Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
mBio ; 10(1)2019 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-30647157

RESUMEN

Streptococcus pneumoniae is commonly carried asymptomatically in the human nasopharynx, but it also causes serious and invasive diseases such as pneumonia, bacteremia, and meningitis, as well as less serious but highly prevalent infections such as otitis media. We have previously shown that closely related pneumococci (of the same capsular serotype and multilocus sequence type [ST]) can display distinct pathogenic profiles in mice that correlate with clinical isolation site (e.g., blood versus ear), suggesting stable niche adaptation within a clonal lineage. This has provided an opportunity to identify determinants of disease tropism. Genomic analysis identified 17 and 27 single nucleotide polymorphisms (SNPs) or insertions/deletions in protein coding sequences between blood and ear isolates of serotype 14 ST15 and serotype 3 ST180, respectively. SNPs in raffinose uptake and utilization genes (rafR or rafK) were detected in both serotypes/lineages. Ear isolates were consistently defective in growth in media containing raffinose as the sole carbon source, as well as in expression of raffinose pathway genes aga, rafG, and rafK, relative to their serotype/ST-matched blood isolates. Similar differences were also seen between serotype 23F ST81 blood and ear isolates. Analysis of rafR allelic exchange mutants of the serotype 14 ST15 blood and ear isolates demonstrated that the SNP in rafR was entirely responsible for their distinct in vitro phenotypes and was also the determinant of differential tropism for the lungs versus ear and brain in a mouse intranasal challenge model. These data suggest that the ability of pneumococci to utilize raffinose determines the nature of disease.IMPORTANCES. pneumoniae is a component of the commensal nasopharyngeal microflora of humans, but from this reservoir, it can progress to localized or invasive disease with a frequency that translates into massive global morbidity and mortality. However, the factors that govern the switch from commensal to pathogen, as well as those that determine disease tropism, are poorly understood. Here we show that capacity to utilize raffinose can determine the nature of the disease caused by a given pneumococcal strain. Moreover, our findings provide an interesting example of convergent evolution, whereby pneumococci belonging to two unrelated serotypes/lineages exhibit SNPs in separate genes affecting raffinose uptake and utilization that correlate with distinct pathogenic profiles in vivo This further underscores the critical role of differential carbohydrate metabolism in the pathogenesis of localized versus invasive pneumococcal disease.


Asunto(s)
Infecciones Neumocócicas/microbiología , Infecciones Neumocócicas/patología , Rafinosa/metabolismo , Streptococcus pneumoniae/fisiología , Tropismo Viral , Animales , Sangre/microbiología , Carbono/metabolismo , Medios de Cultivo/química , Modelos Animales de Enfermedad , Oído/microbiología , Genotipo , Humanos , Redes y Vías Metabólicas/genética , Ratones , Polimorfismo de Nucleótido Simple , Serogrupo , Streptococcus pneumoniae/clasificación , Streptococcus pneumoniae/aislamiento & purificación
2.
PLoS One ; 13(1): e0190402, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29293606

RESUMEN

Streptococcus pneumoniae is the leading cause of community-acquired pneumonia in all ages worldwide, and with ever-increasing antibiotic resistance, the understanding of its pathogenesis and spread is as important as ever. Recently, we reported the presence of a Low Molecular Weight Tyrosine Phosphatase (LMWPTP) Spd1837 in the pneumococcus. This protein is encoded in an operon, OM001 with two other genes, with previous work implicating this operon as important for pneumococcal virulence. Thus, we set out to investigate the role of the individual genes in the operon during pneumococcal pathogenesis. As LMWPTPs play a major role in capsular polysaccharide (CPS) biosynthesis in many bacteria, we tested the effect of mutating spd1837 and its adjacent genes, spd1836 and spd1838 on CPS levels. Our results suggest that individual deletion of the genes, including the LMWPTP, did not modulate CPS levels, in multiple conditions, and in different strain backgrounds. Following in vivo studies, Spd1836 was identified as a novel virulence factor during pneumococcal invasive disease, in both the lungs and blood, with this protein alone responsible for the effects of operon's role in virulence. We also showed that a deletion in spd1836, spd1838 or the overall OM001 operon reduced survival in human saliva during the conditions that mimic transmission compared to the wildtype strain. With studies suggesting that survival in human saliva may be important for transmission, this study identifies Spd1836 and Spd1838 as transmission factors, potentially facilitating the spread of the pneumococcus from person to person. Overall, this study hopes to further our understanding of the bacterial transmission that precedes disease and outbreaks.


Asunto(s)
Operón , Polisacáridos/biosíntesis , Saliva/microbiología , Streptococcus pneumoniae/genética , Factores de Virulencia/metabolismo , Animales , Western Blotting , Electroforesis en Gel de Poliacrilamida , Femenino , Genes Bacterianos , Humanos , Ratones , Streptococcus pneumoniae/metabolismo , Streptococcus pneumoniae/patogenicidad , Virulencia
3.
Sci Rep ; 7(1): 17784, 2017 12 19.
Artículo en Inglés | MEDLINE | ID: mdl-29259314

RESUMEN

Streptococcus pneumoniae is a leading cause of morbidity and mortality globally. The Pilus-1 proteins, RrgA, RrgB and RrgC of S. pneumoniae have been previously assessed for their role in infection, invasive disease and as possible vaccine candidates. In this study we have investigated the glycan binding repertoire of all three Pilus-1 proteins, identifying that the tip adhesin RrgA has the broadest glycan recognition of the three proteins, binding to maltose/cellobiose, α/ß linked galactose and blood group A and H antigens. RrgB only bound mannose, while RrgC bound a subset of glycans also recognized by RrgA. Adherence of S. pneumoniae TIGR4 to epithelial cells was tested using four of the oligosaccharides identified through the glycan array analysis as competitive inhibitors. The blood group H trisaccharide provided the best blocking of S. pneumoniae TIGR4 adherence. Adherence is the first step in disease, and host glycoconjugates are a common target for many adhesins. This study has identified Pilus-1 proteins as new lectins involved in the targeting of host glycosylation by S. pneumoniae.


Asunto(s)
Fimbrias Bacterianas/metabolismo , Lectinas/metabolismo , Streptococcus pneumoniae/metabolismo , Células A549 , Antígenos Bacterianos/metabolismo , Proteínas Bacterianas/metabolismo , Línea Celular Tumoral , Celobiosa/metabolismo , Células Epiteliales/metabolismo , Proteínas Fimbrias/metabolismo , Galactosa/metabolismo , Humanos , Maltosa/metabolismo , Unión Proteica/fisiología , Factores de Virulencia/metabolismo
4.
Sci Rep ; 7(1): 2453, 2017 05 26.
Artículo en Inglés | MEDLINE | ID: mdl-28550292

RESUMEN

Streptococcus pneumoniae (the pneumococcus) is a human pathogen, accounting for massive global morbidity and mortality. Although asymptomatic colonization of the nasopharynx almost invariably precedes disease, the critical determinants enabling pneumococcal progression from this niche to cause invasive disease are poorly understood. One mechanism proposed to be central to this transition involves opacity phase variation, whereby pneumococci harvested from the nasopharynx are typically transparent, while those simultaneously harvested from the blood are opaque. Here, we used two dimensional-differential gel electrophoresis (2D-DIGE) to compare protein expression profiles of transparent and opaque variants of 3 pneumococcal strains, D39 (serotype 2), WCH43 (serotype 4) and WCH16 (serotype 6A) in vitro. One spot comprising a mixture of capsular polysaccharide biosynthesis protein and other proteins was significantly up-regulated in the opaque phenotype in all 3 strains; other proteins were differentially regulated in a strain-specific manner. We conclude that pneumococcal phase variation is a complex and multifactorial process leading to strain-specific pathogenicity.


Asunto(s)
Proteínas Bacterianas/análisis , Infecciones Neumocócicas/sangre , Proteómica/métodos , Streptococcus pneumoniae/metabolismo , Proteínas Bacterianas/metabolismo , Western Blotting , Electroforesis en Gel Bidimensional , Interacciones Huésped-Patógeno , Humanos , Nasofaringe/microbiología , Infecciones Neumocócicas/microbiología , Serogrupo , Serotipificación , Streptococcus pneumoniae/clasificación , Streptococcus pneumoniae/fisiología
5.
Infect Immun ; 84(3): 822-32, 2016 Jan 11.
Artículo en Inglés | MEDLINE | ID: mdl-26755156

RESUMEN

Streptococcus pneumoniae is the leading infectious cause of death in children in the world. However, the mechanisms that drive the progression from asymptomatic colonization to disease are poorly understood. Two virulence-associated genomic accessory regions (ARs) were deleted in a highly virulent serotype 1 clinical isolate (strain 4496) and examined for their contribution to pathogenesis. Deletion of a prophage encoding a platelet-binding protein (PblB) resulted in reduced adherence, biofilm formation, reduced initial infection within the lungs, and a reduction in the number of circulating platelets in infected mice. However, the region's overall contribution to the survival of mice was not significant. In contrast, deletion of the variable region of pneumococcal pathogenicity island 1 (vPPI1) was also responsible for a reduction in adherence and biofilm formation but also reduced survival and invasion of the pleural cavity, blood, and lungs. While the 4496ΔPPI1 strain induced higher expression of the genes encoding interleukin-10 (IL-10) and CD11b in the lungs of challenged mice than the wild-type strain, very few other genes exhibited altered expression. Moreover, while the level of IL-10 protein was increased in the lungs of 4496ΔPPI1 mutant-infected mice compared to strain 4496-infected mice, the levels of gamma interferon (IFN-γ), CXCL10, CCL2, and CCL4 were not different in the two groups. However, the 4496ΔPPI1 mutant was found to be more susceptible than the wild type to phagocytic killing by a macrophage-like cell line. Therefore, our data suggest that vPPI1 may be a major contributing factor to the heightened virulence of certain serotype 1 strains, possibly by influencing resistance to phagocytic killing.


Asunto(s)
Islas Genómicas , Infecciones Neumocócicas/microbiología , Streptococcus pneumoniae/genética , Streptococcus pneumoniae/patogenicidad , Animales , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Femenino , Humanos , Interleucina-10/genética , Interleucina-10/metabolismo , Pulmón/metabolismo , Pulmón/patología , Ratones , Infecciones Neumocócicas/genética , Infecciones Neumocócicas/metabolismo , Infecciones Neumocócicas/patología , Streptococcus pneumoniae/clasificación , Streptococcus pneumoniae/metabolismo , Virulencia
6.
Infect Immun ; 84(2): 407-15, 2016 02.
Artículo en Inglés | MEDLINE | ID: mdl-26573735

RESUMEN

Streptococcus pneumoniae is the world's foremost human pathogen. Acquisition of the first row transition metal ion zinc is essential for pneumococcal colonization and disease. Zinc is acquired via the ATP-binding cassette transporter AdcCB and two zinc-binding proteins, AdcA and AdcAII. We have previously shown that AdcAII is reliant upon the polyhistidine triad (Pht) proteins to aid in zinc recruitment. Pht proteins generally contain five histidine (His) triad motifs that are believed to facilitate zinc binding and therefore play a significant role in pneumococcal metal ion homeostasis. However, the importance and potential redundancy of these motifs have not been addressed. We examined the effects of mutating each of the five His triad motifs of PhtD. The combination of in vitro growth assays, active zinc uptake, and PhtD expression studies show that the His triad closest to the protein's amino terminus is the most important for zinc acquisition. Intriguingly, in vivo competitive infection studies investigating the amino- and carboxyl-terminal His triad mutants indicate that the motifs have similar importance in colonization. Collectively, our new insights into the contributions of the individual His triad motifs of PhtD, and by extension the other Pht proteins, highlight the crucial role of the first His triad site in zinc acquisition. This study also suggests that the Pht proteins likely play a role beyond zinc acquisition in pneumococcal virulence.


Asunto(s)
Secuencias de Aminoácidos , Proteínas Bacterianas/química , Proteínas Bacterianas/metabolismo , Homeostasis , Streptococcus pneumoniae/metabolismo , Zinc/metabolismo , Secuencias de Aminoácidos/genética , Animales , Carga Bacteriana , Proteínas Bacterianas/genética , Aptitud Genética , Histidina/química , Histidina/genética , Humanos , Ratones , Mutación , Streptococcus pneumoniae/genética , Streptococcus pneumoniae/crecimiento & desarrollo , Streptococcus pneumoniae/patogenicidad
7.
Infect Immun ; 83(12): 4781-90, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26416904

RESUMEN

Streptococcus pneumoniae is a diverse species causing invasive as well as localized infections that result in massive global morbidity and mortality. Strains vary markedly in pathogenic potential, but the molecular basis is obscured by the diversity and plasticity of the pneumococcal genome. We have previously reported that S. pneumoniae serotype 3 isolates belonging to the same multilocus sequence type (MLST) differed markedly in in vitro and in vivo phenotypes, in accordance with the clinical site of isolation, suggesting stable niche adaptation within a clonal lineage. In the present study, we have extended our analysis to serotype 14 clinical isolates from cases of sepsis or otitis media that belong to the same MLST (ST15). In a murine intranasal challenge model, five ST15 isolates (three from blood and two from ears) colonized the nasopharynx to similar extents. However, blood and ear isolates exhibited significant differences in bacterial loads in other host niches (lungs, ear, and brain) at both 24 and 72 h postchallenge. In spite of these differences, blood and ear isolates were present in the lungs at similar levels at 6 h postchallenge, suggesting that early immune responses may underpin the distinct virulence phenotypes. Transcriptional analysis of lung tissue from mice infected for 6 h with blood isolates versus ear isolates revealed 8 differentially expressed genes. Two of these were exclusively expressed in response to infection with the ear isolate. These results suggest a link between the differential capacities to elicit early innate immune responses and the distinct virulence phenotypes of clonally related S. pneumoniae strains.


Asunto(s)
Genoma Bacteriano/inmunología , Pulmón/inmunología , Nasofaringe/inmunología , Otitis Media/inmunología , Infecciones Neumocócicas/inmunología , Sepsis/inmunología , Streptococcus pneumoniae/genética , Animales , Células Clonales , Oído/microbiología , Oído/patología , Femenino , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Interacciones Huésped-Patógeno , Inmunidad Innata , Pulmón/microbiología , Pulmón/patología , Ratones , Tipificación de Secuencias Multilocus , Nasofaringe/microbiología , Nasofaringe/patología , Otitis Media/microbiología , Otitis Media/patología , Fenotipo , Infecciones Neumocócicas/microbiología , Infecciones Neumocócicas/patología , Sepsis/microbiología , Sepsis/patología , Serotipificación , Transducción de Señal , Streptococcus pneumoniae/clasificación , Streptococcus pneumoniae/inmunología , Streptococcus pneumoniae/patogenicidad , Virulencia
8.
Proc Natl Acad Sci U S A ; 111(49): E5312-20, 2014 Dec 09.
Artículo en Inglés | MEDLINE | ID: mdl-25422425

RESUMEN

The cholesterol-dependent cytolysin (CDC) pneumolysin (Ply) is a key virulence factor of Streptococcus pneumoniae. Membrane cholesterol is required for the cytolytic activity of this toxin, but it is not clear whether cholesterol is the only cellular receptor. Analysis of Ply binding to a glycan microarray revealed that Ply has lectin activity and binds glycans, including the Lewis histo-blood group antigens. Surface plasmon resonance analysis showed that Ply has the highest affinity for the sialyl LewisX (sLeX) structure, with a K(d) of 1.88 × 10(-5) M. Ply hemolytic activity against human RBCs showed dose-dependent inhibition by sLeX. Flow cytometric analysis and Western blots showed that blocking binding of Ply to the sLeX glycolipid on RBCs prevents deposition of the toxin in the membrane. The lectin domain responsible for sLeX binding is in domain 4 of Ply, which contains candidate carbohydrate-binding sites. Mutagenesis of these predicted carbohydrate-binding residues of Ply resulted in a decrease in hemolytic activity and a reduced affinity for sLeX. This study reveals that this archetypal CDC requires interaction with the sLeX glycolipid cellular receptor as an essential step before membrane insertion. A similar analysis conducted on streptolysin O from Streptococcus pyogenes revealed that this CDC also has glycan-binding properties and that hemolytic activity against RBCs can be blocked with the glycan lacto-N-neotetraose by inhibiting binding to the cell surface. Together, these data support the emerging paradigm shift that pore-forming toxins, including CDCs, have cellular receptors other than cholesterol that define target cell tropism.


Asunto(s)
Eritrocitos/metabolismo , Hemólisis , Polisacáridos/química , Estreptolisinas/química , Secuencia de Aminoácidos , Proteínas Bacterianas/química , Sitios de Unión , Carbohidratos/química , Línea Celular Tumoral , Membrana Celular/metabolismo , Citometría de Flujo , Glucolípidos/química , Humanos , Antígeno Lewis X/química , Datos de Secuencia Molecular , Mutagénesis , Oligosacáridos/química , Unión Proteica , Conformación Proteica , Homología de Secuencia de Aminoácido , Resonancia por Plasmón de Superficie
9.
PLoS One ; 9(8): e103625, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25105894

RESUMEN

Streptococcus pneumoniae is the world's leading cause of pneumonia, bacteremia, meningitis and otitis media. A major pneumococcal virulence factor is the cholesterol-dependent cytolysin, which has the defining property of forming pores in cholesterol-containing membranes. In recent times a clinically significant and internationally successful serotype 1 ST306 clone has been found to express a non-cytolytic variant of Ply (Ply306). However, while the pneumococcus is a naturally transformable organism, strains of the ST306 clonal group have to date been virtually impossible to transform, severely restricting efforts to understand the role of non-cytolytic Ply in the success of this clone. In this study isogenic Ply mutants were constructed in the D39 background and for the first time in the ST306 background (A0229467) to enable direct comparisons between Ply variants for their impact on the immune response in a macrophage-like cell line. Strains that expressed cytolytic Ply were found to induce a significant increase in IL-1ß release from macrophage-like cells compared to the non-cytolytic and Ply-deficient strains in a background-independent manner, confirming the requirement for pore formation in the Ply-dependent activation of the NLRP3 inflammasome. However, cytolytic activity in the D39 background was found to induce increased expression of the genes encoding GM-CSF (CSF2), p19 subunit of IL-23 (IL23A) and IFNß (IFNB1) compared to non-cytolytic and Ply-deficient D39 mutants, but had no effect in the A0229467 background. The impact of Ply on the immune response to the pneumococcus is highly dependent on the strain background, thus emphasising the importance of the interaction between specific virulence factors and other components of the genetic background of this organism.


Asunto(s)
Macrófagos/inmunología , Streptococcus pneumoniae/inmunología , Estreptolisinas/genética , Estreptolisinas/inmunología , Análisis de Varianza , Animales , Proteínas Bacterianas/genética , Proteínas Bacterianas/inmunología , Western Blotting , Células Cultivadas , Cartilla de ADN/genética , Ensayo de Inmunoadsorción Enzimática , Factor Estimulante de Colonias de Granulocitos y Macrófagos/metabolismo , Inflamasomas/metabolismo , Interleucina-1beta/metabolismo , Ratones , Reacción en Cadena de la Polimerasa , Análisis de Secuencia de ADN , Especificidad de la Especie , Streptococcus pneumoniae/genética
10.
Infect Immun ; 82(10): 4315-24, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25069983

RESUMEN

Streptococcus pneumoniae is a globally significant pathogen that causes a range of diseases, including pneumonia, sepsis, meningitis, and otitis media. Its ability to cause disease depends upon the acquisition of nutrients from its environment, including transition metal ions such as zinc. The pneumococcus employs a number of surface proteins to achieve this, among which are four highly similar polyhistidine triad (Pht) proteins. It has previously been established that these proteins collectively aid in the delivery of zinc to the ABC transporter substrate-binding protein AdcAII. Here we have investigated the contribution of each individual Pht protein to pneumococcal zinc homeostasis by analyzing mutant strains expressing only one of the four pht genes. Under conditions of low zinc availability, each of these mutants showed superior growth and zinc accumulation profiles relative to a mutant strain lacking all four genes, indicating that any of the four Pht proteins are able to facilitate delivery of zinc to AdcAII. However, optimal growth and zinc accumulation in vitro and pneumococcal survival and proliferation in vivo required production of all four Pht proteins, indicating that, despite their overlapping functionality, the proteins are not dispensable without incurring a fitness cost. We also show that surface-attached forms of the Pht proteins are required for zinc recruitment and that they do not contribute to defense against extracellular zinc stress.


Asunto(s)
Proteínas Bacterianas/metabolismo , Homeostasis , Streptococcus pneumoniae/metabolismo , Zinc/metabolismo , Animales , Proteínas Bacterianas/genética , Femenino , Eliminación de Gen , Ratones , Viabilidad Microbiana , Streptococcus pneumoniae/genética , Streptococcus pneumoniae/crecimiento & desarrollo , Streptococcus pneumoniae/patogenicidad , Virulencia
11.
Infect Immun ; 82(9): 3919-26, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-25001606

RESUMEN

The pneumococcus is the world's foremost respiratory pathogen, but the mechanisms allowing this pathogen to proceed from initial asymptomatic colonization to invasive disease are poorly understood. We have examined the early stages of invasive pneumococcal disease (IPD) by comparing host transcriptional responses to an invasive strain and a noninvasive strain of serotype 1 Streptococcus pneumoniae in the mouse lung. While the two strains were present in equal numbers in the lung 6 h after intranasal challenge, only the invasive strain (strain 1861) had invaded the pleural cavity at that time point; this correlated with subsequent development of bacteremia in mice challenged with strain 1861 but not the noninvasive strain (strain 1). Progression beyond the lung was associated with stronger induction of the type I interferon (IFN-I) response in the lung at 6 h. Suppression of the IFN-I response through administration of neutralizing antibody to IFNAR1 (the receptor for type I interferons) led to significantly reduced invasion of the pleural cavity by strain 1861 at 6 h postchallenge. Our data suggest that strong induction of the IFN-I response is a key factor in early progression of invasive serotype 1 strain 1861 beyond the lung during development of IPD.


Asunto(s)
Interferón Tipo I/inmunología , Pulmón/inmunología , Infecciones Neumocócicas/inmunología , Streptococcus pneumoniae/inmunología , Animales , Anticuerpos Antibacterianos/inmunología , Anticuerpos Neutralizantes/inmunología , Bacteriemia/inmunología , Bacteriemia/microbiología , Femenino , Pulmón/microbiología , Ratones , Cavidad Pleural/inmunología , Cavidad Pleural/microbiología , Infecciones Neumocócicas/microbiología
12.
mBio ; 4(5): e00655-13, 2013 Sep 10.
Artículo en Inglés | MEDLINE | ID: mdl-24023386

RESUMEN

UNLABELLED: Streptococcus pneumoniae is an important commensal and pathogen responsible for almost a million deaths annually in children under five. The formation of biofilms by S. pneumoniae is important in nasopharyngeal colonization, pneumonia, and otitis media. Pneumolysin (Ply) is a toxin that contributes significantly to the virulence of S. pneumoniae and is an important candidate as a serotype-independent vaccine target. Having previously demonstrated that a luxS knockout mutant was unable to form early biofilms and expressed less ply mRNA than the wild type, we conducted a study to investigate the role of Ply in biofilm formation. We found that Ply was expressed in early phases of biofilm development and localized to cellular aggregates as early as 4 h postinoculation. S. pneumoniae ply knockout mutants in D39 and TIGR4 backgrounds produced significantly less biofilm biomass than wild-type strains at early time points, both on polystyrene and on human respiratory epithelial cells, cultured under static or continuous-flow conditions. Ply's role in biofilm formation appears to be independent of its hemolytic activity, as S. pneumoniae serotype 1 strains, which produce a nonhemolytic variant of Ply, were still able to form biofilms. Transmission electron microscopy of biofilms grown on A549 lung cells using immunogold demonstrated that Ply was located both on the surfaces of pneumococcal cells and in the extracellular biofilm matrix. Altogether, our studies demonstrate a novel role for pneumolysin in the assembly of S. pneumoniae biofilms that is likely important during both carriage and disease and therefore significant for pneumolysin-targeting vaccines under development. IMPORTANCE: The bacterium Streptococcus pneumoniae (commonly known as the pneumococcus) is commonly carried in the human nasopharynx and can spread to other body sites to cause disease. In the nasopharynx, middle ear, and lungs, the pneumococcus forms multicellular surface-associated structures called biofilms. Pneumolysin is an important toxin produced by almost all S. pneumoniae strains, extensively studied for its ability to cause damage to human tissue. In this paper, we demonstrate that pneumolysin has a previously unrecognized role in biofilm formation by showing that strains without pneumolysin are unable to form the same amount of biofilm on plastic and human cell substrates. Furthermore, we show that the role of pneumolysin in biofilm formation is separate from the hemolytic activity responsible for tissue damage during pneumococcal diseases. This novel role for pneumolysin suggests that pneumococcal vaccines directed against this protein should be investigated for their potential impact on biofilms formed during carriage and disease.


Asunto(s)
Biopelículas , Infecciones Neumocócicas/microbiología , Streptococcus pneumoniae/fisiología , Estreptolisinas/metabolismo , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Biopelículas/crecimiento & desarrollo , Células Epiteliales/microbiología , Regulación Bacteriana de la Expresión Génica , Proteínas Hemolisinas/genética , Proteínas Hemolisinas/metabolismo , Humanos , Pulmón/microbiología , Streptococcus pneumoniae/genética , Streptococcus pneumoniae/crecimiento & desarrollo , Estreptolisinas/genética
13.
Infect Immun ; 79(10): 4122-30, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21788389

RESUMEN

Streptococcus pneumoniae is a leading cause of human diseases such as pneumonia, bacteremia, meningitis, and otitis media. Pneumolysin (Ply) is an important virulence factor of S. pneumoniae and a promising future vaccine target. However, the expansion of clones carrying ply alleles with reduced hemolytic activity has been observed in serotypes associated with outbreaks of invasive disease and includes an allele identified in a highly virulent serotype 1 isolate (ply4496). The virulence of Ply-deficient and ply allelic-replacement derivatives of S. pneumoniae D39 was compared with that of wild-type D39. In addition, the protective immunogenicity of Ply against pneumococci with low versus high hemolytic activity was also investigated. Replacement of D39 ply with ply4496 resulted in a small but statistically significant reduction of virulence. However, both native Ply- and Ply4496-expressing strains were significantly more virulent than a Ply-deficient mutant. While the numbers of both Ply- and Ply4496-expressing isolate cells were higher in the blood than the numbers of Ply-deficient mutant cells, the growth of the Ply4496-expressing strain was superior to that of the wild type in the first 15 h postchallenge. Ply immunization provided protection regardless of the hemolytic activity of the challenge strain. In summary, we show that low-hemolytic-activity Ply alleles contribute to systemic virulence and may provide a survival advantage in the blood. Moreover, pneumococci expressing such alleles remain vulnerable to Ply-based vaccines.


Asunto(s)
Bacteriemia/microbiología , Sangre/microbiología , Streptococcus pneumoniae/crecimiento & desarrollo , Streptococcus pneumoniae/patogenicidad , Estreptolisinas/genética , Secuencia de Aminoácidos , Animales , Animales no Consanguíneos , Proteínas Bacterianas/genética , Proteínas Bacterianas/inmunología , Femenino , Hemólisis , Humanos , Inmunización , Ratones , Datos de Secuencia Molecular , Mutación , Infecciones Neumocócicas/epidemiología , Infecciones Neumocócicas/prevención & control , Alineación de Secuencia , Análisis de Secuencia de ADN , Streptococcus pneumoniae/inmunología , Estreptolisinas/deficiencia , Estreptolisinas/inmunología , Virulencia
14.
PLoS One ; 6(5): e19650, 2011 May 05.
Artículo en Inglés | MEDLINE | ID: mdl-21573186

RESUMEN

The bacterial factors responsible for the variation in invasive potential between different clones and serotypes of Streptococcus pneumoniae are largely unknown. Therefore, the isolation of rare serotype 1 carriage strains in Indigenous Australian communities provided a unique opportunity to compare the genomes of non-invasive and invasive isolates of the same serotype in order to identify such factors. The human virulence status of non-invasive, intermediately virulent and highly virulent serotype 1 isolates was reflected in mice and showed that whilst both human non-invasive and highly virulent isolates were able to colonize the murine nasopharynx equally, only the human highly virulent isolates were able to invade and survive in the murine lungs and blood. Genomic sequencing comparisons between these isolates identified 8 regions >1 kb in size that were specific to only the highly virulent isolates, and included a version of the pneumococcal pathogenicity island 1 variable region (PPI-1v), phage-associated adherence factors, transporters and metabolic enzymes. In particular, a phage-associated endolysin, a putative iron/lead permease and an operon within PPI-1v exhibited niche-specific changes in expression that suggest important roles for these genes in the lungs and blood. Moreover, in vivo competition between pneumococci carrying PPI-1v derivatives representing the two identified versions of the region showed that the version of PPI-1v in the highly virulent isolates was more competitive than the version from the less virulent isolates in the nasopharyngeal tissue, blood and lungs. This study is the first to perform genomic comparisons between serotype 1 isolates with distinct virulence profiles that correlate between mice and humans, and has highlighted the important role that hypervariable genomic loci, such as PPI-1v, play in pneumococcal disease. The findings of this study have important implications for understanding the processes that drive progression from colonization to invasive disease and will help direct the development of novel therapeutic strategies.


Asunto(s)
Variación Genética , Genoma Bacteriano/genética , Islas Genómicas/genética , Streptococcus pneumoniae/genética , Streptococcus pneumoniae/patogenicidad , Animales , Recuento de Colonia Microbiana , Modelos Animales de Enfermedad , Regulación Bacteriana de la Expresión Génica , Genes Bacterianos/genética , Humanos , Pulmón/microbiología , Ratones , Ratones Endogámicos BALB C , Mutación/genética , Nasofaringe/microbiología , Fenotipo , Infecciones Neumocócicas/microbiología , Reproducibilidad de los Resultados , Análisis de Secuencia de ADN , Serotipificación , Especificidad de la Especie , Streptococcus pneumoniae/clasificación , Streptococcus pneumoniae/aislamiento & purificación , Virulencia/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA