Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
FASEB J ; 33(11): 12324-12335, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31442078

RESUMEN

GNA2091 is one of the components of the 4-component meningococcal serogroup B vaccine (4CMenB) vaccine and is highly conserved in all meningococcal strains. However, its functional role has not been fully characterized. Here we show that nmb2091 is part of an operon and is cotranscribed with the nmb2089, nmb2090, and nmb2092 adjacent genes, and a similar but reduced operon arrangement is conserved in many other gram-negative bacteria. Deletion of the nmb2091 gene causes an aggregative phenotype with a mild defect in cell separation; differences in the outer membrane composition and phospholipid profile, in particular in the phosphoethanolamine levels; an increased level of outer membrane vesicles; and deregulation of the zinc-responsive genes such as znuD. Finally, the ∆2091 strain is attenuated with respect to the wild-type strain in competitive index experiments in the infant rat model of meningococcal infection. Altogether these data suggest that GNA2091 plays important roles in outer membrane architecture, biogenesis, homeostasis, and in meningococcal survival in vivo, and a model for its role is discussed. These findings highlight the importance of GNA2091 as a vaccine component.-Seib, K. L., Haag, A. F., Oriente, F., Fantappiè, L., Borghi, S., Semchenko, E. A., Schulz, B. L., Ferlicca, F., Taddei, A. R., Giuliani, M. M., Pizza, M., Delany, I. The meningococcal vaccine antigen GNA2091 is an analogue of YraP and plays key roles in outer membrane stability and virulence.


Asunto(s)
Antígenos Bacterianos/fisiología , Membrana Externa Bacteriana/química , Vacunas Meningococicas , Animales , Antígenos Bacterianos/genética , Membrana Externa Bacteriana/fisiología , Infecciones Meningocócicas/mortalidad , Vacunas Meningococicas/genética , Neisseria meningitidis Serogrupo B/genética , Neisseria meningitidis Serogrupo B/patogenicidad , Operón , Proteínas Periplasmáticas/fisiología , Ratas , Ratas Wistar , Regulón , Virulencia , Zinc/farmacología
2.
Proc Natl Acad Sci U S A ; 113(10): 2714-9, 2016 Mar 08.
Artículo en Inglés | MEDLINE | ID: mdl-26888286

RESUMEN

Factor H binding protein (fHbp) is a lipoprotein of Neisseria meningitidis important for the survival of the bacterium in human blood and a component of two recently licensed vaccines against serogroup B meningococcus (MenB). Based on 866 different amino acid sequences this protein is divided into three variants or two families. Quantification of the protein is done by immunoassays such as ELISA or FACS that are susceptible to the sequence variation and expression level of the protein. Here, selected reaction monitoring mass spectrometry was used for the absolute quantification of fHbp in a large panel of strains representative of the population diversity of MenB. The analysis revealed that the level of fHbp expression can vary at least 15-fold and that variant 1 strains express significantly more protein than variant 2 or variant 3 strains. The susceptibility to complement-mediated killing correlated with the amount of protein expressed by the different meningococcal strains and this could be predicted from the nucleotide sequence of the promoter region. Finally, the absolute quantification allowed the calculation of the number of fHbp molecules per cell and to propose a mechanistic model of the engagement of C1q, the recognition component of the complement cascade.


Asunto(s)
Antígenos Bacterianos/metabolismo , Proteínas Bacterianas/metabolismo , Neisseria meningitidis Serogrupo B/metabolismo , Secuencia de Aminoácidos , Antígenos Bacterianos/genética , Antígenos Bacterianos/inmunología , Proteínas Bacterianas/genética , Proteínas Bacterianas/inmunología , Ensayo de Inmunoadsorción Enzimática , Citometría de Flujo , Variación Genética , Humanos , Espectrometría de Masas/métodos , Meningitis Meningocócica/inmunología , Meningitis Meningocócica/microbiología , Vacunas Meningococicas/inmunología , Neisseria meningitidis Serogrupo B/clasificación , Neisseria meningitidis Serogrupo B/genética , Filogenia , Especificidad de la Especie
3.
J Bacteriol ; 198(4): 644-54, 2015 Dec 07.
Artículo en Inglés | MEDLINE | ID: mdl-26644430

RESUMEN

UNLABELLED: Neisseria meningitidis, an exclusively human pathogen and the leading cause of bacterial meningitis, must adapt to different host niches during human infection. N. meningitidis can utilize a restricted range of carbon sources, including lactate, glucose, and pyruvate, whose concentrations vary in host niches. Microarray analysis of N. meningitidis grown in a chemically defined medium in the presence or absence of glucose allowed us to identify genes regulated by carbon source availability. Most such genes are implicated in energy metabolism and transport, and some are implicated in virulence. In particular, genes involved in glucose catabolism were upregulated, whereas genes involved in the tricarboxylic acid cycle were downregulated. Several genes encoding surface-exposed proteins, including the MafA adhesins and Neisseria surface protein A, were upregulated in the presence of glucose. Our microarray analysis led to the identification of a glucose-responsive hexR-like transcriptional regulator that controls genes of the central carbon metabolism of N. meningitidis in response to glucose. We characterized the HexR regulon and showed that the hexR gene is accountable for some of the glucose-responsive regulation; in vitro assays with the purified protein showed that HexR binds to the promoters of the central metabolic operons of the bacterium. Based on DNA sequence alignment of the target sites, we propose a 17-bp pseudopalindromic consensus HexR binding motif. Furthermore, N. meningitidis strains lacking hexR expression were deficient in establishing successful bacteremia in an infant rat model of infection, indicating the importance of this regulator for the survival of this pathogen in vivo. IMPORTANCE: Neisseria meningitidis grows on a limited range of nutrients during infection. We analyzed the gene expression of N. meningitidis in response to glucose, the main energy source available in human blood, and we found that glucose regulates many genes implicated in energy metabolism and nutrient transport, as well as some implicated in virulence. We identified and characterized a transcriptional regulator (HexR) that controls metabolic genes of N. meningitidis in response to glucose. We generated a mutant lacking HexR and found that the mutant was impaired in causing systemic infection in animal models. Since N. meningitidis lacks known bacterial regulators of energy metabolism, our findings suggest that HexR plays a major role in its biology by regulating metabolism in response to environmental signals.


Asunto(s)
Proteínas Bacterianas/metabolismo , Carbono/metabolismo , Regulación Bacteriana de la Expresión Génica , Glucosa/metabolismo , Meningitis Meningocócica/microbiología , Neisseria meningitidis/genética , Neisseria meningitidis/metabolismo , Animales , Proteínas Bacterianas/genética , Secuencia de Bases , Metabolismo Energético , Humanos , Datos de Secuencia Molecular , Operón , Regiones Promotoras Genéticas , Ratas , Ratas Wistar , Regulón
4.
Proc Natl Acad Sci U S A ; 108(27): 11169-74, 2011 Jul 05.
Artículo en Inglés | MEDLINE | ID: mdl-21690334

RESUMEN

Oil-in-water emulsions have been successfully used to increase the efficacy, immunogenicity, and cross-protection of human vaccines; however, their mechanism of action is still largely unknown. Nlrp3 inflammasome has been previously associated to the activity of alum, another adjuvant broadly used in human vaccines, and MyD88 adaptor protein is required for the adjuvanticity of most Toll-like receptor agonists. We compared the contribution of Nlrp3 and MyD88 to the adjuvanticity of alum, the oil-in-water emulsion MF59, and complete Freund's adjuvant in mice using a three-component vaccine against serogroup B Neisseria meningitidis (rMenB). Although the basal antibody responses to the nonadjuvanted rMenB vaccine were largely dependent on Nlrp3, the high-level antibody responses induced by alum, MF59, or complete Freund's adjuvant did not require Nlrp3. Surprisingly, we found that MF59 requires MyD88 to enhance bactericidal antibody responses to the rMenB vaccine. Because MF59 did not activate any of the Toll-like receptors in vitro, we propose that MF59 requires MyD88 for a Toll-like receptor-independent signaling pathway.


Asunto(s)
Adyuvantes Inmunológicos/farmacología , Proteínas Portadoras/metabolismo , Factor 88 de Diferenciación Mieloide/metabolismo , Polisorbatos/farmacología , Escualeno/farmacología , Adyuvantes Inmunológicos/administración & dosificación , Compuestos de Alumbre/administración & dosificación , Animales , Anticuerpos Antibacterianos/biosíntesis , Vacunas Bacterianas/administración & dosificación , Proteínas Portadoras/antagonistas & inhibidores , Proteínas Portadoras/genética , Emulsiones , Femenino , Adyuvante de Freund/administración & dosificación , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Factor 88 de Diferenciación Mieloide/deficiencia , Factor 88 de Diferenciación Mieloide/genética , Proteína con Dominio Pirina 3 de la Familia NLR , Neisseria meningitidis Serogrupo B/inmunología , Polisorbatos/administración & dosificación , Transducción de Señal , Escualeno/administración & dosificación , Receptores Toll-Like/metabolismo , Vacunas Sintéticas/administración & dosificación
5.
Proc Natl Acad Sci U S A ; 107(8): 3770-5, 2010 Feb 23.
Artículo en Inglés | MEDLINE | ID: mdl-20133713

RESUMEN

GNA2132 is a Neisseria meningitidis antigen of unknown function, discovered by reverse vaccinology, which has been shown to induce bactericidal antibodies in animal models. Here we show that this antigen induces protective immunity in humans and it is recognized by sera of patients after meningococcal disease. The protein binds heparin in vitro through an Arg-rich region and this property correlates with increased survival of the unencapsulated bacterium in human serum. Furthermore, two proteases, the meningococcal NalP and human lactoferrin, cleave the protein upstream and downstream from the Arg-rich region, respectively. We conclude that GNA2132 is an important protective antigen of N. meningitidis and we propose to rename it, Neisserial Heparin Binding Antigen (NHBA).


Asunto(s)
Antígenos Bacterianos/inmunología , Péptidos Catiónicos Antimicrobianos/inmunología , Proteínas Sanguíneas/inmunología , Proteínas Portadoras/inmunología , Vacunas Meningococicas/inmunología , Neisseria meningitidis/inmunología , Factores de Virulencia/inmunología , Secuencia de Aminoácidos , Anticuerpos Antibacterianos/sangre , Antígenos Bacterianos/química , Antígenos Bacterianos/genética , Péptidos Catiónicos Antimicrobianos/química , Péptidos Catiónicos Antimicrobianos/genética , Proteínas Sanguíneas/química , Proteínas Sanguíneas/genética , Proteínas Portadoras/química , Proteínas Portadoras/genética , Humanos , Lactoferrina/química , Infecciones Meningocócicas/inmunología , Infecciones Meningocócicas/prevención & control , Vacunas Meningococicas/química , Vacunas Meningococicas/genética , Neisseria meningitidis/patogenicidad , Factores de Virulencia/química , Factores de Virulencia/genética
6.
NPJ Vaccines ; 8(1): 54, 2023 Apr 12.
Artículo en Inglés | MEDLINE | ID: mdl-37045859

RESUMEN

The ability of Neisseria meningitidis Outer Membrane Vesicles (OMV) to induce protective responses in humans is well established and mainly attributed to Porin A (PorA). However, the contribution of additional protein antigens to protection remains to be elucidated. In this study we dissected the immunogenicity of antigens originating from the OMV component of the 4CMenB vaccine in mice and humans. We collected functional data on a panel of strains for which bactericidal responses to 4CMenB in infants was attributable to the OMV component and evaluated the role of 30 OMV-specific protein antigens in cross-coverage. By using tailor-made protein microarrays, the immunosignature of OMV antigens was determined. Three of these proteins, OpcA, NspA, and PorB, triggered mouse antibodies that were bactericidal against several N. meningitidis strains. Finally, by genetic deletion and/or serum depletion studies, we demonstrated the ability of OpcA and PorB to induce functional immune responses in infant sera after vaccination. In conclusion, while confirming the role of PorA in eliciting protective immunity, we identified two OMV antigens playing a key role in protection of infants vaccinated with the 4CMenB vaccine against different N. meningitidis serogroup B strains.

7.
Infect Immun ; 79(2): 970-81, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-21149595

RESUMEN

Neisseria meningitidis is a commensal of the human nasopharynx but is also a major cause of septicemia and meningitis. The meningococcal factor H binding protein (fHbp) binds human factor H (fH), enabling downregulation of complement activation on the bacterial surface. fHbp is a component of two serogroup B meningococcal vaccines currently in clinical development. Here we characterize 12 fHbp subvariants for their level of surface exposure and ability to bind fH, to mediate serum resistance, and to induce bactericidal antibodies. Flow cytometry and Western analysis revealed that all strains examined expressed fHbp on their surface to different extents and bound fH in an fHbp-dependent manner. However, differences in fH binding did not always correlate with the level of fHbp expression, indicating that this is not the only factor affecting the amount of fH bound. To overcome the issue of strain variability in fHbp expression, the MC58ΔfHbp strain was genetically engineered to express different subvariants from a constitutive heterologous promoter. These recombinant strains were characterized for fH binding, and the data confirmed that each subvariant binds different levels of fH. Surface plasmon resonance revealed differences in the stability of the fHbp-fH complexes that ranged over 2 orders of magnitude, indicating that differences in residues between and within variant groups can influence fH binding. Interestingly, the level of survival in human sera of recombinant MC58 strains expressing diverse subvariants did not correlate with the level of fH binding, suggesting that the interaction of fHbp with fH is not the only function of fHbp that influences serum resistance. Furthermore, cross-reactive bactericidal activity was seen within each variant group, although the degree of activity varied, suggesting that amino acid differences within each variant group influence the bactericidal antibody response.


Asunto(s)
Anticuerpos Antibacterianos/metabolismo , Proteínas Bacterianas/metabolismo , Factor H de Complemento/metabolismo , Neisseria meningitidis/genética , Neisseria meningitidis/metabolismo , Secuencia de Aminoácidos , Animales , Proteínas del Sistema Complemento , Femenino , Variación Genética , Humanos , Infecciones Meningocócicas/inmunología , Infecciones Meningocócicas/microbiología , Ratones , Datos de Secuencia Molecular , Filogenia , Unión Proteica , Conejos
8.
Infect Dis Ther ; 10(1): 307-316, 2021 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-33185849

RESUMEN

INTRODUCTION: Invasive meningococcal disease (IMD) is an important public health concern. In developed countries, most IMD is caused by meningococcal serogroup B (MenB) and two protein-based MenB vaccines are currently available: the four-component vaccine 4CMenB (Bexsero, GSK) and the bivalent vaccine MenB-FHbp (Trumenba, Pfizer). Genes encoding the 4CMenB vaccine antigens are also present in strains belonging to other meningococcal serogroups. METHODS: To evaluate the potential of 4CMenB vaccination to protect adolescents against non-MenB IMD, we tested the bactericidal activity of sera from immunized adolescents on 147 (127 European and 20 Brazilian) non-MenB IMD isolates, with a serum bactericidal antibody assay using human complement (hSBA). Serum pools were prepared using samples from randomly selected participants in various clinical trials, pre- and post-vaccination: 12 adolescents who received two doses of 4CMenB 2 months apart, and 10 adolescents who received a single dose of a MenACWY conjugate vaccine (as positive control). RESULTS: 4CMenB pre-immune sera killed 7.5% of the 147 non-MenB isolates at hSBA titers ≥ 1:4. In total, 91 (61.9%) tested isolates were killed by post-dose 2 pooled sera at hSBA titers ≥ 1:4, corresponding to 44/80 (55.0%) MenC, 26/35 (74.3%) MenW, and 21/32 (65.6%) MenY isolates killed. CONCLUSION: 4CMenB vaccination in adolescents induces bactericidal killing of non-MenB isolates, suggesting that mass vaccination could impact IMD due to serogroups other than MenB.

9.
Glycoconj J ; 27(7-9): 643-8, 2010 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-20922476

RESUMEN

We investigated the immune responses of rabbits that were immunised with lipopolysaccharide (LPS)-based glycoconjugates by measuring the reactivity of the derived sera to a panel of selected wild-type and mutant strains of Neisseria meningitidis. In all cases, high titers of antibodies capable of recognising LPS elaborating the identical structure as presented on the immunising glycoconjugate were obtained, and in most cases the derived sera also recognised heterologous strains including wild-type, but at lower titers. However, although serum bactericidal antibodies were consistently obtained against strains elaborating the same LPS structure as the immunising antigen, this functional response was not observed against wild-type strains. We identified several potentially competing neo-epitopes that had been introduced via our conjugation strategies, which might compete with the conserved inner core oligosaccharide target region, thus reducing the antibody titers to epitopes which could facilitate bactericidal killing. This study has therefore identified key factors that are crucial to control in order to increase the likelihood of obtaining bactericidal antibodies to wild-type meningococcal cells with LPS-derived glycoconjugates. Glycoconjugates utilised in this study, have been found to contain epitopes that do not contribute to the derivation of antibodies that may facilitate bactericidal killing of wild-type strains and must be avoided in future LPS-based glycoconjugate preparations.


Asunto(s)
Anticuerpos Antibacterianos/biosíntesis , Glicoconjugados/inmunología , Lipopolisacáridos/inmunología , Infecciones Meningocócicas/prevención & control , Vacunas Meningococicas/química , Neisseria meningitidis/inmunología , Animales , Sueros Inmunes/inmunología , Lipopolisacáridos/genética , Vacunas Meningococicas/biosíntesis , Neisseria meningitidis/genética , Conejos
10.
Vaccine ; 38(47): 7542-7550, 2020 11 03.
Artículo en Inglés | MEDLINE | ID: mdl-33036804

RESUMEN

BACKGROUND: The multicomponent meningococcal serogroup B vaccine (4CMenB) is currently indicated for active immunization against invasive meningococcal disease caused by Neisseria meningitidis serogroup B (MenB). However, genes encoding the 4CMenB antigens are also variably present and expressed in strains belonging to other meningococcal serogroups. In this study, we evaluated the ability of antibodies raised by 4CMenB immunisation to induce complement-mediated bactericidal killing of non-MenB strains. METHODS: A total of 227 invasive non-MenB disease isolates were collected between 1 July 2007 and 30 June 2008 from England and Wales, France, and Germany; 41 isolates were collected during 2012 from Brazil. The isolates were subjected to genotypic analyses. A subset of 147 isolates (MenC, MenW and MenY) representative of the meningococcal genetic diversity of the total sample were tested in the human complement serum bactericidal antibody assay (hSBA) using sera from infants immunised with 4CMenB. RESULTS: Serogroup and clonal complex repertoires of non-MenB isolates were different for each country. For the European panel, MenC, MenW and MenY isolates belonged mainly to ST-11, ST-22 and ST-23 complexes, respectively. For the Brazilian panel, most MenC and MenW isolates belonged to the ST-103 and ST-11 complexes, respectively, and most MenY isolates were not assigned to clonal complexes. Of the 147 non-MenB isolates, 109 were killed in hSBA, resulting in an overall coverage of 74%. CONCLUSION: This is the first study in which 147 non-MenB serogroup isolates have been analysed in hSBA to evaluate the potential of a MenB vaccine to cover strains belonging to other serogroups. These data demonstrate that antibodies raised by 4CMenB are able to induce bactericidal killing of 109 non-MenB isolates, representative of non-MenB genetic and geographic diversity. These findings support previous evidence that 4CMenB immunisation can provide cross-protection against non-MenB strains in infants, which represents an added benefit of 4CMenB vaccination.


Asunto(s)
Infecciones Meningocócicas , Vacunas Meningococicas , Neisseria meningitidis Serogrupo B , Antígenos Bacterianos/genética , Brasil , Inglaterra , Francia , Alemania , Humanos , Lactante , Infecciones Meningocócicas/prevención & control , Neisseria meningitidis Serogrupo B/genética , Serogrupo , Vacunación , Gales
11.
Infect Immun ; 77(5): 1842-53, 2009 May.
Artículo en Inglés | MEDLINE | ID: mdl-19223479

RESUMEN

The well-conserved protein Hfq has emerged as the key modulator of riboregulation in bacteria. This protein is thought to function as an RNA chaperone and to facilitate base pairing between small regulatory RNA (sRNA) and mRNA targets, and many sRNAs are dependent on the Hfq protein for their regulatory functions. To address the possible role of Hfq in riboregulated circuits in Neisseria meningitidis, we generated an Hfq mutant of the MC58 strain, and the knockout mutant has pleiotropic phenotypes; it has a general growth phenotype in vitro in culture media, and it is sensitive to a wide range of stresses, including those that it may encounter in the host. Furthermore, the expression profile of a vast number of proteins is clearly altered in the mutant, and we have identified 27 proteins by proteomics. All of the phenotypes tested to date are also restored by complementation of Hfq expression in the mutant strain. Importantly, in ex vivo and in vivo models of infection the Hfq mutant is attenuated. These data indicate that Hfq plays a key role in stress response and virulence, and we propose a major role for Hfq in regulation of gene expression. Moreover, this study suggests that in meningococcus there is a large Hfq-mediated sRNA network which so far is largely unexplored.


Asunto(s)
Regulación Bacteriana de la Expresión Génica , Proteína de Factor 1 del Huésped/fisiología , Neisseria meningitidis/fisiología , Estrés Fisiológico , Factores de Virulencia/biosíntesis , Animales , Proteínas Bacterianas/biosíntesis , Proteínas Bacterianas/aislamiento & purificación , Sangre/microbiología , Recuento de Colonia Microbiana , Electroforesis en Gel Bidimensional , Perfilación de la Expresión Génica , Técnicas de Inactivación de Genes , Prueba de Complementación Genética , Proteína de Factor 1 del Huésped/genética , Humanos , Espectrometría de Masas , Infecciones Meningocócicas/microbiología , Viabilidad Microbiana , Neisseria meningitidis/genética , Neisseria meningitidis/crecimiento & desarrollo , Ratas , Virulencia , Factores de Virulencia/aislamiento & purificación
12.
Trends Biotechnol ; 23(2): 84-91, 2005 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-15661345

RESUMEN

After 200 years of practice, vaccinology has gained new perspectives for preventing infectious diseases. Sequencing of complete bacterial genomes led to the development of new large-scale technologies, such as bioinformatics, proteomics and DNA microarrays. By examining genetic content, as well as transcription and expression profiles, a more detailed understanding of bacterial pathogenesis can be reached. Moreover, the whole-genome perspective is expected to provide an instrumental contribution to vaccine development, particularly to target those pathogens for which the traditional approaches have failed so far. In this review, we describe how genomic approaches can be used to identify novel vaccine candidates or create safer live-attenuated vaccines.


Asunto(s)
Vacunas Bacterianas , Genoma Bacteriano , Genómica , Vacunas Bacterianas/genética , Biología Computacional , Análisis de Secuencia por Matrices de Oligonucleótidos , Proteómica
13.
J Drug Target ; 13(8-9): 489-98, 2005.
Artículo en Inglés | MEDLINE | ID: mdl-16332574

RESUMEN

Chitosan and its derivative N-trimethyl chitosan chloride (TMC), given as microparticles or powder suspensions, and the non-toxic mucosal adjuvant LTK63, were evaluated for intranasal immunization with the group C meningococcal conjugated vaccine (CRM-MenC). Mice immunized intranasally with CRM-MenC formulated with chitosan or TMC and the LTK63 mutant, showed high titers of serum and mucosal antibodies specific for the MenC polysaccharide. Neither significant differences were observed between microparticle formulations and powder suspensions nor when LTK63 was pre-associated to the delivery system or not. The bactericidal activity measured in serum of mice immunized intranasally with the conjugated vaccine formulated with the delivery systems and the LT mutant was superior to the activity in serum of mice immunized sub-cutaneously. Importantly, intranasal but not parenteral immunization, induced bactericidal antibodies at the nasal level, when formulated with both delivery system and adjuvant.


Asunto(s)
Toxinas Bacterianas/inmunología , Quitosano/inmunología , Sistemas de Liberación de Medicamentos/métodos , Enterotoxinas/inmunología , Proteínas de Escherichia coli/inmunología , Vacunas Meningococicas/inmunología , Administración Intranasal , Animales , Toxinas Bacterianas/administración & dosificación , Toxinas Bacterianas/genética , Quitosano/administración & dosificación , Quitosano/química , Enterotoxinas/administración & dosificación , Enterotoxinas/genética , Proteínas de Escherichia coli/administración & dosificación , Proteínas de Escherichia coli/genética , Femenino , Inmunización , Ratones , Ratones Endogámicos BALB C , Mutación , Tamaño de la Partícula
14.
PLoS One ; 10(3): e0120807, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25786110

RESUMEN

Long pentraxin 3 (PTX3) is a non-redundant component of the humoral arm of innate immunity. The present study was designed to investigate the interaction of PTX3 with Neisseria meningitidis. PTX3 bound acapsular meningococcus, Neisseria-derived outer membrane vesicles (OMV) and 3 selected meningococcal antigens (GNA0667, GNA1030 and GNA2091). PTX3-recognized microbial moieties are conserved structures which fulfil essential microbial functions. Ptx3-deficient mice had a lower antibody response in vaccination protocols with OMV and co-administration of PTX3 increased the antibody response, particularly in Ptx3-deficient mice. Administration of PTX3 reduced the bacterial load in infant rats challenged with Neisseria meningitidis. These results suggest that PTX3 recognizes a set of conserved structures from Neisseria meningitidis and acts as an amplifier/endogenous adjuvant of responses to this bacterium.


Asunto(s)
Adyuvantes Inmunológicos/genética , Anticuerpos Antibacterianos/biosíntesis , Antígenos Bacterianos/inmunología , Proteína C-Reactiva/inmunología , Meningitis Meningocócica/prevención & control , Vacunas Meningococicas/inmunología , Neisseria meningitidis/inmunología , Componente Amiloide P Sérico/inmunología , Adyuvantes Inmunológicos/administración & dosificación , Adyuvantes Inmunológicos/deficiencia , Animales , Animales Recién Nacidos , Anticuerpos Antibacterianos/inmunología , Antígenos Bacterianos/genética , Carga Bacteriana/efectos de los fármacos , Proteína C-Reactiva/administración & dosificación , Proteína C-Reactiva/deficiencia , Proteína C-Reactiva/genética , Femenino , Expresión Génica , Inmunidad Humoral/efectos de los fármacos , Inmunidad Innata/efectos de los fármacos , Masculino , Meningitis Meningocócica/inmunología , Meningitis Meningocócica/virología , Vacunas Meningococicas/administración & dosificación , Vacunas Meningococicas/genética , Ratones , Ratones Noqueados , Neisseria meningitidis/efectos de los fármacos , Neisseria meningitidis/genética , Ovalbúmina/administración & dosificación , Ratas , Ratas Wistar , Componente Amiloide P Sérico/administración & dosificación , Componente Amiloide P Sérico/deficiencia , Componente Amiloide P Sérico/genética , Vacunación
15.
Lancet Infect Dis ; 13(5): 416-25, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23414709

RESUMEN

BACKGROUND: A novel multicomponent vaccine against meningococcal capsular group B (MenB) disease contains four major components: factor-H-binding protein, neisserial heparin binding antigen, neisserial adhesin A, and outer-membrane vesicles derived from the strain NZ98/254. Because the public health effect of the vaccine, 4CMenB (Novartis Vaccines and Diagnostics, Siena, Italy), is unclear, we assessed the predicted strain coverage in Europe. METHODS: We assessed invasive MenB strains isolated mainly in the most recent full epidemiological year in England and Wales, France, Germany, Italy, and Norway. Meningococcal antigen typing system (MATS) results were linked to multilocus sequence typing and antigen sequence data. To investigate whether generalisation of coverage applied to the rest of Europe, we also assessed isolates from the Czech Republic and Spain. FINDINGS: 1052 strains collected from July, 2007, to June, 2008, were assessed from England and Wales, France, Germany, Italy, and Norway. All MenB strains contained at least one gene encoding a major antigen in the vaccine. MATS predicted that 78% of all MenB strains would be killed by postvaccination sera (95% CI 63-90, range of point estimates 73-87% in individual country panels). Half of all strains and 64% of covered strains could be targeted by bactericidal antibodies against more than one vaccine antigen. Results for the 108 isolates from the Czech Republic and 300 from Spain were consistent with those for the other countries. INTERPRETATION: MATS analysis showed that a multicomponent vaccine could protect against a substantial proportion of invasive MenB strains isolated in Europe. Monitoring of antigen expression, however, will be needed in the future. FUNDING: Novartis Vaccines and Diagnostics.


Asunto(s)
Genes Bacterianos , Meningitis Meningocócica/prevención & control , Vacunas Meningococicas/uso terapéutico , Neisseria meningitidis Serogrupo B/aislamiento & purificación , Adhesinas Bacterianas/análisis , Antígenos Bacterianos/genética , Técnicas de Tipificación Bacteriana/métodos , ADN Bacteriano/análisis , Ensayo de Inmunoadsorción Enzimática , Europa (Continente)/epidemiología , Genotipo , Geografía , Humanos , Meningitis Meningocócica/epidemiología , Meningitis Meningocócica/microbiología , Tipificación de Secuencias Multilocus/métodos , Neisseria meningitidis Serogrupo B/clasificación , Neisseria meningitidis Serogrupo B/genética , Neisseria meningitidis Serogrupo B/patogenicidad , Vigilancia de la Población/métodos , Valor Predictivo de las Pruebas , Reproducibilidad de los Resultados
16.
Vaccine ; 30 Suppl 2: B87-97, 2012 May 30.
Artículo en Inglés | MEDLINE | ID: mdl-22607904

RESUMEN

Neisseria meningitidis is a major cause of endemic cases and epidemics of meningitis and devastating septicemia. Although effective vaccines exist for several serogroups of pathogenic N. meningitidis, conventional vaccinology approaches have failed to provide a universal solution for serogroup B (MenB) which consequently remains an important burden of disease worldwide. The advent of whole-genome sequencing changed the approach to vaccine development, enabling the identification of potential vaccine candidates starting directly with the genomic information, with a process named reverse vaccinology. The application of reverse vaccinology to MenB allowed the identification of new protein antigens able to induce bactericidal antibodies. Three highly immunogenic antigens (fHbp, NadA and NHBA) were combined with outer membrane vesicles and formulated for human use in a multicomponent vaccine, named 4CMenB. This is the first MenB vaccine based on recombinant proteins able to elicit a robust bactericidal immune response in adults, adolescents and infants against a broad range of serogroup B isolates. This review describes the successful story of the development of the 4CMenB vaccine, with particular emphasis on the functional, immunological and structural characterization of the protein antigens included in the vaccine.


Asunto(s)
Antígenos Bacterianos/genética , Antígenos Bacterianos/inmunología , Vacunas Meningococicas/inmunología , Neisseria meningitidis Serogrupo B/genética , Neisseria meningitidis Serogrupo B/inmunología , Adhesinas Bacterianas/genética , Adhesinas Bacterianas/inmunología , Adhesinas Bacterianas/metabolismo , Antígenos Bacterianos/metabolismo , Proteínas Bacterianas/genética , Proteínas Bacterianas/inmunología , Proteínas Bacterianas/metabolismo , Humanos , Vacunas Meningococicas/genética , Vacunas Sintéticas/genética , Vacunas Sintéticas/inmunología
17.
Vaccine ; 29(5): 1072-81, 2011 Jan 29.
Artículo en Inglés | MEDLINE | ID: mdl-21130753

RESUMEN

Factor H binding protein (fHbp), one of the main antigens of new vaccines against serogroup B meningococcus, varies in amino acid sequence and level of expression in different clinical isolates. To evaluate the contribution of amino acid sequence variability to vaccine coverage, we constructed a strain that is susceptible to bactericidal killing only by anti-fHbp antibodies and engineered it to express equal levels of 10 different fHbp sub-variants from a constitutive promoter. Testing of these isogenic strains showed that sera from mice or adult volunteers vaccinated with fHbp variant 1.1 were bactericidal against all sub-variants 1 sequences, however the titer against the most distant sequences were several times lower. Sera from vaccinated infants were more susceptible to amino acid variations and they had lower or no bactericidal activity against the distant sub-variants 1 sequences in comparison with sera from adults given the same vaccines. The low coverage provided by fHbp could be overcome using a multicomponent vaccine. We conclude that fHbp is a very important antigen that induces bactericidal antibodies in animals, adults and infants. However, given its high variability of sequence and expression level, it is unlikely that fHbp alone can provide good protection in infants against the distant amino acid sequence variants and therefore multicomponent vaccines inducing protective immunity also against other antigens are more likely to induce a broad protective immunity in all age groups.


Asunto(s)
Antígenos Bacterianos/genética , Antígenos Bacterianos/inmunología , Proteínas Bacterianas/genética , Proteínas Bacterianas/inmunología , Actividad Bactericida de la Sangre/inmunología , Sueros Inmunes/inmunología , Vacunas Meningococicas/inmunología , Neisseria meningitidis/inmunología , Polimorfismo Genético , Adulto , Animales , Femenino , Humanos , Lactante , Ratones , Viabilidad Microbiana , Neisseria meningitidis/genética
18.
Vaccine ; 28(31): 5023-30, 2010 Jul 12.
Artículo en Inglés | MEDLINE | ID: mdl-20493284

RESUMEN

Serum bactericidal activity using human complement is the basis for established correlates of protection against invasive meningococcal disease. During the development of multicomponent protein-based vaccines against meningococcus B, it is necessary to measure antigen-specific bactericidal responses. This is not straightforward because each strain may be killed by antibodies to multiple antigens. We characterized a large panel of strains and, using a competitive inhibition SBA, we identified four strains that are each specifically killed by bactericidal antibodies to one of the major vaccine components. These strains provide a straightforward approach to demonstrate protective responses to each component of the vaccine and demonstrate that each of the antigens in the vaccine is sufficient to provide a potentially protective level of bactericidal activity.


Asunto(s)
Anticuerpos Antibacterianos/inmunología , Infecciones Meningocócicas/inmunología , Vacunas Meningococicas/inmunología , Neisseria meningitidis Serogrupo B/inmunología , Animales , Actividad Bactericida de la Sangre , Proteínas del Sistema Complemento/inmunología , Femenino , Humanos , Infecciones Meningocócicas/prevención & control , Ratones , Neisseria meningitidis Serogrupo B/clasificación , Vacunas Sintéticas/inmunología
19.
Vaccine ; 28(12): 2416-27, 2010 Mar 11.
Artículo en Inglés | MEDLINE | ID: mdl-20067752

RESUMEN

A novel vaccine against serogroup B meningococcal disease - containing a combination of protein antigens identified by reverse vaccinology: fHBP fused to GNA2091, GNA2132 fused to GNA1030, and NadA - is currently in Phase III clinical trials. In order to determine the role of these antigens in the growth, survival and fitness of the meningococcus, we generated a mutant lacking the expression of all five protein antigens (5KO), a mutant lacking the three main antigens (fHBP, GNA2132 and NadA; 3KO), as well as strains lacking the single antigens. Our results show that abrogation of expression of these antigens in Neisseria meningitidis results in reduced growth in vitro, increased sensitivity of the bacterium to stresses it may encounter in the host, as well as reduced fitness in ex vivo models of infection and in an in vivo infant rat competitive index assay. These results support a multivalent vaccine approach, which was undertaken to strengthen the protective activity of the vaccine antigens, increase the breadth of MenB strains targeted by the vaccine, and limit the potential for selection of vaccine escape mutants.


Asunto(s)
Antígenos Bacterianos/fisiología , Viabilidad Microbiana , Neisseria meningitidis Serogrupo B/patogenicidad , Animales , Antígenos Bacterianos/genética , Biomasa , Actividad Bactericida de la Sangre , Ensayos Clínicos como Asunto , Recuento de Colonia Microbiana , Eliminación de Gen , Humanos , Vacunas Meningococicas/inmunología , Mutagénesis Insercional , Neisseria meningitidis Serogrupo B/genética , Nefelometría y Turbidimetría , Ratas , Ratas Wistar , Estrés Fisiológico , Virulencia
20.
J Mol Biol ; 386(1): 97-108, 2009 Feb 13.
Artículo en Inglés | MEDLINE | ID: mdl-19100746

RESUMEN

The factor H binding protein (fHbp) is a 27-kDa membrane-anchored lipoprotein of Neisseria meningitidis that allows the survival of the bacterium in human plasma; it is also a major component of a universal vaccine against meningococcus B. In this study, we used nuclear magnetic resonance spectroscopy, mutagenesis, and in silico modeling to map the epitope recognized by MAb502, a bactericidal monoclonal antibody elicited by fHbp. The data show that the antibody recognizes a conformational epitope within a well-defined area of the immunodominant C-terminal domain of the protein that is formed by two loops connecting different beta-strands of a beta-barrel and a short alpha-helix brought in spatial proximity by the protein folding. The identification of the protective epitopes of fHbp is an important factor for understanding the mechanism(s) of an effective immune response and provides valuable guidelines for designing variants of the protein able to induce broadly protective immunity.


Asunto(s)
Anticuerpos Antibacterianos/química , Anticuerpos Monoclonales/química , Antígenos Bacterianos/química , Antígenos Bacterianos/inmunología , Proteínas Bacterianas/química , Proteínas Bacterianas/inmunología , Secuencia de Aminoácidos , Anticuerpos Antibacterianos/inmunología , Anticuerpos Monoclonales/inmunología , Antígenos Bacterianos/metabolismo , Mapeo Epitopo , Infecciones Meningocócicas/inmunología , Modelos Moleculares , Datos de Secuencia Molecular , Neisseria meningitidis/inmunología , Resonancia Magnética Nuclear Biomolecular , Conformación Proteica , Alineación de Secuencia
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA