Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Sci Rep ; 8(1): 2593, 2018 02 07.
Artículo en Inglés | MEDLINE | ID: mdl-29416049

RESUMEN

Group B Streptococcus (GBS) is a normal inhabitant of recto-vaginal mucosae in up to 30% of healthy women. Colonization is a major risk factor for perinatal infection which can lead to severe complications such as stillbirth and neonatal invasive disease. Intra-partum antibiotic prophylaxis in colonized women is a safe and cost-effective preventive measure against early-onset disease in the first days of life, but has no effect on late-onset manifestations or on early maternal infection. Maternal immunization with capsular polysaccharide-based vaccines shows promise for the prevention of both early-onset and late-onset neonatal infections, although ability to prevent maternal colonization and ascending infection has been less studied. Here we investigated the effect of a GBS glycoconjugate vaccine since the very early stage of maternal GBS acquisition to neonatal outcome by rodent models of vaginal colonization and ascending infection. Immunization of female mice and rats with a type III glycoconjugate reduced vaginal colonization, infection of chorioamniotic/ placental membranes and bacterial transmission to fetuses and pups. Type III specific antibodies were detected in the blood and vagina of vaccinated mothers and their offspring. The obtained data support a potential preventive effect of GBS glycoconjugate vaccines during the different stages of pregnancy.


Asunto(s)
Transmisión Vertical de Enfermedad Infecciosa/prevención & control , Polisacáridos Bacterianos/inmunología , Infecciones Estreptocócicas/prevención & control , Vacunas Estreptocócicas/inmunología , Vagina/microbiología , Animales , Modelos Animales de Enfermedad , Femenino , Ratones , Polisacáridos Bacterianos/administración & dosificación , Ratas , Infecciones Estreptocócicas/microbiología , Vacunas Estreptocócicas/administración & dosificación , Vacunación
2.
Clin Vaccine Immunol ; 23(6): 442-50, 2016 06.
Artículo en Inglés | MEDLINE | ID: mdl-27030589

RESUMEN

Staphylococcus aureus alpha-hemolysin (Hla) assembles into heptameric pores on the host cell membrane, causing lysis, apoptosis, and junction disruption. Herein, we present the design of a newly engineered S. aureus alpha-toxin, HlaPSGS, which lacks the predicted membrane-spanning stem domain. This protein is able to form heptamers in aqueous solution in the absence of lipophilic substrata, and its structure, obtained by transmission electron microscopy and single-particle reconstruction analysis, resembles the cap of the wild-type cytolytic Hla pore. HlaPSGS was found to be impaired in binding to host cells and to its receptor ADAM10 and to lack hemolytic and cytotoxic activity. Immunological studies using human sera as well as sera from mice convalescent from S. aureus infection suggested that the heptameric conformation of HlaPSGS mimics epitopes exposed by the cytolytic Hla pore during infection. Finally, immunization with this newly engineered Hla generated high protective immunity against staphylococcal infection in mice. Overall, this study provides unprecedented data on the natural immune response against Hla and suggests that the heptameric HlaPSGS is a highly valuable vaccine candidate against S. aureus.


Asunto(s)
Toxinas Bacterianas/química , Toxinas Bacterianas/inmunología , Proteínas Hemolisinas/química , Proteínas Hemolisinas/inmunología , Imitación Molecular , Infecciones Estafilocócicas/prevención & control , Staphylococcus aureus , Proteína ADAM10/metabolismo , Animales , Toxinas Bacterianas/administración & dosificación , Toxinas Bacterianas/genética , Línea Celular , Citotoxinas , Epítopos/inmunología , Escherichia coli/genética , Proteínas Hemolisinas/administración & dosificación , Proteínas Hemolisinas/genética , Humanos , Proteínas de la Membrana/metabolismo , Ratones , Microscopía Electrónica de Transmisión , Modelos Moleculares , Ingeniería de Proteínas , Proteínas Recombinantes/administración & dosificación , Proteínas Recombinantes/genética , Proteínas Recombinantes/inmunología , Proteínas Recombinantes/aislamiento & purificación , Vacunas Estafilocócicas/inmunología , Staphylococcus aureus/química , Staphylococcus aureus/metabolismo , Vacunación
3.
PLoS One ; 11(1): e0147767, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26812180

RESUMEN

A rapidly acting, single dose vaccine against Staphylococcus aureus would be highly beneficial for patients scheduled for major surgeries or in intensive care units. Here we show that one immunization with a multicomponent S. aureus candidate vaccine, 4C-Staph, formulated with a novel TLR7-dependent adjuvant, T7-alum, readily protected mice from death and from bacterial dissemination, both in kidney abscess and peritonitis models, outperforming alum-formulated vaccine. This increased efficacy was paralleled by higher vaccine-specific and α-hemolysin-neutralizing antibody titers and Th1/Th17 cell responses. Antibodies played a crucial protective role, as shown by the lack of protection of 4C-Staph/T7-alum vaccine in B-cell-deficient mice and by serum transfer experiments. Depletion of effector CD4+ T cells not only reduced survival but also increased S. aureus load in kidneys of mice immunized with 4C-Staph/T7-alum. The role of IL-17A in the control of bacterial dissemination in 4C-Staph/T7-alum vaccinated mice was indicated by in vivo neutralization experiments. We conclude that single dose 4C-Staph/T7-alum vaccine promptly and efficiently protected mice against S. aureus through the combined actions of antibodies, CD4+ effector T cells, and IL-17A. These data suggest that inclusion of an adjuvant that induces not only fast antibody responses but also IL-17-producing cell-mediated effector responses could efficaciously protect patients scheduled for major surgeries or in intensive care units.


Asunto(s)
Anticuerpos Antibacterianos/inmunología , Linfocitos T CD4-Positivos/inmunología , Interleucina-17/metabolismo , Infecciones Estafilocócicas/prevención & control , Vacunas Estafilocócicas/inmunología , Staphylococcus aureus/inmunología , Receptor Toll-Like 7/metabolismo , Adyuvantes Inmunológicos , Animales , Anticuerpos Neutralizantes/inmunología , Linfocitos T CD4-Positivos/citología , Citocinas/metabolismo , Femenino , Ratones , Ratones Endogámicos C57BL , Bazo/metabolismo , Bazo/patología , Infecciones Estafilocócicas/inmunología , Infecciones Estafilocócicas/mortalidad , Staphylococcus aureus/genética , Tasa de Supervivencia , Células TH1/inmunología , Células Th17/inmunología , Receptor Toll-Like 7/inmunología
4.
Front Immunol ; 6: 439, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26441955

RESUMEN

Staphylococcus aureus (S. aureus) is an important opportunistic pathogen that may cause invasive life-threatening infections, like sepsis and pneumonia. Due to the increasing antibiotic resistance, the development of an effective vaccine against S. aureus is needed. Although a correlate of protection against staphylococcal diseases is not yet established, several findings suggest that both antibodies and CD4 T cells might contribute to optimal immunity. In this study, we show that adjuvanting a multivalent vaccine (4C-Staph) with MF59, an oil-in-water emulsion licensed in human vaccines, further potentiated antigen-specific IgG titers and CD4 T-cell responses compared to alum and conferred protection in the peritonitis model of S. aureus infection. Moreover, we showed that MF59- and alum-adjuvanted 4C-Staph vaccines induced persistent antigen-specific humoral and T-cell responses, and protected mice from infection up to 4 months after immunization. Furthermore, 4C-Staph formulated with MF59 was used to investigate which immune compartment is involved in vaccine-induced protection. Using CD4 T cell-depleted mice or B cell-deficient mice, we demonstrated that both T and B-cell responses contributed to 4C-Staph vaccine-mediated protective immunity. However, the role of CD4 T cells seemed more evident in the presence of low-antibody responses. This study provides preclinical data further supporting the use of the adjuvanted 4C-Staph vaccines against S. aureus diseases, and provides critical insights on the correlates of protective immunity necessary to combat this pathogen.

5.
Infect Immun ; 83(8): 3157-63, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-26015481

RESUMEN

Staphylococcus aureus is a human bacterial pathogen causing a variety of diseases. The occurrence of multidrug-resistant strains of Staphylococcus aureus underlines the need for a vaccine. Defining immune correlates of protection may support the design of an effective vaccine. We used a murine Staphylococcus aureus infection model, in which bacteria were inoculated in an air pouch generated on the back of the animal. Analysis of the air-pouch content in mice immunized or not with an adjuvanted multiantigen vaccine formulation, four-component S. aureus vaccine (4C-Staph), prior to infection allowed us to measure bacteria, cytokines, and 4C-Staph-specific antibodies and to analyze host immune cells recruited to the infection site. Immunization with 4C-Staph resulted in accumulation of antigen-specific antibodies in the pouch and mitigated the infection. Neutrophils were the most abundant cells in the pouch, and they showed the upregulation of Fcγ receptor (FcγR) following immunization with 4C-Staph. Reduction of the infection was also obtained in mice immunized with 4C-Staph and depleted of neutrophils; these mice showed an increase in monocytes and macrophages. Upregulation of the FcγR and the presence of antigen-specific antibodies induced by immunization with 4C-Staph may contribute to increase bacterial opsonophagocytosis. Protection in neutropenic mice indicated that an effective vaccine could activate alternative protection mechanisms compensating for neutropenia, a condition often occurring in S. aureus-infected patients.


Asunto(s)
Monocitos/inmunología , Neutropenia/inmunología , Neutrófilos/inmunología , Receptores de IgG/genética , Infecciones Estafilocócicas/inmunología , Vacunas Estafilocócicas/inmunología , Staphylococcus aureus/inmunología , Animales , Anticuerpos Antibacterianos/inmunología , Modelos Animales de Enfermedad , Femenino , Humanos , Inmunización , Ratones , Ratones Endogámicos C57BL , Neutropenia/genética , Neutropenia/microbiología , Receptores de IgG/inmunología , Infecciones Estafilocócicas/genética , Infecciones Estafilocócicas/microbiología , Vacunas Estafilocócicas/administración & dosificación , Vacunas Estafilocócicas/genética , Staphylococcus aureus/genética
6.
Proc Natl Acad Sci U S A ; 112(12): 3680-5, 2015 Mar 24.
Artículo en Inglés | MEDLINE | ID: mdl-25775551

RESUMEN

Both active and passive immunization strategies against Staphylococcus aureus have thus far failed to show efficacy in humans. With the attempt to develop an effective S. aureus vaccine, we selected five conserved antigens known to have different roles in S. aureus pathogenesis. They include the secreted factors α-hemolysin (Hla), ess extracellular A (EsxA), and ess extracellular B (EsxB) and the two surface proteins ferric hydroxamate uptake D2 and conserved staphylococcal antigen 1A. The combined vaccine antigens formulated with aluminum hydroxide induced antibodies with opsonophagocytic and functional activities and provided consistent protection in four mouse models when challenged with a panel of epidemiologically relevant S. aureus strains. The importance of antibodies in protection was demonstrated by passive transfer experiments. Furthermore, when formulated with a toll-like receptor 7-dependent (TLR7) agonist recently designed and developed in our laboratories (SMIP.7-10) adsorbed to alum, the five antigens provided close to 100% protection against four different staphylococcal strains. The new formulation induced not only high antibody titers but also a Th1 skewed immune response as judged by antibody isotype and cytokine profiles. In addition, low frequencies of IL-17-secreting T cells were also observed. Altogether, our data demonstrate that the rational selection of mixtures of conserved antigens combined with Th1/Th17 adjuvants can lead to promising vaccine formulations against S. aureus.


Asunto(s)
Adyuvantes Inmunológicos/farmacología , Infecciones Estafilocócicas/prevención & control , Vacunas Estafilocócicas/química , Receptor Toll-Like 7/química , Absceso/patología , Inmunidad Adaptativa , Animales , Antibacterianos/química , Anticuerpos Antibacterianos/inmunología , Antígenos/inmunología , Humanos , Ratones , Modelos Animales , Infecciones Estafilocócicas/inmunología , Staphylococcus aureus , Células TH1/inmunología
7.
Mol Cell Proteomics ; 14(2): 418-29, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25368410

RESUMEN

New generation vaccines are in demand to include only the key antigens sufficient to confer protective immunity among the plethora of pathogen molecules. In the last decade, large-scale genomics-based technologies have emerged. Among them, the Reverse Vaccinology approach was successfully applied to the development of an innovative vaccine against Neisseria meningitidis serogroup B, now available on the market with the commercial name BEXSERO® (Novartis Vaccines). The limiting step of such approaches is the number of antigens to be tested in in vivo models. Several laboratories have been trying to refine the original approach in order to get to the identification of the relevant antigens straight from the genome. Here we report a new bioinformatics tool that moves a first step in this direction. The tool has been developed by identifying structural/functional features recurring in known bacterial protective antigens, the so called "Protectome space," and using such "protective signatures" for protective antigen discovery. In particular, we applied this new approach to Staphylococcus aureus and Group B Streptococcus and we show that not only already known protective antigens were re-discovered, but also two new protective antigens were identified.


Asunto(s)
Vacunas Bacterianas/inmunología , Biología Computacional/métodos , Proteoma/inmunología , 5'-Nucleotidasa/metabolismo , Animales , Proteínas Bacterianas/inmunología , Línea Celular , Modelos Animales de Enfermedad , Femenino , Ratones , Neisseria meningitidis Serogrupo B/inmunología , Señales de Clasificación de Proteína , Reproducibilidad de los Resultados , Staphylococcus aureus/inmunología , Streptococcus agalactiae/inmunología
8.
PLoS Pathog ; 10(5): e1004124, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24809621

RESUMEN

SslE, the Secreted and surface-associated lipoprotein from Escherichia coli, has recently been associated to the M60-like extracellular zinc-metalloprotease sub-family which is implicated in glycan recognition and processing. SslE can be divided into two main variants and we recently proposed it as a potential vaccine candidate. By applying a number of in vitro bioassays and comparing wild type, knockout mutant and complemented strains, we have now demonstrated that SslE specifically contributes to degradation of mucin substrates, typically present in the intestine and bladder. Mutation of the zinc metallopeptidase motif of SslE dramatically impaired E. coli mucinase activity, confirming the specificity of the phenotype observed. Moreover, antibodies raised against variant I SslE, cloned from strain IHE3034 (SslEIHE3034), are able to inhibit translocation of E. coli strains expressing different variants through a mucin-based matrix, suggesting that SslE induces cross-reactive functional antibodies that affect the metallopeptidase activity. To test this hypothesis, we used well-established animal models and demonstrated that immunization with SslEIHE3034 significantly reduced gut, kidney and spleen colonization by strains producing variant II SslE and belonging to different pathotypes. Taken together, these data strongly support the importance of SslE in E. coli colonization of mucosal surfaces and reinforce the use of this antigen as a component of a broadly protective vaccine against pathogenic E. coli species.


Asunto(s)
Anticuerpos Antibacterianos/farmacología , Formación de Anticuerpos , Infecciones por Escherichia coli , Proteínas de Escherichia coli/inmunología , Polisacárido Liasas/antagonistas & inhibidores , Factores de Virulencia/inmunología , Animales , Animales no Consanguíneos , Anticuerpos Antibacterianos/metabolismo , Células Cultivadas , Escherichia coli Enteropatógena/crecimiento & desarrollo , Escherichia coli Enteropatógena/inmunología , Escherichia coli Enteropatógena/metabolismo , Activación Enzimática/efectos de los fármacos , Escherichia coli/crecimiento & desarrollo , Escherichia coli/inmunología , Escherichia coli/metabolismo , Infecciones por Escherichia coli/inmunología , Infecciones por Escherichia coli/metabolismo , Infecciones por Escherichia coli/microbiología , Proteínas de Escherichia coli/antagonistas & inhibidores , Proteínas de Escherichia coli/metabolismo , Femenino , Intestinos/microbiología , Ratones , Ratones Endogámicos CBA , Polisacárido Liasas/inmunología , Polisacárido Liasas/metabolismo , Factores de Virulencia/antagonistas & inhibidores , Factores de Virulencia/metabolismo
9.
PLoS One ; 8(9): e74718, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24069334

RESUMEN

Staphylococcus aureus is an opportunistic pathogen, commensal of the human skin and nares, but also responsible for invasive nosocomial as well as community acquired infections. Staphylococcus aureus adheres to the host tissues by means of surface adhesins, such as SdrC, SdrD, and SdrE proteins. The Sdr family of proteins together with a functional A domain, contain respectively two, three or five repeated sequences called B motifs which comprise the CnaB domains. SdrD and SdrE proteins were reported to be protective in animal models against invasive diseases or lethal challenge with human clinical S. aureus isolates. In this study we identified a 126 amino acid sequence containing a CnaB domain, conserved among the three Sdr proteins. The three fragments defined here as CnaBC2, D5 and E3 domains even though belonging to phylogenetically distinct strains, displayed high sequence similarity. Based on the sequence conservation data, we selected the CnaBE3 domain for further analysis and characterization. Polyclonal antibodies raised against the recombinant CnaBE3 domain recognized SdrE, SdrC and SdrD proteins of different S. aureus lineages. Moreover, we demonstrated that the CnaBE3 domain was expressed in vivo during S. aureus infections, and that immunization of this domain alone significantly reduces the bacterial load in mice challenged with S. aureus. Furthermore, we show that the reduction of bacteria by CnaBE3 vaccination is due to functional antibodies. Finally, we demonstrated that the region of the SdrE protein containing the CnaBE3 domain was resistant to trypsin digestion, a characteristic often associated with the presence of an isopeptide bond.


Asunto(s)
Adhesinas Bacterianas/metabolismo , Dominios y Motivos de Interacción de Proteínas , Staphylococcus aureus/metabolismo , Adhesinas Bacterianas/química , Adhesinas Bacterianas/genética , Adhesinas Bacterianas/inmunología , Secuencia de Aminoácidos , Animales , Anticuerpos Antibacterianos/inmunología , Especificidad de Anticuerpos/inmunología , Citotoxicidad Celular Dependiente de Anticuerpos , Carga Bacteriana , Proteínas Bacterianas/química , Proteínas Bacterianas/genética , Proteínas Bacterianas/inmunología , Proteínas Bacterianas/metabolismo , Secuencia Conservada , Femenino , Humanos , Ratones , Datos de Secuencia Molecular , Filogenia , Dominios y Motivos de Interacción de Proteínas/genética , Dominios y Motivos de Interacción de Proteínas/inmunología , Alineación de Secuencia , Infecciones Estafilocócicas/microbiología , Staphylococcus aureus/clasificación , Staphylococcus aureus/genética
10.
Biochem J ; 455(3): 273-84, 2013 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-23895222

RESUMEN

In the human pathogen Staphylococcus aureus, there exists an enormous diversity of proteins containing DUFs (domains of unknown function). In the present study, we characterized the family of conserved staphylococcal antigens (Csa) classified as DUF576 and taxonomically restricted to Staphylococci. The 18 Csa paralogues in S. aureus Newman are highly similar at the sequence level, yet were found to be expressed in multiple cellular locations. Extracellular Csa1A was shown to be post-translationally processed and released. Molecular interaction studies revealed that Csa1A interacts with other Csa paralogues, suggesting that these proteins are involved in the same cellular process. The structures of Csa1A and Csa1B were determined by X-ray crystallography, unveiling a peculiar structure with limited structural similarity to other known proteins. Our results provide the first detailed biological characterization of this family and confirm the uniqueness of this family also at the structural level. We also provide evidence that Csa family members elicit protective immunity in in vivo animal models of staphylococcal infections, indicating a possible important role for these proteins in S. aureus biology and pathogenesis. These findings identify the Csa family as new potential vaccine candidates, and underline the importance of mining the bacterial unknown proteome to identify new targets for preventive vaccines.


Asunto(s)
Antígenos Bacterianos/química , Proteínas Bacterianas/química , Proteoma/química , Staphylococcus aureus/metabolismo , Animales , Antígenos Bacterianos/inmunología , Antígenos Bacterianos/metabolismo , Proteínas Bacterianas/inmunología , Proteínas Bacterianas/metabolismo , Cristalografía por Rayos X , Minería de Datos , Ratones , Ratones Endogámicos , Proteoma/genética , Proteoma/metabolismo , Infecciones Estafilocócicas/inmunología , Infecciones Estafilocócicas/metabolismo , Staphylococcus aureus/inmunología
11.
J Infect Dis ; 206(7): 1041-9, 2012 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-22829645

RESUMEN

Iron availability plays an essential role in staphylococcal pathogenesis. We selected FhuD2, a lipoprotein involved in iron-hydroxamate uptake, as a novel vaccine candidate against Staphylococcus aureus. Unprecedented for staphylococcal lipoproteins, the protein was demonstrated to have a discrete, punctate localization on the bacterial surface. FhuD2 vaccination generated protective immunity against diverse clinical S. aureus isolates in murine infection models. Protection appeared to be associated with functional antibodies that were shown to mediate opsonophagocytosis, to be effective in passive transfer experiments, and to potentially block FhuD2-mediated siderophore uptake. Furthermore, the protein was found to be up-regulated in infected tissues and was required for staphylococcal dissemination and abscess formation. Herein we show that the staphylococcal iron-hydroxamate uptake system is important in invasive infection and functions as an efficacious vaccine target.


Asunto(s)
Antígenos Bacterianos/metabolismo , Proteínas de la Membrana Bacteriana Externa/metabolismo , Proteínas de Transporte de Membrana/metabolismo , Infecciones Estafilocócicas/prevención & control , Staphylococcus aureus/metabolismo , Vacunación , Absceso/inmunología , Absceso/prevención & control , Secuencia de Aminoácidos , Animales , Anticuerpos Antibacterianos/sangre , Antígenos Bacterianos/genética , Antígenos Bacterianos/inmunología , Proteínas de la Membrana Bacteriana Externa/genética , Proteínas de la Membrana Bacteriana Externa/inmunología , Compuestos Férricos/metabolismo , Regulación Bacteriana de la Expresión Génica , Técnicas de Inactivación de Genes , Células HL-60 , Humanos , Ácidos Hidroxámicos/metabolismo , Proteínas de Transporte de Membrana/genética , Proteínas de Transporte de Membrana/inmunología , Ratones , Datos de Secuencia Molecular , Transporte de Proteínas , Conejos , Sepsis/inmunología , Sepsis/prevención & control , Infecciones Estafilocócicas/inmunología , Vacunas Estafilocócicas/administración & dosificación , Staphylococcus aureus/crecimiento & desarrollo , Staphylococcus aureus/inmunología
12.
mBio ; 3(2)2012.
Artículo en Inglés | MEDLINE | ID: mdl-22496310

RESUMEN

UNLABELLED: The increasing antibiotic resistance of pathogenic Escherichia coli species and the absence of a pan-protective vaccine pose major health concerns. We recently identified, by subtractive reverse vaccinology, nine Escherichia coli antigens that protect mice from sepsis. In this study, we characterized one of them, ECOK1_0290, named FdeC (factor adherence E. coli) for its ability to mediate E. coli adhesion to mammalian cells and extracellular matrix. This adhesive propensity was consistent with the X-ray structure of one of the FdeC domains that shows a striking structural homology to Yersinia pseudotuberculosis invasin and enteropathogenic E. coli intimin. Confocal imaging analysis revealed that expression of FdeC on the bacterial surface is triggered by interaction of E. coli with host cells. This phenotype was also observed in bladder tissue sections derived from mice infected with an extraintestinal strain. Indeed, we observed that FdeC contributes to colonization of the bladder and kidney, with the wild-type strain outcompeting the fdeC mutant in cochallenge experiments. Finally, intranasal mucosal immunization with recombinant FdeC significantly reduced kidney colonization in mice challenged transurethrally with uropathogenic E. coli, supporting a role for FdeC in urinary tract infections. IMPORTANCE: Pathogenic Escherichia coli strains are involved in a diverse spectrum of diseases, including intestinal and extraintestinal infections (urinary tract infections and sepsis). The absence of a broadly protective vaccine against all these E. coli strains is a major problem for modern society due to high costs to health care systems. Here, we describe the structural and functional properties of a recently reported protective antigen, named FdeC, and elucidated its putative role during extraintestinal pathogenic E. coli infection by using both in vitro and in vivo infection models. The conservation of FdeC among strains of different E. coli pathotypes highlights its potential as a component of a broadly protective vaccine against extraintestinal and intestinal E. coli infections.


Asunto(s)
Adhesinas de Escherichia coli/inmunología , Infecciones por Escherichia coli/prevención & control , Escherichia coli/inmunología , Escherichia coli/patogenicidad , Infecciones Urinarias/prevención & control , Adhesinas de Escherichia coli/química , Adhesinas de Escherichia coli/genética , Administración Intranasal , Animales , Carga Bacteriana , Cristalografía por Rayos X , Modelos Animales de Enfermedad , Infecciones por Escherichia coli/inmunología , Infecciones por Escherichia coli/microbiología , Vacunas contra Escherichia coli/administración & dosificación , Vacunas contra Escherichia coli/inmunología , Proteínas de la Matriz Extracelular , Femenino , Perfilación de la Expresión Génica , Regulación Bacteriana de la Expresión Génica , Riñón/microbiología , Ratones , Ratones Endogámicos CBA , Microscopía Confocal , Modelos Moleculares , Unión Proteica , Conformación Proteica , Vejiga Urinaria/microbiología , Infecciones Urinarias/inmunología , Infecciones Urinarias/microbiología , Vacunas Sintéticas/administración & dosificación , Vacunas Sintéticas/inmunología
13.
Proc Natl Acad Sci U S A ; 107(20): 9072-7, 2010 May 18.
Artículo en Inglés | MEDLINE | ID: mdl-20439758

RESUMEN

Extraintestinal pathogenic Escherichia coli (ExPEC) are a common cause of disease in both mammals and birds. A vaccine to prevent such infections would be desirable given the increasing antibiotic resistance of these bacteria. We have determined the genome sequence of ExPEC IHE3034 (ST95) isolated from a case of neonatal meningitis and compared this to available genome sequences of other ExPEC strains and a few nonpathogenic E. coli. We found 19 genomic islands present in the genome of IHE3034, which are absent in the nonpathogenic E. coli isolates. By using subtractive reverse vaccinology we identified 230 antigens present in ExPEC but absent (or present with low similarity) in nonpathogenic strains. Nine antigens were protective in a mouse challenge model. Some of them were also present in other pathogenic non-ExPEC strains, suggesting that a broadly protective E. coli vaccine may be possible. The gene encoding the most protective antigen was detected in most of the E. coli isolates, highly conserved in sequence and found to be exported by a type II secretion system which seems to be nonfunctional in nonpathogenic strains.


Asunto(s)
Antígenos Bacterianos/genética , Infecciones por Escherichia coli/prevención & control , Vacunas contra Escherichia coli/genética , Escherichia coli/genética , Genoma Bacteriano/genética , Meningitis por Escherichia coli/microbiología , Animales , Secuencia de Bases , Biología Computacional , Escherichia coli/inmunología , Finlandia , Islas Genómicas/genética , Humanos , Ratones , Datos de Secuencia Molecular , Vías Secretoras/genética , Análisis de Secuencia de ADN
14.
Mol Cell Proteomics ; 7(3): 473-85, 2008 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-17982123

RESUMEN

Extraintestinal pathogenic Escherichia coli are the cause of a diverse spectrum of invasive infections in humans and animals, leading to urinary tract infections, meningitis, or septicemia. In this study, we focused our attention on the identification of the outer membrane proteins of the pathogen in consideration of their important biological role and of their use as potential targets for prophylactic and therapeutic interventions. To this aim, we generated a DeltatolR mutant of the pathogenic IHE3034 strain that spontaneously released a large quantity of outer membrane vesicles in the culture supernatant. The vesicles were analyzed by two-dimensional electrophoresis coupled to mass spectrometry. The analysis led to the identification of 100 proteins, most of which are localized to the outer membrane and periplasmic compartments. Interestingly based on the genome sequences available in the current public database, seven of the identified proteins appear to be specific for pathogenic E. coli and enteric bacteria and therefore are potential targets for vaccine and drug development. Finally we demonstrated that the cytolethal distending toxin, a toxin exclusively produced by pathogenic bacteria, is released in association with the vesicles, supporting the recently proposed role of bacterial vesicles in toxin delivery to host cells. Overall, our data demonstrated that outer membrane vesicles represent an ideal tool to study Gram-negative periplasm and outer membrane compartments and to shed light on new mechanisms of bacterial pathogenesis.


Asunto(s)
Membrana Celular/química , Proteínas de Escherichia coli/genética , Escherichia coli/citología , Escherichia coli/genética , Proteínas de la Membrana/genética , Mutación/genética , Proteómica/métodos , Proteínas de la Membrana Bacteriana Externa/química , Proteínas de la Membrana Bacteriana Externa/metabolismo , Toxinas Bacterianas/química , Toxinas Bacterianas/metabolismo , Membrana Celular/ultraestructura , Escherichia coli/química , Escherichia coli/ultraestructura , Genoma Bacteriano , Péptidos , Proteínas Periplasmáticas/química , Proteínas Periplasmáticas/metabolismo , Unión Proteica , Subunidades de Proteína , Programas Informáticos
15.
Mol Microbiol ; 64(5): 1391-403, 2007 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-17542928

RESUMEN

Outer membrane protein As (OmpAs) are highly conserved proteins within the Enterobacteriaceae family. OmpA contributes to the maintenance of structural membrane integrity and invasion into mammalian cells. In Escherichia coli K1 OmpA also contributes to serum resistance and is involved in the virulence of the bacterium. Here we describe the identification of an OmpA-like protein in Neisseria gonorrhoeae (Ng-OmpA). We show that the gonococcal OmpA-like protein, similarly to E. coli OmpA, plays a significant role in the adhesion and invasion into human cervical carcinoma and endometrial cells and is required for entry into macrophages and intracellular survival. Furthermore, the isogenic knockout ompA mutant demonstrates reduced recovery in a mouse model of infection when compared with the wild-type strain, suggesting that Ng-OmpA plays an important role in the in vivo colonization. All together, these data suggest that the newly identified surface exposed protein Ng-OmpA represents a novel virulence factor of gonococcus.


Asunto(s)
Proteínas de la Membrana Bacteriana Externa/genética , Proteínas de la Membrana Bacteriana Externa/metabolismo , Proteínas Bacterianas/metabolismo , Células Epiteliales/metabolismo , Neisseria gonorrhoeae/química , Secuencias de Aminoácidos , Secuencia de Aminoácidos , Animales , Adhesión Bacteriana/fisiología , Proteínas de la Membrana Bacteriana Externa/química , Proteínas de la Membrana Bacteriana Externa/fisiología , Proteínas Bacterianas/genética , Línea Celular , Línea Celular Tumoral , Secuencia de Consenso , Secuencia Conservada , Células Epiteliales/microbiología , Humanos , Macrófagos Peritoneales/microbiología , Macrófagos Peritoneales/fisiología , Ratones , Modelos Genéticos , Datos de Secuencia Molecular , Mutación , Fagocitosis , Unión Proteica , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Homología de Secuencia de Aminoácido
16.
Mol Microbiol ; 58(3): 669-81, 2005 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-16238618

RESUMEN

Macrophage infectivity potentiators (MIPs) are a family of surface-exposed virulence factors of intracellular microorganisms such as Legionella, Chlamydia and Trypanosoma. These proteins display peptidyl-prolyl cis/trans isomerase (PPIase) activity that is inhibited by immunosuppressants FK506 and rapamycin. Here we describe the identification and characterization in Neisseria gonorrhoeae of Ng-MIP, a surface-exposed lipoprotein with high homology to MIPs. The protein is an homodimer with rapamycin-inhibited PPIase activity confirming that it is a functional member of the MIP family. A knock-out strain, generated by deletion of the mip gene in N. gonorrhoeae F62 strain, was evaluated for its role in infection of mouse and human macrophages. We show that Ng-MIP promotes the intracellular survival of N. gonorrhoeae in macrophages, highlighting a possible role of this protein in promoting the persistence of gonococcal infection.


Asunto(s)
Proteínas Bacterianas/metabolismo , Inmunofilinas/metabolismo , Lipoproteínas/metabolismo , Macrófagos/microbiología , Proteínas de la Membrana/metabolismo , Neisseria gonorrhoeae/enzimología , Isomerasa de Peptidilprolil/metabolismo , Secuencia de Aminoácidos , Animales , Antibacterianos/metabolismo , Proteínas Bacterianas/química , Proteínas Bacterianas/genética , Células Cultivadas , Dimerización , Eliminación de Gen , Gonorrea/microbiología , Humanos , Inmunofilinas/química , Inmunofilinas/genética , Lipoproteínas/química , Lipoproteínas/genética , Macrófagos/citología , Macrófagos/metabolismo , Proteínas de la Membrana/química , Proteínas de la Membrana/genética , Ratones , Datos de Secuencia Molecular , Neisseria gonorrhoeae/inmunología , Isomerasa de Peptidilprolil/química , Isomerasa de Peptidilprolil/genética , Unión Proteica , Estructura Cuaternaria de Proteína , Alineación de Secuencia , Sirolimus/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...