Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 111
Filter
1.
PLoS One ; 18(11): e0294868, 2023.
Article in English | MEDLINE | ID: mdl-38033043

ABSTRACT

Streptococcus gallolyticus sp. gallolyticus (SGG) is a gut pathobiont involved in the development of colorectal cancer (CRC). To decipher SGG contribution in tumor initiation and/or acceleration respectively, a global transcriptome was performed in human normal colonic cells (FHC) and in human tumoral colonic cells (HT29). To identify SGG-specific alterations, we chose the phylogenetically closest relative, Streptococcus gallolyticus subsp. macedonicus (SGM) as control bacterium. We show that SGM, a bacterium generally considered as safe, did not induce any transcriptional changes on the two human colonic cells. The transcriptional reprogramming induced by SGG in normal FHC and tumoral HT29 cells was significantly different, although most of the genes up- and down-regulated were associated with cancer disease. Top up-regulated genes related to cancer were: (i) IL-20, CLK1, SORBS2, ERG1, PIM1, SNORD3A for normal FHC cells and (ii) TSLP, BHLHA15, LAMP3, ZNF27B, KRT17, ATF3 for cancerous HT29 cells. The total number of altered genes were much higher in cancerous than in normal colonic cells (2,090 vs 128 genes being affected, respectively). Gene set enrichment analysis reveals that SGG-induced strong ER- (endoplasmic reticulum) stress and UPR- (unfolded protein response) activation in colonic epithelial cells. Our results suggest that SGG induces a pro-tumoral shift in human colonic cells particularly in transformed cells potentially accelerating tumor development in the colon.


Subject(s)
Colorectal Neoplasms , Streptococcal Infections , Streptococcus gallolyticus subspecies gallolyticus , Humans , Colorectal Neoplasms/microbiology , Streptococcus , Gene Expression Profiling , Streptococcal Infections/microbiology , Streptococcus gallolyticus/genetics
3.
Sci Rep ; 13(1): 14960, 2023 09 11.
Article in English | MEDLINE | ID: mdl-37696912

ABSTRACT

In this work, we investigated the oncogenic role of Streptococcus gallolyticus subsp. gallolyticus (SGG), a gut bacterium associated with colorectal cancer (CRC). We showed that SGG UCN34 accelerates colon tumor development in a chemically induced CRC murine model. Full proteome and phosphoproteome analysis of murine colons chronically colonized by SGG UCN34 revealed that 164 proteins and 725 phosphorylation sites were differentially regulated. Ingenuity Pathway Analysis (IPA) indicates a pro-tumoral shift specifically induced by SGG UCN34, as ~ 90% of proteins and phosphoproteins identified were associated with digestive cancer. Comprehensive analysis of the altered phosphoproteins using ROMA software revealed up-regulation of several cancer hallmark pathways such as MAPK, mTOR and integrin/ILK/actin, affecting epithelial and stromal colonic cells. Importantly, an independent analysis of protein arrays of human colon tumors colonized with SGG showed up-regulation of PI3K/Akt/mTOR and MAPK pathways, providing clinical relevance to our findings. To test SGG's capacity to induce pre-cancerous transformation of the murine colonic epithelium, we grew ex vivo organoids which revealed unusual structures with compact morphology. Taken together, our results demonstrate the oncogenic role of SGG UCN34 in a murine model of CRC associated with activation of multiple cancer-related signaling pathways.


Subject(s)
Colonic Neoplasms , Streptococcus gallolyticus subspecies gallolyticus , Humans , Animals , Mice , Disease Models, Animal , Phosphatidylinositol 3-Kinases , Proteomics , TOR Serine-Threonine Kinases , Phosphoproteins , Proteome , Signal Transduction
4.
Microbiol Spectr ; : e0508522, 2023 Mar 23.
Article in English | MEDLINE | ID: mdl-36951576

ABSTRACT

Streptococcus gallolyticus subsp. gallolyticus (SGG) is an opportunistic gut pathogen associated with colorectal cancer. We previously showed that colonization of the murine colon by SGG in tumoral conditions was strongly enhanced by the production of gallocin A, a two-peptide bacteriocin. Here, we aimed to characterize the mechanisms of its action and resistance. Using a genetic approach, we demonstrated that gallocin A is composed of two peptides, GllA1 and GllA2, which are inactive alone and act together to kill "target" bacteria. We showed that gallocin A can kill phylogenetically close relatives of the pathogen. Importantly, we demonstrated that gallocin A peptides can insert themselves into membranes and permeabilize lipid bilayer vesicles. Next, we showed that the third gene of the gallocin A operon, gip, is necessary and sufficient to confer immunity to gallocin A. Structural modeling of GllA1 and GllA2 mature peptides suggested that both peptides form alpha-helical hairpins stabilized by intramolecular disulfide bridges. The presence of a disulfide bond in GllA1 and GllA2 was confirmed experimentally. Addition of disulfide-reducing agents abrogated gallocin A activity. Likewise, deletion of a gene encoding a surface protein with a thioredoxin-like domain impaired the ability of gallocin A to kill Enterococcus faecalis. Structural modeling of GIP revealed a hairpin-like structure strongly resembling those of the GllA1 and GllA2 mature peptides, suggesting a mechanism of immunity by competition with GllA1/2. Finally, identification of other class IIb bacteriocins exhibiting a similar alpha-helical hairpin fold stabilized with an intramolecular disulfide bridge suggests the existence of a new subclass of class IIb bacteriocins. IMPORTANCE Streptococcus gallolyticus subsp. gallolyticus (SGG), previously named Streptococcus bovis biotype I, is an opportunistic pathogen responsible for invasive infections (septicemia, endocarditis) in elderly people and is often associated with colon tumors. SGG is one of the first bacteria to be associated with the occurrence of colorectal cancer in humans. Previously, we showed that tumor-associated conditions in the colon provide SGG with an ideal environment to proliferate at the expense of phylogenetically and metabolically closely related commensal bacteria such as enterococci (1). SGG takes advantage of CRC-associated conditions to outcompete and substitute commensal members of the gut microbiota using a specific bacteriocin named gallocin, recently renamed gallocin A following the discovery of gallocin D in a peculiar SGG isolate. Here, we showed that gallocin A is a two-peptide bacteriocin and that both GllA1 and GllA2 peptides are required for antimicrobial activity. Gallocin A was shown to permeabilize bacterial membranes and kill phylogenetically closely related bacteria such as most streptococci, lactococci, and enterococci, probably through membrane pore formation. GllA1 and GllA2 secreted peptides are unusually long (42 and 60 amino acids long) and have very few charged amino acids compared to well-known class IIb bacteriocins. In silico modeling revealed that both GllA1 and GllA2 exhibit a similar hairpin-like conformation stabilized by an intramolecular disulfide bond. We also showed that the GIP immunity peptide forms a hairpin-like structure similar to GllA1/GllA2. Thus, we hypothesize that GIP blocks the formation of the GllA1/GllA2 complex by interacting with GllA1 or GllA2. Gallocin A may constitute the first class IIb bacteriocin which displays disulfide bridges important for its structure and activity and might be the founding member of a subtype of class IIb bacteriocins.

5.
FEBS J ; 290(11): 2968-2992, 2023 06.
Article in English | MEDLINE | ID: mdl-36629470

ABSTRACT

Cyclic di-AMP is an essential signalling molecule in Gram-positive bacteria. This second messenger regulates the osmotic pressure of the cell by interacting directly with the regulatory domains, either RCK_C or CBS domains, of several potassium and osmolyte uptake membrane protein systems. Cyclic di-AMP also targets stand-alone CBS domain proteins such as DarB in Bacillus subtilis and CbpB in Listeria monocytogenes. We show here that the CbpB protein of Group B Streptococcus binds c-di-AMP with a very high affinity. Crystal structures of CbpB reveal the determinants of binding specificity and significant conformational changes occurring upon c-di-AMP binding. Deletion of the cbpB gene alters bacterial growth in low potassium conditions most likely due to a decrease in the amount of ppGpp caused by a loss of interaction between CbpB and Rel, the GTP/GDP pyrophosphokinase.


Subject(s)
Carrier Proteins , Streptococcus agalactiae , Streptococcus agalactiae/genetics , Streptococcus agalactiae/metabolism , Guanosine Pentaphosphate , Guanosine Tetraphosphate , Bacterial Proteins/metabolism , Cyclic AMP , Dinucleoside Phosphates/metabolism , Potassium/metabolism
6.
Front Cell Infect Microbiol ; 12: 794391, 2022.
Article in English | MEDLINE | ID: mdl-35360109

ABSTRACT

Purpose: Streptococcus gallolyticus subspecies gallolyticus (SGG) is an opportunistic pathogen causing invasive infections in the elderly often associated with colon neoplasia. The prevalence of SGG in the stools of patients with normal colonoscopy (control) was compared with patients with colorectal adenomas (CRA) or with carcinomas (CRC) from stages I to IV. The presence of the pks island encoding colibactin as well as other CRC-associated bacteria such as toxicogenic Bacteroides fragilis, Fusobacterium nucleatum, and Parvimonas micra was also investigated. Patients and Methods: Fecal samples collected in France between 2011 and 2016 from patients with normal colonoscopy (n = 25), adenoma (n = 23), or colorectal cancer at different stages (n = 81) were tested by PCR for the presence of SGG, B. fragilis, F. nucleatum, P. micra, and the pks island. Relative quantification of SGG, F. nucleatum, and P. micra in stools was performed by qPCR. Results: SGG prevalence was significantly increased in the CRC group. Our results also revealed i) a strong and significant increase of toxinogenic B. fragilis in patients with early-stage adenoma and of pks island at late-stage CRC and ii) increased levels of F. nucleatum and P. micra in the stools of CRC patients. Furthermore, the simultaneous detection of these five bacterial markers was only found in CRC patients. Conclusions: Our results indicate that the prevalence or relative levels of CRC-associated bacteria vary during CRC development. Among them, B. fragilis (bft+) was singled out as the sole pathobiont detected at the early adenoma stage.


Subject(s)
Bacterial Infections , Carcinoma , Aged , Bacteria , Bacteroides fragilis/genetics , Humans , Streptococcus gallolyticus
7.
Brain Behav Immun ; 103: 186-201, 2022 07.
Article in English | MEDLINE | ID: mdl-35427758

ABSTRACT

Group B Streptococcus (GBS) remains a major neonatal life-threatening pathogen. We initially identified glyceraldehyde-3-phosphate dehydrogenase (GAPDH) as a promising vaccine candidate against GBS. Since GAPDH is highly conserved, we investigate whether GBS GAPDH maternal vaccination interferes with the intestinal colonization of the offspring and the development of its mucosal immune system and central nervous system. An altered gut microbiome with increased Proteobacteria is observed in pups born from vaccinated dams during early life. These pups present decreased relative expression of IL-1ß, IL-17A, RegIIIγ and MUC2 in the distal colon. They also display increased CD11b, F4/80 and MHC class II expression on microglia in early life and marked reduction of Ly6C+ cells and neutrophils. Importantly, male mice born from vaccinated mothers present behavioral abnormalities during adulthood, including decreased exploratory behavior, a subtle anxious-like phenotype and global alterations in spatial learning and memory strategies, and higher sensitivity to a stressful stimulus. Our study highlights the danger of using ubiquitous antigens in maternal human vaccines against neonatal pathogens.


Subject(s)
Dysbiosis , Gastrointestinal Microbiome , Prenatal Exposure Delayed Effects , Streptococcal Vaccines , Animals , Dysbiosis/chemically induced , Female , Glyceraldehyde-3-Phosphate Dehydrogenases/genetics , Male , Mice , Pregnancy , Prenatal Exposure Delayed Effects/microbiology , Streptococcal Vaccines/adverse effects , Streptococcus agalactiae , Vaccination
8.
PLoS Genet ; 17(9): e1009761, 2021 09.
Article in English | MEDLINE | ID: mdl-34491998

ABSTRACT

Virulence of the neonatal pathogen Group B Streptococcus is under the control of the master regulator CovR. Inactivation of CovR is associated with large-scale transcriptome remodeling and impairs almost every step of the interaction between the pathogen and the host. However, transcriptome analyses suggested a plasticity of the CovR signaling pathway in clinical isolates leading to phenotypic heterogeneity in the bacterial population. In this study, we characterized the CovR regulatory network in a strain representative of the CC-17 hypervirulent lineage responsible of the majority of neonatal meningitis. Transcriptome and genome-wide binding analysis reveal the architecture of the CovR network characterized by the direct repression of a large array of virulence-associated genes and the extent of co-regulation at specific loci. Comparative functional analysis of the signaling network links strain-specificities to the regulation of the pan-genome, including the two specific hypervirulent adhesins and horizontally acquired genes, to mutations in CovR-regulated promoters, and to variability in CovR activation by phosphorylation. This regulatory adaptation occurs at the level of genes, promoters, and of CovR itself, and allows to globally reshape the expression of virulence genes. Overall, our results reveal the direct, coordinated, and strain-specific regulation of virulence genes by the master regulator CovR and suggest that the intra-species evolution of the signaling network is as important as the expression of specific virulence factors in the emergence of clone associated with specific diseases.


Subject(s)
Bacterial Proteins/physiology , Gene Regulatory Networks , Streptococcus agalactiae/pathogenicity , Virulence Factors/physiology , Virulence/genetics , Bacterial Proteins/genetics , Chromosomes, Bacterial , Genes, Bacterial , Host-Pathogen Interactions , Humans , Promoter Regions, Genetic , Prophages/genetics , Streptococcus agalactiae/genetics , Transcription, Genetic/physiology , Virulence Factors/genetics
9.
Am J Trop Med Hyg ; 105(5): 1339-1346, 2021 08 30.
Article in English | MEDLINE | ID: mdl-34460418

ABSTRACT

Maternal group B Streptococcus (GBS) colonization is a major risk factor for neonatal GBS infection. However, data on GBS are scarce in low- and middle-income countries. Using sociodemographic data and vaginal swabs collected from an international cohort of mothers and newborns, this study aimed to estimate the prevalence of GBS colonization among pregnant women in Madagascar (n = 1,603) and Senegal (n = 616). The prevalence was 5.0% (95% CI, 3.9-6.1) and 16.1% (95% CI, 13.1-19.0) in Madagascar and Senegal, respectively. No factors among sociodemographic characteristics, living conditions, and obstetric history were found to be associated independently with GBS colonization in both countries. This community-based study provides one of the first estimates of maternal GBS colonization among pregnant women from Madagascar and Senegal.


Subject(s)
Maternal Exposure/statistics & numerical data , Mothers/statistics & numerical data , Pregnancy Complications, Infectious/diagnosis , Pregnancy Complications, Infectious/epidemiology , Streptococcal Infections/diagnosis , Streptococcal Infections/epidemiology , Streptococcus/isolation & purification , Adult , Cohort Studies , Female , Humans , Infant, Newborn , Madagascar/epidemiology , Population Surveillance , Pregnancy , Pregnant Women , Prevalence , Senegal/epidemiology
10.
mBio ; 12(1)2021 02 02.
Article in English | MEDLINE | ID: mdl-33531402

ABSTRACT

Fatty acid biosynthesis (FASII) enzymes are considered valid targets for antimicrobial drug development against the human pathogen Staphylococcus aureus However, incorporation of host fatty acids confers FASII antibiotic adaptation that compromises prospective treatments. S. aureus adapts to FASII inhibitors by first entering a nonreplicative latency period, followed by outgrowth. Here, we used transcriptional fusions and direct metabolite measurements to investigate the factors that dictate the duration of latency prior to outgrowth. We show that stringent response induction leads to repression of FASII and phospholipid synthesis genes. (p)ppGpp induction inhibits synthesis of malonyl-CoA, a molecule that derepresses FapR, a key regulator of FASII and phospholipid synthesis. Anti-FASII treatment also triggers transient expression of (p)ppGpp-regulated genes during the anti-FASII latency phase, with concomitant repression of FapR regulon expression. These effects are reversed upon outgrowth. GTP depletion, a known consequence of the stringent response, also occurs during FASII latency, and is proposed as the common signal linking these responses. We next showed that anti-FASII treatment shifts malonyl-CoA distribution between its interactants FapR and FabD, toward FapR, increasing expression of the phospholipid synthesis genes plsX and plsC during outgrowth. We conclude that components of the stringent response dictate malonyl-CoA availability in S. aureus FASII regulation, and contribute to latency prior to anti-FASII-adapted outgrowth. A combinatory approach, coupling a (p)ppGpp inducer and an anti-FASII, blocks S. aureus outgrowth, opening perspectives for bi-therapy treatment.IMPORTANCEStaphylococcus aureus is a major human bacterial pathogen for which new inhibitors are urgently needed. Antibiotic development has centered on the fatty acid synthesis (FASII) pathway, which provides the building blocks for bacterial membrane phospholipids. However, S. aureus overcomes FASII inhibition and adapts to anti-FASII by using exogenous fatty acids that are abundant in host environments. This adaptation mechanism comprises a transient latency period followed by bacterial outgrowth. Here, we use metabolite sensors and promoter reporters to show that responses to stringent conditions and to FASII inhibition intersect, in that both involve GTP and malonyl-CoA. These two signaling molecules contribute to modulating the duration of latency prior to S. aureus adaptation outgrowth. We exploit these novel findings to propose a bi-therapy treatment against staphylococcal infections.


Subject(s)
Anti-Bacterial Agents/pharmacology , Fatty Acids/antagonists & inhibitors , Guanosine Pentaphosphate/physiology , Guanosine Triphosphate/physiology , Malonyl Coenzyme A/physiology , Staphylococcus aureus/drug effects , Adaptation, Physiological/drug effects , Fatty Acids/biosynthesis , Humans , Malonyl Coenzyme A/analysis , Mupirocin/pharmacology , Phospholipids/biosynthesis , Staphylococcal Infections/drug therapy , Staphylococcus aureus/physiology
11.
mBio ; 12(1)2021 01 05.
Article in English | MEDLINE | ID: mdl-33402539

ABSTRACT

Bacteriocins are natural antimicrobial peptides produced by bacteria to kill closely related competitors. The opportunistic pathogen Streptococcus gallolyticus subsp. gallolyticus was recently shown to outcompete commensal enterococci of the murine microbiota under tumoral conditions thanks to the production of a two-peptide bacteriocin named gallocin. Here, we identified four genes involved in the regulatory control of gallocin in S. gallolyticus subsp. gallolyticus UCN34 that encode a histidine kinase/response regulator two-component system (BlpH/BlpR), a secreted peptide (GSP [gallocin-stimulating peptide]), and a putative regulator of unknown function (BlpS). While BlpR is a typical 243-amino-acid (aa) response regulator possessing a phospho-receiver domain and a LytTR DNA-binding domain, BlpS is a 108-aa protein containing only a LytTR domain. Our results showed that the secreted peptide GSP activates the dedicated two-component system BlpH/BlpR to induce gallocin transcription. A genome-wide transcriptome analysis indicates that this regulatory system (GSP-BlpH/BlpR) is specific for bacteriocin production. Importantly, as opposed to BlpR, BlpS was shown to repress gallocin gene transcription. A conserved operator DNA sequence of 30 bp was found in all promoter regions regulated by BlpR and BlpS. Electrophoretic mobility shift assays (EMSA) and footprint assays showed direct and specific binding of BlpS and BlpR to various regulated promoter regions in a dose-dependent manner on this conserved sequence. Gallocin expression appears to be tightly controlled in S. gallolyticus subsp. gallolyticus by quorum sensing and antagonistic activity of 2 LytTR-containing proteins. Competition experiments in gut microbiota medium and 5% CO2 to mimic intestinal conditions demonstrate that gallocin is functional under these in vivo-like conditions.IMPORTANCEStreptococcus gallolyticus subsp. gallolyticus, formerly known as Streptococcus bovis biotype I, is an opportunistic pathogen causing septicemia and endocarditis in the elderly often associated with asymptomatic colonic neoplasia. Recent studies indicate that S. gallolyticus subsp. gallolyticus is both a driver and a passenger of colorectal cancer. We previously showed that S. gallolyticus subsp. gallolyticus produces a bacteriocin, termed gallocin, enabling colonization of the colon under tumoral conditions by outcompeting commensal members of the murine microbiota such as Enterococcus faecalis Here, we identified and extensively characterized a four-component system that regulates gallocin production. Gallocin gene transcription is activated by a secreted peptide pheromone (GSP) and a two-component signal transduction system composed of a transmembrane histidine kinase receptor (BlpH) and a cytosolic response regulator (BlpR). Finally, a DNA-binding protein (BlpS) was found to repress gallocin genes transcription, likely by antagonizing BlpR. Understanding gallocin regulation is crucial to prevent S. gallolyticus subsp. gallolyticus colon colonization under tumoral conditions.


Subject(s)
Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Bacteriocins/genetics , Gene Expression Regulation, Bacterial , Streptococcus gallolyticus/genetics , Streptococcus gallolyticus/metabolism , DNA-Binding Proteins/metabolism , Gastrointestinal Microbiome , Gene Expression Profiling , Genes, Bacterial/genetics , Genome, Bacterial , Histidine Kinase/genetics , Histidine Kinase/metabolism , Quorum Sensing , Streptococcal Infections/microbiology , Transcriptome
12.
Lancet Infect Dis ; 21(7): e191-e201, 2021 07.
Article in English | MEDLINE | ID: mdl-33347816

ABSTRACT

In a 1945 Nobel Lecture, Sir Alexander Fleming warned against the overuse of antibiotics, particularly in response to public pressure. In the subsequent decades, evidence has shown that bacteria can become resistant to almost any available molecule. One key question is how the emergence and dissemination of resistant bacteria or resistance genes can be delayed. Although some clinicians remain sceptical, in this Personal View, we argue that the prescription of fewer antibiotics and shorter treatment duration is just as effective as longer regimens that remain the current guideline. Additionally, we discuss the fact that shorter antibiotic treatments exert less selective pressure on microorganisms, preventing the development of resistance. By contrast, longer treatments associated with a strong selective pressure favour the emergence of resistant clones within commensal organisms. We also emphasise that more studies are needed to identify the optimal duration of antibiotic therapy for common infections, which is important for making changes to the current guidelines, and to identify clinical biomarkers to guide antibiotic treatment in both hospital and ambulatory settings.


Subject(s)
Anti-Bacterial Agents/therapeutic use , Drug Resistance, Bacterial , Practice Patterns, Physicians' , European Union , Humans , Methicillin-Resistant Staphylococcus aureus
13.
Microbes Infect ; 22(1): 55-59, 2020.
Article in English | MEDLINE | ID: mdl-31837399

ABSTRACT

Streptococcus gallolyticus is an opportunistic pathogen responsible for septicemia and endocarditis. We report that S. gallolyticus UCN34 adheres and crosses epithelial monolayers in a Pil3 dependent manner. Confocal images revealed a paracellular passage. Both the Δpil3 mutant and the Pil3+ overexpressing variant were unable to cross Caco-2 and T84 barriers. However, combining live Δpil3 mutant with fixed Pil3+ variant in a 9:1 ratio allowed efficient translocation of the Δpil3 mutant. These results demonstrate that heterogeneous expression of Pil3 plays a key role for UCN34 translocation across the intestinal barrier. Through this skilful strategy, S. gallolyticus probably evade host immune responses.


Subject(s)
Bacterial Translocation , Epithelial Cells/microbiology , Fimbriae, Bacterial/genetics , Streptococcus gallolyticus/physiology , Bacterial Adhesion , Caco-2 Cells , Cell Line, Tumor , Fimbriae, Bacterial/metabolism , Genetic Heterogeneity , Humans , Mutation
14.
Clin Infect Dis ; 69(10): 1740-1748, 2019 10 30.
Article in English | MEDLINE | ID: mdl-30946447

ABSTRACT

BACKGROUND: In infants, the mode of acquisition of CC17 group B Streptococcus (GBS), the hypervirulent clone responsible for late-onset disease (LOD), remains elusive. METHODS: In a prospective multicenter study in France, we evaluated GBS colonization in mother-baby pairs with 2 months of follow-up between 2012 and 2015. Criteria included positivity for GBS colonization at antenatal screening or at delivery. Maternal vaginal samples and infant oral cavity and stool samples were analyzed at delivery, 21 ± 7 days (D21), and 60 ± 7 days (D60) post-delivery. RESULTS: A total of 890 mother-baby pairs were analyzed. GBS colonized 7%, 21%, and 23% of the infants at birth, D21, and D60, respectively, of which 10%, 11%, and 13% were identified as CC17 GBS. Concordance between maternal and infant GBS type was 96%. At D21, the main risk factors for infant colonization by GBS were simultaneous maternal colonization of the vagina (odds ratio [OR], 4.50; 95% confidence interval [CI], 1.69-15.61) and breast milk (OR, 7.93; 95% CI, 3.81-17.14). Importantly, 38% (95% CI, 23%-56%) of infants colonized by CC17 GBS appeared colonized for the first time at D60 vs 18% (95% CI, 14%-24%; P < .049) of infants colonized by non-CC17 GBS. Multivariate analysis showed a higher risk for de novo infant colonization by CC17 at D60 than by other GBS (OR, 2.45; 95% CI, 1.02-5.88). CONCLUSIONS: The high incidence of CC17 GBS in LOD is likely due to an enhanced post-delivery mother-to-infant transmission.


Subject(s)
Infectious Disease Transmission, Vertical , Streptococcal Infections/microbiology , Streptococcus agalactiae/pathogenicity , Adult , Feces/microbiology , Female , France , Humans , Incidence , Infant , Longitudinal Studies , Male , Mothers , Mouth/microbiology , Pregnancy , Prospective Studies , Risk Factors , Streptococcus agalactiae/genetics , Vagina/microbiology , Virulence
15.
Microbes Infect ; 21(2): 99-103, 2019 03.
Article in English | MEDLINE | ID: mdl-30419351

ABSTRACT

The core PI-2b pilus present in "hypervirulent" ST-17 Streptococcus agalactiae strains consists of three pilin subunits (Spb1, Ap1 and Ap2) assembled by sortase SrtC1 and cell-wall anchored by Srt2. Spb1 was shown to be the major pilin and Ap2 the anchor pilin. Ap1 is a putative adhesin. Two additional genes, orf and lep, are part of this operon. The contribution of Lep and Ap1 to the biogenesis of the PI-2b pilus was investigated. Concerning the role of PI-2b, we found that higher PI-2b expression resulted in higher adherence to human brain endothelial cells and higher phagocytosis by human THP1 macrophages.


Subject(s)
Adhesins, Bacterial/metabolism , Bacterial Adhesion , Fimbriae, Bacterial/metabolism , Operon/genetics , Streptococcal Infections/microbiology , Streptococcus agalactiae/genetics , Adhesins, Bacterial/genetics , Cell Wall/metabolism , Endothelial Cells/microbiology , Humans , Macrophages/microbiology , Phagocytosis , Streptococcus agalactiae/pathogenicity , Streptococcus agalactiae/physiology
16.
Sci Rep ; 8(1): 14322, 2018 09 25.
Article in English | MEDLINE | ID: mdl-30254272

ABSTRACT

Streptococcus agalactiae (Group B Streptococcus or GBS) is a frequent cause of serious disease in newborns and adults. Epidemiological evidence indicates a strong association between GBS strains belonging to the hypervirulent CC17 clonal complex and the occurrence of meningitis in neonates. We investigate here the role of PbsP, a cell wall plasminogen binding protein, in colonization of the central nervous system by CC17 GBS. Deletion of pbsP selectively impaired the ability of the CC17 strain BM110 to colonize the mouse brain after intravenous challenge, despite its unchanged capacity to persist at high levels in the blood and to invade the kidneys. Moreover, immunization with a recombinant form of PbsP considerably reduced brain infection and lethality. In vitro, pbsP deletion markedly decreased plasmin-dependent transmigration of BM110 through brain microvascular endothelial cells. Although PbsP was modestly expressed in bacteria grown under standard laboratory conditions, pbsP expression was markedly upregulated during in vivo infection or upon contact with cultured brain endothelial cells. Collectively, our studies indicate that PbsP is a highly conserved Plg binding adhesin, which is functionally important for invasion of the central nervous system by the hypervirulent CC17 GBS. Moreover, this antigen is a promising candidate for inclusion in a universal GBS vaccine.


Subject(s)
Bacterial Proteins/metabolism , Brain/microbiology , Streptococcus agalactiae/metabolism , Streptococcus agalactiae/pathogenicity , Animals , Brain/cytology , Cell Movement , Endothelial Cells/cytology , Fibrinolysin/metabolism , Gene Expression Regulation, Bacterial , Mice , Streptococcus agalactiae/genetics , Virulence
17.
Nat Commun ; 9(1): 3138, 2018 08 07.
Article in English | MEDLINE | ID: mdl-30087335

ABSTRACT

Group B streptococcal (GBS) meningitis remains a devastating disease. The absence of an animal model reproducing the natural infectious process has limited our understanding of the disease and, consequently, delayed the development of effective treatments. We describe here a mouse model in which bacteria are transmitted to the offspring from vaginally colonised pregnant females, the natural route of infection. We show that GBS strain BM110, belonging to the CC17 clonal complex, is more virulent in this vertical transmission model than the isogenic mutant BM110∆cylE, which is deprived of hemolysin/cytolysin. Pups exposed to the more virulent strain exhibit higher mortality rates and lung inflammation than those exposed to the attenuated strain. Moreover, pups that survive to BM110 infection present neurological developmental disability, revealed by impaired learning performance and memory in adulthood. The use of this new mouse model, that reproduces key steps of GBS infection in newborns, will promote a better understanding of the physiopathology of GBS-induced meningitis.


Subject(s)
Disease Models, Animal , Infectious Disease Transmission, Vertical , Streptococcal Infections/physiopathology , Animals , Animals, Newborn , Behavior, Animal , Body Weight , Female , Hemolysin Proteins/chemistry , Inflammation , Male , Maze Learning , Meningitis/microbiology , Meningitis, Bacterial , Mice , Mice, Inbred BALB C , Perforin/chemistry , Pregnancy , Pregnancy, Animal , Streptococcal Infections/transmission , Streptococcus agalactiae/pathogenicity , Vagina/microbiology
18.
Mol Microbiol ; 110(1): 82-94, 2018 10.
Article in English | MEDLINE | ID: mdl-30030946

ABSTRACT

Binding of microbial pathogens to host vitronectin (Vtn) is a common theme in the pathogenesis of invasive infections. In this study, we characterized the role of Vtn in the invasion of mucosal epithelial cells by Streptococcus agalactiae (i.e. group B streptococcus or GBS), a frequent human pathogen. Moreover, we identified PbsP, a previously described plasminogen-binding protein of GBS, as a dual adhesin that can also interact with human Vtn through its streptococcal surface repeat (SSURE) domains. Deletion of the pbsP gene decreases both bacterial adhesion to Vtn-coated inert surfaces and the ability of GBS to interact with epithelial cells. Bacterial adherence to and invasion of epithelial cells were either inhibited or enhanced by cell pretreatment with, respectively, anti-Vtn antibodies or Vtn, confirming the role of Vtn as a GBS ligand on host cells. Finally, antibodies directed against the integrin αv subunit inhibited Vtn-dependent cell invasion by GBS. Collectively, these results indicate that Vtn acts as a bridge between the SSURE domains of PbsP on the GBS surface and host integrins to promote bacterial invasion of epithelial cells. Therefore, inhibition of interactions between PbsP and extracellular matrix components could represent a viable strategy to prevent colonization and invasive disease by GBS.


Subject(s)
Bacterial Proteins/metabolism , Integrin alphaV/metabolism , Streptococcal Infections/microbiology , Streptococcus agalactiae/metabolism , Streptococcus agalactiae/pathogenicity , Vitronectin/metabolism , A549 Cells , Bacterial Adhesion/genetics , Bacterial Proteins/genetics , Caco-2 Cells , Cell Wall/metabolism , Epithelial Cells/microbiology , Humans , Integrin alphaV/genetics , Protein Domains , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Streptococcus agalactiae/genetics , Vitronectin/genetics
19.
PLoS Genet ; 14(4): e1007342, 2018 04.
Article in English | MEDLINE | ID: mdl-29659565

ABSTRACT

Cyclic nucleotides are universally used as secondary messengers to control cellular physiology. Among these signalling molecules, cyclic di-adenosine monophosphate (c-di-AMP) is a specific bacterial second messenger recognized by host cells during infections and its synthesis is assumed to be necessary for bacterial growth by controlling a conserved and essential cellular function. In this study, we sought to identify the main c-di-AMP dependent pathway in Streptococcus agalactiae, the etiological agent of neonatal septicaemia and meningitis. By conditionally inactivating dacA, the only diadenyate cyclase gene, we confirm that c-di-AMP synthesis is essential in standard growth conditions. However, c-di-AMP synthesis becomes rapidly dispensable due to the accumulation of compensatory mutations. We identified several mutations restoring the viability of a ΔdacA mutant, in particular a loss-of-function mutation in the osmoprotectant transporter BusAB. Identification of c-di-AMP binding proteins revealed a conserved set of potassium and osmolyte transporters, as well as the BusR transcriptional factor. We showed that BusR negatively regulates busAB transcription by direct binding to the busAB promoter. Loss of BusR repression leads to a toxic busAB expression in absence of c-di-AMP if osmoprotectants, such as glycine betaine, are present in the medium. In contrast, deletion of the gdpP c-di-AMP phosphodiesterase leads to hyperosmotic susceptibility, a phenotype dependent on a functional BusR. Taken together, we demonstrate that c-di-AMP is essential for osmotic homeostasis and that the predominant mechanism is dependent on the c-di-AMP binding transcriptional factor BusR. The regulation of osmotic homeostasis is likely the conserved and essential function of c-di-AMP, but each species has evolved specific c-di-AMP mechanisms of osmoregulation to adapt to its environment.


Subject(s)
Dinucleoside Phosphates/metabolism , Osmoregulation/physiology , Streptococcus agalactiae/metabolism , ATP-Binding Cassette Transporters/genetics , ATP-Binding Cassette Transporters/metabolism , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Genes, Bacterial , Homeostasis/physiology , Host-Pathogen Interactions/physiology , Humans , Mutation , Osmoregulation/genetics , Phosphorus-Oxygen Lyases/genetics , Phosphorus-Oxygen Lyases/metabolism , Potassium/metabolism , Second Messenger Systems/physiology , Streptococcus agalactiae/genetics , Streptococcus agalactiae/growth & development
20.
Proc Natl Acad Sci U S A ; 115(2): E283-E291, 2018 01 09.
Article in English | MEDLINE | ID: mdl-29279402

ABSTRACT

Colonization by Streptococcus gallolyticus subsp. gallolyticus (SGG) is strongly associated with the occurrence of colorectal cancer (CRC). However, the factors leading to its successful colonization are unknown, and whether SGG influences the oncogenic process or benefits from the tumor-prone environment to prevail remains an open question. Here, we elucidate crucial steps that explain how CRC favors SGG colonization. By using mice genetically prone to CRC, we show that SGG colonization is 1,000-fold higher in tumor-bearing mice than in normal mice. This selective advantage occurs at the expense of resident intestinal enterococci. An SGG-specific locus encoding a bacteriocin ("gallocin") is shown to kill enterococci in vitro. Importantly, bile acids strongly enhance this bacteriocin activity in vivo, leading to greater SGG colonization. Constitutive activation of the Wnt pathway, one of the earliest signaling alterations in CRC, and the decreased expression of the bile acid apical transporter gene Slc10A2, as an effect of the Apc founding mutation, may thereby sustain intestinal colonization by SGG. We conclude that CRC-specific conditions promote SGG colonization of the gut by replacing commensal enterococci in their niche.


Subject(s)
Colorectal Neoplasms/metabolism , Gastrointestinal Tract/microbiology , Streptococcus gallolyticus/physiology , Adenoma , Animals , Bacteriocins/genetics , Bacteriocins/metabolism , Bile Acids and Salts/metabolism , Gene Expression Regulation , Humans , Mice , Organic Anion Transporters, Sodium-Dependent/genetics , Organic Anion Transporters, Sodium-Dependent/metabolism , Receptors, Notch/genetics , Receptors, Notch/metabolism , Symporters/genetics , Symporters/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...