Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 469
Filter
1.
Vet Res ; 55(1): 119, 2024 Sep 27.
Article in English | MEDLINE | ID: mdl-39334407

ABSTRACT

Mobile genetic elements (MGEs) enable bacteria to acquire novel genes and traits. However, the functions of cargo genes within MGEs remain poorly understood. The cadmium resistance operon cadDX is present in many gram-positive bacteria. Although cadDX has been reported to be involved in metal detoxification, its regulatory mechanisms and functions in bacterial pathogenesis are poorly understood. This study revealed that cadDX contributes to cadmium resistance, oxidative stress resistance, and virulence in Streptococcus suis, an important zoonotic pathogen in pigs and humans. CadX represses cadD expression by binding to the cadDX promoter. Notably, cadX responds to H2O2 stress through an additional promoter within the cadDX operon, mitigating the harmful effect of excessive cadD expression during oxidative stress. cadDX resides within an 11 K integrative and mobilizable element that can autonomously form circular structures. Moreover, cadDX is found in diverse MGEs, accounting for its widespread distribution across various bacteria, especially among pathogenic streptococci. Transferring cadDX into another zoonotic pathogen, Streptococcus agalactiae, results in similar phenotypes, including resistance to cadmium and oxidative stresses and increased virulence of S. agalactiae in mice. The new functions and regulatory mechanisms of cadDX shed light on the importance of the cadDX system in driving evolutionary adaptations and survival strategies across diverse gram-positive bacteria.


Subject(s)
Cadmium , Operon , Oxidative Stress , Streptococcal Infections , Streptococcus suis , Virulence , Streptococcus suis/genetics , Streptococcus suis/pathogenicity , Streptococcus suis/drug effects , Streptococcus suis/physiology , Animals , Streptococcal Infections/veterinary , Streptococcal Infections/microbiology , Mice , Streptococcus agalactiae/physiology , Streptococcus agalactiae/genetics , Streptococcus agalactiae/pathogenicity , Streptococcus agalactiae/drug effects , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Drug Resistance, Bacterial/genetics
2.
Microbiology (Reading) ; 170(9)2024 Sep.
Article in English | MEDLINE | ID: mdl-39287974

ABSTRACT

The sit-and-wait hypothesis predicts that bacteria can become more virulent when they survive and transmit outside of their hosts due to circumventing the costs of host mortality. While this hypothesis is largely supported theoretically and through comparative analysis, experimental validation is limited. Here we test this hypothesis in Streptococcus suis, an opportunistic zoonotic pig pathogen, where a pathogenic ecotype proliferated during the change to intensive pig farming that amplifies opportunities for fomite transmission. We show in an in vitro environmental survival experiment that pathogenic ecotypes survive for longer than commensal ecotypes, despite similar rates of decline. The presence of a polysaccharide capsule has no consistent effect on survival. Our findings suggest that extended survival in the food chain may augment the zoonotic capability of S. suis. Moreover, eliminating the long-term environmental survival of bacteria could be a strategy that will both enhance infection control and curtail the evolution of virulence.


Subject(s)
Streptococcus suis , Animals , Swine , Streptococcus suis/pathogenicity , Streptococcus suis/genetics , Virulence , Streptococcal Infections/microbiology , Streptococcal Infections/veterinary , Streptococcal Infections/transmission , Microbial Viability , Swine Diseases/microbiology , Ecotype
3.
Virulence ; 15(1): 2401963, 2024 Dec.
Article in English | MEDLINE | ID: mdl-39282964

ABSTRACT

Streptococcus suis (S. suis) is an important swine bacterial pathogen and causes human infections, leading to a wide range of diseases. However, the role of 5'-nucleotidases in its virulence remains to be fully elucidated. Herein, we identified four cell wall-anchored 5'-nucleotidases (Snts) within S. suis, named SntA, SntB, SntC, and SntD, each displaying similar domains yet exhibiting low sequence homology. The malachite green reagent and HPLC assays demonstrated that these recombinant enzymes are capable of hydrolysing ATP, ADP, and AMP into adenosine (Ado), with the hierarchy of catalytic efficiency being SntC>SntB>SntA>SntD. Moreover, comprehensive enzymatic activity assays illustrated slight variances in substrate specificity, pH tolerance, and metal ion requirements, yet highlighted a conserved substrate-binding pocket, His-Asp catalytic dyad, metal, and phosphate-binding sites across Snts, with the exception of SntA. Through bactericidal assays and murine infection assays involving in site-mutagenesis strains, it was demonstrated that SntB and SntC collaboratively enhance bacterial survivability within whole blood and polymorphonuclear leukocytes (PMNs) via the Ado-A2aR pathway in vitro, and within murine blood and organs in vivo. This suggests a direct correlation between enzymatic activity and enhancement of bacterial survival and virulence. Collectively, S. suis 5'-nucleotidases additively contribute to the generation of adenosine, influencing susceptibility within blood and PMNs, and enhancing survival within blood and organs in vivo. This elucidation of their integral functions in the pathogenic process of S. suis not only enhances our comprehension of bacterial virulence mechanisms, but also illuminates new avenues for therapeutic intervention aimed at curbing S. suis infections.


Subject(s)
5'-Nucleotidase , Adenosine , Disease Models, Animal , Immune Evasion , Streptococcal Infections , Streptococcus suis , Animals , Streptococcus suis/pathogenicity , Streptococcus suis/enzymology , Streptococcus suis/immunology , Streptococcus suis/genetics , 5'-Nucleotidase/genetics , 5'-Nucleotidase/immunology , 5'-Nucleotidase/metabolism , Mice , Adenosine/metabolism , Virulence , Streptococcal Infections/microbiology , Streptococcal Infections/immunology , Female , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Bacterial Proteins/immunology , Neutrophils/immunology , Neutrophils/microbiology , Mice, Inbred BALB C , Substrate Specificity
4.
Vet Microbiol ; 297: 110212, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39111202

ABSTRACT

The complement system is the first defense line of the immune system. However, pathogens have evolved numerous strategies to evade complement attacks. Streptococcus suis is an important zoonotic bacterium, harmful to both the pig industry and human health. ApuA has been reported as a bifunctional amylopullulanase and also contributed to virulence of S. suis. Herein, we found that ApuA could activate both classical and alternative pathways of the complement system. Furthermore, by using bacterial two-hybrid, far-western blot and ELISA assays, it was confirmed that ApuA could interact with complement C3b. The interaction domain of ApuA with C3b was found to be its α-Amylase domain (ApuA_N). After construction of an apuA mutant (ΔapuA) and its complementary strain, it was found that compared to the wild-type strain (WT), ΔapuA had significantly increased C3b deposition and membrane attack complex formation. Additionally, ΔapuA showed significantly lower survival rates in human serum and blood and was more susceptible to engulfment by neutrophils and macrophages. Mice infected with ΔapuA had significantly higher survival rates and lower bacterial loads in their blood, lung and brains, compared to those infected with WT. In summary, this study identified ApuA as a novel factor involved in the complement evasion of S. suis and suggested its multifunctional role in the pathogenesis of S. suis.


Subject(s)
Bacterial Proteins , Complement C3b , Immune Evasion , Streptococcal Infections , Streptococcus suis , Streptococcus suis/pathogenicity , Streptococcus suis/genetics , Streptococcus suis/immunology , Streptococcus suis/enzymology , Animals , Complement C3b/immunology , Mice , Streptococcal Infections/immunology , Streptococcal Infections/veterinary , Streptococcal Infections/microbiology , Humans , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Bacterial Proteins/immunology , Glycoside Hydrolases/genetics , Glycoside Hydrolases/metabolism , Female , Virulence
5.
BMC Vet Res ; 20(1): 337, 2024 Jul 30.
Article in English | MEDLINE | ID: mdl-39080654

ABSTRACT

Streptococcus suis (S. suis) is an important gram-positive pathogen and an emerging zoonotic pathogen that causes meningitis in swine and humans. Although several virulence factors have been characterized in S. suis, the underlying mechanisms of pathogenesis are not fully understood. In this study, we identified Zinc metalloproteinase C (ZmpC) probably as a critical virulence factor widely distributed in S. suis strains. ZmpC was identified as a critical facilitator in the development of bacterial meningitis, as evidenced by the detection of increased expression of TNF-α, IL-8, and matrix metalloprotease 9 (MMP-9). Subcellular localization analysis further revealed that ZmpC was localized to the cell wall surface and gelatin zymography analysis showed that ZmpC could cleave human MMP-9. Mice challenge demonstrated that ZmpC provided protection against S. suis CZ130302 (serotype Chz) and ZY05719 (serotype 2) infection. In conclusion, these results reveal that ZmpC plays an important role in promoting CZ130302 to cause mouse meningitis and may be a potential candidate for a S. suis CZ130302 vaccine.


Subject(s)
Meningitis, Bacterial , Serogroup , Streptococcal Infections , Streptococcus suis , Swine Diseases , Streptococcus suis/pathogenicity , Streptococcus suis/enzymology , Animals , Streptococcal Infections/veterinary , Streptococcal Infections/microbiology , Swine , Swine Diseases/microbiology , Mice , Meningitis, Bacterial/veterinary , Meningitis, Bacterial/microbiology , Female , Virulence Factors/metabolism , Virulence Factors/genetics , Bacterial Proteins/metabolism , Bacterial Proteins/genetics , Humans , Matrix Metalloproteinase 9/metabolism , Matrix Metalloproteinase 9/genetics , Mice, Inbred BALB C , Metalloendopeptidases/metabolism , Metalloendopeptidases/genetics
6.
J Basic Microbiol ; 64(9): e2400030, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39031597

ABSTRACT

Streptococcus suis is an important zoonotic pathogen, causing cytokine storms of Streptococcal toxic shock-like syndrome amongst humans after a wound infection into the bloodstream. To overcome the challenges of fever and leukocyte recruitment, invasive S. suis must deploy multiple stress responses forming a network and utilize proteases to degrade short-lived regulatory and misfolded proteins induced by adverse stresses, thereby adapting and evading host immune responses. In this study, we found that S. suis encodes multiple ATP-dependent proteases, including single-chain FtsH and double-subunit Clp protease complexes ClpAP, ClpBP, ClpCP, and ClpXP, which were activated as the fever of infected mice in vivo. The expression of genes ftsH, clpA/B/C, and clpP, but not clpX, were significantly upregulated in S. suis in response to heat stress, while were not changed notably under the treatments with several other stresses, including oxidative, acidic, and cold stimulation. FtsH and ClpP were required for S. suis survival within host blood under heat stress in vitro and in vivo. Deletion of ftsH or clpP attenuated the tolerance of S. suis to heat, oxidative and acidic stresses, and significantly impaired the bacterial survival within macrophages. Further analysis identified that repressor CtsR directly binds and controls the clpA/B/C and clpP operons and is relieved by heat stress. In summary, the deployments of multiple ATP-dependent proteases form a flexible heat stress response network that appears to allow S. suis to fine-tune the degradation or refolding of the misfolded proteins to maintain cellular homeostasis and optimal survival during infection.


Subject(s)
Bacterial Proteins , Streptococcal Infections , Streptococcus suis , Streptococcus suis/enzymology , Streptococcus suis/genetics , Streptococcus suis/pathogenicity , Animals , Mice , Bacterial Proteins/metabolism , Bacterial Proteins/genetics , Streptococcal Infections/microbiology , Heat-Shock Response , ATP-Dependent Proteases/metabolism , ATP-Dependent Proteases/genetics , Gene Expression Regulation, Bacterial , Macrophages/microbiology , Macrophages/immunology , Endopeptidase Clp/metabolism , Endopeptidase Clp/genetics , Female
7.
Vet Microbiol ; 296: 110166, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38968694

ABSTRACT

Streptococcus suis (S. suis) disease is a prevalent zoonotic infectious threat that elicits a systemic inflammatory response in both swine and humans, frequently culminating in high mortality rates. The excessive inflammation triggered by S. suis infection can precipitate tissue damage and sudden death; however, a comprehensive strategy to mitigate this inflammatory response remains elusive. Our study examines the role of NLRP6 in S. suis infection, with a particular focus on its involvement in pathogen regulation. A marked upregulation of NLRP6 was observed in peritoneal macrophages post-infection with S. suis SC19 strain, consequently activating the NLRP6 inflammasome. Furthermore, SC19 infection was found to augment the secretion of pro-inflammatory cytokines IL-1ß via NLRP6 activation, while NLRP6 deficiency mitigates the invasion and adhesion of SC19 to macrophages. In vivo models revealed that NLRP6 deletion enhanced survival rates of SC19-infected mice, alongside a reduction in tissue bacterial load and inflammatory cytokine levels. NLRP6-/- mice were shown to exhibit attenuated inflammatory responses in pulmonary, hepatic, and splenic tissues post-SC19 infection, as evidenced by lower inflammation scores. Flow cytometry analyses further substantiated that NLRP6 is involved in modulating macrophage and neutrophil recruitment during infection. Our findings suggest that NLRP6 negatively regulates host resistance against S. suis infection; its absence results in reduced mortality, bacterial colonization, and a milder inflammatory response. Elucidating the mechanism of NLRP6 in S. suis-induced inflammation provides novel insights and theoretical underpinnings for the prophylaxis and therapeutics of S. suis diseases.


Subject(s)
Mice, Inbred C57BL , Streptococcal Infections , Streptococcus suis , Streptococcus suis/immunology , Streptococcus suis/pathogenicity , Streptococcus suis/genetics , Animals , Streptococcal Infections/immunology , Streptococcal Infections/microbiology , Mice , Mice, Knockout , Macrophages, Peritoneal/immunology , Macrophages, Peritoneal/microbiology , Inflammasomes/immunology , Inflammasomes/genetics , Cytokines/metabolism , Cytokines/genetics , Inflammation/immunology , Female , Interleukin-1beta/metabolism , Interleukin-1beta/genetics , Receptors, Cell Surface
8.
Microbiol Spectr ; 12(8): e0061224, 2024 Aug 06.
Article in English | MEDLINE | ID: mdl-38980021

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.


Subject(s)
Blood-Brain Barrier , Endothelial Cells , Hemolysin Proteins , Hemolysis , Interleukin-33 , Streptococcus suis , Blood-Brain Barrier/metabolism , Blood-Brain Barrier/microbiology , Animals , Mice , Interleukin-33/metabolism , Humans , Hemolysin Proteins/metabolism , Streptococcus suis/pathogenicity , Endothelial Cells/microbiology , Endothelial Cells/metabolism , Streptococcal Infections/microbiology , Interleukin-6/metabolism , Interleukin-6/genetics , Interleukin-8/metabolism , Swine , Matrix Metalloproteinase 9/metabolism
9.
J Infect Dis ; 230(1): 188-197, 2024 Jul 25.
Article in English | MEDLINE | ID: mdl-39052722

ABSTRACT

The subtilisin-like protease-1 (SspA-1) plays an important role in the pathogenesis of a highly virulent strain of Streptococcus suis 2. However, the mechanism of SspA-1-triggered excessive inflammatory response is still unknown. In this study, we demonstrated that activation of type I IFN signaling is required for SspA-1-induced excessive proinflammatory cytokine production. Further experiments showed that the TLR2 endosomal pathway mediates SspA-1-induced type I IFN signaling and the inflammatory response. Finally, we mapped the major signaling components of the related pathway and found that the TIR adaptor proteins Mal, TRAM, and MyD88 and the downstream activation of IRF1 and IRF7 were involved in this pathway. These results explain the molecular mechanism by which SspA-1 triggers an excessive inflammatory response and reveal a novel effect of type I IFN in S. suis 2 infection, possibly providing further insights into the pathogenesis of this highly virulent S. suis 2 strain.


Subject(s)
Cytokines , Endosomes , Interferon Type I , Signal Transduction , Streptococcus suis , Toll-Like Receptor 2 , Streptococcus suis/immunology , Streptococcus suis/pathogenicity , Streptococcus suis/metabolism , Interferon Type I/metabolism , Toll-Like Receptor 2/metabolism , Cytokines/metabolism , Animals , Endosomes/metabolism , Mice , Streptococcal Infections/immunology , Streptococcal Infections/microbiology , Streptococcal Infections/metabolism , Bacterial Proteins/metabolism , Type IV Secretion Systems/metabolism , Type IV Secretion Systems/genetics , Humans , Myeloid Differentiation Factor 88/metabolism , Myeloid Differentiation Factor 88/genetics , Mice, Inbred C57BL
10.
Methods Mol Biol ; 2815: 15-21, 2024.
Article in English | MEDLINE | ID: mdl-38884907

ABSTRACT

Streptococcus suis is an important zoonotic pathogen causing severe infections in pigs and humans. Serotyping of S. suis strains is crucial for epidemiological surveillance, outbreak investigations, and understanding the pathogenesis of this bacterium. Here, we describe a step-by-step approach that enhances a previously developed pipeline by utilizing a computational script for efficient and accurate typing of S. suis strains. The pipeline is implemented in Perl programming language and leverages the Short Read Sequence Typing for Bacterial Pathogens (SRST2) tool. It integrates various bioinformatics techniques and utilizes multiple databases, including a serotype database, cpsH confirmation database, multi-locus sequence typing (MLST) database, recN species-specific gene database, and virulence gene database. These databases contain comprehensive information on S. suis serotypes, genetic markers, and virulence factors. The script can utilize paired-end or single-end fastq files as input and first confirms the species by sequence read data aligning to the recN gene, ensuring the accurate identification of S. suis strains. The pipeline next performs MLST typing and virulence factor identification using SRST2 while in a parallel processes it performs in silico serotyping of the strains. The pipeline offers a streamlined and semiautomated approach to serotyping S. suis strains, facilitating large-scale studies and reducing the manual effort required for data analysis.


Subject(s)
Computational Biology , Multilocus Sequence Typing , Software , Streptococcus suis , Streptococcus suis/genetics , Streptococcus suis/classification , Streptococcus suis/pathogenicity , Streptococcus suis/isolation & purification , Multilocus Sequence Typing/methods , Computational Biology/methods , Animals , Virulence Factors/genetics , Humans , Swine , Serotyping/methods , Bacterial Typing Techniques/methods , Computer Simulation , Databases, Genetic , Streptococcal Infections/microbiology
11.
Methods Mol Biol ; 2815: 115-119, 2024.
Article in English | MEDLINE | ID: mdl-38884914

ABSTRACT

Streptococcus suis is a swine bacterial pathogen that predominantly causes disease in weaned piglets characterized by swelling of joints, arthritis, septicemia, meningitis, and sudden death. Intravenous, intramuscular, intraperitoneal, and intranasal infection models were developed to study the bacterial pathogenicity and efficacy of vaccines and various therapeutics. The selection of the appropriate infection model is a critical step in any study, as it may impact the outcomes of the study. Here we describe a method for infecting weaned piglets with S. suis using intraperitoneal route as a reliable, consistent, and reproducible animal model to evaluate vaccine protection against systemic bacterial infection.


Subject(s)
Disease Models, Animal , Streptococcal Infections , Streptococcus suis , Swine Diseases , Animals , Swine , Streptococcus suis/pathogenicity , Streptococcal Infections/veterinary , Streptococcal Infections/microbiology , Streptococcal Infections/immunology , Swine Diseases/microbiology , Swine Diseases/immunology , Injections, Intraperitoneal
12.
Emerg Microbes Infect ; 13(1): 2352435, 2024 Dec.
Article in English | MEDLINE | ID: mdl-38703011

ABSTRACT

Streptococcus suis is a major bacterial pathogen in pigs and an emerging zoonotic pathogen. Different S. suis serotypes exhibit diverse characteristics in population structure and pathogenicity. Surveillance data highlight the significance of S. suis serotype 4 (SS4) in swine streptococcusis, a pathotype causing human infections. However, except for a few epidemiologic studies, the information on SS4 remains limited. In this study, we investigated the population structure, pathogenicity, and antimicrobial characteristics of SS4 based on 126 isolates, including one from a patient with septicemia. We discovered significant diversities within this population, clustering into six minimum core genome (MCG) groups (1, 2, 3, 4, 7-2, and 7-3) and five lineages. Two main clonal complexes (CCs), CC17 and CC94, belong to MCG groups 1 and 3, respectively. Numerous important putative virulence-associated genes are present in these two MCG groups, and 35.00% (7/20) of pig isolates from CC17, CC94, and CC839 (also belonging to MCG group 3) were highly virulent (mortality rate ≥ 80%) in zebrafish and mice, similar to the human isolate ID36054. Cytotoxicity assays showed that the human and pig isolates of SS4 strains exhibit significant cytotoxicity to human cells. Antimicrobial susceptibility testing showed that 95.83% of strains isolated from our labs were classified as multidrug-resistant. Prophages were identified as the primary vehicle for antibiotic resistance genes. Our study demonstrates the public health threat posed by SS4, expanding the understanding of SS4 population structure and pathogenicity characteristics and providing valuable information for its surveillance and prevention.


Subject(s)
Serogroup , Streptococcal Infections , Streptococcus suis , Swine Diseases , Streptococcus suis/pathogenicity , Streptococcus suis/genetics , Streptococcus suis/classification , Streptococcus suis/drug effects , Streptococcus suis/isolation & purification , Animals , Swine , Humans , Streptococcal Infections/microbiology , Streptococcal Infections/veterinary , Swine Diseases/microbiology , Virulence , Mice , Genome, Bacterial , Zebrafish , Anti-Bacterial Agents/pharmacology , Phylogeny , Microbial Sensitivity Tests , Virulence Factors/genetics
13.
Int J Biol Macromol ; 268(Pt 2): 131839, 2024 May.
Article in English | MEDLINE | ID: mdl-38663699

ABSTRACT

Streptococcus suis (S. suis) is a significant zoonotic microorganism that causes a severe illness in both pigs and humans and is characterized by severe meningitis and septicemia. Suilysin (SLY), which is secreted by S. suis, plays a crucial role as a virulence factor in the disease. To date, the interaction between SLY and host cells is not fully understood. In this study, we identified the interacting proteins between SLY and human brain microvascular endothelial cells (HBMECs) using the TurboID-mediated proximity labeling method. 251 unique proteins were identified in TurboID-SLY treated group, of which six plasma membrane proteins including ARF6, GRK6, EPB41L5, DSC1, TJP2, and PNN were identified. We found that the proteins capable of interacting with SLY are ARF6 and PNN. Subsequent investigations revealed that ARF6 substantially increased the invasive ability of S. suis in HBMECs. Furthermore, ARF6 promoted SLY-induced the activation of p38 MAPK signaling pathway in HBMECs. Moreover, ARF6 promoted the apoptosis in HBMECs through the activation of p38 MAPK signaling pathway induced by SLY. Finally, we confirmed that ARF6 could increase the virulence of SLY in C57BL/6 mice. These findings offer valuable insights that contribute to a deeper understanding of the pathogenic mechanism of SLY.


Subject(s)
ADP-Ribosylation Factor 6 , ADP-Ribosylation Factors , Apoptosis , Endothelial Cells , Hemolysin Proteins , Streptococcus suis , Streptococcus suis/pathogenicity , Streptococcus suis/metabolism , Humans , Animals , Apoptosis/drug effects , Mice , ADP-Ribosylation Factors/metabolism , ADP-Ribosylation Factors/genetics , Endothelial Cells/metabolism , Endothelial Cells/drug effects , Endothelial Cells/microbiology , Hemolysin Proteins/metabolism , Hemolysin Proteins/pharmacology , p38 Mitogen-Activated Protein Kinases/metabolism , Streptococcal Infections/microbiology , Streptococcal Infections/metabolism , Virulence , Brain/metabolism
14.
Vet Microbiol ; 293: 110084, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38608374

ABSTRACT

Streptococcus suis is an important pathogen causing severe disease in pigs and humans, giving rise to economic losses in the pig production industry. Out of 65 S. suis isolates collected from diseased pigs in Switzerland between 2019 and 2022, 57 isolates were thoroughly examined by phenotypic and whole genome sequence (WGS) based characterization. The isolates' genomes were sequenced allowing for a comprehensive analysis of their distribution in terms of serovar, sequence type (ST), clonal complex (CC), and classical virulence markers. Antimicrobial resistance (AMR) genes were screened, and phenotypic susceptibility to eight classes of antimicrobial agents was examined. Serovar 6, devoid of any resistance genes, was found to be most prevalent, followed by serovars 1, 3, 1/2, and 9. Thirty STs were identified, with ST1104 being the most prevalent. Serovar 2 and serovar 1/2 were associated with CC1, potentially containing the most virulent variants. Based on single nucleotide polymorphism (SNP) analyses, fifteen isolates belonged to one of seven putative transmission clusters each consisting of two or three isolates. High phenotypic AMR rates were detected for tetracyclines (80%) and macrolides (35%) and associated with the resistance genes tet(O) and erm(B), respectively. In contrast, susceptibility to ß-lactam antibiotics and phenicols was high. Determination of phenotypic AMR profiling, including the minimum inhibitory concentrations (MICs) of the tested antimicrobial agents, sets a baseline for future studies. The study provides valuable insights into the genetic diversity and antimicrobial susceptibility of Swiss S. suis isolates, facilitating the identification of emerging clones relevant to public health concerns.


Subject(s)
Anti-Bacterial Agents , Genetic Variation , Microbial Sensitivity Tests , Streptococcal Infections , Streptococcus suis , Swine Diseases , Animals , Streptococcus suis/genetics , Streptococcus suis/drug effects , Streptococcus suis/pathogenicity , Streptococcus suis/classification , Streptococcus suis/isolation & purification , Swine , Swine Diseases/microbiology , Switzerland/epidemiology , Streptococcal Infections/veterinary , Streptococcal Infections/microbiology , Anti-Bacterial Agents/pharmacology , Whole Genome Sequencing , Drug Resistance, Bacterial/genetics , Virulence/genetics , Serogroup , Polymorphism, Single Nucleotide
15.
Emerg Microbes Infect ; 13(1): 2339946, 2024 Dec.
Article in English | MEDLINE | ID: mdl-38578304

ABSTRACT

Streptococcus suis is a significant and emerging zoonotic pathogen. ST1 and ST7 strains are the primary agents responsible for S. suis human infections in China, including the Guangxi Zhuang Autonomous Region (GX). To enhance our understanding of S. suis ST1 population characteristics, we conducted an investigation into the phylogenetic structure, genomic features, and virulence levels of 73 S. suis ST1 human strains from GX between 2005 and 2020. The ST1 GX strains were categorized into three lineages in phylogenetic analysis. Sub-lineage 3-1a exhibited a closer phylogenetic relationship with the ST7 epidemic strain SC84. The strains from lineage 3 predominantly harboured 89K-like pathogenicity islands (PAIs) which were categorized into four clades based on sequence alignment. The acquirement of 89K-like PAIs increased the antibiotic resistance and pathogenicity of corresponding transconjugants. We observed significant diversity in virulence levels among the 37 representative ST1 GX strains, that were classified as follows: epidemic (E)/highly virulent (HV) (32.4%, 12/37), virulent plus (V+) (29.7%, 11/37), virulent (V) (18.9%, 7/37), and lowly virulent (LV) (18.9%, 7/37) strains based on survival curves and mortality rates at different time points in C57BL/6 mice following infection. The E/HV strains were characterized by the overproduction of tumour necrosis factor (TNF)-α in serum and promptly established infection at the early phase of infection. Our research offers novel insights into the population structure, evolution, genomic features, and pathogenicity of ST1 strains. Our data also indicates the importance of establishing a scheme for characterizing and subtyping the virulence levels of S. suis strains.


Subject(s)
Genome, Bacterial , Genomic Islands , Phylogeny , Streptococcal Infections , Streptococcus suis , Streptococcus suis/genetics , Streptococcus suis/pathogenicity , Streptococcus suis/classification , Streptococcus suis/isolation & purification , Streptococcal Infections/microbiology , Streptococcal Infections/veterinary , Streptococcal Infections/epidemiology , China/epidemiology , Humans , Virulence , Animals , Mice , Female , Genomics , Virulence Factors/genetics
16.
Virulence ; 15(1): 2306719, 2024 12.
Article in English | MEDLINE | ID: mdl-38251714

ABSTRACT

The arginine deiminase system (ADS) has been identified in various bacteria and functions to supplement energy production and enhance biological adaptability. The current understanding of the regulatory mechanism of ADS and its effect on bacterial pathogenesis is still limited. Here, we found that the XRE family transcriptional regulator XtrSs negatively affected Streptococcus suis virulence and significantly repressed ADS transcription when the bacteria were incubated in blood. Electrophoretic mobility shift (EMSA) and lacZ fusion assays further showed that XtrSs directly bind to the promoter of ArgR, an acknowledged positive regulator of bacterial ADS, to repress ArgR transcription. Moreover, we provided compelling evidence that S. suis could utilize arginine via ADS to adapt to acid stress, while ΔxtrSs enhanced this acid resistance by upregulating the ADS operon. Moreover, whole ADS-knockout S. suis increased arginine and antimicrobial NO in the infected macrophage cells, decreased intracellular survival, and even caused significant attenuation of bacterial virulence in a mouse infection model, while ΔxtrSs consistently presented the opposite results. Our experiments identified a novel ADS regulatory mechanism in S. suis, whereby XtrSs regulated ADS to modulate NO content in macrophages, promoting S. suis intracellular survival. Meanwhile, our findings provide a new perspective on how Streptococci evade the host's innate immune system.


Subject(s)
Bacterial Proteins , Streptococcal Infections , Streptococcus suis , Animals , Mice , Arginine , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Gene Expression Regulation, Bacterial , Hydrolases/genetics , Hydrolases/metabolism , Macrophages , Streptococcal Infections/microbiology , Streptococcus suis/pathogenicity , Streptococcus suis/physiology
17.
Vet Microbiol ; 288: 109928, 2024 Jan.
Article in English | MEDLINE | ID: mdl-38056180

ABSTRACT

Streptococcus suis serotype 2 (SS2) has been reported to be a highly invasive pathogen in swine and a zoonotic agent for humans. Although many bacterial virulence factors have been identified, our an insightful understanding of SS2 pathogenicity is lacking. The gene nadR, encoding nicotinamide-nucleotide adenylyltransferase, was first reported as a regulator and transporter of the intracellular NAD synthesis pathway in Salmonella typhimurium. In this study, we constructed a mutant strain of nadR (ΔnadR) to test the phenotypic and virulence variations between the deletion mutant and the wild-type strain ZY05719. The phenotypic experimental results showed that ΔnadR obviously inhibited cell growth and exhibited shorter chains than WT. The growth defect of ΔnadR was caused by the loss of the function of nadR for transporting the substrates nicotinamide mononucleotide and nicotinamide riboside in the intracellular NAD synthesis pathway. In the process of interaction with the host, ΔnadR participated in adherence and invasion to the host cells, and it was more easily cleared by RAW264.7 macrophages. More importantly, both zebrafish and BALB/c mice in vivo virulence experimental results showed that ΔnadR dramatically attenuated the virulence of SS2, and the ability of ΔnadR to colonize tissues was notably reduced in comparison with that of WT in the BALB/c mice infection model. To the best of our knowledge, this is the first time to demonstrate that nadR not only plays an important role in bacterial growth, but also in connection with the virulence of SS2 as a global transcriptional regulator.


Subject(s)
Streptococcal Infections , Streptococcus suis , Swine Diseases , Animals , Mice , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , NAD/metabolism , Serogroup , Streptococcal Infections/veterinary , Streptococcal Infections/microbiology , Streptococcus suis/pathogenicity , Swine , Virulence/genetics , Zebrafish
18.
J Microbiol ; 61(4): 433-448, 2023 Apr.
Article in English | MEDLINE | ID: mdl-37010796

ABSTRACT

Streptococcus suis serotype 2 (SS2) is an important zoonotic pathogen that can infect humans in contact with infected pigs or their byproducts. It can employ different types of genes to defend against oxidative stress and ensure its survival. The thioredoxin (Trx) system is a key antioxidant system that contributes adversity adaptation and pathogenicity. SS2 has been shown to encode putative thioredoxin genes, but the biological roles, coding sequence, and underlying mechanisms remains uncharacterized. Here, we demonstrated that SSU05_0237-ORF, from a clinical SS2 strain, ZJ081101, encodes a protein of 104 amino acids with a canonical CGPC active motif and an identity 70-85% similar to the thioredoxin A (TrxA) in other microorganisms. Recombinant TrxA efficiently catalyzed the thiol-disulfide oxidoreduction of insulin. The deletion of TrxA led to a significantly slow growth and markedly compromised tolerance of the pathogen to temperature stress, as well as impaired adhesion ability to pig intestinal epithelial cells (IPEC-J2). However, it was not involved in H2O2 and paraquat-induced oxidative stress. Compared with the wild-type strain, the ΔTrxA strain was more susceptible to killing by macrophages through increasing NO production. Treatment with TrxA mutant strain also significantly attenuated cytotoxic effects on RAW 264.7 cells by inhibiting inflammatory response and apoptosis. Knockdown of pentraxin 3 in RAW 264.7 cells was more vulnerable to phagocytic activity, and TrxA promoted SS2 survival in phagocytic cells depending on pentraxin 3 activity compared with the wild-type strain. Moreover, a co-inoculation experiment in mice revealed that TrxA mutant strain is far more easily cleared from the body than the wild type strain in the period from 8-24 h, and exhibits significantly attenuated oxidative stress and liver injury. In summary, we reveal the important role of TrxA in the pathogenesis of SS2.


Subject(s)
Macrophages , Streptococcal Infections , Streptococcus suis , Animals , Humans , Mice , Bacterial Proteins/metabolism , Hydrogen Peroxide/pharmacology , Hydrogen Peroxide/metabolism , Macrophages/metabolism , Macrophages/microbiology , Serogroup , Streptococcus suis/metabolism , Streptococcus suis/pathogenicity , Swine , Thioredoxins/genetics , Thioredoxins/metabolism , Thioredoxins/pharmacology , Virulence
19.
Sci Rep ; 11(1): 23919, 2021 12 14.
Article in English | MEDLINE | ID: mdl-34907269

ABSTRACT

Streptococcus suis has been well-recognized as a zoonotic pathogen worldwide, and the diversity and unpredictable adaptive potential of sporadic human strains represent a great risk to the public health. In this study, S. suis LSM178, isolated from a patient in contact with pigs and raw pork, was assessed as a hyper-virulent strain and interpreted for the virulence based on its genetic information. The strain was more invasive for Caco-2 cells than two other S. suis strains, SC19 and P1/7. Sequence analysis designated LSM178 with serotype 2 and a novel sequence type 1005. Phylogenetic analysis showed that LSM178 clustered with highly virulent strains including all human strains and epidemic strains. Compared with other strains, these S. suis have the most and the same virulent factors and a type I-89 K pathogenicity island. Further, groups of genes were identified to distinguish these highly virulent strains from other generally virulent strains, emphasizing the key roles of genes modeling transcription, cell barrier, replication, recombination and repair on virulence regulation. Additionally, LSM178 contains a novel prophage conducive potentially to pathogenicity.


Subject(s)
Genome, Bacterial , Genomic Islands , Phylogeny , Streptococcal Infections , Streptococcus suis , Virulence Factors , Animals , Humans , Sequence Analysis, DNA , Streptococcal Infections/genetics , Streptococcal Infections/metabolism , Streptococcus suis/genetics , Streptococcus suis/isolation & purification , Streptococcus suis/metabolism , Streptococcus suis/pathogenicity , Swine , Virulence Factors/biosynthesis , Virulence Factors/genetics
20.
Vet Res ; 52(1): 145, 2021 Dec 19.
Article in English | MEDLINE | ID: mdl-34924012

ABSTRACT

Streptococcus suis is a zoonotic pathogen of swine involved in arthritis, polyserositis, and meningitis. Colonization of piglets by S. suis is very common and occurs early in life. The clinical outcome of infection is influenced by the virulence of the S. suis strains and the immunity of the animals. Here, the role of innate immunity was studied in cesarean-derived colostrum-deprived piglets inoculated intranasally with either virulent S. suis strain 10 (S10) or non-virulent S. suis strain T15. Colonization of the inoculated piglets was confirmed at the end of the study by PCR and immunohistochemistry. Fever (≥40.5 °C) was more prevalent in piglets inoculated with S10 compared to T15 at 4 h after inoculation. During the 3 days of monitoring, no other major clinical signs were detected. Accordingly, only small changes in transcription of genes associated with the antibacterial innate immune response were observed at systemic sites, with S10 inducing an earlier response than T15 in blood. Local inflammatory response to the inoculation, evaluated by transcriptional analysis of selected genes in nasal swabs, was more sustained in piglets inoculated with the virulent S10, as demonstrated by transcription of inflammation-related genes, such as IL1B, IL1A, and IRF7. In contrast, most of the gene expression changes in trachea, lungs, and associated lymph nodes were observed in response to the non-virulent T15 strain. Thus, S. suis colonization in the absence of systemic infection induces an innate immune response in piglets that appears to be related to the virulence potential of the colonizing strain.


Subject(s)
Immunity, Innate , Streptococcal Infections , Streptococcus suis , Swine Diseases , Virulence , Animals , Immunity, Innate/immunology , Streptococcal Infections/immunology , Streptococcal Infections/veterinary , Streptococcal Infections/virology , Streptococcus suis/pathogenicity , Swine , Swine Diseases/immunology , Swine Diseases/virology
SELECTION OF CITATIONS
SEARCH DETAIL