Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 85
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Int J Mol Sci ; 25(7)2024 Mar 27.
Artículo en Inglés | MEDLINE | ID: mdl-38612547

RESUMEN

Protein self-assembling nanoparticles (NPs) can be used as carriers for antigen delivery to increase vaccine immunogenicity. NPs mimic the majority of invading pathogens, inducing a robust adaptive immune response and long-lasting protective immunity. In this context, we investigated the potential of NPs of different sizes and shapes-ring-, rod-like, and spherical particles-as carriers for bacterial oligosaccharides by evaluating in murine models the role of these parameters on the immune response. Oligosaccharides from Neisseria meningitidis type W capsular polysaccharide were conjugated to ring-shape or nanotubes of engineered Pseudomonas aeruginosa Hemolysin-corregulated protein 1 (Hcp1cc) and to spherical Helicobacter pylori ferritin. Glycoconjugated NPs were characterized using advanced technologies such as High-Performance Liquid Chromatography (HPLC), Asymmetric Flow-Field Flow fractionation (AF4), and Transmission electron microscopy (TEM) to verify their correct assembly, dimensions, and glycosylation degrees. Our results showed that spherical ferritin was able to induce the highest immune response in mice against the saccharide antigen compared to the other glycoconjugate NPs, with increased bactericidal activity compared to benchmark MenW-CRM197. We conclude that shape is a key attribute over size to be considered for glycoconjugate vaccine development.


Asunto(s)
Antiinfecciosos , Nanopartículas , Animales , Ratones , Glicoconjugados , Ferritinas , Oligosacáridos
2.
Int J Mol Sci ; 24(7)2023 Mar 24.
Artículo en Inglés | MEDLINE | ID: mdl-37047152

RESUMEN

The presentation of viral antigens on nanoparticles in multivalent arrays has emerged as a valuable technology for vaccines. On the nanoparticle surface, highly ordered, repetitive arrays of antigens can mimic their geometric arrangement on virion surfaces and elicit stronger humoral responses than soluble viral antigens. More recently, bacterial antigens have been presented on self-assembling protein nanoparticles and have elicited protective antibody and effective T-helper responses, further supporting the nanoparticle platform as a universal approach for stimulating potent immunogenicity. Here, we present the rational design, structural analysis, and immunogenicity of self-assembling ferritin nanoparticles displaying eight copies of the Neisseria meningitidis trimeric adhesin NadA. We engineered constructs consisting of two different NadA fragments, head only and head with stalk, that we fused to ferritin and expressed in Escherichia coli. Both fusion constructs self-assembled into the expected nanoparticles as determined by Cryo electron microscopy. In mice, the two nanoparticles elicited comparable NadA antibody levels that were 10- to 100-fold higher than those elicited by the corresponding NadA trimer subunits. Further, the NadAferritin nanoparticles potently induced complement-mediated serum bactericidal activity. These findings confirm the value of self-assembling nanoparticles for optimizing the immunogenicity of bacterial antigens and support the broad applicability of the approach to vaccine programs, especially for the presentation of trimeric antigens.


Asunto(s)
Nanopartículas , Neisseria meningitidis , Ratones , Animales , Ferritinas , Antígenos Bacterianos , Antígenos Virales , Anticuerpos Bloqueadores , Vacunas Combinadas , Nanopartículas/química
3.
Anal Chem ; 93(34): 11669-11678, 2021 08 31.
Artículo en Inglés | MEDLINE | ID: mdl-34308633

RESUMEN

Epitope mapping of antibodies (Abs) is crucial for understanding adaptive immunity, as well as studying the mode of action of therapeutic antibodies and vaccines. Especially insights into the binding of the entire polyclonal antibody population (pAb) raised upon vaccination would be of unique value to vaccine development. However, very few methods for epitope mapping can tolerate the complexity of a pAb sample. Here we show how hydrogen-deuterium exchange mass spectrometry (HDX-MS) can be used to map epitopes recognized by pAb samples. Our approach involves measuring the HDX of the antigen in absence or presence of varied amounts of pAbs, as well as dissociating additives. We apply the HDX-MS workflow to pAbs isolated from rabbit immunized with factor H-binding protein (fHbp), a Neisseria meningitidis vaccine antigen. We identify four immunogenic regions located on the N- and C-terminal region of fHbp and provide insights into the relative abundance and avidity of epitope binding Abs present in the sample. Overall, our results show that HDX-MS can provide a unique and relatively fast method for revealing the binding impact of the entire set of pAbs present in blood samples after vaccination. Such information provides a rare view into effective immunity and can guide the design of improved vaccines against viruses or bacteria.


Asunto(s)
Medición de Intercambio de Deuterio , Espectrometría de Masas de Intercambio de Hidrógeno-Deuterio , Animales , Anticuerpos Monoclonales , Deuterio , Mapeo Epitopo , Espectrometría de Masas , Conejos
4.
FASEB J ; 34(8): 10329-10341, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32725956

RESUMEN

The classical complement pathway is triggered when antigen-bound immunoglobulins bind to C1q through their Fc region. While C1q binds to a single Fc with low affinity, a higher avidity stable binding of two or more of C1q globular heads initiates the downstream reactions of the complement cascade ultimately resulting in bacteriolysis. Synergistic bactericidal activity has been demonstrated when monoclonal antibodies recognize nonoverlapping epitopes of the same antigen. The aim of the present work was to investigate the synergistic effect between antibodies directed toward different antigens. To this purpose, we investigated the bactericidal activity induced by combinations of monoclonal antibodies (mAbs) raised against factor H-binding protein (fHbp) and Neisserial Heparin-Binding Antigen (NHBA), two major antigens included in Bexsero, the vaccine against Meningococcus B, for prevention from this devastating disease in infants and adolescents. Collectively, our results show that mAbs recognizing different antigens can synergistically activate complement even when each single Mab is not bactericidal, reinforcing the evidence that cooperative immunity induced by antigen combinations can represent a remarkable added value of multicomponent vaccines. Our study also shows that the synergistic effect of antibodies is modulated by the nature of the respective epitopes, as well as by the antigen density on the bacterial cell surface.


Asunto(s)
Anticuerpos Antibacterianos/inmunología , Anticuerpos Monoclonales/inmunología , Proteínas del Sistema Complemento/inmunología , Antígenos Bacterianos/inmunología , Proteínas de la Membrana Bacteriana Externa/inmunología , Proteínas Bacterianas/inmunología , Proteínas Portadoras/inmunología , Factor H de Complemento/inmunología , Epítopos/inmunología , Neisseria meningitidis/inmunología , Determinación de Anticuerpos Séricos Bactericidas/métodos
5.
Int J Mol Sci ; 22(9)2021 May 06.
Artículo en Inglés | MEDLINE | ID: mdl-34066555

RESUMEN

Traditional antimicrobial treatments consist of drugs which target different essential functions in pathogens. Nevertheless, bacteria continue to evolve new mechanisms to evade this drug-mediated killing with surprising speed on the deployment of each new drug and antibiotic worldwide, a phenomenon called antimicrobial resistance (AMR). Nowadays, AMR represents a critical health threat, for which new medical interventions are urgently needed. By 2050, it is estimated that the leading cause of death will be through untreatable AMR pathogens. Although antibiotics remain a first-line treatment, non-antibiotic therapies such as prophylactic vaccines and therapeutic monoclonal antibodies (mAbs) are increasingly interesting alternatives to limit the spread of such antibiotic resistant microorganisms. For the discovery of new vaccines and mAbs, the search for effective antigens that are able to raise protective immune responses is a challenging undertaking. In this context, outer membrane vesicles (OMV) represent a promising approach, as they recapitulate the complete antigen repertoire that occurs on the surface of Gram-negative bacteria. In this review, we present Escherichia coli and Pseudomonas aeruginosa as specific examples of key AMR threats caused by Gram-negative bacteria and we discuss the current status of mAbs and vaccine approaches under development as well as how knowledge on OMV could benefit antigen discovery strategies.


Asunto(s)
Farmacorresistencia Bacteriana , Escherichia coli/fisiología , Pseudomonas aeruginosa/fisiología , Animales , Vacunas Bacterianas/inmunología , Infecciones por Escherichia coli/epidemiología , Infecciones por Escherichia coli/inmunología , Infecciones por Escherichia coli/microbiología , Humanos , Infecciones por Pseudomonas/epidemiología , Infecciones por Pseudomonas/inmunología , Infecciones por Pseudomonas/microbiología
6.
Infect Immun ; 87(3)2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30530621

RESUMEN

Clostridium difficile is a major cause of hospital-acquired antibiotic-associated diarrhea. C. difficile produces two cytotoxins, TcdA and TcdB; both toxins are multidomain proteins that lead to cytotoxicity through the modification and inactivation of small GTPases of the Rho/Rac family. Previous studies have indicated that host glycans are targets for TcdA and TcdB, with interactions thought to be with both α- and ß-linked galactose. In the current study, screening of glycan arrays with different domains of TcdA and TcdB revealed that the binding regions of both toxins interact with a wider range of host glycoconjugates than just terminal α- and ß-linked galactose, including blood groups, Lewis antigens, N-acetylglucosamine, mannose, and glycosaminoglycans. The interactions of TcdA and TcdB with ABO blood group and Lewis antigens were assessed by surface plasmon resonance (SPR). The blood group A antigen was the highest-affinity ligand for both toxins. Free glycans alone or in combination were unable to abolish Vero cell cytotoxicity by TcdB. SPR competition assays indicate that there is more than one glycan binding site on TcdB. Host glycoconjugates are common targets of bacterial toxins, but typically this binding is to a specific structure or related structures. The binding of TcdA and TcdB is to a wide range of host glycans providing a wide range of target cells and tissues in vivo.


Asunto(s)
Proteínas Bacterianas/metabolismo , Toxinas Bacterianas/metabolismo , Clostridioides difficile/metabolismo , Enterotoxinas/metabolismo , Lectinas/metabolismo , Animales , Supervivencia Celular , Chlorocebus aethiops , Clonación Molecular , Polisacáridos , Células Vero
7.
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
8.
J Proteome Res ; 17(5): 1794-1800, 2018 05 04.
Artículo en Inglés | MEDLINE | ID: mdl-29619829

RESUMEN

Hydrogen-deuterium exchange (HDx) associated with mass spectrometry (MS) is emerging as a powerful tool to provide conformational information about membrane proteins. Unfortunately, as for X-ray diffraction and NMR, HDx performed on reconstituted in vitro systems might not always reflect the in vivo environment. Outer-membrane vesicles naturally released by Escherichia coli were used to carry out analysis of native OmpF through HDx-MS. A new protocol compatible with HDx analysis that avoids hindrance from the lipid contents was setup. The extent of deuterium incorporation was in good agreement with the X-ray diffraction data of OmpF as the buried ß-barrels incorporated a low amount of deuterium, whereas the internal loop L3 and the external loops incorporated a higher amount of deuterium. Moreover, the kinetics of incorporation clearly highlights that peptides segregate well in two distinct groups based exclusively on a trimeric organization of OmpF in the membrane: peptides presenting fast kinetics of labeling are facing the complex surrounding environment, whereas those presenting slow kinetics are located in the buried core of the trimer. The data show that HDx-MS applied to a complex biological system is able to reveal solvent accessibility and spatial arrangement of an integral outer-membrane protein complex.


Asunto(s)
Proteínas Bacterianas/química , Medición de Intercambio de Deuterio/métodos , Espectrometría de Masas/métodos , Porinas/química , Proteínas de la Membrana Bacteriana Externa/química , Proteínas de Escherichia coli/química , Cinética , Conformación Proteica
9.
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
10.
Arch Biochem Biophys ; 635: 102-109, 2017 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-29097311

RESUMEN

Mono ADP-ribosyltransferases are a class of functionally conserved enzymes present in prokaryotic and eukaryotic organisms. In prokaryotes, mono ADP-ribose transfer enzymes often represent a family of exotoxins that display activity in a variety of bacteria responsible for causing disease in plants and animals. A bioinformatic approach has allowed us to identify that CagL gene from some Helicobacter pylori strains shares a sequence pattern with ADP-ribosylating toxins of the CT-group. In this manuscript we show that recombinant CagL from Shi470 is catalytically active showing ADP-ribosyltransferase, NAD-glycohydrolase, and auto-ADP-ribosylation activities. This is the first time that a catalytically active member of the ADP-ribosyltransferase family is identified in Helicobacter pylori. This observation may lead to the discovery of novel functions exerted by CagL in the pathogenesis of Helicobacter pylori. Indeed, we have shown that vaccination with CagL has protective efficacy in mice indicating that CagL may be considered as potential component of a Helicobacter pylori vaccine.


Asunto(s)
ADP Ribosa Transferasas/metabolismo , ADP-Ribosilación , Proteínas Bacterianas/farmacocinética , Proteínas Bacterianas/uso terapéutico , Infecciones por Helicobacter/microbiología , Infecciones por Helicobacter/prevención & control , NAD+ Nucleosidasa/metabolismo , Animales , Proteínas Bacterianas/química , Sitios de Unión , Femenino , Ratones , Unión Proteica , Resultado del Tratamiento
11.
Biochem J ; 473(24): 4699-4713, 2016 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-27784765

RESUMEN

Factor H-binding protein (fHbp) is an important antigen of Neisseria meningitidis that is capable of eliciting a robust protective immune response in humans. Previous studies on the interactions of fHbp with antibodies revealed that some anti-fHbp monoclonal antibodies that are unable to trigger complement-mediated bacterial killing in vitro are highly co-operative and become bactericidal if used in combination. Several factors have been shown to influence such co-operativity, including IgG subclass and antigen density. To investigate the structural basis of the anti-fHbp antibody synergy, we determined the crystal structure of the complex between fHbp and the Fab (fragment antigen-binding) fragment of JAR5, a specific anti-fHbp murine monoclonal antibody known to be highly co-operative with other monoclonal antibodies. We show that JAR5 is highly synergic with monoclonal antibody (mAb) 12C1, whose structure in complex with fHbp has been previously solved. Structural analyses of the epitopes recognized by JAR5 and 12C1, and computational modeling of full-length IgG mAbs of JAR5 and 12C1 bound to the same fHbp molecule, provide insights into the spatial orientation of Fc (fragment crystallizable) regions and into the possible implications for the susceptibility of meningococci to complement-mediated killing.


Asunto(s)
Anticuerpos Monoclonales/metabolismo , Antígenos Bacterianos/inmunología , Antígenos Bacterianos/metabolismo , Proteínas Bacterianas/inmunología , Proteínas Bacterianas/metabolismo , Neisseria meningitidis/metabolismo , Anticuerpos Monoclonales/química , Anticuerpos Monoclonales/inmunología , Antígenos Bacterianos/química , Proteínas Bacterianas/química , Factor H de Complemento/inmunología , Factor H de Complemento/metabolismo , Epítopos/inmunología , Epítopos/metabolismo , Fragmentos Fab de Inmunoglobulinas/inmunología , Fragmentos Fab de Inmunoglobulinas/metabolismo , Inmunoglobulina G/inmunología , Inmunoglobulina G/metabolismo , Unión Proteica , Estructura Secundaria de Proteína
12.
Proc Natl Acad Sci U S A ; 111(48): 17128-33, 2014 Dec 02.
Artículo en Inglés | MEDLINE | ID: mdl-25404323

RESUMEN

Serogroup B Neisseria meningitidis (MenB) is a major cause of severe sepsis and invasive meningococcal disease, which is associated with 5-15% mortality and devastating long-term sequelae. Neisserial adhesin A (NadA), a trimeric autotransporter adhesin (TAA) that acts in adhesion to and invasion of host epithelial cells, is one of the three antigens discovered by genome mining that are part of the MenB vaccine that recently was approved by the European Medicines Agency. Here we present the crystal structure of NadA variant 5 at 2 Å resolution and transmission electron microscopy data for NadA variant 3 that is present in the vaccine. The two variants show similar overall topology with a novel TAA fold predominantly composed of trimeric coiled-coils with three protruding wing-like structures that create an unusual N-terminal head domain. Detailed mapping of the binding site of a bactericidal antibody by hydrogen/deuterium exchange MS shows that a protective conformational epitope is located in the head of NadA. These results provide information that is important for elucidating the biological function and vaccine efficacy of NadA.


Asunto(s)
Adhesinas Bacterianas/inmunología , Anticuerpos Antibacterianos/inmunología , Antígenos Bacterianos/inmunología , Mapeo Epitopo/métodos , Vacunas Meningococicas/inmunología , Neisseria meningitidis Serogrupo B/inmunología , Adhesinas Bacterianas/química , Adhesinas Bacterianas/genética , Secuencia de Aminoácidos , Antígenos Bacterianos/química , Antígenos Bacterianos/genética , Sitios de Unión de Anticuerpos/genética , Sitios de Unión de Anticuerpos/inmunología , Cristalografía por Rayos X , Medición de Intercambio de Deuterio , Microscopía Electrónica de Transmisión , Modelos Moleculares , Datos de Secuencia Molecular , Neisseria meningitidis Serogrupo B/genética , Isoformas de Proteínas/química , Isoformas de Proteínas/genética , Isoformas de Proteínas/inmunología , Multimerización de Proteína , Estabilidad Proteica , Estructura Terciaria de Proteína , Homología de Secuencia de Aminoácido , Espectrometría de Masa por Ionización de Electrospray , Temperatura
13.
J Biol Inorg Chem ; 21(2): 185-96, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26711661

RESUMEN

Proteases are commonly secreted by microorganisms. In some pathogens, they can play a series of functional roles during infection, including maturation of cell surface or extracellular virulence factors, interference with host cell signaling, massive host tissue destruction, and dissolution of infection-limiting clots through degradation of the host proteins devoted to the coagulation cascade. We previously reported the identification and characterization of Zmp1, a zinc-dependent metalloprotease secreted by Clostridium difficile, demonstrated that Zmp1 is able to degrade fibrinogen in vitro, and identified two residues necessary to the catalytic activity. In the present work, we solved the solution structure of Zmp1 by Nuclear Magnetic Resonance (NMR) and compared it with the recently solved X-ray structures of substrate-bound and substrate-free Zmp1, highlighting similarities and differences. We also combined the structural characterization to biochemical assays and site-directed mutagenesis, to provide new insights into the catalytic site and on the residues responsible for substrate specificity. The Zmp1 structure showed similarity to the catalytic domain of Anthrax Lethal Factor of Bacillus anthracis. Analogies and differences in the catalytic and in the substrate-binding sites of the two proteins are discussed.


Asunto(s)
Clostridioides difficile/enzimología , Metaloproteasas/química , Secuencia de Aminoácidos , Espectroscopía de Resonancia Magnética , Conformación Proteica , Homología de Secuencia de Ácido Nucleico
14.
FASEB J ; 28(4): 1644-53, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24371123

RESUMEN

Factor H binding protein (fHbp) is one of the main antigens of the 4-component meningococcus B (4CMenB) multicomponent vaccine against disease caused by serogroup B Neisseria meningitidis (MenB). fHbp binds the complement down-regulating protein human factor H (hfH), thus resulting in immune evasion. fHbp exists in 3 variant groups with limited cross-protective responses. Previous studies have described the generation of monoclonal antibodies (mAbs) targeting variant-specific regions of fHbp. Here we report for the first time the functional characterization of two mAbs that recognize a wide panel of fHbp variants and subvariants on the MenB surface and that are able to inhibit fHbp binding to hfH. The antigenic regions targeted by the two mAbs were accurately mapped by hydrogen-deuterium exchange mass spectrometry (HDX-MS), revealing partially overlapping epitopes on the N terminus of fHbp. Furthermore, while none of the mAbs had bactericidal activity on its own, a synergistic effect was observed for each of them when tested by the human complement serum bactericidal activity (hSBA) assay in combination with a second nonbactericidal mAb. The bases underlying fHbp variant cross-reactivity, as well as inhibition of hfH binding and cooperativity effect observed for the two mAbs, are discussed in light of the mapped epitopes.


Asunto(s)
Anticuerpos Monoclonales/inmunología , Antígenos Bacterianos/inmunología , Proteínas Bacterianas/inmunología , Reacciones Cruzadas/inmunología , Epítopos/inmunología , Neisseria meningitidis Serogrupo B/inmunología , Anticuerpos Monoclonales/química , Antígenos Bacterianos/genética , Proteínas Bacterianas/genética , Factor H de Complemento/inmunología , Medición de Intercambio de Deuterio , Mapeo Epitopo/métodos , Epítopos/química , Epítopos/genética , Variación Genética , Humanos , Espectrometría de Masas , Infecciones Meningocócicas/inmunología , Infecciones Meningocócicas/microbiología , Vacunas Meningococicas/inmunología , Modelos Moleculares , Neisseria meningitidis Serogrupo B/genética , Neisseria meningitidis Serogrupo B/fisiología , Unión Proteica/inmunología , Conformación Proteica , Resonancia por Plasmón de Superficie
15.
Cell Microbiol ; 15(10): 1674-87, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23517059

RESUMEN

Clostridium difficile is the leading cause of antibiotic-associated diarrhoea and pseudomembranous colitis. While the role of toxins in pathogenesis has been extensively described, the contribution of surface determinants to intestinal colonization is still poorly understood. We focused our study on a novel member of the MSCRAMM family, named CbpA (Collagen binding protein A), for its adhesive properties towards collagen. We demonstrate that CbpA, which carries an LPXTG-like cell wall anchoring domain, is expressed on the bacterial surface of C. difficile and that the recombinant protein binds at high affinity to collagens I and V (apparent Kd in the order of 10(-9 ) M). These findings were validated by confocal microscopy studies showing the colocalization of the protein with type I and V collagen fibres produced by human fibroblasts and mouse intestinal tissues. However, the collagen binding activity of the wild-type C. difficile 630 strain was indistinguishable to the cbpA knock-out strain. To overcome this apparent clostridial adherence redundancy, we engineered a Lactococcus lactis strain for the heterologous expression of CbpA. When exposed on the surface of L. lactis, CbpA significantly enhances the ability of the bacterium to interact with collagen and to adhere to ECM-producing cells. The binding activity of L. lactis-CbpA strain was prevented by an antiserum raised against CbpA, demonstrating the specificity of the interaction. These results suggest that CbpA is a newsurface-exposed adhesin contributing to the C. difficile interaction with the host.


Asunto(s)
Adhesinas Bacterianas/metabolismo , Adhesión Bacteriana , Proteínas Bacterianas/metabolismo , Proteínas Portadoras/metabolismo , Clostridioides difficile/fisiología , Colágeno/metabolismo , Interacciones Huésped-Patógeno , Animales , Fibroblastos/metabolismo , Fibroblastos/microbiología , Humanos , Mucosa Intestinal/metabolismo , Mucosa Intestinal/microbiología , Cinética , Lactococcus lactis/genética , Lactococcus lactis/fisiología , Ratones , Microscopía Confocal , Unión Proteica
16.
Mol Cell Proteomics ; 11(6): M111.015693, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22286755

RESUMEN

We propose an experimental strategy for highly accurate selection of candidates for bacterial vaccines without using in vitro and/or in vivo protection assays. Starting from the observation that efficacious vaccines are constituted by conserved, surface-associated and/or secreted components, the strategy contemplates the parallel application of three high throughput technologies, i.e. mass spectrometry-based proteomics, protein array, and flow-cytometry analysis, to identify this category of proteins, and is based on the assumption that the antigens identified by all three technologies are the protective ones. When we tested this strategy for Group A Streptococcus, we selected a total of 40 proteins, of which only six identified by all three approaches. When the 40 proteins were tested in a mouse model, only six were found to be protective and five of these belonged to the group of antigens in common to the three technologies. Finally, a combination of three protective antigens conferred broad protection against a panel of four different Group A Streptococcus strains. This approach may find general application as an accelerated and highly accurate path to bacterial vaccine discovery.


Asunto(s)
Antígenos Bacterianos/inmunología , Vacunas Bacterianas/administración & dosificación , Infecciones Estreptocócicas/prevención & control , Streptococcus pyogenes/inmunología , Animales , Anticuerpos Antibacterianos/sangre , Antígenos Bacterianos/genética , Antígenos Bacterianos/metabolismo , Proteínas de la Membrana Bacteriana Externa/genética , Proteínas de la Membrana Bacteriana Externa/inmunología , Proteínas de la Membrana Bacteriana Externa/metabolismo , Análisis por Conglomerados , Femenino , Citometría de Flujo , Hemólisis , Humanos , Ratones , Faringitis/sangre , Faringitis/inmunología , Faringitis/microbiología , Análisis por Matrices de Proteínas , Proteoma/inmunología , Proteoma/metabolismo , Ovinos , Infecciones Estreptocócicas/sangre , Infecciones Estreptocócicas/inmunología , Infecciones Estreptocócicas/microbiología , Streptococcus pyogenes/metabolismo , Vacunación
17.
Proc Natl Acad Sci U S A ; 108(24): 9969-74, 2011 Jun 14.
Artículo en Inglés | MEDLINE | ID: mdl-21628568

RESUMEN

Natural immunity against obligate and/or facultative intracellular pathogens is usually mediated by both humoral and cellular immunity. The identification of those antigens stimulating both arms of the immune system is instrumental for vaccine discovery. Although high-throughput technologies have been applied for the discovery of antibody-inducing antigens, few examples of their application for T-cell antigens have been reported. We describe how the compilation of the immunome, here defined as the pool of immunogenic antigens inducing T- and B-cell responses in vivo, can lead to vaccine candidates against Chlamydia trachomatis. We selected 120 C. trachomatis proteins and assessed their immunogenicity using two parallel high-throughput approaches. Protein arrays were generated and screened with sera from C. trachomatis-infected patients to identify antibody-inducing antigens. Splenocytes from C. trachomatis-infected mice were stimulated with 79 proteins, and the frequency of antigen-specific CD4(+)/IFN-γ(+) T cells was analyzed by flow cytometry. We identified 21 antibody-inducing antigens, 16 CD4(+)/IFN-γ(+)-inducing antigens, and five antigens eliciting both types of responses. Assessment of their protective activity in a mouse model of Chlamydia muridarum lung infection led to the identification of seven antigens conferring partial protection when administered with LTK63/CpG adjuvant. Protection was largely the result of cellular immunity as assessed by CD4(+) T-cell depletion. The seven antigens provided robust additive protection when combined in four-antigen combinations. This study paves the way for the development of an effective anti-Chlamydia vaccine and provides a general approach for the discovery of vaccines against other intracellular pathogens.


Asunto(s)
Antígenos Bacterianos/inmunología , Linfocitos B/inmunología , Vacunas Bacterianas/inmunología , Chlamydia trachomatis/inmunología , Linfocitos T/inmunología , Animales , Anticuerpos Antibacterianos/inmunología , Antígenos Bacterianos/metabolismo , Proteínas Bacterianas/inmunología , Proteínas Bacterianas/metabolismo , Vacunas Bacterianas/uso terapéutico , Western Blotting , Linfocitos T CD4-Positivos/inmunología , Línea Celular , Infecciones por Chlamydia/inmunología , Infecciones por Chlamydia/microbiología , Infecciones por Chlamydia/prevención & control , Chlamydia muridarum/inmunología , Chlamydia trachomatis/metabolismo , Femenino , Células HeLa , Humanos , Sueros Inmunes/inmunología , Inmunización , Interferón gamma/inmunología , Ratones , Ratones Endogámicos BALB C , Microscopía Confocal , Células TH1/inmunología
18.
J Bacteriol ; 195(3): 545-55, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23175653

RESUMEN

Bacteria within biofilms are protected from multiple stresses, including immune responses and antimicrobial agents. The biofilm-forming ability of bacterial pathogens has been associated with increased antibiotic resistance and chronic recurrent infections. Although biofilms have been well studied for several gut pathogens, little is known about biofilm formation by anaerobic gut species. The obligate anaerobe Clostridium difficile causes C. difficile infection (CDI), a major health care-associated problem primarily due to the high incidence of recurring infections. C. difficile colonizes the gut when the normal intestinal microflora is disrupted by antimicrobial agents; however, the factors or processes involved in gut colonization during infection remain unclear. We demonstrate that clinical C. difficile strains, i.e., strain 630 and the hypervirulent strain R20291, form structured biofilms in vitro, with R20291 accumulating substantially more biofilm. Microscopic and biochemical analyses show multiple layers of bacteria encased in a biofilm matrix containing proteins, DNA, and polysaccharide. Employing isogenic mutants, we show that virulence-associated proteins, Cwp84, flagella, and a putative quorum-sensing regulator, LuxS, are all required for maximal biofilm formation by C. difficile. Interestingly, a mutant in Spo0A, a transcription factor that controls spore formation, was defective for biofilm formation, indicating a possible link between sporulation and biofilm formation. Furthermore, we demonstrate that bacteria in clostridial biofilms are more resistant to high concentrations of vancomycin, a drug commonly used for treatment of CDI. Our data suggest that biofilm formation by C. difficile is a complex multifactorial process and may be a crucial mechanism for clostridial persistence in the host.


Asunto(s)
Biopelículas/crecimiento & desarrollo , Clostridioides difficile/fisiología , Antibacterianos/farmacología , Adhesión Bacteriana , Biopelículas/efectos de los fármacos , Clostridioides difficile/clasificación , Clostridioides difficile/efectos de los fármacos , Flagelos , Proteínas de la Membrana/fisiología , Pruebas de Sensibilidad Microbiana , Percepción de Quorum , Esporas Bacterianas , Factores de Tiempo , Vancomicina/farmacología , Resistencia a la Vancomicina
19.
Infect Immun ; 81(8): 2851-60, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23716610

RESUMEN

Clostridium difficile is a spore-forming bacterium that can reside in animals and humans. C. difficile infection causes a variety of clinical symptoms, ranging from diarrhea to fulminant colitis. Disease is mediated by TcdA and TcdB, two large enterotoxins released by C. difficile during colonization of the gut. In this study, we evaluated the ability of recombinant toxin fragments to induce neutralizing antibodies in mice. The protective efficacies of the most promising candidates were then evaluated in a hamster model of disease. While limited protection was observed with some combinations, coadministration of a cell binding domain fragment of TcdA (TcdA-B1) and the glucosyltransferase moiety of TcdB (TcdB-GT) induced systemic IgGs which neutralized both toxins and protected vaccinated animals from death following challenge with two strains of C. difficile. Further characterization revealed that despite high concentrations of toxin in the gut lumens of vaccinated animals during the acute phase of the disease, pathological damage was minimized. Assessment of gut contents revealed the presence of TcdA and TcdB antibodies, suggesting that systemic vaccination with this pair of recombinant polypeptides can limit the disease caused by toxin production during C. difficile infection.


Asunto(s)
Proteínas Bacterianas/inmunología , Toxinas Bacterianas/inmunología , Vacunas Bacterianas/inmunología , Infecciones por Clostridium/inmunología , Enterotoxinas/inmunología , Animales , Anticuerpos Antibacterianos/inmunología , Anticuerpos Neutralizantes/inmunología , Antígenos Bacterianos/inmunología , Clostridioides difficile/inmunología , Infecciones por Clostridium/prevención & control , Cricetinae , Modelos Animales de Enfermedad , Electroforesis en Gel de Poliacrilamida , Ensayo de Inmunoadsorción Enzimática , Humanos , Immunoblotting , Ratones , Proteínas Recombinantes/inmunología
20.
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
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA