Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 39
Filter
Add more filters










Publication year range
1.
Mol Cell Proteomics ; 23(3): 100734, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38342408

ABSTRACT

Antigen-antibody interactions play a key role in the immune response post vaccination and the mechanism of action of antibody-based biopharmaceuticals. 4CMenB is a multicomponent vaccine against Neisseria meningitidis serogroup B in which factor H binding protein (fHbp) is one of the key antigens. In this study, we use hydrogen/deuterium exchange mass spectrometry (HDX-MS) to identify epitopes in fHbp recognized by polyclonal antibodies (pAb) from two human donors (HDs) vaccinated with 4CMenB. Our HDX-MS data reveal several epitopes recognized by the complex mixture of human pAb. Furthermore, we show that the pAb from the two HDs recognize the same epitope regions. Epitope mapping of total pAb and purified fHbp-specific pAb from the same HD reveals that the two antibody samples recognize the same main epitopes, showing that HDX-MS based epitope mapping can, in this case at least, be performed directly using total IgG pAb samples that have not undergone Ab-selective purification. Two monoclonal antibodies (mAb) were previously produced from B-cell repertoire sequences from one of the HDs and used for epitope mapping of fHbp with HDX-MS. The epitopes identified for the pAb from the same HD in this study, overlap with the epitopes recognized by the two individual mAbs. Overall, HDX-MS epitope mapping appears highly suitable for simultaneous identification of epitopes recognized by pAb from human donors and to thus both guide vaccine development and study basic human immunity to pathogens, including viruses.


Subject(s)
Meningococcal Infections , Meningococcal Vaccines , Neisseria meningitidis , Humans , Epitope Mapping/methods , Neisseria meningitidis/metabolism , Deuterium/metabolism , Bacterial Proteins/metabolism , Meningococcal Infections/prevention & control , Carrier Proteins , Deuterium Exchange Measurement , Complement Factor H , Antigens, Bacterial , Epitopes , Antibodies, Monoclonal/metabolism , Hydrogen Deuterium Exchange-Mass Spectrometry
2.
FASEB J ; 38(1): e23326, 2024 01.
Article in English | MEDLINE | ID: mdl-38019196

ABSTRACT

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.


Subject(s)
Membrane Proteins , Vaccines , Epitopes/genetics , Antigens, Bacterial/genetics , Lipid Bilayers
3.
Int J Mol Sci ; 24(7)2023 Mar 24.
Article in English | MEDLINE | ID: mdl-37047152

ABSTRACT

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.


Subject(s)
Nanoparticles , Neisseria meningitidis , Mice , Animals , Ferritins , Antigens, Bacterial , Antigens, Viral , Antibodies, Blocking , Vaccines, Combined , Nanoparticles/chemistry
4.
Comput Struct Biotechnol J ; 20: 2070-2081, 2022.
Article in English | MEDLINE | ID: mdl-35601959

ABSTRACT

Invasive meningococcal disease can cause fatal sepsis and meningitis and is a global health threat. Factor H binding protein (fHbp) is a protective antigen included in the two currently available vaccines against serogroup B meningococcus (MenB). FHbp is a remarkably variable surface-exposed meningococcal virulence factor with over 1300 different amino acid sequences identified so far. Based on this variability, fHbp has been classified into three variants, two subfamilies or nine modular groups, with low degrees of cross-protective activity. Here, we report the crystal structure of a natural fHbp cross-variant chimera, named variant1-2,3.x expressed by the MenB clinical isolate NL096, at 1.2 Å resolution, the highest resolution of any fHbp structure reported to date. We combined biochemical, site-directed mutagenesis and computational biophysics studies to deeply characterize this rare chimera. We determined the structure to be composed of two adjacent domains deriving from the three variants and determined the molecular basis of its stability, ability to bind Factor H and to adopt the canonical three-dimensional fHbp structure. These studies guided the design of loss-of-function mutations with potential for even greater immunogenicity. Moreover, this study represents a further step in the understanding of the fHbp biological and immunological evolution in nature. The chimeric variant1-2,3.x fHbp protein emerges as an intriguing cross-protective immunogen and suggests that identification of such naturally occurring hybrid proteins may result in stable and cross-protective immunogens when seeking to design and develop vaccines against highly variable pathogens.

5.
Chem Sci ; 13(8): 2440-2449, 2022 Feb 23.
Article in English | MEDLINE | ID: mdl-35310500

ABSTRACT

The introduction of glycoconjugate vaccines marks an important point in the fight against various infectious diseases. The covalent conjugation of relevant polysaccharide antigens to immunogenic carrier proteins enables the induction of a long-lasting and robust IgG antibody response, which is not observed for pure polysaccharide vaccines. Although there has been remarkable progress in the development of glycoconjugate vaccines, many crucial parameters remain poorly understood. In particular, the influence of the conjugation site and strategy on the immunogenic properties of the final glycoconjugate vaccine is the focus of intense research. Here, we present a comparison of two cysteine selective conjugation strategies, elucidating the impact of both modifications on the structural integrity of the carrier protein, as well as on the immunogenic properties of the resulting glycoconjugate vaccine candidates. Our work suggests that conjugation chemistries impairing structurally relevant elements of the protein carrier, such as disulfide bonds, can have a dramatic effect on protein immunogenicity.

6.
J Control Release ; 330: 933-944, 2021 02 10.
Article in English | MEDLINE | ID: mdl-33152394

ABSTRACT

A range of cationic delivery systems have been investigated as vaccine adjuvants, though few direct comparisons exist. To investigate the impact of the delivery platform, we prepared four cationic systems (emulsions, liposomes, polymeric nanoparticles and solid lipid nanoparticles) all containing equal concentrations of the cationic lipid dimethyldioctadecylammonium bromide in combination with the Neisseria adhesin A variant 3 subunit antigen. The formulations were physicochemically characterized and their ability to associate with cells and promote antigen processing (based on degradation of DQ-OVA, a substrate for proteases which upon hydrolysis is fluorescent) was compared in vitro and their vaccine efficacy (antigen-specific antibody responses and IFN-γ production) and biodistribution (antigen and adjuvant) were evaluated in vivo. Due to their cationic nature, all delivery systems gave high antigen loading (> 85%) with liposomes, lipid nanoparticles and emulsions being <200 nm, whilst polymeric nanoparticles were larger (~350 nm). In vitro, the particulate systems tended to promote cell uptake and antigen processing, whilst emulsions were less effective. Similarly, whilst the particulate delivery systems induced a depot (of both delivery system and antigen) at the injection site, the cationic emulsions did not. However, out of the systems tested the cationic emulsions induced the highest antibody responses. These results demonstrate that while cationic lipids can have strong adjuvant activity, their formulation platform influences their immunogenicity.


Subject(s)
Antibody Formation , Vaccines , Adjuvants, Immunologic , Antigens , Liposomes , Tissue Distribution , Vaccines, Subunit
7.
PLoS Pathog ; 16(10): e1008882, 2020 10.
Article in English | MEDLINE | ID: mdl-33007046

ABSTRACT

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.


Subject(s)
Antibodies/immunology , Antigens, Bacterial/metabolism , Bacterial Proteins/metabolism , Meningitis, Meningococcal/immunology , Meningococcal Vaccines/administration & dosage , Neisseria meningitidis/immunology , Adult , Antibodies/blood , Antigens, Bacterial/immunology , Bacterial Proteins/immunology , Complement Factor H/immunology , Complement Factor H/metabolism , Humans , Meningitis, Meningococcal/metabolism , Meningitis, Meningococcal/microbiology , Meningitis, Meningococcal/prevention & control
8.
FASEB J ; 34(8): 10329-10341, 2020 08.
Article in English | MEDLINE | ID: mdl-32725956

ABSTRACT

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.


Subject(s)
Antibodies, Bacterial/immunology , Antibodies, Monoclonal/immunology , Complement System Proteins/immunology , Antigens, Bacterial/immunology , Bacterial Outer Membrane Proteins/immunology , Bacterial Proteins/immunology , Carrier Proteins/immunology , Complement Factor H/immunology , Epitopes/immunology , Neisseria meningitidis/immunology , Serum Bactericidal Antibody Assay/methods
9.
FASEB J ; 33(11): 12099-12111, 2019 11.
Article in English | MEDLINE | ID: mdl-31442074

ABSTRACT

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.


Subject(s)
Antibodies, Monoclonal/immunology , Antigens, Bacterial/immunology , Bacterial Proteins/immunology , Complement Factor H/immunology , Epitopes/immunology , Meningococcal Vaccines/immunology , Animals , Antibodies, Monoclonal/chemistry , Antibodies, Monoclonal/metabolism , Antigens, Bacterial/genetics , Antigens, Bacterial/metabolism , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Complement Factor H/genetics , Complement Factor H/metabolism , Crystallography, X-Ray , Epitopes/genetics , Epitopes/metabolism , Genetic Variation , Humans , Meningococcal Infections/microbiology , Meningococcal Infections/prevention & control , Meningococcal Vaccines/administration & dosage , Models, Molecular , Neisseria meningitidis/drug effects , Neisseria meningitidis/immunology , Neisseria meningitidis/physiology , Protein Binding , Protein Conformation
10.
PLoS One ; 13(8): e0201922, 2018.
Article in English | MEDLINE | ID: mdl-30133484

ABSTRACT

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.


Subject(s)
Antigens, Bacterial/chemistry , Antigens, Bacterial/immunology , Bacterial Outer Membrane Proteins/chemistry , Bacterial Outer Membrane Proteins/immunology , Carrier Proteins/chemistry , Carrier Proteins/immunology , Epitopes/chemistry , Epitopes/immunology , Antibodies, Bacterial/immunology , Bacterial Vaccines/immunology , Humans , Models, Molecular , Protein Conformation , Structure-Activity Relationship
11.
J Proteome Res ; 17(5): 1794-1800, 2018 05 04.
Article in English | MEDLINE | ID: mdl-29619829

ABSTRACT

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.


Subject(s)
Bacterial Proteins/chemistry , Deuterium Exchange Measurement/methods , Mass Spectrometry/methods , Porins/chemistry , Bacterial Outer Membrane Proteins/chemistry , Escherichia coli Proteins/chemistry , Kinetics , Protein Conformation
12.
Acta Crystallogr F Struct Biol Commun ; 73(Pt 6): 305-314, 2017 06 01.
Article in English | MEDLINE | ID: mdl-28580917

ABSTRACT

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.


Subject(s)
Antibodies, Bacterial/chemistry , Antigens, Bacterial/chemistry , Bacterial Outer Membrane Proteins/chemistry , Carrier Proteins/chemistry , Immunoglobulin Fab Fragments/chemistry , Meningococcal Vaccines/chemistry , Neisseria meningitidis/chemistry , Amino Acid Sequence , Antibodies, Bacterial/genetics , Antigens, Bacterial/genetics , Antigens, Bacterial/immunology , Bacterial Outer Membrane Proteins/genetics , Bacterial Outer Membrane Proteins/immunology , Binding Sites , Carrier Proteins/genetics , Carrier Proteins/immunology , Cloning, Molecular , Crystallography, X-Ray , Escherichia coli/genetics , Escherichia coli/metabolism , Gene Expression , Genetic Vectors/chemistry , Genetic Vectors/metabolism , HEK293 Cells , Humans , Immunoglobulin Fab Fragments/genetics , Intrinsically Disordered Proteins/chemistry , Intrinsically Disordered Proteins/genetics , Intrinsically Disordered Proteins/immunology , Kinetics , Meningitis, Meningococcal/immunology , Meningitis, Meningococcal/microbiology , Meningitis, Meningococcal/prevention & control , Meningococcal Vaccines/immunology , Models, Molecular , Neisseria meningitidis/immunology , Peptides/chemical synthesis , Peptides/chemistry , Protein Binding , Protein Conformation, alpha-Helical , Protein Conformation, beta-Strand , Protein Interaction Domains and Motifs , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/immunology
13.
Proc Natl Acad Sci U S A ; 114(19): 5017-5022, 2017 05 09.
Article in English | MEDLINE | ID: mdl-28439022

ABSTRACT

Despite substantial progress in the prevention of group B Streptococcus (GBS) disease with the introduction of intrapartum antibiotic prophylaxis, this pathogen remains a leading cause of neonatal infection. Capsular polysaccharide conjugate vaccines have been tested in phase I/II clinical studies, showing promise for further development. Mapping of epitopes recognized by protective antibodies is crucial for understanding the mechanism of action of vaccines and for enabling antigen design. In this study, we report the structure of the epitope recognized by a monoclonal antibody with opsonophagocytic activity and representative of the protective response against type III GBS polysaccharide. The structure and the atomic-level interactions were determined by saturation transfer difference (STD)-NMR and X-ray crystallography using oligosaccharides obtained by synthetic and depolymerization procedures. The GBS PSIII epitope is made by six sugars. Four of them derive from two adjacent repeating units of the PSIII backbone and two of them from the branched galactose-sialic acid disaccharide contained in this sequence. The sialic acid residue establishes direct binding interactions with the functional antibody. The crystal structure provides insight into the molecular basis of antibody-carbohydrate interactions and confirms that the conformational epitope is not required for antigen recognition. Understanding the structural basis of immune recognition of capsular polysaccharide epitopes can aid in the design of novel glycoconjugate vaccines.


Subject(s)
Bacterial Capsules/chemistry , Epitopes/chemistry , Oligosaccharides/chemistry , Polysaccharides, Bacterial/chemistry , Streptococcus agalactiae/chemistry , Animals , Carbohydrate Conformation , Crystallography, X-Ray , Mice , Rabbits
14.
Biochem J ; 473(24): 4699-4713, 2016 Dec 15.
Article in English | MEDLINE | ID: mdl-27784765

ABSTRACT

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.


Subject(s)
Antibodies, Monoclonal/metabolism , Antigens, Bacterial/immunology , Antigens, Bacterial/metabolism , Bacterial Proteins/immunology , Bacterial Proteins/metabolism , Neisseria meningitidis/metabolism , Antibodies, Monoclonal/chemistry , Antibodies, Monoclonal/immunology , Antigens, Bacterial/chemistry , Bacterial Proteins/chemistry , Complement Factor H/immunology , Complement Factor H/metabolism , Epitopes/immunology , Epitopes/metabolism , Immunoglobulin Fab Fragments/immunology , Immunoglobulin Fab Fragments/metabolism , Immunoglobulin G/immunology , Immunoglobulin G/metabolism , Protein Binding , Protein Structure, Secondary
15.
Clin Vaccine Immunol ; 22(7): 769-77, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25947148

ABSTRACT

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.


Subject(s)
Neisseria gonorrhoeae/genetics , Neisseria gonorrhoeae/immunology , Neisseria meningitidis/immunology , Protein Engineering , Virulence Factors/genetics , Animals , Antibodies, Bacterial/blood , Antigens, Bacterial/genetics , Bacterial Proteins/genetics , Bacterial Vaccines/administration & dosage , Bacterial Vaccines/immunology , Blood Bactericidal Activity , Mice , Neisseria meningitidis/genetics , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/immunology , Sequence Homology, Amino Acid , Vaccines, Synthetic/administration & dosage , Vaccines, Synthetic/immunology
16.
FASEB J ; 29(6): 2260-7, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25713028

ABSTRACT

Bexsero, a new vaccine against Neisseria meningitidis serogroup B (MenB), is composed of 3 main recombinant proteins and an outer membrane vesicle component. One of the main bactericidal antigens, neisseria heparin binding antigen (NHBA), is present as a fusion protein with the accessory protein genome-derived neisserial antigen (GNA) 1030 to further increase its immunogenicity. The gene encoding for GNA1030 is present and highly conserved in all Neisseria strains, and although orthologs are present in numerous species, its biologic function is unknown. Native mass spectrometry was used to demonstrate that GNA1030 forms a homodimer associated with 2 molecules of ubiquinone-8 (Ub8), a cofactor mainly involved in the electron transport chain and with antioxidant properties. Disc diffusion assays on the wild-type and knockout mutant of GNA1030, in the presence of various compounds, suggested that GNA1030 is not involved in oxidative stress or electron chain transport per se, although it contributes to constitutive refilling of the inner membrane with Ub8. These studies shed light on an accessory protein present in Bexsero and reveal functional insights into the family of related proteins. On the basis of our findings, we propose to name the protein neisseria ubiquinone binding protein (NUbp).


Subject(s)
Antigens, Bacterial/metabolism , Bacterial Proteins/metabolism , Neisseria meningitidis/metabolism , Ubiquinone/metabolism , Amino Acid Sequence , Anti-Bacterial Agents/pharmacology , Antigens, Bacterial/chemistry , Antigens, Bacterial/genetics , Antimycin A/pharmacology , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Blotting, Western , Cell Survival/drug effects , Cell Survival/genetics , Cloning, Molecular , Disulfides/metabolism , Electron Transport Complex III/antagonists & inhibitors , Electron Transport Complex III/metabolism , Hydrogen Peroxide/pharmacology , Mass Spectrometry/methods , Meningococcal Vaccines/metabolism , Methacrylates/pharmacology , Molecular Sequence Data , Mutation , Neisseria meningitidis/genetics , Neisseria meningitidis/growth & development , Oxidants/pharmacology , Periplasmic Proteins/chemistry , Periplasmic Proteins/genetics , Periplasmic Proteins/metabolism , Protein Binding , Protein Multimerization , Thiazoles/pharmacology
17.
Proc Natl Acad Sci U S A ; 111(48): 17128-33, 2014 Dec 02.
Article in English | MEDLINE | ID: mdl-25404323

ABSTRACT

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.


Subject(s)
Adhesins, Bacterial/immunology , Antibodies, Bacterial/immunology , Antigens, Bacterial/immunology , Epitope Mapping/methods , Meningococcal Vaccines/immunology , Neisseria meningitidis, Serogroup B/immunology , Adhesins, Bacterial/chemistry , Adhesins, Bacterial/genetics , Amino Acid Sequence , Antigens, Bacterial/chemistry , Antigens, Bacterial/genetics , Binding Sites, Antibody/genetics , Binding Sites, Antibody/immunology , Crystallography, X-Ray , Deuterium Exchange Measurement , Microscopy, Electron, Transmission , Models, Molecular , Molecular Sequence Data , Neisseria meningitidis, Serogroup B/genetics , Protein Isoforms/chemistry , Protein Isoforms/genetics , Protein Isoforms/immunology , Protein Multimerization , Protein Stability , Protein Structure, Tertiary , Sequence Homology, Amino Acid , Spectrometry, Mass, Electrospray Ionization , Temperature
18.
FASEB J ; 27(12): 4723-30, 2013 Dec.
Article in English | MEDLINE | ID: mdl-23964075

ABSTRACT

NarE is an arginine-specific mono-ADP-ribosyltransferase identified in Neisseria meningitidis that requires the presence of iron in a structured cluster for its enzymatic activities. In this study, we show that NarE can perform auto-ADP-ribosylation. This automodification occurred in a time- and NAD-concentration-dependent manner; was inhibited by novobiocin, an ADP-ribosyltransferase inhibitor; and did not occur when NarE was heat inactivated. No reduction in incorporation was evidenced in the presence of high concentrations of ATP, GTP, ADP-ribose, or nicotinamide, which inhibits NAD-glycohydrolase, impeding the formation of free ADP-ribose. Based on the electrophoretic profile of NarE on auto-ADP-ribosylation and on the results of mutagenesis and mass spectrometry analysis, the auto-ADP-ribosylation appeared to be restricted to the addition of a single ADP-ribose. Chemical stability experiments showed that the ADP-ribosyl linkage was sensitive to hydroxylamine, which breaks ADP-ribose-arginine bonds. Site-directed mutagenesis suggested that the auto-ADP-ribosylation site occurred preferentially on the R(7) residue, which is located in the region I of the ADP-ribosyltransferase family. After auto-ADP-ribosylation, NarE showed a reduction in ADP-ribosyltransferase activity, while NAD-glycohydrolase activity was increased. Overall, our findings provide evidence for a novel intramolecular mechanism used by NarE to regulate its enzymatic activities.


Subject(s)
ADP Ribose Transferases/metabolism , Adenosine Diphosphate Ribose/metabolism , Catalytic Domain , Neisseria meningitidis/enzymology , ADP Ribose Transferases/chemistry , ADP Ribose Transferases/genetics , Amino Acid Motifs , Amino Acid Sequence , Molecular Sequence Data , Mutation , NAD+ Nucleosidase/metabolism , Protein Stability
19.
Infect Immun ; 81(8): 2851-60, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23716610

ABSTRACT

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.


Subject(s)
Bacterial Proteins/immunology , Bacterial Toxins/immunology , Bacterial Vaccines/immunology , Clostridium Infections/immunology , Enterotoxins/immunology , Animals , Antibodies, Bacterial/immunology , Antibodies, Neutralizing/immunology , Antigens, Bacterial/immunology , Clostridioides difficile/immunology , Clostridium Infections/prevention & control , Cricetinae , Disease Models, Animal , Electrophoresis, Polyacrylamide Gel , Enzyme-Linked Immunosorbent Assay , Humans , Immunoblotting , Mice , Recombinant Proteins/immunology
20.
Proc Natl Acad Sci U S A ; 110(9): 3304-9, 2013 Feb 26.
Article in English | MEDLINE | ID: mdl-23396847

ABSTRACT

Mapping of epitopes recognized by functional monoclonal antibodies (mAbs) is essential for understanding the nature of immune responses and designing improved vaccines, therapeutics, and diagnostics. In recent years, identification of B-cell epitopes targeted by neutralizing antibodies has facilitated the design of peptide-based vaccines against highly variable pathogens like HIV, respiratory syncytial virus, and Helicobacter pylori; however, none of these products has yet progressed into clinical stages. Linear epitopes identified by conventional mapping techniques only partially reflect the immunogenic properties of the epitope in its natural conformation, thus limiting the success of this approach. To investigate antigen-antibody interactions and assess the potential of the most common epitope mapping techniques, we generated a series of mAbs against factor H binding protein (fHbp), a key virulence factor and vaccine antigen of Neisseria meningitidis. The interaction of fHbp with the bactericidal mAb 12C1 was studied by various epitope mapping methods. Although a 12-residue epitope in the C terminus of fHbp was identified by both Peptide Scanning and Phage Display Library screening, other approaches, such as hydrogen/deuterium exchange mass spectrometry (MS) and X-ray crystallography, showed that mAb 12C1 occupies an area of ∼1,000 Å(2) on fHbp, including >20 fHbp residues distributed on both N- and C-terminal domains. Collectively, these data show that linear epitope mapping techniques provide useful but incomplete descriptions of B-cell epitopes, indicating that increased efforts to fully characterize antigen-antibody interfaces are required to understand and design effective immunogens.


Subject(s)
Antigens, Bacterial/immunology , Bacterial Proteins/immunology , Epitopes/immunology , Meningococcal Vaccines/immunology , Neisseria meningitidis/immunology , Neisseria meningitidis/pathogenicity , Virulence Factors/immunology , Antibodies, Monoclonal/immunology , Antibody Specificity/immunology , Antigens, Bacterial/chemistry , Bacterial Proteins/chemistry , Cell Surface Display Techniques , Crystallography, X-Ray , Deuterium Exchange Measurement , Epitope Mapping , Epitopes/chemistry , Mass Spectrometry , Meningococcal Infections/immunology , Meningococcal Infections/microbiology , Meningococcal Infections/prevention & control , Models, Molecular , Peptides/chemistry , Peptides/immunology , Protein Binding/immunology , Surface Plasmon Resonance , Virulence Factors/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...