Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
1.
Mol Cell Proteomics ; 17(2): 205-215, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-29203497

RESUMEN

Despite high vaccination coverage world-wide, whooping cough, a highly contagious disease caused by Bordetella pertussis, is recently increasing in occurrence suggesting that novel vaccine formulations targeted at the prevention of colonization and transmission should be investigated. To identify new candidates for inclusion in the acellular formulation, we used spontaneously released outer membrane vesicles (OMV)1 as a potential source of key adhesins. The enrichment of Bvg+ OMV with adhesins and the ability of anti-OMV serum to inhibit the adhesion of B. pertussis to lung epithelial cells in vitro were demonstrated. We employed a proteomic approach to identify the differentially expressed proteins in OMV purified from bacteria in the Bvg+ and Bvg- virulence phases, thus comparing the outer membrane protein pattern of this pathogen in its virulent or avirulent state. Six of the most abundant outer membrane proteins were selected as candidates to be evaluated for their adhesive properties and vaccine potential. We generated E. coli strains singularly expressing the selected proteins and assessed their ability to adhere to lung epithelial cells in vitro Four out of the selected proteins conferred adhesive ability to E. coli Three of the candidates were specifically detected by anti-OMV mouse serum suggesting that these proteins are immunogenic antigens able to elicit an antibody response when displayed on the OMV. Anti-OMV serum was able to inhibit only BrkA-expressing E. coli adhesion to lung epithelial cells. Finally, stand-alone immunization of mice with recombinant BrkA resulted in significant protection against infection of the lower respiratory tract after challenge with B. pertussis Taken together, these data support the inclusion of BrkA and possibly further adhesins to the current acellular pertussis vaccines to improve the impact of vaccination on the bacterial clearance.


Asunto(s)
Antígenos Bacterianos/inmunología , Proteínas de la Membrana Bacteriana Externa/inmunología , Bordetella pertussis/patogenicidad , Membrana Celular/inmunología , Células Epiteliales/fisiología , Interacciones Huésped-Patógeno , Células A549 , Animales , Vacunas Bacterianas , Adhesión Celular , Células Epiteliales/microbiología , Femenino , Humanos , Pulmón/citología , Ratones Endogámicos BALB C , Proteómica , Tos Ferina/prevención & control
2.
Cell Microbiol ; 17(9): 1365-75, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-25801707

RESUMEN

Translocation of the nasopharyngeal barrier by Neisseria meningitidis occurs via an intracellular microtubule-dependent pathway and represents a crucial step in its pathogenesis. Despite this fact, the interaction of invasive meningococci with host subcellular compartments and the resulting impact on their organization and function have not been investigated. The influence of serogroup B strain MC58 on host cell polarity and intracellular trafficking system was assessed by confocal microscopy visualization of different plasma membrane-associated components (such as E-cadherin, ZO-1 and transferrin receptor) and evaluation of the transferrin uptake and recycling in infected Calu-3 monolayers. Additionally, the association of N. meningitidis with different endosomal compartments was evaluated through the concomitant staining of bacteria and markers specific for Rab11, Rab22a, Rab25 and Rab3 followed by confocal microscopy imaging. Subversion of the host cell architecture and intracellular trafficking system, denoted by mis-targeting of cell plasma membrane components and perturbations of transferrin transport, was shown to occur in response to N. meningitidis infection. Notably, the appearance of all of these events seems to positively correlate with the efficiency of N. meningitidis to cross the epithelial barrier. Our data reveal for the first time that N. meningitidis is able to modulate the host cell architecture and function, which might serve as a strategy of this pathogen for overcoming the nasopharyngeal barrier without affecting the monolayer integrity.


Asunto(s)
Polaridad Celular , Células Epiteliales/microbiología , Células Epiteliales/fisiología , Interacciones Huésped-Patógeno , Neisseria meningitidis Serogrupo B/fisiología , Transcitosis , Línea Celular , Humanos , Microscopía Confocal
3.
BMC Microbiol ; 15: 87, 2015 Apr 18.
Artículo en Inglés | MEDLINE | ID: mdl-25927946

RESUMEN

BACKGROUND: Non-typeable Haemophilus influenzae (NTHi) is a Gram negative microorganism residing in the human nasopharyngeal mucosa and occasionally causing infections of both middle ear and lower respiratory airways. A broadly protective vaccine against NTHi has been a long-unmet medical need, as the high genetic variability of this bacterium has posed great challenges. RESULTS: In this study, we developed a robust serum bactericidal assay (SBA) to optimize the selection of protective antigens against NTHi. SBA takes advantage of the complement-mediated lysis of bacterial cells and is a key in vitro method for measuring the functional activity of antibodies. As a proof of concept, we assessed the bactericidal activity of antibodies directed against antigens known to elicit a protective response, including protein D used as carrier protein in the Synflorix pneumococcal polysaccharide conjugate vaccine. Prior to SBA screening, the accessibility of antigens to antibodies and the capacity of the latter to induce C3 complement deposition was verified by flow cytometry. Using baby rabbit serum as a source of complement, the proposed assay not only confirmed the bactericidal activity of the antibodies against the selected vaccine candidates, but also showed a significant reproducibility. CONCLUSIONS: Considering the rapidity and cost-effectiveness of this novel SBA protocol, we conclude that it is likely to become an important tool to prove the capability of antibodies directed against recombinant antigens to induce NTHi in vitro killing and to both select new protective vaccine candidates, and predict vaccine efficacy.


Asunto(s)
Anticuerpos Antibacterianos/inmunología , Antígenos Bacterianos/inmunología , Actividad Bactericida de la Sangre , Haemophilus influenzae/inmunología , Haemophilus influenzae/fisiología , Viabilidad Microbiana/efectos de los fármacos , Animales , Cobayas , Haemophilus influenzae/efectos de los fármacos , Inmunoensayo/métodos , Ratones , Conejos
4.
Cell Microbiol ; 16(6): 925-37, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24397470

RESUMEN

Neisseria meningitidis is a human pathogen that can cause fatal sepsis and meningitis once it reaches the blood stream and the nervous system. Here we demonstrate that a fragment, released upon proteolysis of the surface-exposed protein Neisserial Heparin Binding Antigen (NHBA), by the bacterial protease NalP, alters the endothelial permeability by inducing the internalization of the adherens junction protein VE-cadherin. We found that C2 rapidly accumulates in mitochondria where it induces the production of reactive oxygen species: the latter are required for the phosphorylation of the junctional protein and for its internalization that, in turn, is responsible for the endothelial leakage. Our data support the notion that the NHBA-derived fragment C2 might contribute to the extensive vascular leakage typically associated with meningococcal sepsis.


Asunto(s)
Uniones Adherentes/efectos de los fármacos , Antígenos Bacterianos/metabolismo , Células Endoteliales/efectos de los fármacos , Neisseria meningitidis/fisiología , Permeabilidad/efectos de los fármacos , Antígenos Bacterianos/genética , Línea Celular , Humanos , Factores de Virulencia/genética , Factores de Virulencia/metabolismo
5.
Cell Microbiol ; 14(3): 368-85, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22066472

RESUMEN

NadA (N eisseria meningitidisadhesin A), a meningococcal surface protein, mediates adhesion to and invasion of human cells, an activity in which host membrane proteins have been implicated. While investigating these host factors in human epithelial cells by affinity chromatography, we discovered an unanticipated interaction of NadA with heat shock protein (Hsp) 90, a molecular chaperone. The specific in vitro interaction of recombinant soluble NadA and Hsp90 was confirmed by co-immunoprecipitations, dot and far-Western blot. Intriguingly, ADP, but not ATP, was required for this association, and the Hsp90 inhibitor 17-AAG promoted complex formation. Hsp90 binding to an Escherichia coli strain used as carrier to express surface exposed NadA confirmed these results in live bacteria. We also examined RNA interference, plasmid-driven overexpression, addition of exogenous rHsp90 and 17-AAG inhibition in human epithelial cells to further elucidate the involvement of Hsp90 in NadA-mediated adhesion and invasion. Together, these data suggest an inverse correlation between the amount of host Hsp90 and the NadA adhesive/invasive phenotype. Confocal microscopy also demonstrated that meningococci interact with cellular Hsp90, a completely novel finding. Altogether our results show that variation of host Hsp90 expression or activity interferes with adhesive and invasive events driven by NadA.


Asunto(s)
Adhesinas Bacterianas/metabolismo , Adhesión Bacteriana , Proteínas HSP90 de Choque Térmico/metabolismo , Neisseria meningitidis/fisiología , Secuencia de Aminoácidos , Benzoquinonas/farmacología , Células Cultivadas , Escherichia coli/genética , Escherichia coli/fisiología , Proteínas HSP90 de Choque Térmico/antagonistas & inhibidores , Interacciones Huésped-Patógeno , Humanos , Lactamas Macrocíclicas/farmacología , Infecciones Meningocócicas/metabolismo , Infecciones Meningocócicas/microbiología , Datos de Secuencia Molecular , Unión Proteica , Proteínas Recombinantes/metabolismo
6.
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
7.
J Bacteriol ; 193(1): 107-15, 2011 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-20971901

RESUMEN

NadA is a trimeric autotransporter protein of Neisseria meningitidis belonging to the group of oligomeric coiled-coil adhesins. It is implicated in the colonization of the human upper respiratory tract by hypervirulent serogroup B N. meningitidis strains and is part of a multiantigen anti-serogroup B vaccine. Structure prediction indicates that NadA is made by a COOH-terminal membrane anchor (also necessary for autotranslocation to the bacterial surface), an intermediate elongated coiled-coil-rich stalk, and an NH(2)-terminal region involved in cell interaction. Electron microscopy analysis and structure prediction suggest that the apical region of NadA forms a compact and globular domain. Deletion studies proved that the NH(2)-terminal sequence (residues 24 to 87) is necessary for cell adhesion. In this study, to better define the NadA cell binding site, we exploited (i) a panel of NadA mutants lacking sequences along the coiled-coil stalk and (ii) several oligoclonal rabbit antibodies, and their relative Fab fragments, directed to linear epitopes distributed along the NadA ectodomain. We identified two critical regions for the NadA-cell receptor interaction with Chang cells: the NH(2) globular head domain and the NH(2) dimeric intrachain coiled-coil α-helices stemming from the stalk. This raises the importance of different modules within the predicted NadA structure. The identification of linear epitopes involved in receptor binding that are able to induce interfering antibodies reinforces the importance of NadA as a vaccine antigen.


Asunto(s)
Adhesinas Bacterianas/metabolismo , Mapeo Epitopo , Neisseria meningitidis/metabolismo , Estructura Secundaria de Proteína/fisiología , Adhesinas Bacterianas/genética , Animales , Anticuerpos Antibacterianos , Sitios de Unión , Línea Celular , Regulación Bacteriana de la Expresión Génica , Humanos , Microscopía Electrónica , Modelos Moleculares , Neisseria meningitidis/genética , Unión Proteica , Estructura Secundaria de Proteína/genética , Estructura Terciaria de Proteína , Conejos
8.
Infect Immun ; 79(11): 4308-21, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-21844231

RESUMEN

Neisseria meningitidis NhhA (Neisseria hia/hsf homologue A) is an oligomeric outer membrane protein belonging to the family of trimeric autotransporter adhesins. NhhA mediates the interaction of N. meningitidis with human epithelial cells and components of the extracellular matrix. The recombinant protein is able to induce bactericidal antibodies and hence has also been considered a potential vaccine candidate. In this study, we analyzed the production of NhhA in a large panel of N. meningitidis strains belonging to different serogroups and clonal complexes. We found that trimeric NhhA was produced at different levels by the various strains tested. In some strains belonging to the clonal complex ST41/44, the protein is detectable only as a monomer. Sequencing of the nhhA gene and generation of complementing strains in different genetic backgrounds have proved that a single mutation (Gly to Asp) in the translocator domain affected both trimerization and surface localization of NhhA. In vitro infection assays showed that this mutation impairs meningococcal NhhA-mediated adhesion, suggesting that strains carrying the mutation may rely on different strategies or molecules to mediate interaction with host cells. Finally, we demonstrated that N. meningitidis ST41/44 strains producing the mutated form did not induce killing mediated by NhhA-specific bactericidal antibodies. Our data help to elucidate the secretion mechanisms of trimeric autotransporters and to understand the contribution of NhhA in the evolutionary process of host-Neisseria interactions. Also, they might have important implications for the evaluation of NhhA as a vaccine candidate.


Asunto(s)
Adhesinas Bacterianas/metabolismo , Sustitución de Aminoácidos , Adhesión Bacteriana/fisiología , Neisseria meningitidis/metabolismo , Adhesinas Bacterianas/genética , Secuencia de Aminoácidos , Adhesión Bacteriana/genética , Línea Celular , Humanos , Modelos Moleculares , Datos de Secuencia Molecular , Mutación , Neisseria meningitidis/genética , Conformación Proteica , Estructura Terciaria de Proteína , Transporte de Proteínas
9.
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
10.
J Exp Med ; 195(11): 1445-54, 2002 Jun 03.
Artículo en Inglés | MEDLINE | ID: mdl-12045242

RESUMEN

Neisseria meningitidis is a human pathogen, which, in spite of antibiotic therapy, is still a major cause of mortality due to sepsis and meningitis. Here we describe NadA, a novel surface antigen of N. meningitidis that is present in 52 out of 53 strains of hypervirulent lineages electrophoretic types (ET) ET37, ET5, and cluster A4. The gene is absent in the hypervirulent lineage III, in N. gonorrhoeae and in the commensal species N. lactamica and N. cinerea. The guanine/cytosine content, lower than the chromosome, suggests acquisition by horizontal gene transfer and subsequent limited evolution to generate three well-conserved alleles. NadA has a predicted molecular structure strikingly similar to a novel class of adhesins (YadA and UspA2), forms high molecular weight oligomers, and binds to epithelial cells in vitro supporting the hypothesis that NadA is important for host cell interaction. NadA induces strong bactericidal antibodies and is protective in the infant rat model suggesting that this protein may represent a novel antigen for a vaccine able to control meningococcal disease caused by three hypervirulent lineages.


Asunto(s)
Antígenos de Superficie/genética , Antígenos de Superficie/inmunología , Vacunas Meningococicas/inmunología , Neisseria meningitidis/inmunología , Alelos , Secuencia de Aminoácidos , Animales , Afinidad de Anticuerpos , Especificidad de Anticuerpos , Antígenos de Superficie/química , Antígenos de Superficie/metabolismo , Composición de Base , Secuencia de Bases , Western Blotting , Secuencia Conservada/genética , Evolución Molecular , Citometría de Flujo , Transferencia de Gen Horizontal/genética , Humanos , Sueros Inmunes/inmunología , Meningitis Meningocócica/inmunología , Meningitis Meningocócica/microbiología , Meningitis Meningocócica/prevención & control , Ratones , Datos de Secuencia Molecular , Neisseria meningitidis/genética , Neisseria meningitidis/crecimiento & desarrollo , Neisseria meningitidis/patogenicidad , Ratas
11.
J Exp Med ; 197(6): 789-99, 2003 Mar 17.
Artículo en Inglés | MEDLINE | ID: mdl-12642606

RESUMEN

Sepsis and meningitis caused by serogroup B meningococcus are devastating diseases of infants and young adults, which cannot yet be prevented by vaccination. By genome mining, we discovered GNA1870, a new surface-exposed lipoprotein of Neisseria meningitidis that induces high levels of bactericidal antibodies. The antigen is expressed by all strains of N. meningitidis tested. Sequencing of the gene in 71 strains representative of the genetic and geographic diversity of the N. meningitidis population, showed that the protein can be divided into three variants. Conservation within each variant ranges between 91.6 to 100%, while between the variants the conservation can be as low as 62.8%. The level of expression varies between strains, which can be classified as high, intermediate, and low expressors. Antibodies against a recombinant form of the protein elicit complement-mediated killing of the strains that carry the same variant and induce passive protection in the infant rat model. Bactericidal titers are highest against those strains expressing high yields of the protein; however, even the very low expressors are efficiently killed. The novel antigen is a top candidate for the development of a new vaccine against meningococcus.


Asunto(s)
Antígenos Bacterianos/inmunología , Lipoproteínas/inmunología , Neisseria meningitidis/inmunología , Isoformas de Proteínas/inmunología , Vacunación , Adulto , Secuencia de Aminoácidos , Animales , Antígenos Bacterianos/clasificación , Antígenos Bacterianos/genética , Antígenos Bacterianos/metabolismo , Secuencia de Bases , Femenino , Genes Bacterianos , Humanos , Lactante , Lipoproteínas/genética , Lipoproteínas/metabolismo , Ratones , Datos de Secuencia Molecular , Neisseria meningitidis/metabolismo , Filogenia , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Proteínas Recombinantes/genética , Proteínas Recombinantes/inmunología , Proteínas Recombinantes/metabolismo , Alineación de Secuencia
12.
Cell Microbiol ; 11(7): 1044-63, 2009 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-19290916

RESUMEN

The Oca (Oligomeric coiled-coil adhesin) family is a subgroup of the bacterial trimeric autotransporter adhesins, which includes structurally related proteins, such as YadA of Yersinia enterocolitica and NadA of Neisseria meningitidis. In this study, we searched in silico for novel members of this family in bacterial genomes and identified HadA (Haemophilus adhesin A), a trimeric autotransporter expressed only by Haemophilus influenzae biogroup aegyptius causing Brazilian purpuric fever (BPF), a fulminant septicemic disease of children. By comparative genomics and sequence analysis we predicted that the hadA gene is harboured on a mobile genetic element unique to BPF isolates. Biological analysis of HadA in the native background was limited because this organism is not amenable to genetic manipulation. Alternatively, we demonstrated that expression of HadA confers to a non-invasive Escherichia coli strain the ability to adhere to human cells and to extracellular matrix proteins and to induce in vitro bacterial aggregation and microcolony formation. Intriguingly, HadA is predicted to lack the typical N-terminal head domain of Oca proteins generally associated with cellular receptor binding. We propose here a structural model of the HadA coiled-coil stalk and show that the N-terminal region is still responsible of the binding activity and a KGD motif plays a role. Interestingly, HadA promotes bacterial entry into mammalian cells. Our results show a cytoskeleton re-arrangement and an involvement of clathrin in the HadA-mediated internalization. These data give new insights on the structure-function relationship of oligomeric coiled-coil adhesins and suggest a potential role of this protein in the pathogenesis of BPF.


Asunto(s)
Adhesinas Bacterianas/fisiología , Adhesión Bacteriana , Proteínas Bacterianas/fisiología , Haemophilus influenzae/patogenicidad , Adhesinas Bacterianas/química , Adhesinas Bacterianas/genética , Proteínas Bacterianas/química , Proteínas Bacterianas/genética , Sitios de Unión , Línea Celular , Biología Computacional , ADN Bacteriano/química , ADN Bacteriano/genética , Genómica , Haemophilus influenzae/genética , Humanos , Modelos Moleculares , Datos de Secuencia Molecular , Filogenia , Estructura Cuaternaria de Proteína , Análisis de Secuencia de ADN , Homología de Secuencia
13.
Future Microbiol ; 12: 1247-1259, 2017 11.
Artículo en Inglés | MEDLINE | ID: mdl-28980823

RESUMEN

AIM: Bordetella pertussis has been shown to release outer membrane vesicles (OMV) both in vitro and in vivo but little is known about their biological role during the initial phases of B. pertussis infection of the airways. RESULTS: We have demonstrated that OMV are released by B. pertussis in a human ciliated-airway cell model and purified vesicles can interact with host cells. Binding and uptake are strictly Bvg-regulated and OMV-associated pertussis toxin contributes to host-cell intoxication. Furthermore, we have shown that OMV act as iron-delivery systems complementing the B. pertussis growth defect in iron-limiting conditions. CONCLUSION: We have proved that OMV play different roles in B. pertussis physiopathology and we opened new perspectives to be further investigated.


Asunto(s)
Proteínas de la Membrana Bacteriana Externa/metabolismo , Bordetella pertussis/patogenicidad , Membrana Celular/ultraestructura , Toxina del Pertussis/metabolismo , Tos Ferina/microbiología , Células A549 , Animales , Células CHO , Cricetulus , Interacciones Huésped-Patógeno , Humanos , Hierro/metabolismo
14.
PLoS One ; 11(10): e0162878, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27780200

RESUMEN

Neisserial Heparin Binding Antigen (NHBA) is a surface-exposed lipoprotein ubiquitously expressed by Neisseria meningitidis strains and an antigen of the Bexsero® vaccine. NHBA binds heparin through a conserved Arg-rich region that is the target of two proteases, the meningococcal NalP and human lactoferrin (hLf). In this work, in vitro studies showed that recombinant NHBA protein was able to bind epithelial cells and mutations of the Arg-rich tract abrogated this binding. All N-terminal and C-terminal fragments generated by NalP or hLf cleavage, regardless of the presence or absence of the Arg-rich region, did not bind to cells, indicating that a correct positioning of the Arg-rich region within the full length protein is crucial. Moreover, binding was abolished when cells were treated with heparinase III, suggesting that this interaction is mediated by heparan sulfate proteoglycans (HSPGs). N. meningitidis nhba knockout strains showed a significant reduction in adhesion to epithelial cells with respect to isogenic wild-type strains and adhesion of the wild-type strain was inhibited by anti-NHBA antibodies in a dose-dependent manner. Overall, the results demonstrate that NHBA contributes to meningococcal adhesion to epithelial cells through binding to HSPGs and suggest a possible role of anti-Bexsero® antibodies in the prevention of colonization.


Asunto(s)
Anticuerpos Antibacterianos/genética , Anticuerpos Antibacterianos/metabolismo , Adhesión Bacteriana , Proteínas de la Membrana Bacteriana Externa/metabolismo , Proteínas Portadoras/metabolismo , Células Epiteliales/microbiología , Infecciones Meningocócicas/tratamiento farmacológico , Neisseria meningitidis/fisiología , Anticuerpos Antibacterianos/química , Sitios de Unión , Línea Celular , Técnicas de Inactivación de Genes , Proteoglicanos de Heparán Sulfato/metabolismo , Heparina/metabolismo , Humanos , Lactoferrina/metabolismo , Proteínas de Transporte de Membrana/metabolismo , Infecciones Meningocócicas/microbiología , Infecciones Meningocócicas/prevención & control , Mutación , Neisseria meningitidis/inmunología , Proteínas Recombinantes/metabolismo , Serina Endopeptidasas/metabolismo
15.
mBio ; 6(2): e02575, 2015 Feb 24.
Artículo en Inglés | MEDLINE | ID: mdl-25714719

RESUMEN

UNLABELLED: LytM proteins belong to a family of bacterial metalloproteases. In Gram-negative bacteria, LytM factors are mainly reported to have a direct effect on cell division by influencing cleavage and remodeling of peptidoglycan. In this study, mining nontypeable Haemophilus influenzae (NTHI) genomes, three highly conserved open reading frames (ORFs) containing a LytM domain were identified, and the proteins encoded by the ORFs were named YebA, EnvC, and NlpD on the basis of their homology with the Escherichia coli proteins. Immunoblotting and confocal analysis showed that while NTHI NlpD is exposed on the bacterial surface, YebA and EnvC reside in the periplasm. NTHI ΔyebA and ΔnlpD deletion mutants revealed an aberrant division phenotype characterized by an altered cell architecture and extensive membrane blebbing. The morphology of the ΔenvC deletion mutant was identical to that of the wild-type strain, but it showed a drastic reduction of periplasmic proteins, including the chaperones HtrA, SurA, and Skp, and an accumulation of ß-barrel-containing outer membrane proteins comprising the autotransporters Hap, IgA serine protease, and HMW2A, as observed by proteomic analysis. These data suggest that EnvC may influence the bacterial surface protein repertoire by facilitating the passage of the periplasmic chaperones through the peptidoglycan layer to the close vicinity of the inner face of the outer membrane. This hypothesis was further corroborated by the fact that an NTHI envC defective strain had an impaired capacity to adhere to epithelial cells and to form biofilm. Notably, this strain also showed a reduced serum resistance. These results suggest that LytM factors are not only important components of cell division but they may also influence NTHI physiology and pathogenesis by affecting membrane composition. IMPORTANCE: Nontypeable Haemophilus influenzae (NTHI) is an opportunistic pathogen that colonizes the human nasopharynx and can cause serious infections in children (acute otitis media) and adults (chronic obstructive pulmonary disease). Several virulence factors are well studied, but the complete scenario of NTHI pathogenesis is still unclear. We identified and characterized three NTHI LytM factors homologous to the Escherichia coli LytM proteins. Although LytM factors are reported to play a crucial role in the cell division process, in NTHI they are also involved in other bacterial functions. In particular, YebA and NlpD are fundamental for membrane stability: indeed, their absence causes an increased release of outer membrane vesicles (OMVs). On the other hand, our data suggest that EnvC could directly or indirectly affect peptidoglycan permeability and consequently, bacterial periplasmic and outer membrane protein distribution. Interestingly, by modulating the surface composition of virulence determinants, EnvC also has an impact on NTHI pathogenesis.


Asunto(s)
División Celular , Membrana Celular/química , Haemophilus influenzae/enzimología , Haemophilus influenzae/fisiología , Metaloproteasas/metabolismo , Adhesión Bacteriana , Pared Celular/química , Células Cultivadas , Biología Computacional , Células Epiteliales/microbiología , Eliminación de Gen , Haemophilus influenzae/genética , Haemophilus influenzae/patogenicidad , Humanos , Sistemas de Lectura Abierta , Periplasma/química , Homología de Secuencia de Aminoácido , Virulencia , Factores de Virulencia
16.
Clin Vaccine Immunol ; 22(7): 769-77, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25947148

RESUMEN

Knowledge of the sequences and structures of proteins produced by microbial pathogens is continuously increasing. Besides offering the possibility of unraveling the mechanisms of pathogenesis at the molecular level, structural information provides new tools for vaccine development, such as the opportunity to improve viral and bacterial vaccine candidates by rational design. Structure-based rational design of antigens can optimize the epitope repertoire in terms of accessibility, stability, and variability. In the present study, we used epitope mapping information on the well-characterized antigen of Neisseria meningitidis factor H binding protein (fHbp) to engineer its gonococcal homologue, Ghfp. Meningococcal fHbp is typically classified in three distinct antigenic variants. We introduced epitopes of fHbp variant 1 onto the surface of Ghfp, which is naturally able to protect against meningococcal strains expressing fHbp of variants 2 and 3. Heterologous epitopes were successfully transplanted, as engineered Ghfp induced functional antibodies against all three fHbp variants. These results confirm that structural vaccinology represents a successful strategy for modulating immune responses, and it is a powerful tool for investigating the extension and localization of immunodominant epitopes.


Asunto(s)
Neisseria gonorrhoeae/genética , Neisseria gonorrhoeae/inmunología , Neisseria meningitidis/inmunología , Ingeniería de Proteínas , Factores de Virulencia/genética , Animales , Anticuerpos Antibacterianos/sangre , Antígenos Bacterianos/genética , Proteínas Bacterianas/genética , Vacunas Bacterianas/administración & dosificación , Vacunas Bacterianas/inmunología , Actividad Bactericida de la Sangre , Ratones , Neisseria meningitidis/genética , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/inmunología , Homología de Secuencia de Aminoácido , Vacunas Sintéticas/administración & dosificación , Vacunas Sintéticas/inmunología
17.
Res Microbiol ; 154(6): 443-50, 2003.
Artículo en Inglés | MEDLINE | ID: mdl-12892851

RESUMEN

Bordetella pertussis undergoes phenotypic changes modulated by the bvgAS locus, which regulates the expression of many genes related to virulence and immunogenicity. We previously reported the N-terminal sequence of a 90 kDa bvg-regulated outer membrane protein (OMP) of B. pertussis (SWISS-PROT accession No. p81549), a novel potential virulence factor that we named Vir90. The open reading frames (ORFs) which potentially code for Vir90 in B. pertussis, B. parapertussis and B. bronchiseptica were identified by computer analysis of the genomic sequences available for the three Bordetella species. Nucleotide sequence analysis of the vir90 upstream region revealed the presence of a putative promoter, a BvgA binding site and a putative Fur binding site. The B. pertussis Vir90 protein showed significant homology with ferrisiderophore receptors from Gram-negative bacteria. An antiserum raised against Vir90His recombinant protein recognized the 90-kDa protein in immunoblots of OMPs from these three virulent Bordetella species. The accumulation of the Vir90 protein increased 4-fold under low iron growth conditions. Therefore, the vir90 gene is expressed in the tested species and its expression is regulated positively by the BvgAS system and negatively under high iron concentration, likely by Fur.


Asunto(s)
Bordetella pertussis/genética , Factores de Virulencia de Bordetella/genética , Secuencia de Aminoácidos , Proteínas Bacterianas/metabolismo , Secuencia de Bases , Bordetella pertussis/metabolismo , Bordetella pertussis/patogenicidad , Regulación Bacteriana de la Expresión Génica , Hierro/farmacología , Datos de Secuencia Molecular , Regiones Promotoras Genéticas , Conformación Proteica , Proteínas Recombinantes/metabolismo , Homología de Secuencia , Factores de Transcripción/metabolismo , Virulencia , Factores de Virulencia de Bordetella/química , Factores de Virulencia de Bordetella/metabolismo
18.
PLoS One ; 9(10): e110047, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25347845

RESUMEN

Neisseria meningitidis adhesin A (NadA) is a meningococcus surface protein thought to assist in the adhesion of the bacterium to host cells. We have previously shown that NadA also promotes bacterial internalization in a heterologous expression system. Here we have used the soluble recombinant NadA (rNadA) lacking the membrane anchor region to characterize its internalization route in Chang epithelial cells. Added to the culture medium, rNadA internalizes through a PI3K-dependent endocytosis process not mediated by the canonical clathrin or caveolin scaffolds, but instead follows an ARF6-regulated recycling pathway previously described for MHC-I. The intracellular pool of rNadA reaches a steady state level within one hour of incubation and colocalizes in endocytic vesicles with MHC-I and with the extracellularly labeled chaperone Hsp90. Treatment with membrane permeated and impermeable Hsp90 inhibitors 17-AAG and FITC-GA respectively, lead to intracellular accumulation of rNadA, strongly suggesting that the extracellular secreted pool of the chaperone is involved in rNadA intracellular trafficking. A significant number of intracellular vesicles containing rNadA recruit Rab11, a small GTPase associated to recycling endosomes, but do not contain transferrin receptor (TfR). Interestingly, cell treatment with Hsp90 inhibitors, including the membrane-impermeable FITC-GA, abolished Rab11-rNadA colocalization but do not interfere with Rab11-TfR colocalization. Collectively, these results are consistent with a model whereby rNadA internalizes into human epithelial cells hijacking the recycling endosome pathway and recycle back to the surface of the cell via an ARF6-dependent, Rab11 associated and Hsp90-regulated mechanism. The present study addresses for the first time a meningoccoccal adhesin mechanism of endocytosis and suggests a possible entry pathway engaged by N. meningitidis in primary infection of human epithelial cells.


Asunto(s)
Factores de Ribosilacion-ADP/metabolismo , Adhesinas Bacterianas/metabolismo , Células Epiteliales/metabolismo , Proteínas HSP90 de Choque Térmico/metabolismo , Proteínas de Unión al GTP rab/metabolismo , Factor 6 de Ribosilación del ADP , Línea Celular , Humanos , Espacio Intracelular , Neisseria meningitidis/fisiología , Fosfatidilinositol 3-Quinasas/metabolismo , Inhibidores de las Quinasa Fosfoinosítidos-3 , Unión Proteica , Transporte de Proteínas , Proteolisis , Proteínas Recombinantes , Temperatura
19.
Sci Transl Med ; 3(91): 91ra62, 2011 Jul 13.
Artículo en Inglés | MEDLINE | ID: mdl-21753121

RESUMEN

The sequence variability of protective antigens is a major challenge to the development of vaccines. For Neisseria meningitidis, the bacterial pathogen that causes meningitis, the amino acid sequence of the protective antigen factor H binding protein (fHBP) has more than 300 variations. These sequence differences can be classified into three distinct groups of antigenic variants that do not induce cross-protective immunity. Our goal was to generate a single antigen that would induce immunity against all known sequence variants of N. meningitidis. To achieve this, we rationally designed, expressed, and purified 54 different mutants of fHBP and tested them in mice for the induction of protective immunity. We identified and determined the crystal structure of a lead chimeric antigen that was able to induce high levels of cross-protective antibodies in mice against all variant strains tested. The new fHBP antigen had a conserved backbone that carried an engineered surface containing specificities for all three variant groups. We demonstrate that the structure-based design of multiple immunodominant antigenic surfaces on a single protein scaffold is possible and represents an effective way to create broadly protective vaccines.


Asunto(s)
Antígenos Bacterianos/inmunología , Diseño de Fármacos , Inmunidad/inmunología , Neisseria meningitidis/inmunología , Animales , Antibacterianos/farmacología , Antígenos Bacterianos/química , Proteínas Bacterianas/química , Proteínas Bacterianas/inmunología , Cristalografía por Rayos X , Humanos , Inmunidad/efectos de los fármacos , Espectroscopía de Resonancia Magnética , Ratones , Modelos Moleculares , Proteínas Mutantes/química , Proteínas Mutantes/inmunología , Mutación/genética , Neisseria meningitidis/efectos de los fármacos , Ingeniería de Proteínas , Estructura Secundaria de Proteína
20.
J Leukoc Biol ; 86(1): 143-53, 2009 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-19401383

RESUMEN

Hypervirulent MenB causing fatal human infections frequently display the oligomeric-coiled coil adhesin NadA, a 45-kDa intrinsic outer membrane protein implicated in binding to and invasion of respiratory epithelial cells. A recombinant soluble mutant lacking the 10-kDa COOH terminal membrane domain (NadA(Delta351-405)) also activates human monocytes/macrophages/DCs. As NadA is physiologically released during sepsis as part of OMVs, in this study, we tested the hypothesis that NadA(+) OMVs have an enhanced or modified proinflammatory/proimmune action compared with NadA(-) OMVs. To do this we investigated the activity of purified free NadA(Delta351-405) and of OMVs from MenB and Escherichia coli strains, expressing or not full-length NadA. NadA(Delta351-405) stimulated monocytes and macrophages to secrete cytokines (IL-1beta, TNF-alpha, IL-6, IL-12p40, IL-12p70, IL-10) and chemokines (IL-8, MIP-1alpha, MCP-1, RANTES), and full-length NadA improved MenB OMV activity, preferentially on macrophages, and only increased cytokine release. NadA(Delta351-405) induced the lymphocyte costimulant CD80 in monocytes and macrophages, and NadA(+) OMVs induced a wider set of molecules supporting antigen presentation (CD80, CD86, HLA-DR, and ICAM-1) more efficiently than NadA(-) OMVs only in macrophages. Moreover, membrane NadA effects, unlike NadA(Delta351-405) ones, were much less IFN-gamma-sensitive. The activity of NadA-positive E. coli OMVs was similar to that of control OMVs. NadA in MenB OMVs acted at adhesin concentrations approximately 10(6) times lower than those required to stimulate cells with free NadA(Delta351-405).


Asunto(s)
Adhesinas Bacterianas/inmunología , Macrófagos/inmunología , Monocitos/microbiología , Neisseria meningitidis/química , Antígenos de Superficie/análisis , Membrana Celular/microbiología , Células Cultivadas , Quimiocinas/metabolismo , Citocinas/metabolismo , Humanos , Macrófagos/microbiología , Monocitos/inmunología , Neisseria meningitidis/enzimología , Oxo-Ácido-Liasas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA