Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 111
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
PLoS Genet ; 17(9): e1009761, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34491998

RESUMEN

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.


Asunto(s)
Proteínas Bacterianas/fisiología , Redes Reguladoras de Genes , Streptococcus agalactiae/patogenicidad , Factores de Virulencia/fisiología , Virulencia/genética , Proteínas Bacterianas/genética , Cromosomas Bacterianos , Genes Bacterianos , Interacciones Huésped-Patógeno , Humanos , Regiones Promotoras Genéticas , Profagos/genética , Streptococcus agalactiae/genética , Transcripción Genética/fisiología , Factores de Virulencia/genética
2.
Brain Behav Immun ; 103: 186-201, 2022 07.
Artículo en Inglés | MEDLINE | ID: mdl-35427758

RESUMEN

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.


Asunto(s)
Disbiosis , Microbioma Gastrointestinal , Efectos Tardíos de la Exposición Prenatal , Vacunas Estreptocócicas , Animales , Disbiosis/inducido químicamente , Femenino , Gliceraldehído-3-Fosfato Deshidrogenasas/genética , Masculino , Ratones , Embarazo , Efectos Tardíos de la Exposición Prenatal/microbiología , Vacunas Estreptocócicas/efectos adversos , Streptococcus agalactiae , Vacunación
3.
PLoS Genet ; 14(4): e1007342, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29659565

RESUMEN

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.


Asunto(s)
Fosfatos de Dinucleósidos/metabolismo , Osmorregulación/fisiología , Streptococcus agalactiae/metabolismo , Transportadoras de Casetes de Unión a ATP/genética , Transportadoras de Casetes de Unión a ATP/metabolismo , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Genes Bacterianos , Homeostasis/fisiología , Interacciones Huésped-Patógeno/fisiología , Humanos , Mutación , Osmorregulación/genética , Liasas de Fósforo-Oxígeno/genética , Liasas de Fósforo-Oxígeno/metabolismo , Potasio/metabolismo , Sistemas de Mensajero Secundario/fisiología , Streptococcus agalactiae/genética , Streptococcus agalactiae/crecimiento & desarrollo
4.
Proc Natl Acad Sci U S A ; 115(2): E283-E291, 2018 01 09.
Artículo en Inglés | MEDLINE | ID: mdl-29279402

RESUMEN

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.


Asunto(s)
Neoplasias Colorrectales/metabolismo , Tracto Gastrointestinal/microbiología , Streptococcus gallolyticus/fisiología , Adenoma , Animales , Bacteriocinas/genética , Bacteriocinas/metabolismo , Ácidos y Sales Biliares/metabolismo , Regulación de la Expresión Génica , Humanos , Ratones , Transportadores de Anión Orgánico Sodio-Dependiente/genética , Transportadores de Anión Orgánico Sodio-Dependiente/metabolismo , Receptores Notch/genética , Receptores Notch/metabolismo , Simportadores/genética , Simportadores/metabolismo
5.
Clin Infect Dis ; 69(10): 1740-1748, 2019 10 30.
Artículo en Inglés | MEDLINE | ID: mdl-30946447

RESUMEN

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.


Asunto(s)
Transmisión Vertical de Enfermedad Infecciosa , Infecciones Estreptocócicas/microbiología , Streptococcus agalactiae/patogenicidad , Adulto , Heces/microbiología , Femenino , Francia , Humanos , Incidencia , Lactante , Estudios Longitudinales , Masculino , Madres , Boca/microbiología , Embarazo , Estudios Prospectivos , Factores de Riesgo , Streptococcus agalactiae/genética , Vagina/microbiología , Virulencia
6.
Mol Microbiol ; 110(1): 82-94, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-30030946

RESUMEN

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.


Asunto(s)
Proteínas Bacterianas/metabolismo , Integrina alfaV/metabolismo , Infecciones Estreptocócicas/microbiología , Streptococcus agalactiae/metabolismo , Streptococcus agalactiae/patogenicidad , Vitronectina/metabolismo , Células A549 , Adhesión Bacteriana/genética , Proteínas Bacterianas/genética , Células CACO-2 , Pared Celular/metabolismo , Células Epiteliales/microbiología , Humanos , Integrina alfaV/genética , Dominios Proteicos , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Streptococcus agalactiae/genética , Vitronectina/genética
7.
J Immunol ; 196(6): 2733-41, 2016 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-26873993

RESUMEN

Streptococci are common human colonizers with a species-specific mucocutaneous distribution. At the same time, they are among the most important and most virulent invasive bacterial pathogens. Thus, site-specific cellular innate immunity, which is predominantly executed by resident and invading myeloid cells, has to be adapted with respect to streptococcal sensing, handling, and response. In this article, we show that TLR13 is the critical mouse macrophage (MΦ) receptor in the response to group B Streptococcus, both in bone marrow-derived MΦs and in mature tissue MΦs, such as those residing in the lamina propria of the colon and the dermis, as well as in microglia. In contrast, TLR13 and its chaperone UNC-93B are dispensable for a potent cytokine response of blood monocytes to group B Streptococcus, although monocytes serve as the key progenitors of intestinal and dermal MΦs. Furthermore, a specific role for TLR13 with respect to MΦ function is supported by the response to staphylococci, where TLR13 and UNC-93B limit the cytokine response in bone marrow-derived MΦs and microglia, but not in dermal MΦs. In summary, TLR13 is a critical and site-specific receptor in the single MΦ response to ß-hemolytic streptococci.


Asunto(s)
Macrófagos/fisiología , Proteínas de Transporte de Membrana/metabolismo , Infecciones Estreptocócicas/inmunología , Streptococcus agalactiae/inmunología , Receptores Toll-Like/metabolismo , Animales , Colon/patología , Citocinas/metabolismo , Hemólisis , Interacciones Huésped-Patógeno , Inmunidad Mucosa/genética , Inmunidad Mucosa/inmunología , Macrófagos/microbiología , Proteínas de Transporte de Membrana/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Microglía/patología , Especificidad de Órganos , Piel/patología , Receptores Toll-Like/genética
8.
Mol Microbiol ; 101(1): 27-41, 2016 07.
Artículo en Inglés | MEDLINE | ID: mdl-26888569

RESUMEN

Streptococcus agalactiae (Group B Streptococcus or GBS) is a leading cause of invasive infections in neonates whose virulence is dependent on its ability to interact with cells and host components. We here characterized a surface protein with a critical function in GBS pathophysiology. This adhesin, designated PbsP, possesses two Streptococcal Surface Repeat domains, a methionine and lysine-rich region, and a LPXTG cell wall-anchoring motif. PbsP mediates plasminogen (Plg) binding both in vitro and in vivo and we showed that cell surface-bound Plg can be activated into plasmin by tissue plasminogen activator to increase the bacterial extracellular proteolytic activity. Absence of PbsP results in a decreased bacterial transmigration across brain endothelial cells and impaired virulence in a murine model of infection. PbsP is conserved among the main GBS lineages and is a major plasminogen adhesin in non-CC17 GBS strains. Importantly, immunization of mice with recombinant PbsP confers protective immunity. Our results indicate that GBS have evolved different strategies to recruit Plg which indicates that the ability to acquire cell surface proteolytic activity is essential for the invasiveness of this bacterium.


Asunto(s)
Adhesinas Bacterianas/metabolismo , Plasminógeno/metabolismo , Streptococcus agalactiae/metabolismo , Secuencia de Aminoácidos , Animales , Adhesión Bacteriana/fisiología , Pared Celular/metabolismo , Células Endoteliales/metabolismo , Fibrinolisina/metabolismo , Humanos , Ratones , Unión Proteica , Infecciones Estreptocócicas/microbiología , Streptococcus/metabolismo , Streptococcus agalactiae/genética , Streptococcus agalactiae/patogenicidad , Virulencia
9.
Mol Microbiol ; 97(6): 1209-22, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26094503

RESUMEN

The Group B Streptococcus (GBS) 'hypervirulent' ST-17 clone is strongly associated with invasive neonatal meningitis. Comparative genome analyses revealed that the serine-rich repeat (Srr) glycoprotein Srr2 is a cell wall-anchored protein specific for ST-17 strains, the non-ST-17 isolates expressing Srr1. Here, we unravel the binding capacity of GBS Srr proteins to relevant components of the host fibrinolysis pathway. We demonstrate that: (i) Srr2 binds plasminogen and plasmin whereas Srr1 does not; (ii) the ability of ST-17 strains to bind fibrinogen reflects a high level surface display of Srr2 combined with a higher affinity of Srr2 than Srr1 to bind this ligand; and (iii) Srr2 binding to host plasma proteins results in the formation of bacterial aggregates that are efficiently endocytosed by phagocytes. Importantly, we show that Srr2 increased bacterial survival to phagocytic killing and bacterial persistence in a murine model of meningitis. We conclude that Srr2 is a multifaceted adhesin used by the ST-17 clone to hijack ligands of the host coagulation system, thereby contributing to bacterial dissemination and invasiveness, and ultimately to meningitis.


Asunto(s)
Adhesinas Bacterianas/metabolismo , Proteínas Bacterianas/metabolismo , Fibrinógeno/metabolismo , Plasminógeno/metabolismo , Streptococcus agalactiae/metabolismo , Streptococcus agalactiae/patogenicidad , Animales , Femenino , Fibrinolisina/metabolismo , Glicosiltransferasas/metabolismo , Ligandos , Ratones Endogámicos BALB C , Unión Proteica , Virulencia
10.
Antimicrob Agents Chemother ; 60(12): 7424-7430, 2016 12.
Artículo en Inglés | MEDLINE | ID: mdl-27736761

RESUMEN

Group B Streptococcus (GBS) is the leading cause of neonatal invasive infections and an emerging pathogen in the elderly. Our objectives were to describe the evolution of GBS resistance to antibiotics in France and to investigate the emergence of fluoroquinolone (FQ)-resistant isolates. A total of 8,757 unrelated GBS isolates were collected and tested for antibiotic susceptibility from 2007 to 2014 according to EUCAST recommendations. All isolates were susceptible to penicillin G, amoxicillin, and vancomycin. Resistance to macrolides decreased from 47.0% to 30.0%, whereas high-level resistance to aminoglycosides, especially amikacin, increased from 6.4% to 8.8% and 24 isolates (0.3%) were highly resistant to gentamicin. FQ resistance gradually increased from 0.2% in 2007 (n = 1) to 1.5% in 2014 (n = 18, P < 0.01). Capsular polysaccharide (CPS) genotyping, multilocus sequence typing, and sequencing of the quinolone resistance-determining region (QRDR) showed that GBS isolates of sequence type 19 (ST-19) CPS type V were largely overrepresented in FQ-resistant isolates (n = 30, 45.5%). All 30 strains displayed the same QRDR mutations and were often associated with cross-resistance to macrolides (93.3%) and gentamicin (30%). In conclusion, we report the rise of FQ- and aminoglycoside-resistant GBS in France over an 8-year study period, an evolution likely linked to the clonal expansion of ST-19 CPS V-resistant isolates. This study emphasizes the need for a continuous surveillance of GBS epidemiology and antibiotic susceptibility.


Asunto(s)
Antibacterianos/farmacología , Farmacorresistencia Bacteriana Múltiple/genética , Genes Bacterianos , Mutación , Infecciones Estreptocócicas/epidemiología , Streptococcus agalactiae/genética , Adulto , Aminoglicósidos/farmacología , Niño , Células Clonales , Femenino , Fluoroquinolonas/farmacología , Francia/epidemiología , Expresión Génica , Hospitales , Humanos , Lactante , Macrólidos/farmacología , Masculino , Pruebas de Sensibilidad Microbiana , Embarazo , Análisis de Secuencia de ADN , Infecciones Estreptocócicas/tratamiento farmacológico , Infecciones Estreptocócicas/microbiología , Streptococcus agalactiae/efectos de los fármacos , Streptococcus agalactiae/aislamiento & purificación
11.
J Clin Microbiol ; 54(1): 75-82, 2016 01.
Artículo en Inglés | MEDLINE | ID: mdl-26491182

RESUMEN

Group B Streptococcus (GBS) is a common commensal bacterium in adults, but is also the leading cause of invasive bacterial infections in neonates in developed countries. The ß-hemolysin/cytolysin (ß-h/c), which is always associated with the production of an orange-to-red pigment, is a major virulence factor that is also used for GBS diagnosis. A collection of 1,776 independent clinical GBS strains isolated in France between 2006 and 2013 was evaluated on specific medium for ß-h/c activity and pigment production. The genomic sequences of nonhemolytic and nonpigmented (NH/NP) strains were analyzed to identify the molecular basis of this phenotype. Gene deletions or complementations were carried out to confirm the genotype-phenotype association. Sixty-three GBS strains (3.5%) were NH/NP, and 47 of these (74.6%) originated from invasive infections, including bacteremia and meningitis, in neonates or adults. The mutations are localized predominantly in the cyl operon, encoding the ß-h/c pigment biosynthetic pathway and, in the abx1 gene, encoding a CovSR regulator partner. In conclusion, although usually associated with GBS virulence, ß-h/c pigment production is not absolutely required to cause human invasive infections. Caution should therefore be taken in the use of hemolysis and pigmentation as criteria for GBS diagnosis in routine clinical laboratory settings.


Asunto(s)
Proteínas Hemolisinas/análisis , Pigmentos Biológicos/análisis , Infecciones Estreptocócicas/microbiología , Streptococcus agalactiae/genética , Streptococcus agalactiae/aislamiento & purificación , Adulto , Técnicas Bacteriológicas , Medios de Cultivo/química , Francia/epidemiología , Eliminación de Gen , Estudios de Asociación Genética , Prueba de Complementación Genética , Genoma Bacteriano , Humanos , Recién Nacido , Análisis de Secuencia de ADN , Infecciones Estreptocócicas/epidemiología , Streptococcus agalactiae/clasificación
12.
PLoS Pathog ; 10(1): e1003860, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24453966

RESUMEN

The molecular triggers leading to virulence of a number of human-adapted commensal bacteria such as Streptococcus gallolyticus are largely unknown. This opportunistic pathogen is responsible for endocarditis in the elderly and associated with colorectal cancer. Colonization of damaged host tissues with exposed collagen, such as cardiac valves and pre-cancerous polyps, is mediated by appendages referred to as Pil1 pili. Populations of S. gallolyticus are heterogeneous with the majority of cells weakly piliated while a smaller fraction is hyper piliated. We provide genetic evidences that heterogeneous pil1 expression depends on a phase variation mechanism involving addition/deletion of GCAGA repeats that modifies the length of an upstream leader peptide. Synthesis of longer leader peptides potentiates the transcription of the pil1 genes through ribosome-induced destabilization of a premature stem-loop transcription terminator. This study describes, at the molecular level, a new regulatory mechanism combining phase variation in a leader peptide-encoding gene and transcription attenuation. This simple and robust mechanism controls a stochastic heterogeneous pilus expression, which is important for evading the host immune system while ensuring optimal tissue colonization.


Asunto(s)
Proteínas Fimbrias/biosíntesis , Fimbrias Bacterianas/metabolismo , Regulación Bacteriana de la Expresión Génica/fisiología , Streptococcus/metabolismo , Endocarditis Bacteriana/genética , Endocarditis Bacteriana/metabolismo , Proteínas Fimbrias/genética , Fimbrias Bacterianas/genética , Fimbrias Bacterianas/ultraestructura , Humanos , Procesos Estocásticos , Streptococcus/genética , Streptococcus/ultraestructura
13.
Mol Cell Proteomics ; 13(9): 2168-82, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-24797265

RESUMEN

Serine-rich (Srr) proteins exposed at the surface of Gram-positive bacteria are a family of adhesins that contribute to the virulence of pathogenic staphylococci and streptococci. Lectin-binding experiments have previously shown that Srr proteins are heavily glycosylated. We report here the first mass-spectrometry analysis of the glycosylation of Streptococcus agalactiae Srr1. After Srr1 enrichment and trypsin digestion, potential glycopeptides were identified in collision induced dissociation spectra using X! Tandem. The approach was then refined using higher energy collisional dissociation fragmentation which led to the simultaneous loss of sugar residues, production of diagnostic oxonium ions and backbone fragmentation for glycopeptides. This feature was exploited in a new open source software tool (SpectrumFinder) developed for this work. By combining these approaches, 27 glycopeptides corresponding to six different segments of the N-terminal region of Srr1 [93-639] were identified. Our data unambiguously indicate that the same protein residue can be modified with different glycan combinations including N-acetylhexosamine, hexose, and a novel modification that was identified as O-acetylated-N-acetylhexosamine. Lectin binding and monosaccharide composition analysis strongly suggested that HexNAc and Hex correspond to N-acetylglucosamine and glucose, respectively. The same protein segment can be modified with a variety of glycans generating a wide structural diversity of Srr1. Electron transfer dissociation was used to assign glycosylation sites leading to the unambiguous identification of six serines and one threonine residues. Analysis of purified Srr1 produced in mutant strains lacking accessory glycosyltransferase encoding genes demonstrates that O-GlcNAcylation is an initial step in Srr1 glycosylation that is likely required for subsequent decoration with Hex. In summary, our data obtained by a combination of fragmentation mass spectrometry techniques associated to a new software tool, demonstrate glycosylation heterogeneity of Srr1, characterize a new protein modification, and identify six glycosylation sites located in the N-terminal region of the protein.


Asunto(s)
Adhesinas Bacterianas/metabolismo , Adhesinas Bacterianas/química , Cromatografía Liquida , Glicopéptidos/química , Glicopéptidos/metabolismo , Glicosilación , Monosacáridos/análisis , Serina , Programas Informáticos , Streptococcus agalactiae/metabolismo , Espectrometría de Masas en Tándem
14.
J Infect Dis ; 212(10): 1646-55, 2015 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-26014801

RESUMEN

Streptococcus gallolyticus is an increasing cause of bacteremia and infective endocarditis in the elderly. Several epidemiological studies have associated the presence of this bacterium with colorectal cancer. We have studied the interaction of S. gallolyticus with human colonic cells. S. gallolyticus strain UCN34, adhered better to mucus-producing cells such as HT-29-MTX than to the parental HT-29 cells. Attachment to colonic mucus is dependent on the pil3 pilus operon, which is heterogeneously expressed in the wild-type UCN34 population. We constructed a pil3 deletion mutant in a Pil3 overexpressing variant (Pil3+) and were able to demonstrate the role of Pil3 pilus in binding to colonic mucus. Importantly, we showed that pil3 deletion mutant was unable to colonize mice colon as compared to the isogenic Pil3+ variant. Our findings establish for the first time a murine model of intestinal colonization by S. gallolyticus.


Asunto(s)
Adhesión Bacteriana , Colon/microbiología , Células Epiteliales/microbiología , Fimbrias Bacterianas/metabolismo , Moco/microbiología , Infecciones Estreptocócicas/microbiología , Streptococcus/fisiología , Adhesinas Bacterianas/genética , Adhesinas Bacterianas/metabolismo , Animales , Línea Celular , Proteínas Fimbrias/genética , Fimbrias Bacterianas/genética , Eliminación de Gen , Expresión Génica , Genes Bacterianos , Humanos , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Moco/metabolismo , Operón , Streptococcus/genética , Streptococcus/metabolismo
15.
J Biol Chem ; 289(9): 5479-89, 2014 Feb 28.
Artículo en Inglés | MEDLINE | ID: mdl-24429288

RESUMEN

Streptococcus agalactiae (Group B Streptococcus) is a commensal of the human intestine and vagina of adult women but is the leading cause of invasive infection in neonates. This Gram-positive bacterium displays a set of virulence-associated surface proteins involved in the interaction with the host, such as adhesion to host cells, invasion of tissues, or subversion of the immune system. In this study, we characterized a cell wall-localized protein as an ecto-5'-nucleoside diphosphate phosphohydrolase (NudP) involved in the degradation of extracellular nucleotides which are central mediators of the immune response. Biochemical characterization of recombinant NudP revealed a Mn(2+)-dependent ecto-5'-nucleotidase activity on ribo- and deoxyribonucleoside 5'-mono- and 5'-diphosphates with a substrate specificity different from that of known orthologous enzymes. Deletion of the gene coding the housekeeping enzyme sortase A led to the release of NudP into the culture supernatant, confirming that this enzyme is anchored to the cell wall by its non-canonical LPXTN motif. The NudP ecto-5'-nucleotidase activity is reminiscent of the reactions performed by the mammalian ectonucleotidases CD39 and CD73 involved in regulating the extracellular level of ATP and adenosine. We further demonstrated that the absence of NudP activity decreases bacterial survival in mouse blood, a process dependent on extracellular adenosine. In vivo assays in animal models of infection showed that NudP activity is critical for virulence. These results demonstrate that Group B Streptococcus expresses a specific ecto-5'-nucleotidase necessary for its pathogenicity and highlight the diversity of reactions performed by this enzyme family. These results suggest that bacterial pathogens have developed specialized strategies to subvert the mammalian immune response controlled by the extracellular nucleotide signaling pathways.


Asunto(s)
Adenosina/metabolismo , Viabilidad Microbiana , N-Glicosil Hidrolasas/metabolismo , Streptococcus agalactiae/enzimología , Adenosina/genética , Secuencias de Aminoácidos , Animales , Femenino , Humanos , Ratones , Ratones Endogámicos BALB C , N-Glicosil Hidrolasas/genética , N-Glicosil Hidrolasas/inmunología , Ratas , Ratas Sprague-Dawley , Transducción de Señal/fisiología , Streptococcus agalactiae/genética , Streptococcus agalactiae/inmunología
16.
J Biol Chem ; 289(30): 21003-21015, 2014 Jul 25.
Artículo en Inglés | MEDLINE | ID: mdl-24904056

RESUMEN

Streptococcus agalactiae (group B Streptococcus or GBS) is a common cause of invasive infections in newborn infants and adults. The ability of GBS to bind human fibrinogen is of crucial importance in promoting colonization and invasion of host barriers. We characterized here a novel fibrinogen-binding protein of GBS, designated FbsC (Gbs0791), which is encoded by the prototype GBS strain NEM316. FbsC, which bears two bacterial immunoglobulin-like tandem repeat domains and a C-terminal cell wall-anchoring motif (LPXTG), was found to be covalently linked to the cell wall by the housekeeping sortase A. Studies using recombinant FbsC indicated that it binds fibrinogen in a dose-dependent and saturable manner, and with moderate affinity. Expression of FbsC was detected in all clinical GBS isolates, except those belonging to the hypervirulent lineage ST17. Deletion of fbsC decreases NEM316 abilities to adhere to and invade human epithelial and endothelial cells, and to form biofilm in vitro. Notably, bacterial adhesion to fibrinogen and fibrinogen binding to bacterial cells were abolished following fbsC deletion in NEM316. Moreover, the virulence of the fbsC deletion mutant and its ability to colonize the brain were impaired in murine models of infection. Finally, immunization with recombinant FbsC significantly protected mice from lethal GBS challenge. In conclusion, FbsC is a novel fibrinogen-binding protein expressed by most GBS isolates that functions as a virulence factor by promoting invasion of epithelial and endothelial barriers. In addition, the protein has significant immunoprotective activity and may be a useful component of an anti-GBS vaccine.


Asunto(s)
Proteínas Bacterianas/inmunología , Fibrinógeno/inmunología , Interacciones Huésped-Patógeno/inmunología , Infecciones Estreptocócicas/inmunología , Streptococcus agalactiae/fisiología , Factores de Virulencia/inmunología , Animales , Adhesión Bacteriana/genética , Adhesión Bacteriana/inmunología , Proteínas Bacterianas/genética , Células CACO-2 , Modelos Animales de Enfermedad , Células Endoteliales/inmunología , Células Endoteliales/microbiología , Células Endoteliales/patología , Células Epiteliales/inmunología , Células Epiteliales/microbiología , Células Epiteliales/patología , Fibrinógeno/genética , Humanos , Ratones , Unión Proteica/genética , Unión Proteica/inmunología , Infecciones Estreptocócicas/genética , Vacunas Estreptocócicas/genética , Vacunas Estreptocócicas/inmunología , Factores de Virulencia/genética
17.
J Biol Chem ; 289(20): 13701-5, 2014 May 16.
Artículo en Inglés | MEDLINE | ID: mdl-24692555

RESUMEN

The inflammatory cytokine IL-1ß is critical for host responses against many human pathogens. Here, we define Group B Streptococcus (GBS)-mediated activation of the Nod-like receptor-P3 (NLRP3) inflammasome in macrophages. NLRP3 activation requires GBS expression of the cytolytic toxin, ß-hemolysin, lysosomal acidification, and leakage. These processes allow the interaction of GBS RNA with cytosolic NLRP3. The present study supports a model in which GBS RNA, along with lysosomal components including cathepsins, leaks out of lysosomes and interacts with NLRP3 to induce IL-1ß production.


Asunto(s)
Proteínas Bacterianas/metabolismo , Proteínas Portadoras/metabolismo , Proteínas Hemolisinas/metabolismo , Inflamasomas/metabolismo , Interleucina-1beta/biosíntesis , Macrófagos/metabolismo , ARN Bacteriano/metabolismo , Streptococcus agalactiae/fisiología , Animales , Humanos , Interleucina-1beta/metabolismo , Lisosomas/metabolismo , Lisosomas/microbiología , Macrófagos/citología , Macrófagos/microbiología , Ratones , Proteína con Dominio Pirina 3 de la Familia NLR , Fagosomas/metabolismo , Fagosomas/microbiología , Streptococcus agalactiae/metabolismo
18.
BMC Genomics ; 16: 419, 2015 May 30.
Artículo en Inglés | MEDLINE | ID: mdl-26024923

RESUMEN

BACKGROUND: Streptococcus agalactiae, or Group B Streptococcus, is a leading cause of neonatal infections and an increasing cause of infections in adults with underlying diseases. In an effort to reconstruct the transcriptional networks involved in S. agalactiae physiology and pathogenesis, we performed an extensive and robust characterization of its transcriptome through a combination of differential RNA-sequencing in eight different growth conditions or genetic backgrounds and strand-specific RNA-sequencing. RESULTS: Our study identified 1,210 transcription start sites (TSSs) and 655 transcript ends as well as 39 riboswitches and cis-regulatory regions, 39 cis-antisense non-coding RNAs and 47 small RNAs potentially acting in trans. Among these putative regulatory RNAs, ten were differentially expressed in response to an acid stress and two riboswitches sensed directly or indirectly the pH modification. Strikingly, 15% of the TSSs identified were associated with the incorporation of pseudo-templated nucleotides, showing that reiterative transcription is a pervasive process in S. agalactiae. In particular, 40% of the TSSs upstream genes involved in nucleotide metabolism show reiterative transcription potentially regulating gene expression, as exemplified for pyrG and thyA encoding the CTP synthase and the thymidylate synthase respectively. CONCLUSIONS: This comprehensive map of the transcriptome at the single nucleotide resolution led to the discovery of new regulatory mechanisms in S. agalactiae. It also provides the basis for in depth analyses of transcriptional networks in S. agalactiae and of the regulatory role of reiterative transcription following variations of intra-cellular nucleotide pools.


Asunto(s)
Nucleótidos/análisis , ARN Mensajero/análisis , Streptococcus agalactiae/genética , Perfilación de la Expresión Génica/métodos , Regulación Bacteriana de la Expresión Génica , Redes Reguladoras de Genes , Genes Bacterianos , Secuenciación de Nucleótidos de Alto Rendimiento/métodos , ARN Bacteriano/análisis , Análisis de Secuencia de ARN/métodos , Streptococcus agalactiae/crecimiento & desarrollo
19.
PLoS Pathog ; 9(2): e1003179, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23436996

RESUMEN

Group B Streptococcus (GBS), a common commensal of the female genital tract, is the leading cause of invasive infections in neonates. Expression of major GBS virulence factors, such as the hemolysin operon cyl, is regulated directly at the transcriptional level by the CovSR two-component system. Using a random genetic approach, we identified a multi-spanning transmembrane protein, Abx1, essential for the production of the GBS hemolysin. Despite its similarity to eukaryotic CaaX proteases, the Abx1 function is not involved in a post-translational modification of the GBS hemolysin. Instead, we demonstrate that Abx1 regulates transcription of several virulence genes, including those comprising the hemolysin operon, by a CovSR-dependent mechanism. By combining genetic analyses, transcriptome profiling, and site-directed mutagenesis, we showed that Abx1 is a regulator of the histidine kinase CovS. Overexpression of Abx1 is sufficient to activate virulence gene expression through CovS, overcoming the need for an additional signal. Conversely, the absence of Abx1 has the opposite effect on virulence gene expression consistent with CovS locked in a kinase-competent state. Using a bacterial two-hybrid system, direct interaction between Abx1 and CovS was mapped specifically to CovS domains involved in signal processing. We demonstrate that the CovSR two-component system is the core of a signaling pathway integrating the regulation of CovS by Abx1 in addition to the regulation of CovR by the serine/threonine kinase Stk1. In conclusion, our study reports a regulatory function for Abx1, a member of a large protein family with a characteristic Abi-domain, which forms a signaling complex with the histidine kinase CovS in GBS.


Asunto(s)
Proteínas Bacterianas/genética , Regulación Bacteriana de la Expresión Génica , Transducción de Señal , Infecciones Estreptocócicas/microbiología , Streptococcus agalactiae/genética , Secuencia de Aminoácidos , Animales , Proteínas Bacterianas/metabolismo , Epistasis Genética , Femenino , Perfilación de la Expresión Génica , Hemólisis , Histidina Quinasa , Humanos , Modelos Biológicos , Datos de Secuencia Molecular , Mutación , Análisis de Secuencia por Matrices de Oligonucleótidos , Fosfoproteínas Fosfatasas/genética , Fosfoproteínas Fosfatasas/metabolismo , Pigmentos Biológicos/metabolismo , Mapeo de Interacción de Proteínas , Proteínas Quinasas/genética , Proteínas Quinasas/metabolismo , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Estructura Terciaria de Proteína , Ratas , Alineación de Secuencia , Streptococcus agalactiae/metabolismo , Streptococcus agalactiae/patogenicidad , Virulencia/genética , Factores de Virulencia/genética
20.
J Immunol ; 191(9): 4759-68, 2013 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-24078699

RESUMEN

Sepsis is the third most common cause of neonatal death, with Group B Streptococcus (GBS) being the leading bacterial agent. The pathogenesis of neonatal septicemia is still unsolved. We described previously that host susceptibility to GBS infection is due to early IL-10 production. In this study, we investigated whether triggering TLR2 to produce IL-10 is a risk factor for neonatal bacterial sepsis. We observed that, in contrast to wild-type (WT) pups, neonatal TLR2-deficient mice were resistant to GBS-induced sepsis. Moreover, if IL-10 signaling were blocked in WT mice, they also were resistant to sepsis. This increased survival rate was due to an efficient recruitment of neutrophils to infected tissues that leads to bacterial clearance, thus preventing the development of sepsis. To confirm that IL-10 produced through TLR2 activation prevents neutrophil recruitment, WT pups were treated with the TLR2 agonist Pam3CSK4 prior to nebulization with the neutrophil chemotactic agent LTB4. Neutrophil recruitment into the neonatal lungs was inhibited in pups treated with Pam3CSK4. However, the migration was restored in Pam3CSK4-treated pups when IL-10 signaling was blocked (either by anti-IL-10R mAb treatment or by using IL-10-deficient mice). Our findings highlight that TLR2-induced IL-10 production is a key event in neonatal susceptibility to bacterial sepsis.


Asunto(s)
Interleucina-10/metabolismo , Infiltración Neutrófila/inmunología , Neutrófilos/inmunología , Sepsis/inmunología , Receptor Toll-Like 2/metabolismo , Animales , Movimiento Celular/inmunología , Femenino , Interleucina-10/antagonistas & inhibidores , Interleucina-10/genética , Leucotrieno B4 , Lipopéptidos/farmacología , Pulmón/inmunología , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Neutrófilos/metabolismo , Receptores de Interleucina-8B/biosíntesis , Sepsis/microbiología , Sepsis/mortalidad , Infecciones Estreptocócicas/inmunología , Streptococcus agalactiae/inmunología , Receptor Toll-Like 2/agonistas , Receptor Toll-Like 2/deficiencia , Receptor Toll-Like 2/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA