Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
2.
FASEB J ; 38(1): e23326, 2024 01.
Artículo en Inglés | MEDLINE | ID: mdl-38019196

RESUMEN

The identification and recombinant production of functional antigens and/or epitopes of pathogens represent a crucial step for the development of an effective protein-based vaccine. Many vaccine targets are outer membrane proteins anchored into the lipidic bilayer through an extended hydrophobic portion making their recombinant production challenging. Moreover, only the extracellular loops, and not the hydrophobic regions, are naturally exposed to the immune system. In this work, the Domain 3 (D3) from Group B Streptococcus (GBS) pilus 2a backbone protein has been identified and engineered to be used as a scaffold for the display of extracellular loops of two Neisseria gonorrhoeae membrane proteins (PorB.1b and OpaB). A computational structure-based approach has been applied to the design of both the scaffold and the model antigens. Once identified the best D3 engineerable site, several different chimeric D3 displaying PorB.1b and OpaB extracellular loops were produced as soluble proteins. Each molecule has been characterized in terms of solubility, stability, and ability to correctly display the foreign epitope. This antigen dissection strategy allowed the identification of most immunogenic extracellular loops of both PorB.1b and OpaB gonococcal antigens. The crystal structure of chimeric D3 displaying PorB.1b immunodominant loop has been obtained confirming that the engineerization did not alter the predicted native structure of this epitope. Taken together, the reported data suggest that D3 is a novel protein scaffold for epitope insertion and display, and a valid alternative to the production of whole membrane protein antigens. Finally, this work describes a generalized computational structure-based approach for the identification, design, and dissection of epitopes in target antigens through chimeric proteins.


Asunto(s)
Proteínas de la Membrana , Vacunas , Epítopos/genética , Antígenos Bacterianos/genética , Membrana Dobles de Lípidos
3.
NPJ Vaccines ; 8(1): 152, 2023 Oct 06.
Artículo en Inglés | MEDLINE | ID: mdl-37803013

RESUMEN

A maternal vaccine to protect neonates against Group B Streptococcus invasive infection is an unmet medical need. Such a vaccine should ideally be offered during the third trimester of pregnancy and induce strong immune responses after a single dose to maximize the time for placental transfer of protective antibodies. A key target antigen is the capsular polysaccharide, an anti-phagocytic virulence factor that elicits protective antibodies when conjugated to carrier proteins. The most prevalent polysaccharide serotypes conjugated to tetanus or diphtheria toxoids have been tested in humans as monovalent and multivalent formulations, showing excellent safety profiles and immunogenicity. However, responses were suboptimal in unprimed individuals after a single shot, the ideal schedule for vaccination during the third trimester of pregnancy. In the present study, we obtained and optimized self-assembling virus-like particles conjugated to Group B Streptococcus capsular polysaccharides. The resulting glyco-nanoparticles elicited strong immune responses in mice already after one immunization, providing pre-clinical proof of concept for a single-dose vaccine.

4.
PLoS One ; 17(9): e0273322, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36112575

RESUMEN

Recombinant protein-based vaccines are a valid and safer alternative to traditional vaccines based on live-attenuated or killed pathogens. However, the immune response of subunit vaccines is generally lower compared to that elicited by traditional vaccines and usually requires the use of adjuvants. The use of self-assembling protein nanoparticles, as a platform for vaccine antigen presentation, is emerging as a promising approach to enhance the production of protective and functional antibodies. In this work we demonstrated the successful repetitive antigen display of the C-terminal ß-barrel domain of factor H binding protein, derived from serogroup B Meningococcus on the surface of different self-assembling nanoparticles using genetic fusion. Six nanoparticle scaffolds were tested, including virus-like particles with different sizes, geometries, and physicochemical properties. Combining computational and structure-based rational design we were able generate antigen-fused scaffolds that closely aligned with three-dimensional structure predictions. The chimeric nanoparticles were produced as recombinant proteins in Escherichia coli and evaluated for solubility, stability, self-assembly, and antigen accessibility using a variety of biophysical methods. Several scaffolds were identified as being suitable for genetic fusion with the ß-barrel from fHbp, including ferritin, a de novo designed aldolase from Thermotoga maritima, encapsulin, CP3 phage coat protein, and the Hepatitis B core antigen. In conclusion, a systematic screening of self-assembling nanoparticles has been applied for the repetitive surface display of a vaccine antigen. This work demonstrates the capacity of rational structure-based design to develop new chimeric nanoparticles and describes a strategy that can be utilized to discover new nanoparticle-based approaches in the search for vaccines against bacterial pathogens.


Asunto(s)
Vacunas Meningococicas , Nanopartículas , Neisseria meningitidis , Aldehído-Liasas , Antígenos , Vacunas Bacterianas , Proteínas Portadoras , Factor H de Complemento , Ferritinas , Antígenos del Núcleo de la Hepatitis B , Nanopartículas/química , Neisseria meningitidis/genética , Proteínas Recombinantes , Vacunas Combinadas , Vacunas de Subunidad
5.
PLoS Pathog ; 16(10): e1008882, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-33007046

RESUMEN

Neisseria meningitidis serogroup B (MenB) is the leading cause of meningococcal meningitis and sepsis in industrialized countries, with the highest incidence in infants and adolescents. Two recombinant protein vaccines that protect against MenB are now available (i.e. 4CMenB and MenB-fHbp). Both vaccines contain the Factor H Binding Protein (fHbp) antigen, which can bind the Human Factor H (fH), the main negative regulator of the alternative complement pathway, thus enabling bacterial survival in the blood. fHbp is present in meningococcal strains as three main variants which are immunologically distinct. Here we sought to obtain detailed information about the epitopes targeted by anti-fHbp antibodies induced by immunization with the 4CMenB multicomponent vaccine. Thirteen anti-fHbp human monoclonal antibodies (mAbs) were identified in a library of over 100 antibody fragments (Fabs) obtained from three healthy adult volunteers immunized with 4CMenB. Herein, the key cross-reactive mAbs were further characterized for antigen binding affinity, complement-mediated serum bactericidal activity (SBA) and the ability to inhibit binding of fH to live bacteria. For the first time, we identified a subset of anti-fHbp mAbs able to elicit human SBA against strains with all three variants and able to compete with human fH for fHbp binding. We present the crystal structure of fHbp v1.1 complexed with human antibody 4B3. The structure, combined with mutagenesis and binding studies, revealed the critical cross-reactive epitope. The structure also provided the molecular basis of competition for fH binding. These data suggest that the fH binding site on fHbp v1.1 can be accessible to the human immune system upon immunization, enabling elicitation of human mAbs broadly protective against MenB. The novel structural, biochemical and functional data are of great significance because the human vaccine-elicited mAbs are the first reported to inhibit the binding of fH to fHbp, and are bactericidal with human complement. Our studies provide molecular insights into the human immune response to the 4CMenB meningococcal vaccine and fuel the rationale for combined structural, immunological and functional studies when seeking deeper understanding of the mechanisms of action of human vaccines.


Asunto(s)
Anticuerpos/inmunología , Antígenos Bacterianos/metabolismo , Proteínas Bacterianas/metabolismo , Meningitis Meningocócica/inmunología , Vacunas Meningococicas/administración & dosificación , Neisseria meningitidis/inmunología , Adulto , Anticuerpos/sangre , Antígenos Bacterianos/inmunología , Proteínas Bacterianas/inmunología , Factor H de Complemento/inmunología , Factor H de Complemento/metabolismo , Humanos , Meningitis Meningocócica/metabolismo , Meningitis Meningocócica/microbiología , Meningitis Meningocócica/prevención & control
6.
FASEB J ; 33(11): 12099-12111, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31442074

RESUMEN

The 4 component meningococcus B vaccine (4CMenB) vaccine is the first vaccine containing recombinant proteins licensed for the prevention of invasive meningococcal disease caused by meningococcal serogroup B strains. 4CMenB contains 3 main recombinant proteins, including the Neisseria meningitidis factor H binding protein (fHbp), a lipoprotein able to bind the human factor H. To date, over 1000 aa sequences of fHbp have been identified, and they can be divided into variant groups 1, 2, and 3, which are usually not crossprotective. Nevertheless, previous characterizations of a small set (n = 10) of mAbs generated in humans after 4CMenB immunization revealed 2 human Fabs (huFabs) (1A12, 1G3) with some crossreactivity for variants 1, 2, and 3. This unexpected result prompted us to examine a much larger set of human mAbs (n = 110), with the aim of better understanding the extent and nature of crossreactive anti-fHbp antibodies. In this study, we report an analysis of the human antibody response to fHbp, by the characterization of 110 huFabs collected from 3 adult vaccinees during a 6-mo study. Although the 4CMenB vaccine contains fHbp variant 1, 13 huFabs were also found to be crossreactive with variants 2 and 3. The crystal structure of the crossreactive huFab 1E6 in complex with fHbp variant 3 was determined, revealing a novel, highly conserved epitope distinct from the epitopes recognized by 1A12 or 1G3. Further, functional characterization shows that human mAb 1E6 is able to elicit rabbit, but not human, complement-mediated bactericidal activity against meningococci displaying fHbp from any of the 3 different variant groups. This functional and structural information about the human antibody response upon 4CMenB immunization contributes to further unraveling the immunogenic properties of fHbp. Knowledge gained about the epitope profile recognized by the human antibody repertoire could guide future vaccine design.-Bianchi, F., Veggi, D., Santini, L., Buricchi, F., Bartolini, E., Lo Surdo, P., Martinelli, M., Finco, O., Masignani, V., Bottomley, M. J., Maione, D., Cozzi, R. Cocrystal structure of meningococcal factor H binding protein variant 3 reveals a new crossprotective epitope recognized by human mAb 1E6.


Asunto(s)
Anticuerpos Monoclonales/inmunología , Antígenos Bacterianos/inmunología , Proteínas Bacterianas/inmunología , Factor H de Complemento/inmunología , Epítopos/inmunología , Vacunas Meningococicas/inmunología , Animales , Anticuerpos Monoclonales/química , Anticuerpos Monoclonales/metabolismo , Antígenos Bacterianos/genética , Antígenos Bacterianos/metabolismo , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Factor H de Complemento/genética , Factor H de Complemento/metabolismo , Cristalografía por Rayos X , Epítopos/genética , Epítopos/metabolismo , Variación Genética , Humanos , Infecciones Meningocócicas/microbiología , Infecciones Meningocócicas/prevención & control , Vacunas Meningococicas/administración & dosificación , Modelos Moleculares , Neisseria meningitidis/efectos de los fármacos , Neisseria meningitidis/inmunología , Neisseria meningitidis/fisiología , Unión Proteica , Conformación Proteica
7.
PLoS One ; 13(8): e0201922, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30133484

RESUMEN

Neisserial heparin binding antigen (NHBA) is one of three main recombinant protein antigens in 4CMenB, a vaccine for the prevention of invasive meningococcal disease caused by Neisseria meningitidis serogroup B. NHBA is a surface-exposed lipoprotein composed of a predicted disordered N-terminal region, an arginine-rich region that binds heparin, and a C-terminal domain that folds as an anti-parallel ß-barrel and that upon release after cleavage by human proteases alters endothelial permeability. NHBA induces bactericidal antibodies in humans, and NHBA-specific antibodies elicited by the 4CMenB vaccine contribute to serum bactericidal activity, the correlate of protection. To better understand the structural bases of the human antibody response to 4CMenB vaccination and to inform antigen design, we used X-ray crystallography to elucidate the structures of two C-terminal fragments of NHBA, either alone or in complex with the Fab derived from the vaccine-elicited human monoclonal antibody 5H2, and the structure of the unbound Fab 5H2. The structures reveal details on the interaction between an N-terminal ß-hairpin fragment and the ß-barrel, and explain how NHBA is capable of generating cross-reactive antibodies through an extensive conserved conformational epitope that covers the entire C-terminal face of the ß-barrel. By providing new structural information on a vaccine antigen and on the human immune response to vaccination, these results deepen our molecular understanding of 4CMenB, and might also aid future vaccine design projects.


Asunto(s)
Antígenos Bacterianos/química , Antígenos Bacterianos/inmunología , Proteínas de la Membrana Bacteriana Externa/química , Proteínas de la Membrana Bacteriana Externa/inmunología , Proteínas Portadoras/química , Proteínas Portadoras/inmunología , Epítopos/química , Epítopos/inmunología , Anticuerpos Antibacterianos/inmunología , Vacunas Bacterianas/inmunología , Humanos , Modelos Moleculares , Conformación Proteica , Relación Estructura-Actividad
8.
Acta Crystallogr F Struct Biol Commun ; 73(Pt 6): 305-314, 2017 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-28580917

RESUMEN

Neisserial heparin-binding antigen (NHBA) is a surface-exposed lipoprotein from Neisseria meningitidis and is a component of the meningococcus B vaccine Bexsero. As part of a study to characterize the three-dimensional structure of NHBA and the molecular basis of the human immune response to Bexsero, the crystal structures of two fragment antigen-binding domains (Fabs) isolated from human monoclonal antibodies targeting NHBA were determined. Through a high-resolution analysis of the organization and the amino-acid composition of the CDRs, these structures provide broad insights into the NHBA epitopes recognized by the human immune system. As expected, these Fabs also show remarkable structural conservation, as shown by a structural comparison of 15 structures of apo Fab 10C3 which were obtained from crystals grown in different crystallization conditions and were solved while searching for a complex with a bound NHBA fragment or epitope peptide. This study also provides indirect evidence for the intrinsically disordered nature of two N-terminal regions of NHBA.


Asunto(s)
Anticuerpos Antibacterianos/química , Antígenos Bacterianos/química , Proteínas de la Membrana Bacteriana Externa/química , Proteínas Portadoras/química , Fragmentos Fab de Inmunoglobulinas/química , Vacunas Meningococicas/química , Neisseria meningitidis/química , Secuencia de Aminoácidos , Anticuerpos Antibacterianos/genética , Antígenos Bacterianos/genética , Antígenos Bacterianos/inmunología , Proteínas de la Membrana Bacteriana Externa/genética , Proteínas de la Membrana Bacteriana Externa/inmunología , Sitios de Unión , Proteínas Portadoras/genética , Proteínas Portadoras/inmunología , Clonación Molecular , Cristalografía por Rayos X , Escherichia coli/genética , Escherichia coli/metabolismo , Expresión Génica , Vectores Genéticos/química , Vectores Genéticos/metabolismo , Células HEK293 , Humanos , Fragmentos Fab de Inmunoglobulinas/genética , Proteínas Intrínsecamente Desordenadas/química , Proteínas Intrínsecamente Desordenadas/genética , Proteínas Intrínsecamente Desordenadas/inmunología , Cinética , Meningitis Meningocócica/inmunología , Meningitis Meningocócica/microbiología , Meningitis Meningocócica/prevención & control , Vacunas Meningococicas/inmunología , Modelos Moleculares , Neisseria meningitidis/inmunología , Péptidos/síntesis química , Péptidos/química , Unión Proteica , Conformación Proteica en Hélice alfa , Conformación Proteica en Lámina beta , Dominios y Motivos de Interacción de Proteínas , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/inmunología
9.
Acta Crystallogr F Struct Biol Commun ; 72(Pt 3): 214-9, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26919525

RESUMEN

Staphylococcus pseudintermedius is a leading cause of disease in dogs, and zoonosis causes human infections. Methicillin-resistant S. pseudintermedius strains are emerging, resembling the global health threat of S. aureus. Therefore, it is increasingly important to characterize potential targets for intervention against S. pseudintermedius. Here, FhuD, an S. pseudintermedius surface lipoprotein implicated in iron uptake, was characterized. It was found that FhuD bound ferrichrome in an iron-dependent manner, which increased the thermostability of FhuD by >15 °C. The crystal structure of ferrichrome-free FhuD was determined via molecular replacement at 1.6 Å resolution. FhuD exhibits the class III solute-binding protein (SBP) fold, with a ligand-binding cavity between the N- and C-terminal lobes, which is here occupied by a PEG molecule. The two lobes of FhuD were oriented in a closed conformation. These results provide the first detailed structural characterization of FhuD, a potential therapeutic target of S. pseudintermedius.


Asunto(s)
Proteínas Bacterianas/química , Proteínas Portadoras/química , Proteínas de Transporte de Membrana/química , Staphylococcus , Secuencia de Aminoácidos , Cristalización , Cristalografía por Rayos X , Ferricromo/química , Modelos Moleculares , Unión Proteica , Estabilidad Proteica , Sideróforos/química , Homología Estructural de Proteína
10.
PLoS One ; 10(8): e0135383, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26267900

RESUMEN

Antibodies (Ab) to neuraminidase (NA) play a role in limiting influenza infection and might help reduce the disease impact. The most widely used serological assay to measure functional anti-NA immune responses is the Enzyme-Linked Lectin Assay (ELLA) which relies on hemagglutinin (HA) mismatched virus reassortants, or detergent treated viruses as the NA source to overcome interference associated with steric hindrance of anti-HA Ab present in sera. The difficulty in producing and handling these reagents, which are not easily adapted for screening large numbers of samples, limits the routine analysis of functional anti-NA Ab in clinical trials. In this study, we produced influenza lentiviral pseudoparticles (PPs) containing only the NA antigen (NA-PPs) with a simple two-plasmid co-transfection system. NA-PPs were characterized and tested as an innovative source of NA in the NA inhibition (NI) assay. Both swine A/California/07/2009 (H1N1) and avian A/turkey/Turkey/01/2005 (H5N1) N1s within NA-PPs retained their sialidase activity and were specifically inhibited by homologous and N1 subtype-specific, heterologous sheep sera. Moreover, A/California/07/2009 N1-PPs were a better source of NA compared to whole live and detergent treated H1N1 viruses in ELLA, likely due to lack of interference by anti-HA Ab, and absence of possible structural modifications caused by treatment with detergent. This innovative assay is safer and applicable to all NAs. Taken together, these results highlight the potential of NA-PPs-based NI assays to be developed as sensitive, flexible, easy to handle and scalable serological tests for routine NA immune response analysis.


Asunto(s)
Anticuerpos Antivirales/inmunología , Técnicas para Inmunoenzimas/métodos , Virus de la Influenza A/inmunología , Neuraminidasa/inmunología , Proteínas Virales/inmunología , Animales , Células HEK293 , Humanos , Virus de la Influenza A/enzimología , Lectinas/química , Ratones , Ratones Endogámicos BALB C
11.
PLoS One ; 10(8): e0135474, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26280677

RESUMEN

Developing a universal influenza vaccine that induces broad spectrum and longer-term immunity has become an important potentially achievable target in influenza vaccine research and development. Hemagglutinin (HA) and neuraminidase (NA) are the two major influenza virus antigens. Although antibody responses against influenza virus are mainly directed toward HA, NA is reported to be more genetically stable; hence NA-based vaccines have the potential to be effective for longer time periods. NA-specific immunity has been shown to limit the spread of influenza virus, thus reducing disease symptoms and providing cross-protection against heterosubtypic viruses in mouse challenge experiments. The production of large quantities of highly pure and stable NA could be beneficial for the development of new antivirals, subunit-based vaccines, and novel diagnostic tools. In this study, recombinant NA (rNA) was produced in mammalian cells at high levels from both swine A/California/07/2009 (H1N1) and avian A/turkey/Turkey/01/2005 (H5N1) influenza viruses. Biochemical, structural, and immunological characterizations revealed that the soluble rNAs produced are tetrameric, enzymatically active and immunogenic, and finally they represent good alternatives to conventionally used sources of NA in the Enzyme-Linked Lectin Assay (ELLA).


Asunto(s)
Antígenos Virales/inmunología , Lectinas/inmunología , Neuraminidasa/inmunología , Proteínas Recombinantes/inmunología , Proteínas Virales/inmunología , Animales , Anticuerpos Antivirales/inmunología , Formación de Anticuerpos/inmunología , Aves , Línea Celular , Protección Cruzada/inmunología , Reacciones Cruzadas/inmunología , Ensayo de Immunospot Ligado a Enzimas/métodos , Femenino , Glicoproteínas Hemaglutininas del Virus de la Influenza/inmunología , Subtipo H1N1 del Virus de la Influenza A/inmunología , Subtipo H5N1 del Virus de la Influenza A/inmunología , Vacunas contra la Influenza/inmunología , Gripe Aviar/inmunología , Ratones , Infecciones por Orthomyxoviridae/inmunología , Porcinos
12.
FASEB J ; 29(11): 4629-40, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26202865

RESUMEN

Group B Streptococcus (GBS) expresses 3 structurally distinct pilus types (1, 2a, and 2b) identified as important virulence factors and vaccine targets. These pili are heterotrimeric polymers, covalently assembled on the cell wall by sortase (Srt) enzymes. We investigated the pilus-2b biogenesis mechanism by using a multidisciplinary approach integrating genetic, biochemical, and structural studies to dissect the role of the 2 pilus-2b-associated Srts. We show that only 1 sortase (SrtC1-2b) is responsible for pilus protein polymerization, whereas the second one (Srt2-2b) does not act as a pilin polymerase, but similarly to the housekeeping class A Srt (SrtA), it is involved in cell-wall pilus anchoring by targeting the minor ancillary subunit. Based on its function and sequence features, Srt2-2b does not belong to class C Srts (SrtCs), nor is it a canonical member of any other known family of Srts. We also report the crystal structure of SrtC1-2b at 1.9 Å resolution. The overall fold resembles the typical structure of SrtCs except for the N-terminal lid region that appears in an open conformation displaced from the active site. Our findings reveal that GBS pilus type 2b biogenesis differs significantly from the current model of pilus assembly in gram-positive pathogens.


Asunto(s)
Proteínas Bacterianas/química , Cisteína Endopeptidasas/química , Fimbrias Bacterianas/enzimología , Streptococcus agalactiae/enzimología , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Cristalografía por Rayos X , Cisteína Endopeptidasas/genética , Cisteína Endopeptidasas/metabolismo , Fimbrias Bacterianas/genética , Estructura Terciaria de Proteína , Streptococcus agalactiae/genética
13.
PLoS One ; 10(5): e0125875, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25942637

RESUMEN

Group B Streptococcus (GBS) is a major cause of invasive disease in infants. Like other Gram-positive bacteria, GBS uses a sortase C-catalyzed transpeptidation mechanism to generate cell surface pili from backbone and ancillary pilin precursor substrates. The three pilus types identified in GBS contain structural subunits that are highly immunogenic and are promising candidates for the development of a broadly-protective vaccine. Here we report the X-ray crystal structure of the backbone protein of pilus 2b (BP-2b) at 1.06Å resolution. The structure reveals a classical IgG-like fold typical of the pilin subunits of other Gram-positive bacteria. The crystallized portion of the protein (residues 185-468) encompasses domains D2 and D3 that together confer high stability to the protein due to the presence of an internal isopeptide bond within each domain. The D2+D3 region, lacking the N-terminal D1 domain, was as potent as the entire protein in conferring protection against GBS challenge in a well-established mouse model. By site-directed mutagenesis and complementation studies in GBS knock-out strains we identified the residues and motives essential for assembly of the BP-2b monomers into high-molecular weight complexes, thus providing new insights into pilus 2b polymerization.


Asunto(s)
Proteínas Bacterianas/química , Fimbrias Bacterianas/metabolismo , Modelos Moleculares , Conformación Proteica , Multimerización de Proteína , Streptococcus agalactiae/metabolismo , Secuencias de Aminoácidos , Animales , Proteínas Bacterianas/genética , Proteínas Bacterianas/inmunología , Proteínas Bacterianas/metabolismo , Sitios de Unión , Modelos Animales de Enfermedad , Femenino , Inmunización , Ratones , Unión Proteica , Subunidades de Proteína/química , Subunidades de Proteína/metabolismo , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/inmunología , Proteínas Recombinantes/metabolismo , Infecciones Estreptocócicas/prevención & control , Streptococcus agalactiae/genética , Streptococcus agalactiae/inmunología
14.
Mol Cell Proteomics ; 14(2): 418-29, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25368410

RESUMEN

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


Asunto(s)
Vacunas Bacterianas/inmunología , Biología Computacional/métodos , Proteoma/inmunología , 5'-Nucleotidasa/metabolismo , Animales , Proteínas Bacterianas/inmunología , Línea Celular , Modelos Animales de Enfermedad , Femenino , Ratones , Neisseria meningitidis Serogrupo B/inmunología , Señales de Clasificación de Proteína , Reproducibilidad de los Resultados , Staphylococcus aureus/inmunología , Streptococcus agalactiae/inmunología
15.
Biosci Rep ; 34(6): e00154, 2014 Nov 24.
Artículo en Inglés | MEDLINE | ID: mdl-25311310

RESUMEN

The Gram-positive bacterium Staphylococcus pseudintermedius is a leading cause of canine bacterial pyoderma, resulting in worldwide morbidity in dogs. S. pseudintermedius also causes life-threatening human infections. Furthermore, methicillin-resistant S. pseudintermedius is emerging, resembling the human health threat of methicillin-resistant Staphylococcus aureus. Therefore it is increasingly important to characterize targets for intervention strategies to counteract S. pseudintermedius infections. Here we used biophysical methods, mutagenesis, and X-ray crystallography, to define the ligand-binding properties and structure of SitA, an S. pseudintermedius surface lipoprotein. SitA was strongly and specifically stabilized by Mn2+ and Zn2+ ions. Crystal structures of SitA complexed with Mn2+ and Zn2+ revealed a canonical class III solute-binding protein with the metal cation bound in a cavity between N- and C-terminal lobes. Unexpectedly, one crystal contained both apo- and holo-forms of SitA, revealing a large side-chain reorientation of His64, and associated structural differences accompanying ligand binding. Such conformational changes may regulate fruitful engagement of the cognate ABC (ATP-binding cassette) transporter system (SitBC) required for metal uptake. These results provide the first detailed characterization and mechanistic insights for a potential therapeutic target of the major canine pathogen S. pseudintermedius, and also shed light on homologous structures in related staphylococcal pathogens afflicting humans.


Asunto(s)
Apolipoproteínas/metabolismo , Proteínas Bacterianas/metabolismo , Manganeso/metabolismo , Staphylococcus/metabolismo , Zinc/metabolismo , Secuencia de Aminoácidos , Animales , Apolipoproteínas/química , Apolipoproteínas/genética , Proteínas Bacterianas/química , Proteínas Bacterianas/genética , Rastreo Diferencial de Calorimetría , Dicroismo Circular , Cristalografía por Rayos X , Enfermedades de los Perros/microbiología , Perros , Humanos , Manganeso/química , Modelos Moleculares , Datos de Secuencia Molecular , Mutación Missense , Unión Proteica , Multimerización de Proteína , Estabilidad Proteica , Estructura Cuaternaria de Proteína , Estructura Terciaria de Proteína , Homología de Secuencia de Aminoácido , Infecciones Estafilocócicas/microbiología , Staphylococcus/genética , Staphylococcus/fisiología , Zinc/química
16.
Curr Top Med Chem ; 13(20): 2629-37, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24066888

RESUMEN

The Structural Vaccinology (SV) approach is the logical evolution of Reverse Vaccinology: a genome-based approach combined with structural biology, with the idea that protective determinants can be used to selectively engineer the antigens that can be re-designed and simplified for inclusion in vaccine combinations. The final objectives of the rational structure-based antigen optimization are the facilitation of industrial-scale production of the antigens combination, obtain a greater immunogenicity and a greater safety profile and finally expand the breadth of protection. Structural Vaccinology is particularly powerful in case of antigenic variation between closely related strains and species. Several examples are available in literature of how SV has already been applied successfully to several bacterial and viral projects. The examples of structure-based antigens optimization reviewed here describe different template procedures that can be followed to develop improved vaccines against other pathogens and potentially help resolve challenges in manufacturing or efficacy.


Asunto(s)
Vacunas/química , Animales , Antígenos/química , Antígenos/genética , Antígenos/inmunología , Humanos , Vacunas/genética , Vacunas/inmunología
17.
PLoS Comput Biol ; 9(6): e1003115, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23825940

RESUMEN

The pilus 2a backbone protein (BP-2a) is one of the most structurally and functionally characterized components of a potential vaccine formulation against Group B Streptococcus. It is characterized by six main immunologically distinct allelic variants, each inducing variant-specific protection. To investigate the molecular determinants driving the variant immunogenic specificity of BP-2a, in terms of single residue contributions, we generated six monoclonal antibodies against a specific protein variant based on their capability to recognize the polymerized pili structure on the bacterial surface. Three mAbs were also able to induce complement-dependent opsonophagocytosis killing of live GBS and target the same linear epitope present in the structurally defined and immunodominant domain D3 of the protein. Molecular docking between the modelled scFv antibody sequences and the BP-2a crystal structure revealed the potential role at the binding interface of some non-conserved antigen residues. Mutagenesis analysis confirmed the necessity of a perfect balance between charges, size and polarity at the binding interface to obtain specific binding of mAbs to the protein antigen for a neutralizing response.


Asunto(s)
Proteínas Bacterianas/metabolismo , Streptococcus agalactiae/metabolismo , Secuencia de Aminoácidos , Animales , Anticuerpos Monoclonales/inmunología , Proteínas Bacterianas/química , Proteínas Bacterianas/inmunología , Mapeo Epitopo , Ratones , Modelos Moleculares , Datos de Secuencia Molecular , Fagocitosis , Homología de Secuencia de Aminoácido , Streptococcus agalactiae/inmunología
18.
FASEB J ; 27(8): 3144-54, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23631841

RESUMEN

Gram-positive bacteria build pili on their cell surface via a class C sortase-catalyzed transpeptidation mechanism from pilin protein substrates. Despite the availability of several crystal structures, pilus-related C sortases remain poorly characterized to date, and their mechanisms of transpeptidation and regulation need to be further investigated. The available 3-dimensional structures of these enzymes reveal a typical sortase fold, except for the presence of a unique feature represented by an N-terminal highly flexible loop known as the "lid." This region interacts with the residues composing the catalytic triad and covers the active site, thus maintaining the enzyme in an autoinhibited state and preventing the accessibility to the substrate. It is believed that enzyme activation may occur only after lid displacement from the catalytic domain. In this work, we provide the first direct evidence of the regulatory role of the lid, demonstrating that it is possible to obtain in vitro an efficient polymerization of pilin subunits using an active C sortase lid mutant carrying a single residue mutation in the lid region. Moreover, biochemical analyses of this recombinant mutant reveal that the lid confers thermodynamic and proteolytic stability to the enzyme.


Asunto(s)
Aminoaciltransferasas/metabolismo , Proteínas Bacterianas/metabolismo , Cisteína Endopeptidasas/metabolismo , Fimbrias Bacterianas/enzimología , Streptococcus agalactiae/metabolismo , Secuencias de Aminoácidos , Secuencia de Aminoácidos , Aminoaciltransferasas/química , Aminoaciltransferasas/genética , Proteínas Bacterianas/química , Proteínas Bacterianas/genética , Biocatálisis , Western Blotting , Dominio Catalítico , Cisteína Endopeptidasas/química , Cisteína Endopeptidasas/genética , Proteínas Fimbrias/genética , Proteínas Fimbrias/metabolismo , Fluorometría , Cinética , Espectroscopía de Resonancia Magnética , Modelos Moleculares , Mutación , Filogenia , Polimerizacion , Pliegue de Proteína , Estructura Terciaria de Proteína , Proteolisis , Streptococcus agalactiae/genética
19.
PLoS One ; 7(11): e49048, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-23145064

RESUMEN

Gram-positive bacteria assemble pili through class C sortase enzymes specialized in polymerizing pilin subunits into covalently linked, high-molecular-weight, elongated structures. Here we report the crystal structures of two class C sortases (SrtC1 and SrtC2) from Group B Streptococcus (GBS) Pilus Island 1. The structures show that both sortases are comprised of two domains: an 8-stranded ß-barrel catalytic core conserved among all sortase family members and a flexible N-terminal region made of two α-helices followed by a loop, known as the lid, which acts as a pseudo-substrate. In vitro experiments performed with recombinant SrtC enzymes lacking the N-terminal portion demonstrate that this region of the enzyme is dispensable for catalysis but may have key roles in substrate specificity and regulation. Moreover, in vitro FRET-based assays show that the LPXTG motif common to many sortase substrates is not the sole determinant of sortase C specificity during pilin protein recognition.


Asunto(s)
Aminoaciltransferasas/química , Proteínas Bacterianas/química , Cisteína Endopeptidasas/química , Proteínas Fimbrias/química , Fimbrias Bacterianas/química , Streptococcus/enzimología , Catálisis , Dominio Catalítico , Cristalografía por Rayos X/métodos , Modelos Moleculares , Pliegue de Proteína , Estructura Terciaria de Proteína , Streptococcus/química , Especificidad por Sustrato
20.
FASEB J ; 26(5): 2008-18, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-22253480

RESUMEN

Group B Streptococcus pili are covalently linked structures assembled via a sortase-catalyzed transpeptidation mechanism involving specific residues and motifs. A sequence element containing a conserved glutamic acid, called the E-box, has been described to be involved in pilus formation. Although it is known that the glutamic acid is involved in stabilizing the internal isopeptide bonds, its role in pilus assembly still needs to be investigated. Using site-specific mutagenesis and complementation studies of knockout strains, we found that the E-box glutamic residue of the backbone and the major ancillary proteins is essential for pilus protein polymerization. NMR analysis revealed that the mutation of this residue seriously affected the folding of the protein. By contrast, the mutation of the lysine involved in the same isopeptide bond did not engender a structural destabilization, and the native fold was preserved. Moreover, molecular dynamics simulations on the E-box-containing domain of the backbone protein showed that the E-box glutamic acid is necessary to maintain the appropriate dryness of the domain core and that its mutation favors an unfolded state. The data provide the first direct evidence that the E-box has an additional and key role in maintaining the correct protein fold independently of isopeptide bond formation.


Asunto(s)
Fimbrias Bacterianas/fisiología , Ácido Glutámico/fisiología , Streptococcus agalactiae/fisiología , Western Blotting , Simulación de Dinámica Molecular , Resonancia Magnética Nuclear Biomolecular
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...