Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 82
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
PLoS Pathog ; 19(5): e1011421, 2023 05.
Artículo en Inglés | MEDLINE | ID: mdl-37256908

RESUMEN

Streptococcus pneumoniae is an opportunistic pathogen that colonizes the upper respiratory tract asymptomatically and, upon invasion, can lead to severe diseases including otitis media, sinusitis, meningitis, bacteremia, and pneumonia. One of the first lines of defense against pneumococcal invasive disease is inflammation, including the recruitment of neutrophils to the site of infection. The invasive pneumococcus can be cleared through the action of serine proteases generated by neutrophils. It is less clear how serine proteases impact non-invasive pneumococcal colonization, which is the key first step to invasion and transmission. One significant aspect of pneumococcal biology and adaptation in the respiratory tract is its natural competence, which is triggered by a small peptide CSP. In this study, we investigate if serine proteases are capable of degrading CSP and the impact this has on pneumococcal competence. We found that CSP has several potential sites for trypsin-like serine protease degradation and that there were preferential cleavage sites recognized by the proteases. Digestion of CSP with two different trypsin-like serine proteases dramatically reduced competence in a dose-dependent manner. Incubation of CSP with mouse lung homogenate also reduced recombination frequency of the pneumococcus. These ex vivo experiments suggested that serine proteases in the lower respiratory tract reduce pneumococcal competence. This was subsequently confirmed measuring in vivo recombination frequencies after induction of protease production via poly (I:C) stimulation and via co-infection with influenza A virus, which dramatically lowered recombination events. These data shed light on a new mechanism by which the host can modulate pneumococcal behavior and genetic exchange via direct degradation of the competence signaling peptide.


Asunto(s)
Infecciones Neumocócicas , Streptococcus pneumoniae , Animales , Ratones , Streptococcus pneumoniae/genética , Inflamación , Serina Proteasas , Péptidos
2.
PLoS Pathog ; 18(12): e1011020, 2022 12.
Artículo en Inglés | MEDLINE | ID: mdl-36542660

RESUMEN

BACKGROUND: For almost a century, it has been recognized that influenza A virus (IAV) infection can promote the development of secondary bacterial infections (SBI) mainly caused by Streptococcus pneumoniae (Spn). Recent observations have shown that IAV is able to directly bind to the surface of Spn. To gain a foundational understanding of how direct IAV-Spn interaction alters bacterial biological fitness we employed combinatorial multiomic and molecular approaches. RESULTS: Here we show IAV significantly remodels the global transcriptome, proteome and phosphoproteome profiles of Spn independently of host effectors. We identified Spn surface proteins that interact with IAV proteins (hemagglutinin, nucleoprotein, and neuraminidase). In addition, IAV was found to directly modulate expression of Spn virulence determinants such as pneumococcal surface protein A, pneumolysin, and factors associated with antimicrobial resistance among many others. Metabolic pathways were significantly altered leading to changes in Spn growth rate. IAV was also found to drive Spn capsule shedding and the release of pneumococcal surface proteins. Released proteins were found to be involved in evasion of innate immune responses and actively reduced human complement hemolytic and opsonizing activity. IAV also led to phosphorylation changes in Spn proteins associated with metabolism and bacterial virulence. Validation of proteomic data showed significant changes in Spn galactose and glucose metabolism. Furthermore, supplementation with galactose rescued bacterial growth and promoted bacterial invasion, while glucose supplementation led to enhanced pneumolysin production and lung cell apoptosis. CONCLUSIONS: Here we demonstrate that IAV can directly modulate Spn biology without the requirement of host effectors and support the notion that inter-kingdom interactions between human viruses and commensal pathobionts can promote bacterial pathogenesis and microbiome dysbiosis.


Asunto(s)
Virus de la Influenza A , Gripe Humana , Infecciones por Orthomyxoviridae , Humanos , Streptococcus pneumoniae/metabolismo , Virus de la Influenza A/genética , Virulencia , Galactosa/metabolismo , Multiómica , Proteómica , Gripe Humana/genética , Gripe Humana/complicaciones
3.
Proc Natl Acad Sci U S A ; 117(21): 11703-11714, 2020 05 26.
Artículo en Inglés | MEDLINE | ID: mdl-32393645

RESUMEN

Patients with hematological malignancies or undergoing hematopoietic stem cell transplantation are vulnerable to colonization and infection with multidrug-resistant organisms, including vancomycin-resistant Enterococcus faecium (VREfm). Over a 10-y period, we collected and sequenced the genomes of 110 VREfm isolates from gastrointestinal and blood cultures of 24 pediatric patients undergoing chemotherapy or hematopoietic stem cell transplantation for hematological malignancy at St. Jude Children's Research Hospital. We used patient-specific reference genomes to identify variants that arose over time in subsequent gastrointestinal and blood isolates from each patient and analyzed these variants for insight into how VREfm adapted during colonization and bloodstream infection within each patient. Variants were enriched in genes involved in carbohydrate metabolism, and phenotypic analysis identified associated differences in carbohydrate utilization among isolates. In particular, a Y585C mutation in the sorbitol operon transcriptional regulator gutR was associated with increased bacterial growth in the presence of sorbitol. We also found differences in biofilm-formation capability between isolates and observed that increased biofilm formation correlated with mutations in the putative E. faecium capsular polysaccharide (cps) biosynthetic locus, with different mutations arising independently in distinct genetic backgrounds. Isolates with cps mutations showed improved survival following exposure to lysozyme, suggesting a possible reason for the selection of capsule-lacking bacteria. Finally, we observed mutations conferring increased tolerance of linezolid and daptomycin in patients who were treated with these antibiotics. Overall, this study documents known and previously undescribed ways that VREfm evolve during intestinal colonization and subsequent bloodstream infection in immunocompromised pediatric patients.


Asunto(s)
Enterococcus faecium , Infecciones por Bacterias Grampositivas/microbiología , Enterococos Resistentes a la Vancomicina , Antibacterianos/farmacología , Bacteriemia/microbiología , Biopelículas , Niño , Enterococcus faecium/efectos de los fármacos , Enterococcus faecium/genética , Enterococcus faecium/patogenicidad , Evolución Molecular , Femenino , Microbioma Gastrointestinal/genética , Genoma Bacteriano/genética , Humanos , Huésped Inmunocomprometido , Masculino , Mutación/genética , Sorbitol/metabolismo , Enterococos Resistentes a la Vancomicina/efectos de los fármacos , Enterococos Resistentes a la Vancomicina/genética , Enterococos Resistentes a la Vancomicina/patogenicidad
4.
Clin Infect Dis ; 75(4): 647-656, 2022 09 10.
Artículo en Inglés | MEDLINE | ID: mdl-34891152

RESUMEN

BACKGROUND: Carriage studies are fundamental to assessing the effects of pneumococcal vaccines. Because a large proportion of oral streptococci carry homologues of pneumococcal genes, non-culture-based detection and serotyping of upper respiratory tract (URT) samples can be problematic. In the current study, we investigated whether culture-free molecular methods could differentiate pneumococci from oral streptococci carried by adults in the URT. METHODS: Paired nasopharyngeal (NP) and oropharyngeal (OP) samples were collected from 100 older adults twice a month for 1 year. Extracts from the combined NP + OP samples (n = 2400) were subjected to lytA real-time polymerase chain reaction (PCR). Positive samples were subjected to pure culture isolation, followed by species confirmation using multiple approaches. Multibead assays and whole-genome sequencing were used for serotyping. RESULTS: In 20 of 301 combined NP + OP extracts with positive lytA PCR results, probable pneumococcus-like colonies grew, based on colony morphology and biochemical tests. Multiple approaches confirmed that 4 isolates were Streptococcus pneumoniae, 3 were Streptococcus pseudopneumoniae, 12 were Streptococcus mitis, and 1 were Streptococcus oralis. Eight nonpneumococcal strains carried pneumococcus-like cps loci (approximate size, 18-25 kb) that showed >70% nucleotide identity with their pneumococcal counterparts. While investigating the antigenic profile, we found that some S. mitis strains (P066 and P107) reacted with both serotype-specific polyclonal (type 39 and FS17b) and monoclonal (Hyp10AG1 and Hyp17FM1) antisera, whereas some strains (P063 and P074) reacted only with polyclonal antisera (type 5 and FS35a). CONCLUSION: The extensive capsular overlap suggests that pneumococcal vaccines could reduce carriage of oral streptococci expressing cross-reactive capsules. Furthermore, direct use of culture-free PCR-based methods in URT samples has limited usefulness for carriage studies.


Asunto(s)
Infecciones Neumocócicas , Streptococcus pneumoniae , Anciano , Portador Sano/diagnóstico , Humanos , Sueros Inmunes , Nasofaringe , Infecciones Neumocócicas/prevención & control , Vacunas Neumococicas , Reacción en Cadena en Tiempo Real de la Polimerasa , Serotipificación , Organización Mundial de la Salud
5.
Mol Biol Evol ; 38(6): 2209-2226, 2021 05 19.
Artículo en Inglés | MEDLINE | ID: mdl-33502519

RESUMEN

Streptococcus pneumoniae is a commensal of the human nasopharynx and a major cause of respiratory and invasive disease. We examined adaptation and evolution of pneumococcus, within nasopharynx and lungs, in an experimental system where the selective pressures associated with transmission were removed. This was achieved by serial passage of pneumococci, separately, in mouse models of nasopharyngeal carriage or pneumonia. Passaged pneumococci became more effective colonizers of the respiratory tract and we observed several examples of potential parallel evolution. The cell wall-modifying glycosyltransferase LafA was under strong selection during lung passage, whereas the surface expressed pneumococcal vaccine antigen gene pvaA and the glycerol-3-phosphate dehydrogenase gene gpsA were frequent targets of mutation in nasopharynx-passaged pneumococci. These mutations were not identified in pneumococci that were separately evolved by serial passage on laboratory agar. We focused on gpsA, in which the same single nucleotide polymorphism arose in two independently evolved nasopharynx-passaged lineages. We describe a new role for this gene in nasopharyngeal carriage and show that the identified single nucleotide change confers resistance to oxidative stress and enhanced nasopharyngeal colonization potential. We demonstrate that polymorphisms in gpsA arise and are retained during human colonization. These findings highlight how within-host environmental conditions can determine trajectories of bacterial evolution. Relative invasiveness or attack rate of pneumococcal lineages may be defined by genes that make niche-specific contributions to bacterial fitness. Experimental evolution in animal infection models is a powerful tool to investigate the relative roles played by pathogen virulence and colonization factors within different host niches.


Asunto(s)
Adaptación Biológica/genética , Evolución Biológica , Infecciones Neumocócicas/microbiología , Streptococcus pneumoniae/patogenicidad , Animales , Femenino , Genoma Bacteriano , Humanos , Pulmón/microbiología , Ratones , Nasofaringe/microbiología , Distribución Aleatoria , Streptococcus pneumoniae/genética , Factores de Virulencia
6.
Infect Immun ; 89(8): e0071320, 2021 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-34031124

RESUMEN

Streptococcus pneumoniae is an opportunistic pathogen that is a common cause of serious invasive diseases such as pneumonia, bacteremia, meningitis, and otitis media. Transmission of this bacterium has classically been thought to occur through inhalation of respiratory droplets and direct contact with nasal secretions. However, the demonstration that S. pneumoniae is desiccation tolerant and, therefore, environmentally stable for extended periods of time opens up the possibility that this pathogen is also transmitted via contaminated surfaces (fomites). To better understand the molecular mechanisms that enable S. pneumoniae to survive periods of desiccation, we performed a high-throughput transposon sequencing (Tn-seq) screen in search of genetic determinants of desiccation tolerance. We identified 42 genes whose disruption reduced desiccation tolerance and 45 genes that enhanced desiccation tolerance. The nucleotide excision repair pathway was the most enriched category in our Tn-seq results, and we found that additional DNA repair pathways are required for desiccation tolerance, demonstrating the importance of maintaining genome integrity after desiccation. Deletion of the nucleotide excision repair gene uvrA resulted in a delay in transmission between infant mice, indicating a correlation between desiccation tolerance and pneumococcal transmssion. Understanding the molecular mechanisms that enable pneumococcal persistence in the environment may enable targeting of these pathways to prevent fomite transmission, thereby preventing the establishment of new colonization and any resulting invasive disease.


Asunto(s)
Reparación del ADN , Elementos Transponibles de ADN , Infecciones Neumocócicas/microbiología , Streptococcus pneumoniae/genética , Adaptación Biológica , Animales , Susceptibilidad a Enfermedades , Interacciones Huésped-Patógeno , Ratones , Infecciones Neumocócicas/inmunología , Infecciones Neumocócicas/transmisión , Transducción de Señal , Streptococcus pneumoniae/inmunología , Streptococcus pneumoniae/patogenicidad
7.
Mol Microbiol ; 114(4): 536-552, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32495474

RESUMEN

Streptococcus pneumoniae is a major human pathogen that must adapt to unique nutritional environments in several host niches. The pneumococcus can metabolize a range of carbohydrates that feed into glycolysis ending in pyruvate, which is catabolized by several enzymes. We investigated how the pneumococcus utilizes these enzymes to metabolize different carbohydrates and how this impacts survival in the host. Loss of ldh decreased bacterial burden in the nasopharynx and enhanced bacteremia in mice. Loss of spxB, pdhC or pfl2 decreased bacteremia and increased host survival. In glucose or galactose, loss of ldh increased capsule production, whereas loss of spxB and pdhC reduced capsule production. The pfl2 mutant exhibited reduced capsule production only in galactose. In glucose, pyruvate was metabolized primarily by LDH to generate lactate and NAD+ and by SpxB and PDHc to generate acetyl-CoA. In galactose, pyruvate metabolism was shunted toward acetyl-CoA production. The majority of acetyl-CoA generated by PFL was used to regenerate NAD+ with a subset used in capsule production, while the acetyl-CoA generated by SpxB and PDHc was utilized primarily for capsule biosynthesis. These data suggest that the pneumococcus can alter flux of pyruvate metabolism dependent on the carbohydrate present to succeed in distinct host niches.


Asunto(s)
Metabolismo de los Hidratos de Carbono/fisiología , Ácido Pirúvico/metabolismo , Streptococcus pneumoniae/metabolismo , Animales , Proteínas Bacterianas/metabolismo , Carbohidratos/fisiología , Femenino , Galactosa/metabolismo , Glucólisis , Peróxido de Hidrógeno/metabolismo , Redes y Vías Metabólicas , Ratones , Ratones Endogámicos BALB C , Streptococcus pneumoniae/enzimología , Virulencia/fisiología
8.
Bioessays ; 41(12): e1900128, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31693223

RESUMEN

The synergies between viral and bacterial infections are well established. Most studies have been focused on the indirect mechanisms underlying this phenomenon, including immune modulation and alterations to the mucosal structures that promote pathogen outgrowth. A growing body of evidence implicates direct binding of virus to bacterial surfaces being an additional mechanism of synergy at the host-pathogen interface. These cross-kingdom interactions enhance bacterial and viral adhesion and can alter tissue tropism. These bacterial-viral complexes play unique roles in pathogenesis and can alter virulence potential. The bacterial-viral complexes may also play important roles in pathogen transmission. Additionally, the complexes are recognized by the host immune system in a distinct manner, thus presenting novel routes for vaccine development. These synergies are active for multiple species in both the respiratory and gastrointestinal tract, indicating that direct interactions between bacteria and virus to modulate host interactions are used by a diverse array of species.


Asunto(s)
Interacciones Huésped-Patógeno/fisiología , Animales , Infecciones Bacterianas/genética , Infecciones Bacterianas/metabolismo , Coinfección/genética , Coinfección/metabolismo , Interacciones Huésped-Patógeno/genética , Humanos , Streptococcus/patogenicidad , Virulencia , Virus/patogenicidad
9.
Antimicrob Agents Chemother ; 64(11)2020 10 20.
Artículo en Inglés | MEDLINE | ID: mdl-32816729

RESUMEN

Coagulase-negative staphylococci (CoNS) are a common etiology of serious and recurrent infections in immunocompromised patients. Although most isolates appear susceptible to vancomycin, a single strain might have a subpopulation of resistant bacteria. This phenomenon is termed heteroresistance and may adversely affect the response to treatment. A retrospective cohort study was performed of pediatric patients with leukemia treated at St. Jude Children's Research Hospital who developed CoNS central line-associated bloodstream infection (CLABSI). Available isolates were sequenced and tested for vancomycin heteroresistance by population analysis profiling. Risk factors for heteroresistance and the association of heteroresistance with treatment failure (death or relapse of infection) or poor clinical response to vancomycin therapy (treatment failure or persistent bacteremia after vancomycin initiation) were evaluated. For 65 participants with CoNS CLABSI, 62 initial isolates were evaluable, of which 24 (39%) were vancomycin heteroresistant. All heteroresistant isolates were of Staphylococcus epidermidis and comprised multiple sequence types. Participants with heteroresistant bacteria had more exposure to vancomycin prophylaxis (P = 0.026) during the 60 days prior to infection. Of the 40 participants evaluable for clinical outcomes, heteroresistance increased the risk of treatment failure (P = 0.012) and poor clinical response (P = 0.001). This effect persisted after controlling for identified confounders. These data indicate that vancomycin heteroresistance is common in CoNS isolates from CLABSIs in pediatric patients with leukemia and is associated with poor clinical outcomes. Validation of these findings in an independent cohort and evaluation of alternative antibiotic therapy in patients with heteroresistant infections have the potential to improve care for serious CoNS infections.


Asunto(s)
Bacteriemia , Sepsis , Infecciones Estafilocócicas , Antibacterianos/uso terapéutico , Bacteriemia/tratamiento farmacológico , Niño , Coagulasa , Humanos , Pruebas de Sensibilidad Microbiana , Estudios Retrospectivos , Sepsis/tratamiento farmacológico , Infecciones Estafilocócicas/tratamiento farmacológico , Vancomicina/uso terapéutico
10.
Infect Immun ; 87(10)2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31308088

RESUMEN

Acute otitis media is one of the most common childhood infections worldwide. Currently licensed vaccines against the common otopathogen Streptococcus pneumoniae target the bacterial capsular polysaccharide and confer no protection against nonencapsulated strains or capsular types outside vaccine coverage. Mucosal infections such as acute otitis media remain prevalent, even those caused by vaccine-covered serotypes. Here, we report that a protein-based vaccine, a fusion construct of epitopes of CbpA to pneumolysin toxoid, confers effective protection against pneumococcal acute otitis media for non-PCV-13 serotypes and enhances protection for PCV-13 serotypes when coadministered with PCV-13. Having cross-reactive epitopes, the fusion protein also induces potent antibody responses against nontypeable Haemophilus influenzae and S. pneumoniae, engendering protection against acute otitis media caused by emerging unencapsulated otopathogens. These data suggest that augmenting capsule-based vaccination with conserved, cross-reactive protein-based vaccines broadens and enhances protection against acute otitis media.


Asunto(s)
Anticuerpos Antibacterianos/biosíntesis , Infecciones por Haemophilus/prevención & control , Haemophilus influenzae/inmunología , Otitis Media/prevención & control , Vacunas Neumococicas/biosíntesis , Proteínas Recombinantes de Fusión/biosíntesis , Streptococcus pneumoniae/inmunología , Enfermedad Aguda , Animales , Proteínas Bacterianas/biosíntesis , Proteínas Bacterianas/genética , Protección Cruzada , Reacciones Cruzadas , Femenino , Expresión Génica , Infecciones por Haemophilus/inmunología , Infecciones por Haemophilus/microbiología , Haemophilus influenzae/efectos de los fármacos , Haemophilus influenzae/patogenicidad , Humanos , Inmunogenicidad Vacunal , Ratones , Ratones Endogámicos BALB C , Otitis Media/inmunología , Otitis Media/microbiología , Vacunas Neumococicas/administración & dosificación , Vacunas Neumococicas/genética , Proteínas Recombinantes de Fusión/administración & dosificación , Proteínas Recombinantes de Fusión/genética , Streptococcus pneumoniae/efectos de los fármacos , Streptococcus pneumoniae/patogenicidad , Estreptolisinas/biosíntesis , Estreptolisinas/genética , Toxoides/biosíntesis , Toxoides/genética , Vacunación , Vacunas Sintéticas
11.
Artículo en Inglés | MEDLINE | ID: mdl-30858215

RESUMEN

The most frequent ailment for which antibiotics are prescribed is otitis media (ear infections), which is most commonly caused by Haemophilus influenzae, Moraxella catarrhalis, and Streptococcus pneumoniae Treatment of otitis media is complicated by the fact that the bacteria in the middle ear typically form biofilms, which can be recalcitrant to antibiotic treatment. Furthermore, bacterial respiratory infections can be greatly exacerbated by viral coinfection, which is particularly evidenced by the synergy between influenza and S. pneumoniae In this study, we sought to ascertain the in vivo efficacy of aminomethyl spectinomycin lead 1950, an effective antibacterial agent both in vitro and in vivo against Streptococcus pneumoniae in the context of complex respiratory infections and acute otitis media. A single dose of 1950 significantly reduced bacterial burden in the respiratory tract for all three pathogens, even when species were present in a coinfection model. Additionally, a single dose of 1950 effectively reduced pneumococcal acute otitis media from the middle ear. The agent 1950 also proved efficacious in the context of influenza-pneumococcal super infection. These data further support the development of this family of compounds as potential therapeutic agents against the common causes of complex upper respiratory tract infections and acute otitis media.


Asunto(s)
Infecciones del Sistema Respiratorio/tratamiento farmacológico , Espectinomicina/uso terapéutico , Animales , Femenino , Haemophilus influenzae/efectos de los fármacos , Haemophilus influenzae/patogenicidad , Ratones , Ratones Endogámicos BALB C , Moraxella catarrhalis/efectos de los fármacos , Moraxella catarrhalis/patogenicidad , Otitis Media/tratamiento farmacológico , Otitis Media/microbiología , Neumonía/tratamiento farmacológico , Neumonía/microbiología , Espectinomicina/administración & dosificación , Espectinomicina/química , Streptococcus pneumoniae/efectos de los fármacos , Streptococcus pneumoniae/patogenicidad
12.
Clin Infect Dis ; 67(4): 541-548, 2018 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-29518185

RESUMEN

Background: Myelosuppression-related infections remain important causes of morbidity and mortality in children with acute lymphoblastic leukemia (ALL). Methods: By analyzing fecal samples collected at diagnosis and after each of the initial 3 phases of chemotherapy, we evaluated the role of gut microbiota in predicting infections in 199 children with newly diagnosed ALL. The bacterial 16S rRNA gene was analyzed by high-depth sequencing to determine the diversity and composition of the microbiome. Results: After the induction and reinduction I phases of chemotherapy, microbial diversity decreased significantly relative to the prechemotherapy value. After chemotherapy, the relative abundance of certain bacterial taxa (eg, Bacteroidetes) decreased significantly, whereas that of other taxa (eg, Clostridiaceae and Streptococcaceae) increased. A baseline gut microbiome characterized by Proteobacteria predicted febrile neutropenia. Adjusting for the chemotherapy phase and ALL risk level, Enterococcaceae dominance (relative abundance ≥30%) predicted significantly greater risk of subsequent febrile neutropenia and diarrheal illness, whereas Streptococcaceae dominance predicted significantly greater risk of subsequent diarrheal illness. Conclusions: In children undergoing therapy for newly diagnosed ALL, the relative abundance of Proteobacteria before chemotherapy initiation predicts development of febrile neutropenia, and domination of the gut microbiota by Enterococcaceae or Streptococcaceae at any time during chemotherapy predicts infection in subsequent phases of chemotherapy. Clinical Trial Registration: NCT00549848.


Asunto(s)
Antineoplásicos/efectos adversos , Bacterias/efectos de los fármacos , Infecciones Bacterianas/complicaciones , Microbioma Gastrointestinal/efectos de los fármacos , Leucemia-Linfoma Linfoblástico de Células Precursoras/tratamiento farmacológico , Adolescente , Antineoplásicos/uso terapéutico , Bacterias/clasificación , Niño , Preescolar , Heces/microbiología , Femenino , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Lactante , Masculino , Leucemia-Linfoma Linfoblástico de Células Precursoras/complicaciones , Valor Predictivo de las Pruebas , ARN Ribosómico 16S/genética , Factores de Riesgo , Análisis de Secuencia de ADN
13.
PLoS Pathog ; 12(10): e1005951, 2016 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-27760231

RESUMEN

The pneumococcus is one of the most prodigious producers of hydrogen peroxide amongst bacterial pathogens. Hydrogen peroxide production by the pneumococcus has been implicated in antibiotic synergism, competition between other bacterial colonizers of the nasopharynx, and damage to epithelial cells. However, the role during invasive disease has been less clear with mutants defective in hydrogen peroxide production demonstrating both attenuation and heightened invasive disease capacity depending upon strain and serotype background. This work resolves these conflicting observations by demonstrating that the main hydrogen peroxide producing enzyme of the pneumococcus, SpxB, is required for capsule formation in a strain dependent manner. Capsule production by strains harboring capsules with acetylated sugars was dependent upon the presence of spxB while capsule production in serotypes lacking such linkages were not. The spxB mutant had significantly lower steady-state cellular levels of acetyl-CoA, suggesting that loss of capsule arises from dysregulation of this intermediary metabolite. This conclusion is corroborated by deletion of pdhC, which also resulted in lower steady-state acetyl-CoA levels and phenocopied the capsule expression profile of the spxB mutant. Capsule and acetyl-CoA levels were restored in the spxB and lctO (lactate oxidase) double mutant, supporting the connection between central metabolism and capsule formation. Taken together, these data show that the defect in pathogenesis in the spxB mutant is due to a metabolic imbalance that attenuates capsule formation and not to reduced hydrogen peroxide formation.


Asunto(s)
Cápsulas Bacterianas/metabolismo , Piruvato Oxidasa/metabolismo , Streptococcus pneumoniae/patogenicidad , Animales , Ensayo de Inmunoadsorción Enzimática , Femenino , Técnicas de Inactivación de Genes , Peróxido de Hidrógeno/metabolismo , Ratones , Ratones Endogámicos BALB C , Infecciones Neumocócicas/metabolismo , Reacción en Cadena de la Polimerasa , Streptococcus pneumoniae/metabolismo , Virulencia
14.
PLoS Pathog ; 12(8): e1005804, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-27505057

RESUMEN

The healthy lung maintains a steady state of immune readiness to rapidly respond to injury from invaders. Integrins are important for setting the parameters of this resting state, particularly the epithelial-restricted αVß6 integrin, which is upregulated during injury. Once expressed, αVß6 moderates acute lung injury (ALI) through as yet undefined molecular mechanisms. We show that the upregulation of ß6 during influenza infection is involved in disease pathogenesis. ß6-deficient mice (ß6 KO) have increased survival during influenza infection likely due to the limited viral spread into the alveolar spaces leading to reduced ALI. Although the ß6 KO have morphologically normal lungs, they harbor constitutively activated lung CD11b+ alveolar macrophages (AM) and elevated type I IFN signaling activity, which we traced to the loss of ß6-activated transforming growth factor-ß (TGF-ß). Administration of exogenous TGF-ß to ß6 KO mice leads to reduced numbers of CD11b+ AMs, decreased type I IFN signaling activity and loss of the protective phenotype during influenza infection. Protection extended to other respiratory pathogens such as Sendai virus and bacterial pneumonia. Our studies demonstrate that the loss of one epithelial protein, αVß6 integrin, can alter the lung microenvironment during both homeostasis and respiratory infection leading to reduced lung injury and improved survival.


Asunto(s)
Antígenos de Neoplasias/inmunología , Integrinas/inmunología , Interferón Tipo I/biosíntesis , Interferón Tipo I/inmunología , Pulmón/inmunología , Infecciones del Sistema Respiratorio/inmunología , Traslado Adoptivo , Animales , Modelos Animales de Enfermedad , Ensayo de Inmunoadsorción Enzimática , Femenino , Citometría de Flujo , Técnica del Anticuerpo Fluorescente , Immunoblotting , Pulmón/microbiología , Macrófagos Alveolares/inmunología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Análisis de Secuencia por Matrices de Oligonucleótidos , Reacción en Cadena en Tiempo Real de la Polimerasa
15.
Proc Natl Acad Sci U S A ; 111(29): 10532-7, 2014 Jul 22.
Artículo en Inglés | MEDLINE | ID: mdl-25002480

RESUMEN

Extracellular fatty acid incorporation into the phospholipids of Staphylococcus aureus occurs via fatty acid phosphorylation. We show that fatty acid kinase (Fak) is composed of two dissociable protein subunits encoded by separate genes. FakA provides the ATP binding domain and interacts with two distinct FakB proteins to produce acyl-phosphate. The FakBs are fatty acid binding proteins that exchange bound fatty acid/acyl-phosphate with fatty acid/acyl-phosphate presented in detergent micelles or liposomes. The ΔfakA and ΔfakB1 ΔfakB2 strains were unable to incorporate extracellular fatty acids into phospholipid. FakB1 selectively bound saturated fatty acids whereas FakB2 preferred unsaturated fatty acids. Affymetrix array showed a global perturbation in the expression of virulence genes in the ΔfakA strain. The severe deficiency in α-hemolysin protein secretion in ΔfakA and ΔfakB1 ΔfakB2 mutants coupled with quantitative mRNA measurements showed that fatty acid kinase activity was required to support virulence factor transcription. These data reveal the function of two conserved gene families, their essential role in the incorporation of host fatty acids by Gram-positive pathogens, and connects fatty acid kinase to the regulation of virulence factor transcription in S. aureus.


Asunto(s)
Proteínas Bacterianas/metabolismo , Ácidos Grasos/metabolismo , Interacciones Huésped-Patógeno , Fosfotransferasas (aceptor de Grupo Carboxilo)/metabolismo , Staphylococcus aureus/enzimología , Secuencia de Aminoácidos , Proteínas Bacterianas/química , Datos de Secuencia Molecular , Fosfolípidos/metabolismo , Fosfotransferasas (aceptor de Grupo Carboxilo)/química , Staphylococcus aureus/patogenicidad , Especificidad por Sustrato , Transcripción Genética , Factores de Virulencia/genética , Factores de Virulencia/metabolismo
16.
Antimicrob Agents Chemother ; 60(7): 4264-73, 2016 07.
Artículo en Inglés | MEDLINE | ID: mdl-27161626

RESUMEN

Broad-spectrum antibiotic therapy decimates the gut microbiome, resulting in a variety of negative health consequences. Debio 1452 is a staphylococcus-selective enoyl-acyl carrier protein reductase (FabI) inhibitor under clinical development and was used to determine whether treatment with pathogen-selective antibiotics would minimize disturbance to the microbiome. The effect of oral Debio 1452 on the microbiota of mice was compared to the effects of four commonly used broad-spectrum oral antibiotics. During the 10 days of oral Debio 1452 treatment, there was minimal disturbance to the gut bacterial abundance and composition, with only the unclassified S24-7 taxon reduced at days 6 and 10. In comparison, broad-spectrum oral antibiotics caused ∼100- to 4,000-fold decreases in gut bacterial abundance and severely altered the microbial composition. The gut bacterial abundance and composition of Debio 1452-treated mice were indistinguishable from those of untreated mice 2 days after the antibiotic treatment was stopped. In contrast, the bacterial abundance in broad-spectrum-antibiotic-treated mice took up to 7 days to recover, and the gut composition of the broad-spectrum-antibiotic-treated mice remained different from that of the control group 20 days after the cessation of antibiotic treatment. These results illustrate that a pathogen-selective approach to antibiotic development will minimize disturbance to the gut microbiome.


Asunto(s)
Antibacterianos/farmacología , Animales , Heces/microbiología , Femenino , Microbioma Gastrointestinal/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Microbiota/efectos de los fármacos , Microbiota/genética , ARN Ribosómico 16S/genética
17.
BMC Microbiol ; 16(1): 271, 2016 11 09.
Artículo en Inglés | MEDLINE | ID: mdl-27829373

RESUMEN

BACKGROUND: Streptococcus pneumoniae is one of the leading causes of community acquired pneumonia and acute otitis media. Certain aspects of S. pneumoniae's virulence are dependent upon expression and release of the protein toxin pneumolysin (PLY) and upon the activity of the peroxide-producing enzyme, pyruvate oxidase (SpxB). We investigated the possible synergy of these two proteins and identified that release of PLY is enhanced by expression of SpxB prior to stationary phase growth. RESULTS: Mutants lacking the spxB gene were defective in PLY release and complementation of spxB restored PLY release. This was demonstrated by cytotoxic effects of sterile filtered supernatants upon epithelial cells and red blood cells. Additionally, peroxide production appeared to contribute to the mechanism of PLY release since a significant correlation was found between peroxide production and PLY release among a panel of clinical isolates. Exogenous addition of H2O2 failed to induce PLY release and catalase supplementation prevented PLY release in some strains, indicating peroxide may exert its effect intracellularly or in a strain-dependent manner. SpxB expression did not trigger bacterial cell death or LytA-dependent autolysis, but did predispose cells to deoxycholate lysis. CONCLUSIONS: Here we demonstrate a novel link between spxB expression and PLY release. These findings link liberation of PLY toxin to oxygen availability and pneumococcal metabolism.


Asunto(s)
Piruvato Oxidasa/metabolismo , Streptococcus pneumoniae/metabolismo , Estreptolisinas/metabolismo , Autólisis , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Catalasa , ADN Bacteriano/genética , Células Epiteliales/microbiología , Eritrocitos/microbiología , Genes Bacterianos , Peróxido de Hidrógeno/metabolismo , Oxígeno , Piruvato Oxidasa/genética , Eliminación de Secuencia , Streptococcus pneumoniae/enzimología , Streptococcus pneumoniae/genética , Estreptolisinas/genética , Virulencia
18.
J Infect Dis ; 212(2): 195-201, 2015 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-25505300

RESUMEN

BACKGROUND: Infection with influenza A virus (IAV) increases susceptibility to respiratory bacterial infections, resulting in increased bacterial carriage and complications such acute otitis media, pneumonia, bacteremia, and meningitis. Recently, vaccination with live attenuated influenza virus (LAIV) was reported to enhance subclinical bacterial colonization within the nasopharynx, similar to IAV. Although LAIV does not predispose to bacterial pneumonia, whether it may alter bacterial transmigration toward the middle ear, where it could have clinically relevant implications, has not been investigated. METHODS: BALB/c mice received LAIV or phosphate-buffered saline 1 or 7 days before or during pneumococcal colonization with either of 2 clinical isolates, 19F or 7F. Middle ear bacterial titers were monitored daily via in vivo imaging. RESULTS: LAIV increased bacterial transmigration to and persistence within the middle ear. When colonization followed LAIV inoculation, a minimum LAIV incubation period of 4 days was required before bacterial transmigration commenced. CONCLUSIONS: While LAIV vaccination is safe and effective at reducing IAV and coinfection with influenza virus and bacteria, LAIV may increase bacterial transmigration to the middle ear and could thus increase the risk of clinically relevant acute otitis media. These data warrant further investigations into interactions between live attenuated viruses and naturally colonizing bacterial pathogens.


Asunto(s)
Vacunas contra la Influenza/efectos adversos , Otitis Media/microbiología , Infecciones Neumocócicas/microbiología , Vacunación , Animales , Traslocación Bacteriana , Inmunidad Innata , Vacunas contra la Influenza/administración & dosificación , Vacunas contra la Influenza/inmunología , Ratones Endogámicos BALB C , Otitis Media/inmunología , Infecciones Neumocócicas/inmunología , Streptococcus pneumoniae/fisiología , Vacunas Vivas no Atenuadas/administración & dosificación , Vacunas Vivas no Atenuadas/efectos adversos , Vacunas Vivas no Atenuadas/inmunología
19.
Infect Immun ; 83(4): 1684-94, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25667262

RESUMEN

In bacteria, the intracellular levels of metals are mediated by tightly controlled acquisition and efflux systems. This is particularly true of copper, a trace element that is universally toxic in excess. During infection, the toxic properties of copper are exploited by the mammalian host to facilitate bacterial clearance. To better understand the role of copper during infection, we characterized the contribution of the cop operon to copper homeostasis and virulence in Streptococcus pneumoniae. Deletion of either the exporter, encoded by copA, or the chaperone, encoded by cupA, led to hypersensitivity to copper stress. We further demonstrated that loss of the copper exporter encoded by copA led to decreased virulence in pulmonary, intraperitoneal, and intravenous models of infection. Deletion of copA resulted in enhanced macrophage-mediated bacterial clearance in vitro. The attenuation phenotype of the copA mutant in the lung was found to be dependent on pulmonary macrophages, underscoring the importance of copper efflux in evading immune defenses. Overall, these data provide insight into the role of the cop operon in pneumococcal pathogenesis.


Asunto(s)
Proteínas Bacterianas/genética , Proteínas de Transporte de Catión/genética , Cobre/metabolismo , Infecciones Neumocócicas/patología , Streptococcus pneumoniae/patogenicidad , Animales , Bacteriemia/genética , Bacteriemia/patología , Adhesión Bacteriana , Línea Celular , Eliminación de Gen , Regulación Bacteriana de la Expresión Génica , Humanos , Pulmón/inmunología , Pulmón/microbiología , Macrófagos/inmunología , Ratones , Ratones Endogámicos BALB C , Chaperonas Moleculares/metabolismo , Infecciones Neumocócicas/genética , Streptococcus pneumoniae/genética , Streptococcus pneumoniae/inmunología
20.
Microbiology (Reading) ; 161(11): 2127-36, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26311256

RESUMEN

NAD is a necessary cofactor present in all living cells. Some bacteria cannot de novo synthesize NAD and must use the salvage pathway to import niacin or nicotinamide riboside via substrate importers NiaX and PnuC, respectively. Although homologues of these two importers and their substrates have been identified in other organisms, limited data exist in Streptococcus pneumoniae, specifically, on its effect on overall virulence. Here, we sought to characterize the substrate specificity of NiaX and PnuC in Str. pneumoniae TIGR4 and the contribution of these proteins to virulence of the pathogen. Although binding affinity of each importer for nicotinamide mononucleotide may overlap, we found NiaX to specifically import nicotinamide and nicotinic acid, and PnuC to be primarily responsible for nicotinamide riboside import. Furthermore, a pnuC mutant is completely attenuated during both intranasal and intratracheal infections in mice. Taken together, these findings underscore the importance of substrate salvage in pneumococcal pathogenesis and indicate that PnuC could potentially be a viable small-molecule therapeutic target to alleviate disease progression in the host.


Asunto(s)
Proteínas de Transporte de Membrana/metabolismo , NAD/metabolismo , Streptococcus pneumoniae/metabolismo , Streptococcus pneumoniae/patogenicidad , Factores de Virulencia/metabolismo , Animales , Modelos Animales de Enfermedad , Eliminación de Gen , Proteínas de Transporte de Membrana/genética , Ratones , Infecciones Neumocócicas/microbiología , Infecciones Neumocócicas/patología , Streptococcus pneumoniae/genética , Especificidad por Sustrato , Virulencia , Factores de Virulencia/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA