factors of several pathogens. The role of suilysin (SLY), a pore-forming cholesterol-dependent cytolysin of S. suis, as a true virulence factor is under debate. Most of the bacterial toxins have been reported to modulate the host immune system to facilitate invasion and subsequent replication of bacteria within respective host cells. SLY has been demonstrated to play an important role in the pathogenesis of S. suis infection and inflammatory response in vitro and in vivo. This review highlights the contributions of SLY to the pathogenicity of S. suis. It will address its role during the development of S. suis meningitis in pigs, as well as humans, and discuss SLY as a potential vaccine candidate.
Collapse
Affiliation(s)
- Tobias Tenenbaum
- Pediatric Infectious Diseases, Department of Pediatrics, Medical Faculty Mannheim, Heidelberg University, Theodor-Kutzer-Ufer 1-3, Mannheim D-68167, Germany
| | - Tauseef M Asmat
- Center for Advanced Studies in Vaccinology and Biotechnology, Brewery Road, University of Balochistan, 87300 Quetta, Pakistan
| | - Maren Seitz
- Institute for Microbiology, University of Veterinary Medicine Hannover, Bischofsholer Damm 15, Hannover D-30173, Germany
| | - Horst Schroten
- Pediatric Infectious Diseases, Department of Pediatrics, Medical Faculty Mannheim, Heidelberg University, Theodor-Kutzer-Ufer 1-3, Mannheim D-68167, Germany
| | - Christian Schwerk
- Pediatric Infectious Diseases, Department of Pediatrics, Medical Faculty Mannheim, Heidelberg University, Theodor-Kutzer-Ufer 1-3, Mannheim D-68167, Germany
| |
Collapse
|
34
|
Seele J, Nau R, Prajeeth CK, Stangel M, Valentin-Weigand P, Seitz M. Astrocytes Enhance Streptococcus suis-Glial Cell Interaction in Primary Astrocyte-Microglial Cell Co-Cultures. Pathogens 2016; 5:pathogens5020043. [PMID: 27304968 PMCID: PMC4931394 DOI: 10.3390/pathogens5020043] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2016] [Revised: 05/28/2016] [Accepted: 06/07/2016] [Indexed: 01/09/2023] Open
Abstract
Streptococcus (S.) suis infections are the most common cause of meningitis in pigs. Moreover, S. suis is a zoonotic pathogen, which can lead to meningitis in humans, mainly in adults. We assume that glial cells may play a crucial role in host-pathogen interactions during S. suis infection of the central nervous system. Glial cells are considered to possess important functions during inflammation and injury of the brain in bacterial meningitis. In the present study, we established primary astrocyte-microglial cell co-cultures to investigate interactions of S. suis with glial cells. For this purpose, microglial cells and astrocytes were isolated from new-born mouse brains and characterized by flow cytometry, followed by the establishment of astrocyte and microglial cell mono-cultures as well as astrocyte-microglial cell co-cultures. In addition, we prepared microglial cell mono-cultures co-incubated with uninfected astrocyte mono-culture supernatants and astrocyte mono-cultures co-incubated with uninfected microglial cell mono-culture supernatants. After infection of the different cell cultures with S. suis, bacteria-cell association was mainly observed with microglial cells and most prominently with a non-encapsulated mutant of S. suis. A time-dependent induction of NO release was found only in the co-cultures and after co-incubation of microglial cells with uninfected supernatants of astrocyte mono-cultures mainly after infection with the capsular mutant. Only moderate cytotoxic effects were found in co-cultured glial cells after infection with S. suis. Taken together, astrocytes and astrocyte supernatants increased interaction of microglial cells with S. suis. Astrocyte-microglial cell co-cultures are suitable to study S. suis infections and bacteria-cell association as well as NO release by microglial cells was enhanced in the presence of astrocytes.
Collapse
Affiliation(s)
- Jana Seele
- Center for Infection Medicine, Institute for Microbiology, University of Veterinary Medicine Hannover, Bischofsholer Damm 15, Hannover 30173, Germany.
- Institute for Neuropathology, University Medical Center Göttingen, Robert-Koch-Straße 40, Göttingen 37099, Germany.
- Department of Geriatrics, Evangelisches Krankenhaus Göttingen-Weende, An der Lutter 24, Göttingen 37075, Germany.
| | - Roland Nau
- Institute for Neuropathology, University Medical Center Göttingen, Robert-Koch-Straße 40, Göttingen 37099, Germany.
- Department of Geriatrics, Evangelisches Krankenhaus Göttingen-Weende, An der Lutter 24, Göttingen 37075, Germany.
| | - Chittappen K Prajeeth
- Department of Neurology, Center for Systems Neuroscience (ZSN), Hannover Medical School, Carl-Neuberg-Straße 1, Hannover 30625, Germany.
| | - Martin Stangel
- Department of Neurology, Center for Systems Neuroscience (ZSN), Hannover Medical School, Carl-Neuberg-Straße 1, Hannover 30625, Germany.
| | - Peter Valentin-Weigand
- Center for Infection Medicine, Institute for Microbiology, University of Veterinary Medicine Hannover, Bischofsholer Damm 15, Hannover 30173, Germany.
| | - Maren Seitz
- Center for Infection Medicine, Institute for Microbiology, University of Veterinary Medicine Hannover, Bischofsholer Damm 15, Hannover 30173, Germany.
| |
Collapse
|
35
|
Sun Y, Li N, Zhang J, Liu H, Liu J, Xia X, Sun C, Feng X, Gu J, Du C, Han W, Lei L. Enolase of Streptococcus Suis Serotype 2 Enhances Blood-Brain Barrier Permeability by Inducing IL-8 Release. Inflammation 2016; 39:718-726. [PMID: 26732390 DOI: 10.1007/s10753-015-0298-7] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Streptococcus suis serotype 2 (SS2) is an emerging zoonosis, and meningitis is the most frequent clinical manifestation, but mechanism of its virulent factor, enolase (Eno), is unknown in meningitis. In this study, Eno was inducibly expressed and added to an in vitro Transwell co-culture model of the blood-brain barrier (BBB) consisted of porcine brain microvascular endothelial cells (PBMECs) and astrocytes (ACs), the results showed that Eno induces a significant increase in BBB permeability and promotes the release of IL-8 et al. cytokines. Furthermore, IL-8 could significantly destroy the integrity of the BBB model in vitro. In mice models administered Eno for 24 h, Eno could significantly promote Evans blue (EB) moving from the blood to the brain and significantly increased the serum and brain levels of IL-8, as detected by ELISA. While G31P (IL-8 receptor antagonist) significantly decreased the concentration of EB in the brains of mice injected with Eno. The present study demonstrated that SS2 Eno may play an important role in disrupting BBB integrity by prompting IL-8 release.
Collapse
Affiliation(s)
- Yingying Sun
- College of Veterinary Medicine, Jilin University, Xi'an Road 5333, Changchun, 130062, China
| | - Na Li
- College of Veterinary Medicine, Jilin University, Xi'an Road 5333, Changchun, 130062, China
| | - Jing Zhang
- Changchun University of Chinese Medicine, Changchun, People's Republic of China
| | - Hongtao Liu
- College of Veterinary Medicine, Jilin University, Xi'an Road 5333, Changchun, 130062, China
| | - Jianfang Liu
- College of Veterinary Medicine, Jilin University, Xi'an Road 5333, Changchun, 130062, China
| | - Xiaojing Xia
- College of Veterinary Medicine, Jilin University, Xi'an Road 5333, Changchun, 130062, China
| | - Changjiang Sun
- College of Veterinary Medicine, Jilin University, Xi'an Road 5333, Changchun, 130062, China
| | - Xin Feng
- College of Veterinary Medicine, Jilin University, Xi'an Road 5333, Changchun, 130062, China
| | - Jingmin Gu
- College of Veterinary Medicine, Jilin University, Xi'an Road 5333, Changchun, 130062, China
| | - Chongtao Du
- College of Veterinary Medicine, Jilin University, Xi'an Road 5333, Changchun, 130062, China
| | - Wenyu Han
- College of Veterinary Medicine, Jilin University, Xi'an Road 5333, Changchun, 130062, China
| | - Liancheng Lei
- College of Veterinary Medicine, Jilin University, Xi'an Road 5333, Changchun, 130062, China.
| |
Collapse
|
36
|
Auger JP, Christodoulides M, Segura M, Xu J, Gottschalk M. Interactions of Streptococcus suis serotype 2 with human meningeal cells and astrocytes. BMC Res Notes 2015; 8:607. [PMID: 26502903 PMCID: PMC4624383 DOI: 10.1186/s13104-015-1581-2] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2015] [Accepted: 10/14/2015] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND Streptococcus suis serotype 2 is an important porcine pathogen and emerging zoonotic agent responsible for meningitis, of which different sequence types predominate worldwide. Though bacterial meningitis is defined as an exacerbated inflammation of the meninges, the underlying astrocytes of the glia limitans superficialis may also be implicated. However, the interactions between this pathogen and human meningeal cells or astrocytes remain unknown. Furthermore, the roles of well-described virulence factors (capsular polysaccharide, suilysin and cell wall modifications) in these interactions have yet to be studied. Consequently, the interactions between S. suis serotype 2 and human meningeal cells or astrocytes were evaluated for the first time in order to better understand their involvement during meningitis in humans. RESULTS Streptococcus suis serotype 2 adhered to human meningeal cells and astrocytes; invasion of meningeal cells was rare however, whereas invasion of astrocytes was generally more frequent. Regardless of the interaction or cell type, differences were not observed between sequence types. Though the capsular polysaccharide modulated the adhesion to and invasion of meningeal cells and astrocytes, the suilysin and cell wall modifications only influenced astrocyte invasion. Surprising, S. suis serotype 2 induced little or no inflammatory response from both cell types, but this absence of inflammatory response was probably not due to S. suis-induced cell death. CONCLUSIONS Though S. suis serotype 2 interacted with human meningeal cells and astrocytes, there was no correlation between sequence type and interaction. Consequently, the adhesion to and invasion of human meningeal cells and astrocytes are strain-specific characteristics. As such, the meningeal cells of the leptomeninges and the astrocytes of the glia limitans superficialis may not be directly implicated in the inflammatory response observed during meningitis in humans.
Collapse
Affiliation(s)
- Jean-Philippe Auger
- Faculty of Veterinary Medicine, Research Group on Infectious Diseases of Swine (GREMIP) and Swine and Poultry Infectious Diseases Research Center (CRIPA), University of Montreal, 3200 Sicotte Street, Saint-Hyacinthe, QC, J2S 2M2, Canada.
| | - Myron Christodoulides
- Neisseria Research Group, Molecular Microbiology, Clinical and Experimental Sciences, University of Southampton Faculty of Medicine, Southampton, UK.
| | - Mariela Segura
- Faculty of Veterinary Medicine, Research Group on Infectious Diseases of Swine (GREMIP) and Swine and Poultry Infectious Diseases Research Center (CRIPA), University of Montreal, 3200 Sicotte Street, Saint-Hyacinthe, QC, J2S 2M2, Canada.
| | - Jianguo Xu
- Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China.
| | - Marcelo Gottschalk
- Faculty of Veterinary Medicine, Research Group on Infectious Diseases of Swine (GREMIP) and Swine and Poultry Infectious Diseases Research Center (CRIPA), University of Montreal, 3200 Sicotte Street, Saint-Hyacinthe, QC, J2S 2M2, Canada.
| |
Collapse
|
37
|
Wang J, Kong D, Zhang S, Jiang H, Zheng Y, Zang Y, Hao H, Jiang Y. Interaction of fibrinogen and muramidase-released protein promotes the development of Streptococcus suis meningitis. Front Microbiol 2015; 6:1001. [PMID: 26441928 PMCID: PMC4585153 DOI: 10.3389/fmicb.2015.01001] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2015] [Accepted: 09/07/2015] [Indexed: 01/30/2023] Open
Abstract
Muramidase-released protein (MRP) is as an important virulence marker of Streptococcus suis (S. suis) serotype 2. Our previous works have shown that MRP can bind human fibrinogen (hFg); however, the function of this interaction in S. suis meningitis is not known. In this study, we found that the deletion of mrp significantly impairs the hFg-mediated adherence and traversal ability of S. suis across human cerebral microvascular endothelial cells (hCMEC/D3). Measurement of the permeability to Lucifer yellow in vitro and Evans blue extravasation in vivo show that the MRP-hFg interaction significantly increases the permeability of the blood–brain barrier (BBB). In the mouse meningitis model, wild type S. suis caused higher bacterial loads in the brain and more severe histopathological signs of meningitis than the mrp mutant at day 3 post-infection. Western blot analysis and immunofluorescence observations reveal that the MRP-hFg interaction can destroy the cell adherens junction protein p120-catenin of hCMEC/D3. These results indicate that the MRP-hFg interaction is important in the development of S. suis meningitis.
Collapse
Affiliation(s)
- Junping Wang
- State Key Laboratory of Pathogen and Biosecurity, Institute of Microbiology and Epidemiology, Academy of Military Medical Sciences , Beijing, China ; Urumqi Ethnic Cadres' College , Urumqi, China
| | - Decong Kong
- State Key Laboratory of Pathogen and Biosecurity, Institute of Microbiology and Epidemiology, Academy of Military Medical Sciences , Beijing, China
| | - Shengwei Zhang
- State Key Laboratory of Pathogen and Biosecurity, Institute of Microbiology and Epidemiology, Academy of Military Medical Sciences , Beijing, China
| | - Hua Jiang
- State Key Laboratory of Pathogen and Biosecurity, Institute of Microbiology and Epidemiology, Academy of Military Medical Sciences , Beijing, China
| | - Yuling Zheng
- State Key Laboratory of Pathogen and Biosecurity, Institute of Microbiology and Epidemiology, Academy of Military Medical Sciences , Beijing, China
| | - Yating Zang
- State Key Laboratory of Pathogen and Biosecurity, Institute of Microbiology and Epidemiology, Academy of Military Medical Sciences , Beijing, China
| | - Huaijie Hao
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences , Beijing, China
| | - Yongqiang Jiang
- State Key Laboratory of Pathogen and Biosecurity, Institute of Microbiology and Epidemiology, Academy of Military Medical Sciences , Beijing, China
| |
Collapse
|
38
|
Novel variant serotype of streptococcus suis isolated from piglets with meningitis. Appl Environ Microbiol 2014; 81:976-85. [PMID: 25416757 DOI: 10.1128/aem.02962-14] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Streptococcus suis is an emerging zoonotic pathogen causing severe infections in pigs and humans. In previous studies, 33 serotypes of S. suis have been identified using serum agglutination. Here, we describe a novel S. suis strain, CZ130302, isolated from an outbreak of acute piglet meningitis in eastern China. Strong pathogenicity of meningitis caused by strain CZ130302 was reproduced in the BALB/c mouse model. The strain showed a high fatality rate (8/10), higher than those for known virulent serotype 2 strains P1/7 (1/10) and 9801 (2/10). Cell adhesion assay results with bEnd.3 and HEp2 cells showed that CZ130302 was significantly close to P1/7 and 9801. Both the agglutination test and its complementary test showed that strain CZ130302 had no strong cross-reaction with the other 33 S. suis serotypes. The multiplex PCR assays revealed no specified bands for all four sets used to detect the other 33 serotypes. In addition, genetic analysis of the whole cps gene clusters of all serotypes was performed in this study. The results of comparative genomics showed that the cps gene cluster of CZ130302, which was not previously reported, showed no homology to the gene sequences of the other strains. Especially, the wzy, wzx, and acetyltransferase genes of strain CZ130302 are phylogenetically distinct from strains of the other 33 serotypes. Therefore, this study suggested that strain CZ130302 represents a novel variant serotype of S. suis (designated serotype Chz) which has a high potential to be virulent and associated with meningitis in animals.
Collapse
|
39
|
Dando SJ, Mackay-Sim A, Norton R, Currie BJ, St John JA, Ekberg JAK, Batzloff M, Ulett GC, Beacham IR. Pathogens penetrating the central nervous system: infection pathways and the cellular and molecular mechanisms of invasion. Clin Microbiol Rev 2014; 27:691-726. [PMID: 25278572 PMCID: PMC4187632 DOI: 10.1128/cmr.00118-13] [Citation(s) in RCA: 302] [Impact Index Per Article: 27.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
The brain is well protected against microbial invasion by cellular barriers, such as the blood-brain barrier (BBB) and the blood-cerebrospinal fluid barrier (BCSFB). In addition, cells within the central nervous system (CNS) are capable of producing an immune response against invading pathogens. Nonetheless, a range of pathogenic microbes make their way to the CNS, and the resulting infections can cause significant morbidity and mortality. Bacteria, amoebae, fungi, and viruses are capable of CNS invasion, with the latter using axonal transport as a common route of infection. In this review, we compare the mechanisms by which bacterial pathogens reach the CNS and infect the brain. In particular, we focus on recent data regarding mechanisms of bacterial translocation from the nasal mucosa to the brain, which represents a little explored pathway of bacterial invasion but has been proposed as being particularly important in explaining how infection with Burkholderia pseudomallei can result in melioidosis encephalomyelitis.
Collapse
Affiliation(s)
- Samantha J Dando
- Institute for Glycomics, Griffith University, Gold Coast, Queensland, Australia
| | - Alan Mackay-Sim
- Eskitis Institute for Drug Discovery, Griffith University, Brisbane, Queensland, Australia
| | - Robert Norton
- Townsville Hospital, Townsville, Queensland, Australia
| | - Bart J Currie
- Menzies School of Health Research and Royal Darwin Hospital, Darwin, Northern Territory, Australia
| | - James A St John
- Eskitis Institute for Drug Discovery, Griffith University, Brisbane, Queensland, Australia
| | - Jenny A K Ekberg
- Eskitis Institute for Drug Discovery, Griffith University, Brisbane, Queensland, Australia School of Biomedical Sciences, Queensland University of Technology, Brisbane, Queensland, Australia
| | - Michael Batzloff
- Institute for Glycomics, Griffith University, Gold Coast, Queensland, Australia
| | - Glen C Ulett
- School of Medical Science and Griffith Health Institute, Griffith University, Gold Coast, Queensland, Australia
| | - Ifor R Beacham
- Institute for Glycomics, Griffith University, Gold Coast, Queensland, Australia
| |
Collapse
|
40
|
Dang Y, Lachance C, Wang Y, Gagnon CA, Savard C, Segura M, Grenier D, Gottschalk M. Transcriptional approach to study porcine tracheal epithelial cells individually or dually infected with swine influenza virus and Streptococcus suis. BMC Vet Res 2014; 10:86. [PMID: 24708855 PMCID: PMC4022123 DOI: 10.1186/1746-6148-10-86] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2013] [Accepted: 03/31/2014] [Indexed: 11/16/2022] Open
Abstract
Background Swine influenza is a highly contagious viral infection in pigs affecting the respiratory tract that can have significant economic impacts. Streptococcus suis serotype 2 is one of the most important post-weaning bacterial pathogens in swine causing different infections, including pneumonia. Both pathogens are important contributors to the porcine respiratory disease complex. Outbreaks of swine influenza virus with a significant level of co-infections due to S. suis have lately been reported. In order to analyze, for the first time, the transcriptional host response of swine tracheal epithelial (NPTr) cells to H1N1 swine influenza virus (swH1N1) infection, S. suis serotype 2 infection and a dual infection, we carried out a comprehensive gene expression profiling using a microarray approach. Results Gene clustering showed that the swH1N1 and swH1N1/S. suis infections modified the expression of genes in a similar manner. Additionally, infection of NPTr cells by S. suis alone resulted in fewer differentially expressed genes compared to mock-infected cells. However, some important genes coding for inflammatory mediators such as chemokines, interleukins, cell adhesion molecules, and eicosanoids were significantly upregulated in the presence of both pathogens compared to infection with each pathogen individually. This synergy may be the consequence, at least in part, of an increased bacterial adhesion/invasion of epithelial cells previously infected by swH1N1, as recently reported. Conclusion Influenza virus would replicate in the respiratory epithelium and induce an inflammatory infiltrate comprised of mononuclear cells and neutrophils. In a co-infection situation, although these cells would be unable to phagocyte and kill S. suis, they are highly activated by this pathogen. S. suis is not considered a primary pulmonary pathogen, but an exacerbated production of proinflammatory mediators during a co-infection with influenza virus may be important in the pathogenesis and clinical outcome of S. suis-induced respiratory diseases.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Marcelo Gottschalk
- Faculté de Médecine Vétérinaire, Université de Montréal, 3200 Sicotte, St-Hyacinthe, J2S 2M2 Québec, Canada.
| |
Collapse
|
41
|
Role of capsule and suilysin in mucosal infection of complement-deficient mice with Streptococcus suis. Infect Immun 2014; 82:2460-71. [PMID: 24686060 DOI: 10.1128/iai.00080-14] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
Virulent Streptococcus suis serotype 2 strains are invasive extracellular bacteria causing septicemia and meningitis in piglets and humans. One objective of this study was to elucidate the function of complement in innate immune defense against S. suis. Experimental infection of wild-type (WT) and C3(-/-) mice demonstrated for the first time that the complement system protects naive mice against invasive mucosal S. suis infection. S. suis WT but not an unencapsulated mutant caused mortality associated with meningitis and other pathologies in C3(-/-) mice. The capsule contributed also substantially to colonization of the upper respiratory tract. Experimental infection of C3(-/-) mice with a suilysin mutant indicated that suilysin expression facilitated an early disease onset and the pathogenesis of meningitis. Flow cytometric analysis revealed C3 antigen deposition on the surface of ca. 40% of S. suis WT bacteria after opsonization with naive WT mouse serum, although to a significantly lower intensity than on the unencapsulated mutant. Ex vivo multiplication in murine WT and C3(-/-) blood depended on capsule but not suilysin expression. Interestingly, S. suis invasion of inner organs was also detectable in C5aR(-/-) mice, suggesting that chemotaxis and activation of immune cells via the anaphylatoxin receptor C5aR is, in addition to opsonization, a further important function of the complement system in defense against mucosal S. suis infection. In conclusion, we unequivocally demonstrate here the importance of complement against mucosal S. suis serotype 2 infection and that the capsule of this pathogen is also involved in escape from complement-independent immunity.
Collapse
|
42
|
Zhu H, Zhou J, Ni Y, Yu Z, Mao A, Hu Y, Wang W, Zhang X, Wen L, Li B, Wang X, Yu Y, Lv L, Guo R, Lu C, He K. Contribution of eukaryotic-type serine/threonine kinase to stress response and virulence of Streptococcus suis. PLoS One 2014; 9:e91971. [PMID: 24637959 PMCID: PMC3956855 DOI: 10.1371/journal.pone.0091971] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2013] [Accepted: 02/16/2014] [Indexed: 11/18/2022] Open
Abstract
Streptococcus suis serotype 2 (SS2) is an important swine and human pathogen responsible for septicemia and meningitis. The bacterial homologues of eukaryotic-type serine/threonine kinases (ESTKs) have been reported to play critical roles in various cellular processes. To investigate the role of STK in SS2, an isogenic stk mutant strain (Δstk) and a complemented strain (CΔstk) were constructed. The Δstk showed a significant decrease in adherence to HEp-2 cells, compared with the wild-type strain, and a reduced survival ratio in whole blood. In addition, the Δstk exhibited a notable reduced tolerance of environmental stresses including high temperature, acidic pH, oxidative stress, and high osmolarity. More importantly, the Δstk was attenuated in both the CD1 mouse and piglet models of infection. The results of quantitative reverse transcription-PCR (qRT-PCR) analysis indicated that the expressions of a few genes involving in adherence, stress response and virulence were clearly decreased in the Δstk mutant strain. Our data suggest that SsSTK is required for virulence and stress response in SS2.
Collapse
Affiliation(s)
- Haodan Zhu
- Institute of Veterinary Medicine, Jiangsu Academy of Agricultural Sciences, Key Laboratory of Veterinary Biological Engineering and Technology of Ministry of Agriculture, National Center for Engineering Research of Veterinary Bio-products, Nanjing, China
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, China
| | - Junming Zhou
- Institute of Veterinary Medicine, Jiangsu Academy of Agricultural Sciences, Key Laboratory of Veterinary Biological Engineering and Technology of Ministry of Agriculture, National Center for Engineering Research of Veterinary Bio-products, Nanjing, China
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, China
| | - Yanxiu Ni
- Institute of Veterinary Medicine, Jiangsu Academy of Agricultural Sciences, Key Laboratory of Veterinary Biological Engineering and Technology of Ministry of Agriculture, National Center for Engineering Research of Veterinary Bio-products, Nanjing, China
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, China
| | - Zhengyu Yu
- Institute of Veterinary Medicine, Jiangsu Academy of Agricultural Sciences, Key Laboratory of Veterinary Biological Engineering and Technology of Ministry of Agriculture, National Center for Engineering Research of Veterinary Bio-products, Nanjing, China
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, China
| | - Aihua Mao
- Institute of Veterinary Medicine, Jiangsu Academy of Agricultural Sciences, Key Laboratory of Veterinary Biological Engineering and Technology of Ministry of Agriculture, National Center for Engineering Research of Veterinary Bio-products, Nanjing, China
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, China
| | - Yiyi Hu
- Institute of Veterinary Medicine, Jiangsu Academy of Agricultural Sciences, Key Laboratory of Veterinary Biological Engineering and Technology of Ministry of Agriculture, National Center for Engineering Research of Veterinary Bio-products, Nanjing, China
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, China
| | - Wei Wang
- Institute of Veterinary Medicine, Jiangsu Academy of Agricultural Sciences, Key Laboratory of Veterinary Biological Engineering and Technology of Ministry of Agriculture, National Center for Engineering Research of Veterinary Bio-products, Nanjing, China
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, China
| | - Xuehan Zhang
- Institute of Veterinary Medicine, Jiangsu Academy of Agricultural Sciences, Key Laboratory of Veterinary Biological Engineering and Technology of Ministry of Agriculture, National Center for Engineering Research of Veterinary Bio-products, Nanjing, China
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, China
| | - Libin Wen
- Institute of Veterinary Medicine, Jiangsu Academy of Agricultural Sciences, Key Laboratory of Veterinary Biological Engineering and Technology of Ministry of Agriculture, National Center for Engineering Research of Veterinary Bio-products, Nanjing, China
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, China
| | - Bin Li
- Institute of Veterinary Medicine, Jiangsu Academy of Agricultural Sciences, Key Laboratory of Veterinary Biological Engineering and Technology of Ministry of Agriculture, National Center for Engineering Research of Veterinary Bio-products, Nanjing, China
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, China
| | - Xiaomin Wang
- Institute of Veterinary Medicine, Jiangsu Academy of Agricultural Sciences, Key Laboratory of Veterinary Biological Engineering and Technology of Ministry of Agriculture, National Center for Engineering Research of Veterinary Bio-products, Nanjing, China
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, China
| | - Yang Yu
- Institute of Veterinary Medicine, Jiangsu Academy of Agricultural Sciences, Key Laboratory of Veterinary Biological Engineering and Technology of Ministry of Agriculture, National Center for Engineering Research of Veterinary Bio-products, Nanjing, China
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, China
| | - Lixin Lv
- Institute of Veterinary Medicine, Jiangsu Academy of Agricultural Sciences, Key Laboratory of Veterinary Biological Engineering and Technology of Ministry of Agriculture, National Center for Engineering Research of Veterinary Bio-products, Nanjing, China
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, China
| | - Rongli Guo
- Institute of Veterinary Medicine, Jiangsu Academy of Agricultural Sciences, Key Laboratory of Veterinary Biological Engineering and Technology of Ministry of Agriculture, National Center for Engineering Research of Veterinary Bio-products, Nanjing, China
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, China
| | - Chengping Lu
- Key Lab of Animal Bacteriology, Ministry of Agriculture, Nanjing Agricultural University, Nanjing, China
| | - Kongwang He
- Institute of Veterinary Medicine, Jiangsu Academy of Agricultural Sciences, Key Laboratory of Veterinary Biological Engineering and Technology of Ministry of Agriculture, National Center for Engineering Research of Veterinary Bio-products, Nanjing, China
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, China
- * E-mail:
| |
Collapse
|
43
|
Guo CM, Chen RR, Kalhoro DH, Wang ZF, Liu GJ, Lu CP, Liu YJ. Identification of genes preferentially expressed by highly virulent piscine Streptococcus agalactiae upon interaction with macrophages. PLoS One 2014; 9:e87980. [PMID: 24498419 PMCID: PMC3912197 DOI: 10.1371/journal.pone.0087980] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2013] [Accepted: 12/31/2013] [Indexed: 11/19/2022] Open
Abstract
Streptococcus agalactiae, long recognized as a mammalian pathogen, is an emerging concern with regard to fish. In this study, we used a mouse model and in vitro cell infection to evaluate the pathogenetic characteristics of S. agalactiae GD201008-001, isolated from tilapia in China. This bacterium was found to be highly virulent and capable of inducing brain damage by migrating into the brain by crossing the blood–brain barrier (BBB). The phagocytosis assays indicated that this bacterium could be internalized by murine macrophages and survive intracellularly for more than 24 h, inducing injury to macrophages. Further, selective capture of transcribed sequences (SCOTS) was used to investigate microbial gene expression associated with intracellular survival. This positive cDNA selection technique identified 60 distinct genes that could be characterized into 6 functional categories. More than 50% of the differentially expressed genes were involved in metabolic adaptation. Some genes have previously been described as associated with virulence in other bacteria, and four showed no significant similarities to any other previously described genes. This study constitutes the first step in further gene expression analyses that will lead to a better understanding of the molecular mechanisms used by S. agalactiae to survive in macrophages and to cross the BBB.
Collapse
Affiliation(s)
- Chang-Ming Guo
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Rong-Rong Chen
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | | | - Zhao-Fei Wang
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Guang-Jin Liu
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Cheng-Ping Lu
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Yong-Jie Liu
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
- * E-mail:
| |
Collapse
|
44
|
Capsular sialic acid of Streptococcus suis serotype 2 binds to swine influenza virus and enhances bacterial interactions with virus-infected tracheal epithelial cells. Infect Immun 2013; 81:4498-508. [PMID: 24082069 DOI: 10.1128/iai.00818-13] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Streptococcus suis serotype 2 is an important swine bacterial pathogen, and it is also an emerging zoonotic agent. It is unknown how S. suis virulent strains, which are usually found in low quantities in pig tonsils, manage to cross the first host defense lines to initiate systemic disease. Influenza virus produces a contagious infection in pigs which is frequently complicated by bacterial coinfections, leading to significant economic impacts. In this study, the effect of a preceding swine influenza H1N1 virus (swH1N1) infection of swine tracheal epithelial cells (NTPr) on the ability of S. suis serotype 2 to adhere to, invade, and activate these cells was evaluated. Cells preinfected with swH1N1 showed bacterial adhesion and invasion levels that were increased more than 100-fold compared to those of normal cells. Inhibition studies confirmed that the capsular sialic acid moiety is responsible for the binding to virus-infected cell surfaces. Also, preincubation of S. suis with swH1N1 significantly increased bacterial adhesion to/invasion of epithelial cells, suggesting that S. suis also uses swH1N1 as a vehicle to invade epithelial cells when the two infections occur simultaneously. Influenza virus infection may facilitate the transient passage of S. suis at the respiratory tract to reach the bloodstream and cause bacteremia and septicemia. S. suis may also increase the local inflammation at the respiratory tract during influenza infection, as suggested by an exacerbated expression of proinflammatory mediators in coinfected cells. These results give new insight into the complex interactions between influenza virus and S. suis in a coinfection model.
Collapse
|
45
|
Kouki A, Pieters RJ, Nilsson UJ, Loimaranta V, Finne J, Haataja S. Bacterial Adhesion of Streptococcus suis to Host Cells and Its Inhibition by Carbohydrate Ligands. BIOLOGY 2013; 2:918-35. [PMID: 24833053 PMCID: PMC3960878 DOI: 10.3390/biology2030918] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/02/2013] [Revised: 05/22/2013] [Accepted: 05/22/2013] [Indexed: 11/16/2022]
Abstract
Streptococcus suis is a Gram-positive bacterium, which causes sepsis and meningitis in pigs and humans. This review examines the role of known S. suis virulence factors in adhesion and S. suis carbohydrate-based adhesion mechanisms, as well as the inhibition of S. suis adhesion by anti-adhesion compounds in in vitro assays. Carbohydrate-binding specificities of S. suis have been identified, and these studies have shown that many strains recognize Galα1-4Gal-containing oligosaccharides present in host glycolipids. In the era of increasing antibiotic resistance, new means to treat infections are needed. Since microbial adhesion to carbohydrates is important to establish disease, compounds blocking adhesion could be an alternative to antibiotics. The use of oligosaccharides as drugs is generally hampered by their relatively low affinity (micromolar) to compete with multivalent binding to host receptors. However, screening of a library of chemically modified Galα1-4Gal derivatives has identified compounds that inhibit S. suis adhesion in nanomolar range. Also, design of multivalent Galα1-4Gal-containing dendrimers has resulted in a significant increase of the inhibitory potency of the disaccharide. The S. suis adhesin binding to Galα1-4Gal-oligosaccharides, Streptococcal adhesin P (SadP), was recently identified. It has a Galα1-4Gal-binding N-terminal domain and a C-terminal LPNTG-motif for cell wall anchoring. The carbohydrate-binding domain has no homology to E. coli P fimbrial adhesin, which suggests that these Gram-positive and Gram-negative bacterial adhesins recognizing the same receptor have evolved by convergent evolution. SadP adhesin may represent a promising target for the design of anti-adhesion ligands for the prevention and treatment of S. suis infections.
Collapse
Affiliation(s)
- Annika Kouki
- Department of Medical Biochemistry and Genetics, University of Turku, Kiinamyllynkatu 10, Turku FI-20520, Finland.
| | - Roland J Pieters
- Department of Medicinal Chemistry and Chemical Biology, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, P.O. Box 80082, Utrecht 3508 TB, The Netherlands.
| | - Ulf J Nilsson
- Centre for Analysis and Synthesis, Department of Chemistry, Lund University, POB 124, Lund SE-22100, Sweden.
| | - Vuokko Loimaranta
- Department of Medical Biochemistry and Genetics, University of Turku, Kiinamyllynkatu 10, Turku FI-20520, Finland.
| | - Jukka Finne
- Department of Biosciences, Division of Biochemistry and Biotechnology, University of Helsinki, P.O.B. 56, Helsinki FI-00014, Finland.
| | - Sauli Haataja
- Department of Medical Biochemistry and Genetics, University of Turku, Kiinamyllynkatu 10, Turku FI-20520, Finland.
| |
Collapse
|
46
|
Lachance C, Segura M, Gerber PP, Xu J, Gottschalk M. Toll-like receptor 2-independent host innate immune response against an epidemic strain of Streptococcus suis that causes a toxic shock-like syndrome in humans. PLoS One 2013; 8:e65031. [PMID: 23724118 PMCID: PMC3665724 DOI: 10.1371/journal.pone.0065031] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2013] [Accepted: 04/20/2013] [Indexed: 11/18/2022] Open
Abstract
Streptococcus suis is an emerging zoonotic agent causing meningitis and septicemia. Outbreaks in humans in China with atypical cases of streptococcal toxic shock-like syndrome have been described to be caused by a clonal epidemic S. suis strain characterized as sequence type (ST) 7 by multilocus sequence typing, different from the classical ST1 usually isolated in Europe. Previous in vitro studies showed that Toll-like receptor (TLR) 2 plays a major role in S. suis ST1 interactions with host cells. In the present study, the in vivo role of TLR2 in systemic infections caused by S. suis ST1 or ST7 strains using TLR2 deficient (TLR2(-/-)) mice was evaluated. TLR2-mediated recognition significantly contributes to the acute disease caused by the highly virulent S. suis ST1 strain, since the TLR2(-/-) mice remained unaffected when compared to wild type (WT) mice. The lack of mortality could not be associated with a lower bacterial burden; however, a significant decrease in the induction of pro-inflammatory mediators, as evaluated by microarray, real-time PCR and protein assays, was observed. On the other hand, TLR2(-/-) mice infected with the epidemic ST7 strain presented no significant differences regarding survival and expression of pro-inflammatory mediators when compared to the WT mice. Together, these results show a TLR2-independent host innate immune response to S. suis that depends on the strain.
Collapse
Affiliation(s)
- Claude Lachance
- Faculty of Veterinary Medicine, University of Montreal, Saint-Hyacinthe, Québec, Canada
| | - Mariela Segura
- Faculty of Veterinary Medicine, University of Montreal, Saint-Hyacinthe, Québec, Canada
| | - Pehuén Pereyra Gerber
- Faculty of Veterinary Medicine, University of Montreal, Saint-Hyacinthe, Québec, Canada
| | - Jianguo Xu
- State Key Laboratory for Infectious Disease Prevention and Control, National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Marcelo Gottschalk
- Faculty of Veterinary Medicine, University of Montreal, Saint-Hyacinthe, Québec, Canada
- * E-mail:
| |
Collapse
|
47
|
Use of tetravalent galabiose for inhibition of streptococcus suis serotype 2 infection in a mouse model. BIOLOGY 2013; 2:702-18. [PMID: 24832804 PMCID: PMC3960886 DOI: 10.3390/biology2020702] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/14/2013] [Revised: 03/26/2013] [Accepted: 03/28/2013] [Indexed: 11/24/2022]
Abstract
Streptococcus suis is an important swine pathogen associated with a variety of infections such as meningitis, arthritis and septicemia. The bacterium is zoonotic and has been found to cause meningitis especially in humans occupationally exposed to infected pigs. Since adhesion is a prerequisite for colonization and subsequent infection, anti-adhesion treatment seems a natural alternative to traditional treatment with antibiotics. In order to optimize the inhibitory potency a multivalency approach was taken in the inhibitor design. A synthetic tetravalent galabiose compound was chosen which had previously shown promising anti-adhesion effects with S. suis in vitro. The aim of this study was to evaluate the in vivo effects of the compound using an infection peritonitis mouse model. As such S. suis serotype 2 infection and treatment were tested in vivo and the effects were compared to the effect of treatment with penicillin.
Collapse
|
48
|
Lapirattanakul J, Nomura R, Nemoto H, Naka S, Ooshima T, Nakano K. Multilocus sequence typing of Streptococcus mutans strains with the cbm gene encoding a novel collagen-binding protein. Arch Oral Biol 2013; 58:989-96. [PMID: 23506778 DOI: 10.1016/j.archoralbio.2013.02.007] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2012] [Revised: 01/29/2013] [Accepted: 02/15/2013] [Indexed: 11/25/2022]
Abstract
OBJECTIVE Streptococcus mutans, an oral pathogen associated with infective endocarditis (IE), possesses two genes encoding collagen-binding proteins, namely cnm and cbm. In this study, we used multilocus sequence typing (MLST) of S. mutans with the cbm gene. DESIGN Forty-five S. mutans strains including 15 strains with the cnm gene, 15 strains with the cbm gene, and 15 strains without these two genes were analysed by MLST. In addition, the collagen-binding properties as well as the abilities to adhere to and invade human umbilical vein endothelial cells (HUVEC) were also evaluated for all strains. RESULTS In the groups of cnm-positive and cbm-positive strains, all properties, including collagen binding, adhesion, and invasion were significantly greater than those of the cnm-cbm-negative group. Moreover, MLST revealed three clonal complexes of S. mutans possessing the cbm gene. These three clones showed no close relatedness with clones of strains containing the cnm gene. Among three clones harbouring the cbm gene, two clones belong to serotype k, and appeared to be associated with the pathogenesis of IE due to their strong collagen binding and relatively enhanced abilities to adhere to and invade endothelial cells. However, such properties were relatively weak in the other non-serotype k clone possessing the cbm gene. CONCLUSIONS MLST indicated a difference in evolution between S. mutans strains with the cbm gene and those with the cnm gene. In addition, this technique also suggested the importance of cbm-positive S. mutans clones relative to the pathogenesis of IE.
Collapse
Affiliation(s)
- Jinthana Lapirattanakul
- Department of Oral Microbiology, Faculty of Dentistry, Mahidol University, Bangkok 10400, Thailand.
| | | | | | | | | | | |
Collapse
|
49
|
Li L, Zhu DK, Zhou Y, Wang MS, Cheng AC, Jia RY, Chen S, Liu F, Yang QM, Chen XY. Adhesion and invasion to duck embryo fibroblast cells by Riemerella anatipestifer. Poult Sci 2013; 91:3202-8. [PMID: 23155031 DOI: 10.3382/ps.2012-02552] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Here, we investigated adhesion and invasion of Riemerella anatipestifer (RA) to primary duck embryo fibroblast (DEF) cells. The ability of RA to adhere to, and more importantly, to invade DEF cells was demonstrated by using a gentamicin invasion assay and was confirmed by transmission electron microscopy (TEM). Adhesion of RA could be found by TEM after 1 h of inoculation. Both apoptosis and necrocytosis of DEF were indicated by TEM after 10 h of incubation, which suggested a complex mechanism of DEF cell death induced by RA. Our results showed that internalized RA had the ability to leave the DEF cells. Inhibition studies indicated that RA proteins play a role in adhesion. Moreover, invasion of RA to DEF cells was shown to require rearrangement of actin microfilaments and microtubular cytoskeletal elements. Because the adhesion and invasion ability of RA to DEF cells could be demonstrated in vitro, similar processes might occur in vivo, where DEF cells play a crucial role in the diffusion of RA in ducks.
Collapse
Affiliation(s)
- L Li
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Wenjiang, Chengdu City, Sichuan 611130, P.R. China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Fittipaldi N, Segura M, Grenier D, Gottschalk M. Virulence factors involved in the pathogenesis of the infection caused by the swine pathogen and zoonotic agent Streptococcus suis. Future Microbiol 2012; 7:259-79. [PMID: 22324994 DOI: 10.2217/fmb.11.149] [Citation(s) in RCA: 322] [Impact Index Per Article: 24.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Streptococcus suis is a major swine pathogen responsible for important economic losses to the swine industry worldwide. It is also an emerging zoonotic agent of meningitis and streptococcal toxic shock-like syndrome. Since the recent recognition of the high prevalence of S. suis human disease in southeast and east Asia, the interest of the scientific community in this pathogen has significantly increased. In the last few years, as a direct consequence of these intensified research efforts, large amounts of data on putative virulence factors have appeared in the literature. Although the presence of some proposed virulence factors does not necessarily define a S. suis strain as being virulent, several cell-associated or secreted factors are clearly important for the pathogenesis of the S. suis infection. In order to cause disease, S. suis must colonize the host, breach epithelial barriers, reach and survive in the bloodstream, invade different organs, and cause exaggerated inflammation. In this review, we discuss the potential contribution of different described S. suis virulence factors at each step of the pathogenesis of the infection. Finally, we briefly discuss other described virulence factors, virulence factor candidates and virulence markers for which a precise role at specific steps of the pathogenesis of the S. suis infection has not yet been clearly established.
Collapse
Affiliation(s)
- Nahuel Fittipaldi
- Groupe de Recherche sur les Maladies Infectieuses du Porc & Centre de Recherche en Infectiologie Porcine, Faculté de médecine vétérinaire, Université de Montréal, 3200 rue Sicotte, CP5000, St-Hyacinthe, Quebec, J2S 7C6, Canada
| | | | | | | |
Collapse
|