Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 58
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
Cell ; 156(1-2): 97-108, 2014 Jan 16.
Artículo en Inglés | MEDLINE | ID: mdl-24439371

RESUMEN

Successful infection depends on the ability of the pathogen to gain nutrients from the host. The extracellular pathogenic bacterium group A Streptococcus (GAS) causes a vast array of human diseases. By using the quorum-sensing sil system as a reporter, we found that, during adherence to host cells, GAS delivers streptolysin toxins, creating endoplasmic reticulum stress. This, in turn, increases asparagine (ASN) synthetase expression and the production of ASN. The released ASN is sensed by the bacteria, altering the expression of ∼17% of GAS genes of which about one-third are dependent on the two-component system TrxSR. The expression of the streptolysin toxins is strongly upregulated, whereas genes linked to proliferation are downregulated in ASN absence. Asparaginase, a widely used chemotherapeutic agent, arrests GAS growth in human blood and blocks GAS proliferation in a mouse model of human bacteremia. These results delineate a pathogenic pathway and propose a therapeutic strategy against GAS infections.


Asunto(s)
Percepción de Quorum , Infecciones Estreptocócicas/microbiología , Streptococcus/metabolismo , Animales , Asparagina/metabolismo , Aspartatoamoníaco Ligasa/genética , Aspartatoamoníaco Ligasa/metabolismo , Bacteriemia/microbiología , Modelos Animales de Enfermedad , Estrés del Retículo Endoplásmico , Células HeLa , Humanos , Masculino , Ratones , Ratones Endogámicos BALB C , Streptococcus/citología , Streptococcus/patogenicidad , Transcripción Genética , Factores de Virulencia/genética
2.
Infect Immun ; 92(6): e0008324, 2024 Jun 11.
Artículo en Inglés | MEDLINE | ID: mdl-38712951

RESUMEN

Streptococcus pyogenes [group A streptococcus (GAS)] is a human pathogen capable of infecting diverse tissues. To successfully infect these sites, GAS must detect available nutrients and adapt accordingly. The phosphoenolpyruvate transferase system (PTS) mediates carbohydrate uptake and metabolic gene regulation to adapt to the nutritional environment. Regulation by the PTS can occur through phosphorylation of transcriptional regulators at conserved PTS-regulatory domains (PRDs). GAS has several PRD-containing stand-alone regulators with regulons encoding both metabolic genes and virulence factors [PRD-containing virulence regulators (PCVRs)]. One is RofA, which regulates the expression of virulence genes in multiple GAS serotypes. It was hypothesized that RofA is phosphorylated by the PTS in response to carbohydrate levels to coordinate virulence gene expression. In this study, the RofA regulon of M1T1 strain 5448 was determined using RNA sequencing. Two operons were consistently differentially expressed across growth in the absence of RofA; the pilus operon was downregulated, and the capsule operon was upregulated. This correlated with increased capsule production and decreased adherence to keratinocytes. Purified RofA-His was phosphorylated in vitro by PTS proteins EI and HPr, and phosphorylated RofA-FLAG was detected in vivo when GAS was grown in low-glucose C medium. Phosphorylated RofA was not observed when C medium was supplemented 10-fold with glucose. Mutations of select histidine residues within the putative PRDs contributed to the in vivo phosphorylation of RofA, although phosphorylation of RofA was still observed, suggesting other phosphorylation sites exist in the protein. Together, these findings support the hypothesis that RofA is a PCVR that may couple sugar metabolism with virulence regulation.


Asunto(s)
Proteínas Bacterianas , Regulación Bacteriana de la Expresión Génica , Streptococcus pyogenes , Factores de Virulencia , Streptococcus pyogenes/patogenicidad , Streptococcus pyogenes/genética , Streptococcus pyogenes/metabolismo , Proteínas Bacterianas/metabolismo , Proteínas Bacterianas/genética , Factores de Virulencia/genética , Factores de Virulencia/metabolismo , Virulencia , Fosforilación , Humanos , Regulón , Operón , Infecciones Estreptocócicas/microbiología , Sistema de Fosfotransferasa de Azúcar del Fosfoenolpiruvato/metabolismo , Sistema de Fosfotransferasa de Azúcar del Fosfoenolpiruvato/genética , Queratinocitos/microbiología
3.
Mol Microbiol ; 117(2): 525-538, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-34923680

RESUMEN

Streptococcus pyogenes, also known as group A Streptococcus or GAS, is a human-restricted pathogen causing a diverse array of infections. The ability to adapt to different niches requires GAS to adjust gene expression in response to environmental cues. We previously identified the abundance of biometals and carbohydrates led to natural induction of the Rgg2/3 cell-cell communication system (quorum sensing, QS). Here we determined the mechanism by which the Rgg2/3 QS system is stimulated exclusively by mannose and repressed by glucose, a phenomenon known as carbon catabolite repression (CCR). Instead of carbon catabolite protein A, the primary mediator of CCR in Gram-positive bacteria; CCR of Rgg2/3 requires the PTS regulatory domain (PRD)-containing transcriptional regulator Mga. Deletion of Mga led to carbohydrate-independent activation of Rgg2/3 by down-regulating rgg3, the QS repressor. Through phosphoablative and phosphomimetic substitutions within Mga PRDs, we demonstrated that selective phosphorylation of PRD1 conferred repression of the Rgg2/3 system. Moreover, given the carbohydrate specificity mediating Mga-dependent governance over Rgg2/3, we tested mannose-specific PTS components and found the EIIA/B subunit ManL was required for Mga-dependent repression. These findings provide newfound connections between PTSMan , Mga, and QS, and further demonstrate that Mga is a central regulatory nexus for integrating nutritional status and virulence.


Asunto(s)
Represión Catabólica , Streptococcus pyogenes , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Regulación Bacteriana de la Expresión Génica/genética , Humanos , Percepción de Quorum/genética , Streptococcus pyogenes/metabolismo
4.
Infect Immun ; 89(4)2021 03 17.
Artículo en Inglés | MEDLINE | ID: mdl-33397818

RESUMEN

Streptococcus pneumoniae grows in biofilms during both asymptomatic colonization and infection. Pneumococcal biofilms on abiotic surfaces exhibit delayed growth and lower biomass and lack the structures seen on epithelial cells or during nasopharyngeal carriage. We show here that adding hemoglobin to the medium activated unusually early and vigorous biofilm growth in multiple S. pneumoniae serotypes grown in batch cultures on abiotic surfaces. Human blood (but not serum, heme, or iron) also stimulated biofilms, and the pore-forming pneumolysin, ply, was required for this induction. S. pneumoniae transitioning from planktonic into sessile growth in the presence of hemoglobin displayed an extensive transcriptome remodeling within 1 and 2 h. Differentially expressed genes included those involved in the metabolism of carbohydrates, nucleotides, amino acid, and lipids. The switch into adherent states also influenced the expression of several regulatory systems, including the comCDE genes. Inactivation of comC resulted in 67% reduction in biofilm formation, while the deletion of comD or comE had limited or no effect, respectively. These observations suggest a novel route for CSP-1 signaling independent of the cognate ComDE two-component system. Biofilm induction and the associated transcriptome remodeling suggest hemoglobin serves as a signal for host colonization in pneumococcus.


Asunto(s)
Proteínas Bacterianas/metabolismo , Biopelículas/crecimiento & desarrollo , Hemoglobinas/metabolismo , Interacciones Huésped-Patógeno , Infecciones Neumocócicas/microbiología , Streptococcus pneumoniae/fisiología , Células Sanguíneas/metabolismo , Humanos , Infecciones Neumocócicas/sangre , Infecciones Neumocócicas/metabolismo , Streptococcus pneumoniae/patogenicidad
5.
Nat Chem Biol ; 15(5): 463-471, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30936502

RESUMEN

Cell wall glycopolymers on the surface of Gram-positive bacteria are fundamental to bacterial physiology and infection biology. Here we identify gacH, a gene in the Streptococcus pyogenes group A carbohydrate (GAC) biosynthetic cluster, in two independent transposon library screens for its ability to confer resistance to zinc and susceptibility to the bactericidal enzyme human group IIA-secreted phospholipase A2. Subsequent structural and phylogenetic analysis of the GacH extracellular domain revealed that GacH represents an alternative class of glycerol phosphate transferase. We detected the presence of glycerol phosphate in the GAC, as well as the serotype c carbohydrate from Streptococcus mutans, which depended on the presence of the respective gacH homologs. Finally, nuclear magnetic resonance analysis of GAC confirmed that glycerol phosphate is attached to approximately 25% of the GAC N-acetylglucosamine side-chains at the C6 hydroxyl group. This previously unrecognized structural modification impacts host-pathogen interaction and has implications for vaccine design.


Asunto(s)
Glicerol/metabolismo , Fosfatos/metabolismo , Polisacáridos Bacterianos/metabolismo , Streptococcus/metabolismo , Glicerol/química , Fosfatos/química , Polisacáridos Bacterianos/química , Streptococcus/química
6.
Infect Immun ; 88(10)2020 09 18.
Artículo en Inglés | MEDLINE | ID: mdl-32719156

RESUMEN

Streptococcus pyogenes (group A Streptococcus [GAS]), a major human-specific pathogen, relies on efficient nutrient acquisition for successful infection within its host. The phosphotransferase system (PTS) couples the import of carbohydrates with their phosphorylation prior to metabolism and has been linked to GAS pathogenesis. In a screen of an insertional mutant library of all 14 annotated PTS permease (EIIC) genes in MGAS5005, the annotated ß-glucoside PTS transporter (bglP) was found to be crucial for GAS growth and survival in human blood and was validated in another M1T1 GAS strain, 5448. In 5448, bglP was shown to be in an operon with a putative phospho-ß-glucosidase (bglB) downstream and a predicted antiterminator (licT) upstream. Using defined nonpolar mutants of the ß-glucoside permease (bglP) and ß-glucosidase enzyme (bglB) in 5448, we showed that bglB, not bglP, was important for growth in blood. Furthermore, transcription of the licT-blgPB operon was found to be repressed by glucose and induced by the ß-glucoside salicin as the sole carbon source. Investigation of the individual bglP and bglB mutants determined that they influence in vitro growth in the ß-glucoside salicin; however, only bglP was necessary for growth in other non-ß-glucoside PTS sugars, such as fructose and mannose. Additionally, loss of BglP and BglB suggests that they are important for the regulation of virulence-related genes that control biofilm formation, streptolysin S (SLS)-mediated hemolysis, and localized ulcerative lesion progression during subcutaneous infections in mice. Thus, our results indicate that the ß-glucoside PTS transports salicin and its metabolism can differentially influence GAS pathophysiology during soft tissue infection.


Asunto(s)
Alcoholes Bencílicos/metabolismo , Glucósidos/metabolismo , Sistema de Fosfotransferasa de Azúcar del Fosfoenolpiruvato/metabolismo , Infecciones de los Tejidos Blandos/patología , Infecciones Estreptocócicas/patología , Streptococcus pyogenes/metabolismo , Streptococcus pyogenes/patogenicidad , Animales , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Biopelículas/crecimiento & desarrollo , Represión Catabólica , Regulación Bacteriana de la Expresión Génica , Hemólisis/genética , Humanos , Ratones , Viabilidad Microbiana/genética , Mutación , Operón , Sistema de Fosfotransferasa de Azúcar del Fosfoenolpiruvato/genética , Infecciones de los Tejidos Blandos/metabolismo , Infecciones de los Tejidos Blandos/microbiología , Infecciones Estreptocócicas/metabolismo , Infecciones Estreptocócicas/microbiología , Streptococcus pyogenes/genética , Streptococcus pyogenes/crecimiento & desarrollo , Azúcares/metabolismo , Virulencia/genética
7.
PLoS Pathog ; 14(10): e1007348, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-30321240

RESUMEN

Human Group IIA secreted phospholipase A2 (hGIIA) is an acute phase protein with bactericidal activity against Gram-positive bacteria. Infection models in hGIIA transgenic mice have suggested the importance of hGIIA as an innate defense mechanism against the human pathogens Group A Streptococcus (GAS) and Group B Streptococcus (GBS). Compared to other Gram-positive bacteria, GAS is remarkably resistant to hGIIA activity. To identify GAS resistance mechanisms, we exposed a highly saturated GAS M1 transposon library to recombinant hGIIA and compared relative mutant abundance with library input through transposon-sequencing (Tn-seq). Based on transposon prevalence in the output library, we identified nine genes, including dltA and lytR, conferring increased hGIIA susceptibility. In addition, seven genes conferred increased hGIIA resistance, which included two genes, gacH and gacI that are located within the Group A Carbohydrate (GAC) gene cluster. Using GAS 5448 wild-type and the isogenic gacI mutant and gacI-complemented strains, we demonstrate that loss of the GAC N-acetylglucosamine (GlcNAc) side chain in the ΔgacI mutant increases hGIIA resistance approximately 10-fold, a phenotype that is conserved across different GAS serotypes. Increased resistance is associated with delayed penetration of hGIIA through the cell wall. Correspondingly, loss of the Lancefield Group B Carbohydrate (GBC) rendered GBS significantly more resistant to hGIIA-mediated killing. This suggests that the streptococcal Lancefield antigens, which are critical determinants for streptococcal physiology and virulence, are required for the bactericidal enzyme hGIIA to exert its bactericidal function.


Asunto(s)
Antibacterianos/farmacología , Pared Celular/metabolismo , Fosfolipasas A2 Grupo II/inmunología , Inmunidad Innata/efectos de los fármacos , Polisacáridos Bacterianos/farmacología , Infecciones Estreptocócicas/microbiología , Streptococcus/inmunología , Actividad Bactericida de la Sangre , Fosfolipasas A2 Grupo II/sangre , Fosfolipasas A2 Grupo II/genética , Interacciones Huésped-Patógeno , Humanos , Infecciones Estreptocócicas/sangre , Infecciones Estreptocócicas/enzimología , Streptococcus/patogenicidad
8.
Infect Immun ; 87(12)2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31591169

RESUMEN

As a strict human pathogen, Streptococcus pyogenes (group A Streptococcus, or GAS) causes a wide range of infections, from superficial to life-threatening diseases, upon dissemination. Thus, it is necessary to gain a better understanding of how GAS successfully overcomes host-mediated challenges and infects various host niches. We previously identified subcutaneous fitness (scf) genes in the clinically relevant wild-type (WT) GAS M1T1 5448 strain that are critical for fitness during murine soft-tissue infection at both 24 h and 48 h postinfection. The uncharacterized locus scfCDE was transcribed as an operon and is predicted to encode an ABC importer for nutrient uptake (e.g., amino acids). Individual scfCDE deletion mutants grew comparably to WT 5448 in rich medium but exhibited reduced fitness during competitive growth in murine soft tissue and in nutrient-limiting chemically defined medium (CDM). A deletion of the permease gene scfD resulted in a monoculture growth defect in CDM that could be rescued by addition of excess peptides, suggesting a role as an amino acid importer. Interestingly, the ΔscfC substrate-binding and ΔscfD permease mutants, but not the ΔscfE ATPase mutant, were highly attenuated in murine soft tissue. Moreover, all three genes were required for GAS survival in human blood, indicating their impact is not limited to superficial infections. As such, scfCDE plays an integral role in enhancing GAS adaptation during localized infection as well as dissemination to deeper host environments. Since scfCDE is conserved throughout Firmicutes, this work may contribute to the development of therapeutic strategies against GAS and other Gram-positive pathogens.


Asunto(s)
Transportadoras de Casetes de Unión a ATP/genética , Infecciones Estreptocócicas/patología , Streptococcus pyogenes/genética , Streptococcus pyogenes/patogenicidad , Factores de Virulencia/genética , Transportadoras de Casetes de Unión a ATP/metabolismo , Animales , Regulación Bacteriana de la Expresión Génica , Ratones , Infecciones Estreptocócicas/genética , Virulencia/genética
9.
PLoS Pathog ; 13(8): e1006584, 2017 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-28832676

RESUMEN

The Group A Streptococcus remains a significant human pathogen causing a wide array of disease ranging from self-limiting to life-threatening invasive infections. Epithelium (skin or throat) colonization with progression to the subepithelial tissues is the common step in all GAS infections. Here, we used transposon-sequencing (Tn-seq) to define the GAS 5448 genetic requirements for in vivo fitness in subepithelial tissue. A near-saturation transposon library of the M1T1 GAS 5448 strain was injected subcutaneously into mice, producing suppurative inflammation at 24 h that progressed to prominent abscesses with tissue necrosis at 48 h. The library composition was monitored en masse by Tn-seq and ratios of mutant abundance comparing the output (12, 24 and 48 h) versus input (T0) mutant pools were calculated for each gene. We identified a total of 273 subcutaneous fitness (scf) genes with 147 genes (55 of unknown function) critical for the M1T1 GAS 5448 fitness in vivo; and 126 genes (53 of unknown function) potentially linked to in vivo fitness advantage. Selected scf genes were validated in competitive subcutaneous infection with parental 5448. Two uncharacterized genes, scfA and scfB, encoding putative membrane-associated proteins and conserved among Gram-positive pathogens, were further characterized. Defined scfAB mutants in GAS were outcompeted by wild type 5448 in vivo, attenuated for lesion formation in the soft tissue infection model and dissemination to the bloodstream. We hypothesize that scfAB play an integral role in enhancing adaptation and fitness of GAS during localized skin infection, and potentially in propagation to other deeper host environments.


Asunto(s)
Genes Bacterianos/genética , Infecciones de los Tejidos Blandos/microbiología , Infecciones Estreptocócicas/microbiología , Streptococcus pyogenes/genética , Virulencia/genética , Animales , Modelos Animales de Enfermedad , Aptitud Genética/genética , Ratones , Reacción en Cadena de la Polimerasa
10.
J Bacteriol ; 200(8)2018 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-29378883

RESUMEN

Streptococcus pyogenes (group A Streptococcus [GAS]) causes a wide range of human infections. The pathogenesis of GAS infections is dependent on the temporal expression of numerous secreted and surface-associated virulence factors that interact with host proteins. Streptococcal pyrogenic exotoxin B (SpeB) is one of the most extensively studied toxins produced by GAS, and the coordinate growth phase-dependent regulation of speB expression is linked to disease severity phenotypes. Here, we identified the endopeptidase PepO as a novel growth phase-dependent regulator of SpeB in the invasive GAS M1 serotype strain 5448. By using transcriptomics followed by quantitative reverse transcriptase PCR and Western blot analyses, we demonstrate through targeted mutagenesis that PepO influences growth phase-dependent induction of speB gene expression. Compared to wild-type and complemented mutant strains, we demonstrate that the 5448ΔpepO mutant strain is more susceptible to killing by human neutrophils and is attenuated in virulence in a murine model of invasive GAS infection. Our results expand the complex regulatory network that is operating in GAS to control SpeB production and suggest that PepO is a virulence requirement during GAS M1T1 strain 5448 infections.IMPORTANCE Despite the continuing susceptibility of S. pyogenes to penicillin, this bacterial pathogen remains a leading infectious cause of global morbidity and mortality. A particular subclone of the M1 serotype (M1T1) has persisted globally for decades as the most frequently isolated serotype from patients with invasive and noninvasive diseases in Western countries. One of the key GAS pathogenicity factors is the potent broad-spectrum cysteine protease SpeB. Although there has been extensive research interest on the regulatory mechanisms that control speB gene expression, its genetic regulation is not fully understood. Here, we identify the endopeptidase PepO as a new regulator of speB gene expression in the globally disseminated M1T1 clone and as being essential for virulence.


Asunto(s)
Proteínas Bacterianas/metabolismo , Cisteína Endopeptidasas/metabolismo , Exotoxinas/metabolismo , Regulación Bacteriana de la Expresión Génica , Streptococcus pyogenes/enzimología , Streptococcus pyogenes/patogenicidad , Animales , Animales Modificados Genéticamente , Proteínas Bacterianas/genética , Cisteína Endopeptidasas/genética , Modelos Animales de Enfermedad , Exotoxinas/genética , Perfilación de la Expresión Génica , Humanos , Ratones , Mutagénesis , Neutrófilos/microbiología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Infecciones Estreptocócicas/microbiología , Streptococcus pyogenes/genética , Virulencia , Factores de Virulencia/genética , Factores de Virulencia/metabolismo
11.
Mol Microbiol ; 103(3): 518-533, 2017 02.
Artículo en Inglés | MEDLINE | ID: mdl-27862457

RESUMEN

The Group A Streptococcus (GAS, Streptococcus pyogenes) is a Gram-positive human pathogen that must adapt to unique host environments in order to survive. Links between sugar metabolism and virulence have been demonstrated in GAS, where mutants in the phosphoenolpyruvate-dependent phosphotransferase system (PTS) exhibited Streptolysin S (SLS)-mediated hemolysis during exponential growth. This early onset hemolysis correlated with an increased lesion size and severity in a murine soft tissue infection model when compared with parental M1T1 MGAS5005. To identify the PTS components responsible for this phenotype, we insertionally inactivated the 14 annotated PTS EIIC-encoding genes in the GAS MGAS5005 genome and subjected this library to metabolic and hemolysis assays to functionally characterize each EIIC. It was found that a few EIIs had a very limited influence on PTS sugar metabolism, whereas others were fairly promiscuous. The mannose-specific EII locus, encoded by manLMN, was expressed as a mannose-inducible operon that exhibited the most influence on PTS sugar metabolism, including mannose. Importantly, components of the mannose-specific EII also acted to prevent the early onset of SLS-mediated hemolysis. Interestingly, these roles were not identical in two different M1T1 GAS strains, highlighting the possible versatility of the PTS to adapt to strain-specific needs.


Asunto(s)
Sistema de Fosfotransferasa de Azúcar del Fosfoenolpiruvato/genética , Animales , Proteínas Bacterianas , Metabolismo de los Hidratos de Carbono/genética , Metabolismo de los Hidratos de Carbono/fisiología , Femenino , Regulación Bacteriana de la Expresión Génica/genética , Biblioteca de Genes , Glucosa/metabolismo , Hemólisis , Manosa/metabolismo , Proteínas de Transporte de Membrana/metabolismo , Ratones , Operón/genética , Sistema de Fosfotransferasa de Azúcar del Fosfoenolpiruvato/metabolismo , Streptococcus/genética , Streptococcus pyogenes/genética , Estreptolisinas , Virulencia
12.
Infect Immun ; 85(3)2017 03.
Artículo en Inglés | MEDLINE | ID: mdl-27993974

RESUMEN

As an exclusively human pathogen, Streptococcus pyogenes (the group A streptococcus [GAS]) has specifically adapted to evade host innate immunity and survive in multiple tissue niches, including blood. GAS can overcome the metabolic constraints of the blood environment and expresses various immunomodulatory factors necessary for survival and immune cell resistance. Here we present our investigation of one such factor, the predicted LysR family transcriptional regulator CpsY. The encoding gene, cpsY, was initially identified as being required for GAS survival in a transposon-site hybridization (TraSH) screen in whole human blood. CpsY is homologous with transcriptional regulators of Streptococcus mutans (MetR), Streptococcus iniae (CpsY), and Streptococcus agalactiae (MtaR) that regulate methionine transport, amino acid metabolism, resistance to neutrophil-mediated killing, and survival in vivo Our investigation indicated that CpsY is involved in GAS resistance to innate immune cells of its human host. However, GAS CpsY does not manifest the in vitro phenotypes of its homologs in other streptococcal species. GAS CpsY appears to regulate a small set of genes that is markedly different from the regulons of its homologs. The differential expression of these genes depends on the growth medium, and CpsY modestly influences their expression. The GAS CpsY regulon includes known virulence factors (mntE, speB, spd, nga [spn], prtS [SpyCEP], and sse) and cell surface-associated factors of GAS (emm1, mur1.2, sibA [cdhA], and M5005_Spy0500). Intriguingly, the loss of CpsY in GAS does not result in virulence defects in murine models of infection, suggesting that CpsY function in immune evasion is specific to the human host.


Asunto(s)
Proteínas Bacterianas/genética , Interacciones Huésped-Patógeno/inmunología , Inmunidad Innata , Infecciones Estreptocócicas/inmunología , Infecciones Estreptocócicas/microbiología , Streptococcus pyogenes/fisiología , Factores de Transcripción/genética , Animales , Modelos Animales de Enfermedad , Humanos , Macrófagos/inmunología , Macrófagos/metabolismo , Macrófagos/microbiología , Ratones , Viabilidad Microbiana , Mutación , Neutrófilos/inmunología , Neutrófilos/metabolismo , Neutrófilos/microbiología , Infecciones Estreptocócicas/metabolismo , Infecciones Estreptocócicas/mortalidad , Virulencia
13.
Infect Immun ; 84(4): 1016-1031, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26787724

RESUMEN

Bacterial pathogens rely on the availability of nutrients for survival in the host environment. The phosphoenolpyruvate-phosphotransferase system (PTS) is a global regulatory network connecting sugar uptake with signal transduction. Since the fructose PTS has been shown to impact virulence in several streptococci, including the human pathogen Streptococcus pyogenes(the group A Streptococcus[GAS]), we characterized its role in carbon metabolism and pathogenesis in the M1T1 strain 5448. Growth in fructose as a sole carbon source resulted in 103 genes affected transcriptionally, where the frulocus (fruRBA) was the most induced. Reverse transcriptase PCR showed that fruRBA formed an operon which was repressed by FruR in the absence of fructose, in addition to being under carbon catabolic repression. Growth assays and carbon utilization profiles revealed that although the entire fruoperon was required for growth in fructose, FruA was the main transporter for fructose and also was involved in the utilization of three additional PTS sugars: cellobiose, mannitol, and N-acetyl-D-galactosamine. The inactivation of sloR, a fruA homolog that also was upregulated in the presence of fructose, failed to reveal a role as a secondary fructose transporter. Whereas the ability of both ΔfruR and ΔfruB mutants to survive in the presence of whole human blood or neutrophils was impaired, the phenotype was not reproduced in murine whole blood, and those mutants were not attenuated in a mouse intraperitoneal infection. Since the ΔfruA mutant exhibited no phenotype in the human or mouse assays, we propose that FruR and FruB are important for GAS survival in a human-specific environment.


Asunto(s)
Sangre/microbiología , Fructosa/metabolismo , Neutrófilos/fisiología , Operón/fisiología , Streptococcus pyogenes/fisiología , Animales , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Actividad Bactericida de la Sangre/fisiología , Mapeo Cromosómico , Cromosomas Bacterianos , Femenino , Regulación Bacteriana de la Expresión Génica , Humanos , Ratones , Mutación , Infecciones Estreptocócicas/microbiología
14.
Mol Microbiol ; 98(5): 946-62, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26278404

RESUMEN

The sugar nucleotide dTDP-L-rhamnose is critical for the biosynthesis of the Group A Carbohydrate, the molecular signature and virulence determinant of the human pathogen Group A Streptococcus (GAS). The final step of the four-step dTDP-L-rhamnose biosynthesis pathway is catalyzed by dTDP-4-dehydrorhamnose reductases (RmlD). RmlD from the Gram-negative bacterium Salmonella is the only structurally characterized family member and requires metal-dependent homo-dimerization for enzymatic activity. Using a biochemical and structural biology approach, we demonstrate that the only RmlD homologue from GAS, previously renamed GacA, functions in a novel monomeric manner. Sequence analysis of 213 Gram-negative and Gram-positive RmlD homologues predicts that enzymes from all Gram-positive species lack a dimerization motif and function as monomers. The enzymatic function of GacA was confirmed through heterologous expression of gacA in a S. mutans rmlD knockout, which restored attenuated growth and aberrant cell division. Finally, analysis of a saturated mutant GAS library using Tn-sequencing and generation of a conditional-expression mutant identified gacA as an essential gene for GAS. In conclusion, GacA is an essential monomeric enzyme in GAS and representative of monomeric RmlD enzymes in Gram-positive bacteria and a subset of Gram-negative bacteria. These results will help future screens for novel inhibitors of dTDP-L-rhamnose biosynthesis.


Asunto(s)
Proteínas Bacterianas/metabolismo , Deshidrogenasas de Carbohidratos/genética , Deshidrogenasas de Carbohidratos/metabolismo , Streptococcus pyogenes/enzimología , Secuencia de Aminoácidos , Proteínas Bacterianas/química , Proteínas Bacterianas/genética , Deshidrogenasas de Carbohidratos/química , Carbohidrato Epimerasas/metabolismo , Clonación Molecular , Bacterias Grampositivas/enzimología , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Modelos Moleculares , Mutación , Azúcares de Nucleósido Difosfato/biosíntesis , Estructura Terciaria de Proteína , Ramnosa/análogos & derivados , Ramnosa/biosíntesis , Ramnosa/metabolismo , Alineación de Secuencia , Streptococcus pyogenes/genética , Nucleótidos de Timina/biosíntesis , Nucleótidos de Timina/metabolismo
15.
Infect Immun ; 82(9): 3891-9, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-25001603

RESUMEN

Group A streptococcus (GAS) is an important human pathogen that causes a number of diseases with a wide range of severities. While all known strains of GAS are still sensitive to penicillin, there have been reports of antibiotic treatment failure in as many as 20% to 40% of cases. Biofilm formation has been implicated as a possible cause for these failures. A biofilm is a microbially derived, sessile community where cells grow attached to a surface or as a bacterial conglomerate and surrounded by a complex extracellular matrix. While the ability of group A streptococcus to form biofilms in the laboratory has been shown, there is a lack of understanding of the role of GAS biofilms during an infection. We hypothesized that during infections, GAS exhibits a biofilm phenotype, complete with unique protein expression. To test this hypothesis, a rabbit model of GAS osteomyelitis was developed. A rabbit was inoculated with GAS using an infected indwelling device. Following the infection, blood and tissue samples were collected. Histological samples of the infected tibia were prepared, and the formation of a biofilm in vivo was visualized using peptide nucleic acid fluorescent in situ hybridization (PNA-FISH) and confocal microscopy. In addition, Western blotting with convalescent rabbit serum detected cell wall proteins expressed in vitro under biofilm and planktonic growth conditions. Immunogenic proteins were then identified using matrix-assisted laser desorption ionization-time of flight tandem mass spectrometry (MALDI-TOF/TOF MS). These identities, along with the in vivo results, support the hypothesis that GAS forms biofilms during an infection. This unique phenotype should be taken into consideration when designing a vaccine or any other treatment for group A streptococcus infections.


Asunto(s)
Proteínas Bacterianas/genética , Cuerpos Extraños/genética , Infecciones Estreptocócicas/genética , Streptococcus pyogenes/genética , Tibia/microbiología , Animales , Proteínas Bacterianas/inmunología , Biopelículas/crecimiento & desarrollo , Femenino , Cuerpos Extraños/inmunología , Cuerpos Extraños/microbiología , Osteomielitis/genética , Osteomielitis/inmunología , Osteomielitis/microbiología , Conejos , Infecciones Estreptocócicas/metabolismo , Infecciones Estreptocócicas/microbiología , Streptococcus pyogenes/inmunología , Tibia/inmunología
16.
Infect Immun ; 82(3): 1192-204, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24379283

RESUMEN

Obtaining essential nutrients, such as carbohydrates, is an important process for bacterial pathogens to successfully colonize host tissues. The phosphoenolpyruvate phosphotransferase system (PTS) is the primary mechanism by which bacteria transport sugars and sense the carbon state of the cell. The group A streptococcus (GAS) is a fastidious microorganism that has adapted to a variety of niches in the human body to elicit a wide array of diseases. A ΔptsI mutant (enzyme I [EI] deficient) generated in three different strains of M1T1 GAS was unable to grow on multiple carbon sources (PTS and non-PTS). Complementation with ptsI expressed under its native promoter in single copy was able to rescue the growth defect of the mutant. In a mouse model of GAS soft tissue infection, all ΔptsI mutants exhibited a significantly larger and more severe ulcerative lesion than mice infected with the wild type. Increased transcript levels of sagA and streptolysin S (SLS) activity during exponential-phase growth was observed. We hypothesized that early onset of SLS activity would correlate with the severity of the lesions induced by the ΔptsI mutant. In fact, infection of mice with a ΔptsI sagB double mutant resulted in a lesion comparable to that of either the wild type or a sagB mutant alone. Therefore, a functional PTS is not required for subcutaneous skin infection in mice; however, it does play a role in coordinating virulence factor expression and disease progression.


Asunto(s)
Proteínas Bacterianas/metabolismo , Sistema de Fosfotransferasa de Azúcar del Fosfoenolpiruvato/metabolismo , Infecciones de los Tejidos Blandos/metabolismo , Infecciones Estreptocócicas/metabolismo , Streptococcus pyogenes/metabolismo , Estreptolisinas/metabolismo , Animales , Proteínas Bacterianas/genética , Proteínas Bacterianas/inmunología , Carbohidratos/inmunología , Exotoxinas/genética , Exotoxinas/inmunología , Exotoxinas/metabolismo , Femenino , Genes Bacterianos/genética , Genes Bacterianos/inmunología , Ratones , Mutación/genética , Mutación/inmunología , Sistema de Fosfotransferasa de Azúcar del Fosfoenolpiruvato/genética , Sistema de Fosfotransferasa de Azúcar del Fosfoenolpiruvato/inmunología , Infecciones de los Tejidos Blandos/genética , Infecciones de los Tejidos Blandos/inmunología , Infecciones de los Tejidos Blandos/microbiología , Infecciones Cutáneas Estafilocócicas/genética , Infecciones Cutáneas Estafilocócicas/inmunología , Infecciones Cutáneas Estafilocócicas/metabolismo , Infecciones Cutáneas Estafilocócicas/microbiología , Infecciones Estreptocócicas/genética , Infecciones Estreptocócicas/inmunología , Infecciones Estreptocócicas/microbiología , Streptococcus pyogenes/genética , Streptococcus pyogenes/inmunología , Estreptolisinas/genética , Estreptolisinas/inmunología , Virulencia/genética , Virulencia/inmunología , Factores de Virulencia/genética , Factores de Virulencia/inmunología , Factores de Virulencia/metabolismo
17.
Mol Microbiol ; 88(6): 1176-93, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23651410

RESUMEN

The ability of a bacterial pathogen to monitor available carbon sources in host tissues provides a clear fitness advantage. In the group A streptococcus (GAS), the virulence regulator Mga contains homology to phosphotransferase system (PTS) regulatory domains (PRDs) found in sugar operon regulators. Here we show that Mga was phosphorylated in vitro by the PTS components EI/HPr at conserved PRD histidines. A ΔptsI (EI-deficient) GAS mutant exhibited decreased Mga activity. However, PTS-mediated phosphorylation inhibited Mga-dependent transcription of emm in vitro. Using alanine (unphosphorylated) and aspartate (phosphomimetic) mutations of PRD histidines, we establish that a doubly phosphorylated PRD1 phosphomimetic (D/DMga4) is completely inactive in vivo, shutting down expression of the Mga regulon. Although D/DMga4 is still able to bind DNA in vitro, homo-multimerization of Mga is disrupted and the protein is unable to activate transcription. PTS-mediated regulation of Mga activity appears to be important for pathogenesis, as bacteria expressing either non-phosphorylated (A/A) or phosphomimetic (D/D) PRD1 Mga mutants were attenuated in a model of GAS invasive skin disease. Thus, PTS-mediated phosphorylation of Mga may allow the bacteria to modulate virulence gene expression in response to carbohydrate status. Furthermore, PRD-containing virulence regulators (PCVRs) appear to be widespread in Gram-positive pathogens.


Asunto(s)
Proteínas Bacterianas/metabolismo , Regulación Bacteriana de la Expresión Génica , Sistema de Fosfotransferasa de Azúcar del Fosfoenolpiruvato/metabolismo , Regulón , Streptococcus pyogenes/genética , Streptococcus pyogenes/metabolismo , Animales , ADN Bacteriano/metabolismo , Modelos Animales de Enfermedad , Ratones , Fosforilación , Unión Proteica , Multimerización de Proteína , Enfermedades Cutáneas Bacterianas/microbiología , Enfermedades Cutáneas Bacterianas/patología , Infecciones Estreptocócicas/microbiología , Infecciones Estreptocócicas/patología , Streptococcus pyogenes/patogenicidad , Transcripción Genética , Virulencia
18.
Infect Immun ; 81(3): 862-75, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23297387

RESUMEN

The group A streptococcus (GAS) is a strict human pathogen responsible for a wide spectrum of diseases. Although GAS genome sequences are available, functional genomic analyses have been limited. We developed a mariner-based transposon, osKaR, designed to perform Transposon-Site Hybridization (TraSH) in GAS and successfully tested its use in several invasive serotypes. A complex osKaR mutant library in M1T1 GAS strain 5448 was subjected to negative selection in human blood to identify genes important for GAS fitness in this clinically relevant environment. Mutants underrepresented after growth in blood (output pool) compared to growth in rich media (input pool) were identified using DNA microarray hybridization of transposon-specific tags en masse. Using blood from three different donors, we identified 81 genes that met our criteria for reduced fitness in blood from at least two individuals. Genes known to play a role in survival of GAS in blood were found, including those encoding the virulence regulator Mga (mga), the peroxide response regulator PerR (perR), and the RofA-like regulator Ralp-3 (ralp3). We also identified genes previously reported for their contribution to sepsis in other pathogens, such as de novo nucleotide synthesis (purD, purA, pyrB, carA, carB, guaB), sugar metabolism (scrB, fruA), zinc uptake (adcC), and transcriptional regulation (cpsY). To validate our findings, independent mutants with mutations in 10 different genes identified in our screen were confirmed to be defective for survival in blood bactericidal assays. Overall, this work represents the first use of TraSH in GAS to identify potential virulence genes.


Asunto(s)
Sangre/microbiología , Regulación Bacteriana de la Expresión Génica/fisiología , Genoma Bacteriano/genética , Streptococcus pyogenes/genética , Mapeo Cromosómico , Cromosomas Bacterianos , Aptitud Genética , Proteínas Hemolisinas/genética , Proteínas Hemolisinas/metabolismo , Humanos , Mutagénesis Insercional , Mutación
19.
Mol Microbiol ; 83(5): 953-67, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22468267

RESUMEN

The Group A Streptococcus (GAS) is a strict human pathogen that causes a broad spectrum of illnesses. One of the key regulators of virulence in GAS is the transcriptional activator Mga, which co-ordinates the early stages of infection. Although the targets of Mga have been well characterized, basic biochemical analyses have been limited due to difficulties in obtaining purified protein. In this study, high-level purification of soluble Mga was achieved, enabling the first detailed characterization of the protein. Fluorescence titrations coupled with filter-binding assays indicate that Mga binds cognate DNA with nanomolar affinity. Gel filtration analyses, analytical ultracentrifugation and co-immunoprecipitation experiments demonstrate that Mga forms oligomers in solution.Moreover, the ability of the protein to oligomerize in solution was found to correlate with transcriptional activation; DNA binding appears to be necessary but insufficient for full activity. Truncation analyses reveal that the uncharacterized C-terminal region of Mga, possessing similarity to phosphotransferase system EIIB proteins, plays a critical role in oligomerization and in vivo activity. Mga from a divergent serotype was found to behave similarly, suggesting that this study describes a general mechanism for Mga regulation of target virulence genes within GAS and provides insight into related regulators in other Gram-positive pathogens.


Asunto(s)
Proteínas Bacterianas/metabolismo , Streptococcus pyogenes/genética , Factores de Transcripción/metabolismo , Activación Transcripcional , Proteínas Bacterianas/genética , ADN Bacteriano/metabolismo , Regulación Bacteriana de la Expresión Génica , Multimerización de Proteína , Streptococcus pyogenes/patogenicidad , Factores de Transcripción/genética , Virulencia
20.
J Bacteriol ; 194(18): 4904-19, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22773785

RESUMEN

The Mga regulator of Streptococcus pyogenes directly activates the transcription of a core regulon that encodes virulence factors such as M protein (emm), C5a peptidase (scpA), and streptococcal inhibitor of complement (sic) by directly binding to a 45-bp binding site as determined by an electrophoretic mobility shift assay (EMSA) and DNase I protection. However, by comparing the nucleotide sequences of all established Mga binding sites, we found that they exhibit only 13.4% identity with no discernible symmetry. To determine the core nucleotides involved in functional Mga-DNA interactions, the M1T1 Pemm1 binding site was altered and screened for nucleotides important for DNA binding in vitro and for transcriptional activation using a plasmid-based luciferase reporter in vivo. Following this analysis, 34 nucleotides within the Pemm1 binding site that had an effect on Mga binding, Mga-dependent transcriptional activation, or both were identified. Of these critical nucleotides, guanines and cytosines within the major groove were disproportionately identified clustered at the 5' and 3' ends of the binding site and with runs of nonessential adenines between the critical nucleotides. On the basis of these results, a Pemm1 minimal binding site of 35 bp bound Mga at a level comparable to the level of binding of the larger 45-bp site. Comparison of Pemm with directed mutagenesis performed in the M1T1 Mga-regulated PscpA and Psic promoters, as well as methylation interference analysis of PscpA, establish that Mga binds to DNA in a promoter-specific manner.


Asunto(s)
Proteínas Bacterianas/metabolismo , ADN Bacteriano/genética , ADN Bacteriano/metabolismo , Regiones Promotoras Genéticas , Streptococcus pyogenes/patogenicidad , Fusión Artificial Génica , Sitios de Unión , Análisis Mutacional de ADN , Genes Reporteros , Luciferasas/análisis , Luciferasas/genética , Plásmidos , Unión Proteica , Streptococcus pyogenes/metabolismo , Transcripción Genética , Factores de Virulencia/biosíntesis
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA