Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
1.
Sci Rep ; 12(1): 13920, 2022 08 17.
Article in English | MEDLINE | ID: mdl-35978073

ABSTRACT

Streptococcus suis, a common member of the porcine respiratory microbiota, can cause life-threatening diseases in pigs as well as humans. A previous study identified the gene trpX as conditionally essential for in vivo survival by intrathecal infection of pigs with a transposon library of S. suis strain 10. Here, we characterized trpX, encoding a putative tryptophan/tyrosine transport system substrate-binding protein, in more detail. We compared growth capacities of the isogenic trpX-deficient mutant derivative strain 10∆trpX with its parent. Growth experiments in chemically defined media (CDM) revealed that growth of 10∆trpX depended on tryptophan concentration, suggesting TrpX involvement in tryptophan uptake. We demonstrated that trpX is part of an operon structure and co-transcribed with two additional genes encoding a putative permease and ATPase, respectively. Bioinformatics analysis identified a putative tryptophan T-box riboswitch in the 5' untranslated region of this operon. Finally, qRT-PCR and a reporter activation assay revealed trpX mRNA induction under tryptophan-limited conditions. In conclusion, our study showed that TrpX is part of a putative tryptophan ABC transporter system regulated by a T-box riboswitch probably functioning as a substrate-binding protein. Due to the tryptophan auxotrophy of S. suis, TrpX plays a crucial role for metabolic adaptation and growth during infection.


Subject(s)
Riboswitch , Streptococcal Infections , Streptococcus suis , Animals , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Biological Transport , Carrier Proteins/metabolism , Humans , Operon/genetics , Streptococcal Infections/genetics , Streptococcal Infections/veterinary , Streptococcus suis/metabolism , Swine , Tryptophan/metabolism
2.
J Adv Res ; 36: 101-112, 2022 02.
Article in English | MEDLINE | ID: mdl-35127168

ABSTRACT

Introduction: Due to the increase of antibiotic resistant bacterial strains, there is an urgent need for development of alternatives to antibiotics. Cathelicidins can be such an alternative to antibiotics having both a direct antimicrobial capacity as well as an immunomodulatory function. Previously, the full d-enantiomer of chicken cathelicidin-2 (d-CATH-2) has shown to prophylactically protect chickens against infection 7 days post hatch when administered in ovo three days before hatch. Objectives: To further evaluate d-CATH-2 in mammals as a candidate for an alternative to antibiotics.In this study, the prophylactic capacity of d-CATH-2 and two truncated derivatives, d-C(1-21) and d-C(4-21), was determined in mammalian cells. Methods: Antibacterial assays; immune cell differentiation and modulation; cytotoxicity, isothermal titration calorimetry; in vivo prophylactic capacity of peptides in an S. suis infection model. Results: d-CATH-2 and its derivatives were shown to have a strong direct antibacterial capacity against four different S. suis serotype 2 strains (P1/7, S735, D282, and OV625) in bacterial medium and even stronger in cell culture medium. In addition, d-CATH-2 and its derivatives ameliorated the efficiency of mouse bone marrow-derived macrophages (BMDM) and skewed mouse bone marrow-derived dendritic cells (BMDC) towards cells with a more macrophage-like phenotype. The peptides directly bind lipoteichoic acid (LTA) and inhibit LTA-induced activation of macrophages. In addition, S. suis killed by the peptide was unable to further activate mouse macrophages, which indicates that S. suis was eliminated by the previously reported silent killing mechanism. Administration of d-C(1-21) at 24 h or 7 days before infection resulted in a small prophylactic protection with reduced disease severity and reduced mortality of the treated mice. Conclusion: d-enantiomers of CATH-2 show promise as anti-infectives against pathogenic S. suis for application in mammals.


Subject(s)
Streptococcus suis , Animals , Cathelicidins/chemistry , Cathelicidins/metabolism , Cathelicidins/pharmacology , Chickens , Macrophages/metabolism , Mice , Serogroup
3.
Microbiologyopen ; 10(5): e1234, 2021 10.
Article in English | MEDLINE | ID: mdl-34713609

ABSTRACT

Streptococcus suis, an emerging zoonotic pathogen, causes invasive diseases in pigs, including sepsis, meningitis, endocarditis, pneumonia, and arthritis. Importantly, similar pathologies are reported in human S. suis infections. In previous work, the locus SSU0375 of S. suis strain P1.7 had been identified as a conditionally essential gene by intrathecal experimental infection of pigs with a transposon library of S. suis. This study aimed to identify the function of the corresponding gene product. Bioinformatics analysis and homology modeling revealed sequence and structural homologies with the Streptococcus pneumoniae mid-cell-anchored protein Z (MapZ) that is involved in cell division in different bacterial species. Indeed, depletion of this locus in S. suis strain 10 revealed a growth defect as compared to the wild type. Electron microscopy analysis of the corresponding mutant demonstrated morphological growth defects as compared to the wild-type strain, including an irregular cell shape and size as well as mispositioned division septa. Light microscopy and subsequent quantitative image analysis confirmed these morphological alterations. In the genetic rescue strain, the wild-type phenotype was completely restored. In summary, we proposed that SSU0375 or the corresponding locus in strain 10 encode for a S. suis MapZ homolog that guides septum positioning as evidenced for other members of the Streptococci family.


Subject(s)
Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Cell Division , Cytokinesis , Streptococcus suis/genetics , Streptococcus suis/metabolism , Animals , Computational Biology/methods , Genes, Essential , Humans , Mutation , Streptococcal Infections/microbiology , Streptococcus pneumoniae/genetics , Streptococcus suis/cytology
4.
J Anim Sci ; 98(6)2020 Jun 01.
Article in English | MEDLINE | ID: mdl-32479635

ABSTRACT

The objective of this study is to investigate the effect of a maternal antibiotic administration during the last week of gestation on the early life intestinal development in neonatal piglets. Colonization of the gut with bacteria starts during birth and plays a major role in the intestinal and immunological development of the intestine. We demonstrate that maternal interventions induced changes in the sows (n = 6 to 8 per treatment) fecal microbiota diversity around birth (P < 0.001, day 1). Whole-genome microarray analysis in small intestinal samples of 1-d old piglets (n = 6 to 8 per treatment) showed significantly expressed genes (Padj < 0.05) which were involved in processes of tight junction formation and immunoglobulin production. Furthermore, when performing morphometry analysis, the number of goblet cells in jejunum was significantly (P < 0.001) lower in piglets from amoxicillin administered sows compared with the respective control piglets. Both significantly expressed genes (Padj < 0.05) and significant morphometry data (jejunum P < 0.05 and ileum P < 0.01) indicate that the crypts of piglets from amoxicillin administered sows deepen around weaning (day 26) as an effect of the amoxicillin administration in sows. The latter might imply that the intestinal development of piglets was delayed by maternal antibiotic administration. Taken together, these results show that maternally oral antibiotic administration changes in early life can affect intestinal development of the offspring piglets for a period of at least 5 wk after the maternal antibiotic administration was finished. These results show that modulation of the neonatal intestine is possible by maternal interventions.


Subject(s)
Anti-Bacterial Agents/administration & dosage , Bacteria/drug effects , Microbiota/drug effects , Swine/physiology , Animals , Animals, Newborn , Bacteria/growth & development , Feces/microbiology , Female , Gastrointestinal Tract/drug effects , Gastrointestinal Tract/growth & development , Gastrointestinal Tract/microbiology , Pregnancy , Swine/growth & development , Swine/microbiology , Weaning
5.
Virulence ; 11(1): 446-464, 2020 12.
Article in English | MEDLINE | ID: mdl-32419603

ABSTRACT

Streptococcus suis is a Gram-positive bacterium and zoonotic pathogen that causes meningitis and sepsis in pigs and humans. The aim of this study was to identify genes required for S. suis infection. We created Tn-Seq libraries in a virulent S. suis strain 10, which was used to inoculate pigs in an intrathecal experimental infection. Comparative analysis of the relative abundance of mutants recovered from different sites of infection (blood, cerebrospinal fluid, and meninges of the brain) identified 361 conditionally essential genes, i.e. required for infection, which is about 18% of the genome. The conditionally essential genes were primarily involved in metabolic and transport processes, regulation, ribosomal structure and biogenesis, transcription, and cell wall membrane and envelope biogenesis, stress defenses, and immune evasion. Directed mutants were created in a set of 10 genes of different genetic ontologies and their role was determined in ex vivo models. Mutants showed different levels of sensitivity to survival in whole blood, serum, cerebrospinal fluid, thermic shock, and stress conditions, as compared to the wild type. Additionally, the role of three selected mutants was validated in co-infection experiments in which pigs were infected with both wild type and isogenic mutant strains. The genetic determinants of infection identified in this work contribute to novel insights in S. suis pathogenesis and could serve as targets for novel vaccines or antimicrobial drugs.


Subject(s)
Genes, Bacterial , Genes, Essential , Streptococcal Infections/veterinary , Streptococcus suis/genetics , Swine Diseases/microbiology , Animals , Coinfection/microbiology , Disease Models, Animal , Immune Evasion , Meningitis , Mutation , Streptococcal Infections/microbiology , Streptococcus suis/pathogenicity , Swine , Virulence
6.
Sci Rep ; 9(1): 15429, 2019 10 28.
Article in English | MEDLINE | ID: mdl-31659179

ABSTRACT

Streptococcus suis is a porcine pathogen, causing severe invasive infections. S. suis serotype 9 is increasingly causing disease in Dutch and Chinese pig herds, but it is unknown whether all serotype 9 isolates are equally virulent and markers that can identify virulent strains are not available. Therefore, discrimination between virulent isolates and carriage isolates typically not associated with disease, is currently not possible. We collected tonsillar S. suis isolates from 6 herds not previously diagnosed with S. suis infections, and clinical S. suis isolates of previously diseased pigs. We confirmed the virulence of a virulent type strain and one representative clinical isolate, and the lack of virulence of two carriage isolates, in a pig infection model. Phylogenetic analysis of whole genome sequences of 124 isolates resulted in 10 groups, of which two were almost uniquely populated by clinical isolates. The population structure of S. suis serotype 9 appears highly diverse. However, analysis of the capsule loci sequences showed variation in a single region which fully correlated with a virulent genotype. Transmission electron microscopy suggested differences in capsule thickness between carriage and clinical genotypes. In conclusion, we found that that the S. suis serotype 9 population in the Netherlands is diverse. A distinct virulence-associated lineage was identified and could be discriminated based on the capsule locus sequence. Whilst the difference in virulence cannot be directly attributed to the DNA sequence, the correlation of capsule locus sequence with virulence could be used in the development of diagnostic tests to identify potential virulent S. suis serotype 9 in pigs.


Subject(s)
DNA, Bacterial/genetics , Phylogeny , Serogroup , Streptococcal Infections , Streptococcus suis , Swine Diseases , Animals , Humans , Streptococcal Infections/genetics , Streptococcal Infections/microbiology , Streptococcal Infections/veterinary , Streptococcus suis/genetics , Streptococcus suis/pathogenicity , Streptococcus suis/ultrastructure , Swine , Swine Diseases/genetics , Swine Diseases/microbiology
7.
Virulence ; 10(1): 334-351, 2019 12.
Article in English | MEDLINE | ID: mdl-30957693

ABSTRACT

Streptococcus suis is a Gram-positive bacterium and a zoonotic pathogen residing in the nasopharynx or the gastrointestinal tract of pigs with a potential of causing life-threatening invasive disease. It is endemic in the porcine production industry worldwide, and it is also an emerging human pathogen. After invasion, the pathogen adapts to cause bacteremia and disseminates to different organs including the brain. To gain insights in this process, we infected piglets with a highly virulent strain of S. suis, and bacterial transcriptomes were obtained from blood and different organs (brain, joints, and heart) when animals had severe clinical symptoms of infection. Microarrays were used to determine the genome-wide transcriptional profile at different infection sites and during growth in standard growth medium in vitro. We observed differential expression of around 30% of the Open Reading Frames (ORFs) and infection-site specific patterns of gene expression. Genes with major changes in expression were involved in transcriptional regulation, metabolism, nutrient acquisition, stress defenses, and virulence, amongst others, and results were confirmed for a subset of selected genes using RT-qPCR. Mutants were generated in two selected genes, and the encoded proteins, i.e., NADH oxidase and MetQ, were shown to be important virulence factors in coinfection experiments and in vitro assays. The knowledge derived from this study regarding S. suis gene expression in vivo and identification of virulence factors is important for the development of novel diagnostic and therapeutic strategies to control S. suis disease.


Subject(s)
Adaptation, Physiological/genetics , Streptococcal Infections/veterinary , Streptococcus suis/genetics , Streptococcus suis/pathogenicity , Transcriptome , Virulence Factors/genetics , ATP-Binding Cassette Transporters/genetics , ATP-Binding Cassette Transporters/metabolism , Animals , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Microarray Analysis , Multienzyme Complexes/genetics , Multienzyme Complexes/metabolism , Mutation , NADH, NADPH Oxidoreductases/genetics , NADH, NADPH Oxidoreductases/metabolism , Streptococcal Infections/microbiology , Swine , Swine Diseases/microbiology , Virulence Factors/metabolism
8.
PLoS One ; 14(1): e0210801, 2019.
Article in English | MEDLINE | ID: mdl-30653570

ABSTRACT

Streptococcus suis is an economically important pathogen of pigs as well as a zoonotic cause of human disease. Serotyping is used for further characterization of isolates; some serotypes seem to be more virulent and more widely spread than others. This study characterizes a collection of German field isolates of Streptococcus suis from pigs dating from 1996 to 2016 with respect to capsular genes (cps) specific for individual serotypes and pathotype by multiplex PCR and relates results to the clinical background of these isolates. The most prominent finding was the reduction in prevalence of serotype-2/serotype-1/2 among invasive isolates during this sampling period, which might be attributed to widely implemented autogenous vaccination programs in swine against serotype 2 in Germany. In diseased pigs (systemically ill; respiratory disease) isolates of serotype-1/serotype-14, serotype-2/serotype-1/2, serotype 3 to 5 and 7 to 9 were most frequent while in carrier isolates a greater variety of cps types was found. Serotype-1/serotype-14 seemed to be preferentially located in joints, serotype 4 and serotype 3 in the central nervous system, respectively. The virulence associated extracellular protein factor was almost exclusively associated with invasive serotype-1/serotype-14 and serotype-2/serotype-1/2 isolates. In contrast, lung isolates of serotype-2/serotype-1/2 mainly harbored the gene for muramidase-released protein. Serotype 4 and serotype 9 isolates from clinically diseased pigs most frequently carried the muramidase-released protein gene and the suilysin gene. When examined by transmission electron microscopy all but one of the isolates which were non-typable by molecular and serological methods showed various amounts of capsular material indicating potentially new serotypes among these isolates. Given the variety of cps types/serotypes detected in pigs, not only veterinarians but also medical doctors should consider other serotypes than just serotype 2 when investigating potential human cases of Streptococcus suis infection.


Subject(s)
Streptococcal Infections/veterinary , Streptococcus suis/classification , Sus scrofa/microbiology , Swine Diseases/microbiology , Animals , Antigens, Bacterial/genetics , Bacterial Capsules/ultrastructure , Bacterial Proteins/genetics , Carrier State/epidemiology , Carrier State/microbiology , Carrier State/virology , Genes, Bacterial , Germany/epidemiology , Humans , Microscopy, Electron, Transmission , Molecular Typing , Serotyping , Streptococcal Infections/epidemiology , Streptococcal Infections/microbiology , Streptococcus suis/genetics , Streptococcus suis/isolation & purification , Swine , Swine Diseases/epidemiology , Zoonoses/microbiology
9.
Pathogens ; 5(3)2016 Jul 21.
Article in English | MEDLINE | ID: mdl-27455333

ABSTRACT

Streptococcus (S.) suis is a zoonotic pathogen causing septicemia and meningitis in pigs and humans. During infection S. suis must metabolically adapt to extremely diverse environments of the host. CcpA and the FNR family of bacterial transcriptional regulators are important for metabolic gene regulation in various bacteria. The role of CcpA in S. suis is well defined, but the function of the FNR-like protein of S. suis, FlpS, is yet unknown. Transcriptome analyses of wild-type S. suis and a flpS mutant strain suggested that FlpS is involved in the regulation of the central carbon, arginine degradation and nucleotide metabolism. However, isotopologue profiling revealed no substantial changes in the core carbon and amino acid de novo biosynthesis. FlpS was essential for the induction of the arcABC operon of the arginine degrading pathway under aerobic and anaerobic conditions. The arcABC-inducing activity of FlpS could be associated with the level of free oxygen in the culture medium. FlpS was necessary for arcABC-dependent intracellular bacterial survival but redundant in a mice infection model. Based on these results, we propose that the core function of S. suis FlpS is the oxygen-dependent activation of the arginine deiminase system.

10.
Pathogens ; 5(3)2016 Jun 30.
Article in English | MEDLINE | ID: mdl-27376336

ABSTRACT

Invasive Streptococcus suis (S. suis) infections in pigs are often associated with serotypes 2 and 9. Mucosal sites of healthy pigs can be colonized with these serotypes, often multiple serotypes per pig. To unravel the contribution of these serotypes in pathogenesis and epidemiology, simultaneous quantification of serotypes is needed. A quantitative real-time PCR (qPCR) targeting cps2J (serotypes 2 and 1/2) and cps9H (serotype 9) was evaluated with nasal and tonsillar samples from S. suis exposed pigs. qPCR specifically detected serotypes in all pig samples. The serotypes loads in pig samples estimated by qPCR showed, except for serotype 9 in tonsillar samples (correlation coefficient = 0.25), moderate to strong correlation with loads detected by culture (correlation coefficient > 0.65), and also in pigs exposed to both serotypes (correlation coefficient > 0.75). This qPCR is suitable for simultaneous differentiation and quantification of important S. suis serotypes.

11.
BMC Microbiol ; 16: 102, 2016 Jun 08.
Article in English | MEDLINE | ID: mdl-27276874

ABSTRACT

BACKGROUND: Streptococcus pneumoniae, a Gram-positive bacterium carried in the human nasopharynx, is an important human pathogen causing mild diseases such as otitis media and sinusitis as well as severe diseases including pneumonia, meningitis and sepsis. There is a strong resemblance between the anatomy, immunology and physiology of the pig and human species. Furthermore, there are striking similarities between S. suis pathogenesis in piglets and S. pneumoniae pathogenesis in humans. Therefore, we investigated the use of piglets as a model for pneumococcal colonization and invasive disease. RESULTS: Intravenous inoculation of piglets with an invasive pneumococcal isolate led to bacteraemia during 5 days, showing clear bacterial replication in the first two days. Bacteraemia was frequently associated with fever and septic arthritis. Moreover, intranasal inoculation of piglets with a nasopharyngeal isolate led to colonization for at least six consecutive days. CONCLUSIONS: This demonstrates that central aspects of human pneumococcal infections can be modelled in piglets enabling the use of this model for studies on colonization and transmission but also on development of vaccines and host-directed therapies. Moreover this is the first example of an animal model inducing high levels of pneumococcal septic arthritis.


Subject(s)
Bacteremia/pathology , Disease Models, Animal , Pneumococcal Infections/veterinary , Streptococcus pneumoniae/pathogenicity , Swine Diseases/microbiology , Animals , Arthritis, Infectious/microbiology , Bacteremia/microbiology , Fever/etiology , Humans , Nasopharynx/microbiology , Pneumococcal Infections/microbiology , Pneumococcal Infections/pathology , Swine , Swine Diseases/pathology
13.
J Infect Dis ; 212(1): 95-105, 2015 Jul 01.
Article in English | MEDLINE | ID: mdl-25525050

ABSTRACT

BACKGROUND: Streptococcus suis has emerged as an important cause of bacterial meningitis in adults. The ingestion of undercooked pork is a risk factor for human S. suis serotype 2 (SS2) infection. Here we provide experimental evidence indicating that the gastrointestinal tract is an entry site of SS2 infection. METHODS: We developed a noninvasive in vivo model to study oral SS2 infection in piglets. We compared in vitro interaction of S. suis with human and porcine intestinal epithelial cells (IEC). RESULTS: Two out of 15 piglets showed clinical symptoms compatible with S. suis infection 24-48 hours after ingestion of SS2. SS2 was detected in mesenteric lymph nodes of 40% of challenged piglets. SS2 strains isolated from patients showed significantly higher adhesion to human IEC compared to invasive strains isolated from pigs. In contrast, invasive SS9 strains showed significantly higher adhesion to porcine IEC. Translocation across human IEC, which occurred predominately via a paracellular route, was significantly associated with clonal complex 1, the predominant zoonotic genotype. Adhesion and translocation were dependent on capsular polysaccharide production. CONCLUSIONS: SS2 should be considered a food-borne pathogen. S. suis interaction with human and pig IEC correlates with S. suis serotype and genotype, which can explain the zoonotic potential of SS2.


Subject(s)
Host-Pathogen Interactions , Intestinal Mucosa/microbiology , Streptococcal Infections/microbiology , Streptococcal Infections/veterinary , Streptococcus suis/physiology , Zoonoses/microbiology , Adult , Animals , Cell Line , Disease Models, Animal , Epithelial Cells/immunology , Epithelial Cells/microbiology , Humans , Male , Meningitis, Bacterial/microbiology , Meningitis, Bacterial/veterinary , Swine
14.
BMC Microbiol ; 14: 264, 2014 Nov 12.
Article in English | MEDLINE | ID: mdl-25384512

ABSTRACT

BACKGROUND: Streptococcus suis is a major problem in the swine industry causing meningitis, arthritis and pericarditis in piglets. Pathogenesis of S. suis is poorly understood. We previously showed that introduction of a 3 kb genomic fragment from virulent serotype 2 strain 10 into a weakly virulent serotype 2 strain S735, generated a hypervirulent isolate. The 3 kb genomic fragment contained two complete open reading frames (ORF) in an operon-structure of which one ORF showed similarity to folylpolyglutamate synthetase, whereas the function of the second ORF could not be predicted based on database searches for protein similarity. RESULTS: In this study we demonstrate that introduction of orf2 from strain 10 into strain S735 is sufficient to dramatically increase the virulence of S735 in pigs. This increase in virulence could not be associated with changes in pro-inflammatory responses of porcine blood mononucleated cells in response to S. suis in vitro. Sequence analysis of the orf2-folC-operon of S. suis isolates 10 and S735 revealed an SNP in the -35 region of the putative promoter sequence of the operon, as well as several SNPs resulting in amino acid substitutions in the ORF2 protein. Transcript levels of orf2 and folC were significantly higher in the virulent strain 10 than in the weakly virulent strain S735 and in vitro mutagenesis of the orf2 promoter confirmed that this was due to a SNP in the predicted -35 region upstream of the orf2 promoter. In this study, we demonstrated that the stronger promoter was present in all virulent and highly virulent S. suis isolates included in our study. This highlights a correlation between high orf2 expression and virulence. Conversely, the weaker promoter was present in isolates known to be weakly pathogenic or non-pathogenic. CONCLUSION: In summary, we demonstrate the importance of orf2 in the virulence of S. suis.


Subject(s)
Polymorphism, Single Nucleotide , Promoter Regions, Genetic , Streptococcal Infections/microbiology , Streptococcal Infections/pathology , Streptococcus suis/growth & development , Streptococcus suis/genetics , Virulence Factors/genetics , Animals , Gene Expression Profiling , Operon/genetics , Sequence Analysis, DNA , Swine , Virulence
15.
PLoS One ; 9(6): e99394, 2014.
Article in English | MEDLINE | ID: mdl-24968201

ABSTRACT

Here we show that S. suis, a major bacterial pathogen of pigs and emerging pathogen in humans responds to a peptide pheromone by developing competence for DNA transformation. This species does not fall within any of the phylogenetic clusters of streptococci previously shown to regulate competence via peptide pheromones suggesting that more species of streptococci may be naturally competent. Induction of competence was dependent on ComX, a sigma factor that controls the streptococcal late competence regulon, extracellular addition of a comX-inducing peptide (XIP), and ComR, a regulator of comX. XIP was identified as an N-terminally truncated variant of ComS. Different comS alleles are present among strains of S. suis. These comS alleles are not functionally equivalent and appear to operate in conjuction with a cognate ComR to regulate comX through a conserved comR-box promoter. We demonstrate that these 'pherotypes' can be genetically transferred between strains, suggesting that similar approaches might be used to control competence induction in other lactic acid bacteria that lack ComR/ComS homologues but possess comX and the late competence regulon. The approaches described in this paper to identify and optimize peptide-induced competence may also assist other researchers wishing to identify natural competence in other bacteria. Harnessing natural competence is expected to accelerate genetic research on this and other important streptococcal pathogens and to allow high-throughput mutation approaches to be implemented, opening up new avenues for research.


Subject(s)
Bacterial Proteins/genetics , DNA Transformation Competence/genetics , Streptococcus suis/genetics , Transcription Factors/genetics , Amino Acid Sequence , Bacterial Proteins/chemistry , Bacterial Proteins/metabolism , Molecular Sequence Data , Transcription Factors/chemistry , Transcription Factors/metabolism
16.
Future Microbiol ; 9(5): 587-91, 2014.
Article in English | MEDLINE | ID: mdl-24957086

ABSTRACT

First International Workshop on Streptococcus suis, Beijing, China, 12-13 August 2013. This second and final chapter of the report on the First International Workshop on Streptococcus suis follows on from Part 1, published in the April 2014, volume 9, issue 4 of Future Microbiology. S. suis is a swine pathogen and a zoonotic agent afflicting people in close contact with infected pigs or pork meat. Although sporadic cases of human infections had been reported worldwide, deadly S. suis outbreaks emerged in Asia. The severity of the disease underscores the lack of knowledge on the virulence and zoonotic evolution of this human-infecting agent. The pathogenesis of the infection, interactions with host cells and new avenues for treatments were among the topics discussed during the First International Workshop on S. suis (China 2013).


Subject(s)
Streptococcal Infections/pathology , Streptococcus suis/pathogenicity , Swine/microbiology , Zoonoses/microbiology , Zoonoses/transmission , Animals , Anti-Bacterial Agents/therapeutic use , Endocarditis, Bacterial/microbiology , Host-Pathogen Interactions/immunology , Humans , Meningitis, Bacterial/microbiology , Nisin/pharmacology , Sepsis/microbiology , Streptococcal Infections/immunology , Streptococcal Infections/microbiology , Streptococcal Infections/veterinary , Streptococcal Vaccines , Streptococcus suis/classification , Streptococcus suis/genetics , Streptococcus suis/immunology , Swine/immunology , Swine Diseases/microbiology , Vaccination
17.
Future Microbiol ; 9(4): 441-4, 2014.
Article in English | MEDLINE | ID: mdl-24810343

ABSTRACT

First International Workshop on Streptococcus suis, Beijing, China, 12-13 August 2013 The first international workshop on Streptococcus suis, which is an important swine pathogen and emerging zoonotic agent, took place in Beijing, jointly organized by the Faculty of Veterinary Medicine, University of Montreal, Canada and the National Institute for Communicable Disease Control and Prevention, China CDC. The aim of the meeting was to gather together, for the first time, more than 80 researchers working on S. suis, from countries including China, Canada, Japan, The Netherlands, Germany, Thailand, the UK and Vietnam. This article, the first of a two-part report on this First International Workshop, reviews current aspects of the epidemiology and population genomics of S. suis, covers public health concerns and discusses questions about S. suis serotyping and molecular diagnostics.


Subject(s)
Communicable Diseases, Emerging/epidemiology , Streptococcal Infections/epidemiology , Streptococcal Infections/veterinary , Streptococcus suis/isolation & purification , Swine Diseases/epidemiology , Zoonoses/epidemiology , Animals , Communicable Diseases, Emerging/microbiology , Humans , Streptococcal Infections/microbiology , Streptococcus suis/classification , Streptococcus suis/genetics , Swine , Swine Diseases/microbiology , Zoonoses/microbiology
18.
PLoS One ; 9(3): e89334, 2014.
Article in English | MEDLINE | ID: mdl-24642967

ABSTRACT

Streptococcus suis is a major bacterial pathogen of young pigs causing worldwide economic problems for the pig industry. S. suis is also an emerging pathogen of humans. Colonization of porcine oropharynx by S. suis is considered to be a high risk factor for invasive disease. In the oropharyngeal cavity, where glucose is rapidly absorbed but dietary α-glucans persist, there is a profound effect of carbohydrate availability on the expression of virulence genes. Nineteen predicted or confirmed S. suis virulence genes that promote adhesion to and invasion of epithelial cells were expressed at higher levels when S. suis was supplied with the α-glucan starch/pullulan compared to glucose as the single carbon source. Additionally the production of suilysin, a toxin that damages epithelial cells, was increased more than ten-fold when glucose levels were low and S. suis was growing on pullulan. Based on biochemical, bioinformatics and in vitro and in vivo gene expression studies, we developed a biological model that postulates the effect of carbon catabolite repression on expression of virulence genes in the mucosa, organs and blood. This research increases our understanding of S. suis virulence mechanisms and has important implications for the design of future control strategies including the development of anti-infective strategies by modulating animal feed composition.


Subject(s)
Bacterial Proteins/genetics , Gene Expression Regulation, Bacterial , Streptococcal Infections/veterinary , Streptococcus suis/genetics , Streptococcus suis/pathogenicity , Swine Diseases/microbiology , Animals , Bacterial Proteins/metabolism , Base Sequence , Epithelial Cells/microbiology , Gene Expression Profiling , Glucans/metabolism , Glucose/metabolism , Hemolysin Proteins/biosynthesis , Hemolysin Proteins/metabolism , Humans , Lactose/metabolism , Models, Biological , Molecular Sequence Annotation , Molecular Sequence Data , Oropharynx/microbiology , Streptococcal Infections/metabolism , Streptococcal Infections/microbiology , Streptococcus suis/metabolism , Swine , Swine Diseases/metabolism , Trisaccharides/metabolism , Virulence
19.
Mol Microbiol ; 92(1): 61-83, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24673665

ABSTRACT

Streptococcus suis (S. suis) is a neglected zoonotic streptococcus causing fatal diseases in humans and in pigs. The transcriptional regulator CcpA (catabolite control protein A) is involved in the metabolic adaptation to different carbohydrate sources and virulence of S. suis and other pathogenic streptococci. In this study, we determined the DNA binding characteristics of CcpA and identified the CcpA regulon during growth of S. suis. Electrophoretic mobility shift analyses showed promiscuous DNA binding of CcpA to cognate cre sites in vitro. In contrast, sequencing of immunoprecipitated chromatin revealed two specific consensus motifs, a pseudo-palindromic cre motif (WWGAAARCGYTTTCWW) and a novel cre2 motif (TTTTYHWDHHWWTTTY), within the regulatory elements of the genes directly controlled by CcpA. Via these elements CcpA regulates expression of genes involved in carbohydrate uptake and conversion, and in addition in important metabolic pathways of the central carbon metabolism, like glycolysis, mixed-acid fermentation, and the fragmentary TCA cycle. Furthermore, our analyses provide evidence that CcpA regulates the genes of the central carbon metabolism by binding either the pseudo-palindromic cre motif or the cre2 motif in a HPr(Ser)∼P independent conformation.


Subject(s)
Bacterial Proteins/chemistry , Bacterial Proteins/metabolism , Carbohydrate Metabolism , Repressor Proteins/chemistry , Repressor Proteins/metabolism , Streptococcus suis/genetics , Amino Acid Motifs , Amino Acid Sequence , Bacterial Proteins/genetics , Binding Sites , Consensus Sequence , DNA, Bacterial/metabolism , Electrophoretic Mobility Shift Assay , Gene Expression Regulation, Bacterial , Gene Knockout Techniques , Genes, Bacterial , Integrases/chemistry , Integrases/metabolism , Regulon , Repressor Proteins/genetics , Streptococcus suis/growth & development
20.
J Dairy Sci ; 96(6): 3723-36, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23587380

ABSTRACT

Streptococcus uberis is a highly prevalent causative agent of bovine mastitis, which leads to large economic losses in the dairy industry. The aim of this study was to examine the host response during acute inflammation after experimental challenge with capsulated Strep. uberis. Gene expression in response to Strep. uberis was compared between infected and control quarters in 3 animals. All quarters (n=16) were sampled at 16 different locations. Microarray data showed that 239 genes were differentially expressed between infected and control quarters. No differences in gene expression were observed between the different locations. Microarray data were confirmed for several genes using quantitative PCR analysis. Genes differentially expressed due to early Strep. uberis mastitis represented several stages of the process of infection: (1) pathogen recognition; (2) chemoattraction of neutrophils; (3) tissue repair mechanisms; and (4) bactericidal activity. Three different pathogen recognition genes were induced: ficolins, lipopolysaccharide binding protein, and toll-like receptor 2. Calgranulins were found to be the most strongly upregulated genes during early inflammation. By histology and immunohistochemistry, we demonstrated that changes in gene expression in response to Strep. uberis were induced both in infiltrating somatic milk cells and in mammary epithelial cells, demonstrating that the latter cell type plays a role in milk production as well as immune responsiveness. Given the rapid development of inflammation or mastitis after infection, early diagnosis of (Strep. uberis) mastitis is required for prevention of disease and spread of the pathogen. Insight into host responses could help to design immunomodulatory therapies to dampen inflammation after (early) diagnosis of Strep. uberis mastitis. Future research should focus on development of these early diagnostics and immunomodulatory components for mastitis treatment.


Subject(s)
Mammary Glands, Animal/microbiology , Mastitis, Bovine/microbiology , Streptococcal Infections/veterinary , Streptococcus/immunology , Acute-Phase Proteins/genetics , Animals , Carrier Proteins/genetics , Cattle , Cell Count , Epithelial Cells/immunology , Female , Gene Expression , Humans , Inflammation/immunology , Inflammation/microbiology , Lectins/genetics , Mammary Glands, Animal/immunology , Mammary Glands, Animal/pathology , Mastitis, Bovine/immunology , Mastitis, Bovine/metabolism , Membrane Glycoproteins/genetics , Microarray Analysis/veterinary , Milk/cytology , Polymerase Chain Reaction/veterinary , Streptococcal Infections/immunology , Streptococcal Infections/metabolism , Toll-Like Receptor 2/genetics , Ficolins
SELECTION OF CITATIONS
SEARCH DETAIL
...