Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 53
Filter
Add more filters










Publication year range
1.
FEBS J ; 289(3): 730-747, 2022 02.
Article in English | MEDLINE | ID: mdl-34499806

ABSTRACT

Specific antibody interactions with short peptides have made epitope tagging systems a vital tool employed in virtually all fields of biological research. Here, we present a novel epitope tagging system comprised of a monoclonal antibody named GD-26, which recognises the TD peptide (GTGATPADD) derived from Haloarcula marismortui bacteriorhodopsin I (HmBRI) D94N mutant. The crystal structure of the antigen-binding fragment (Fab) of GD-26 complexed with the TD peptide was determined to a resolution of 1.45 Å. The TD peptide was found to adopt a 310 helix conformation within the binding cleft, providing a characteristic peptide structure for recognition by GD-26 Fab. Based on the structure information, polar and nonpolar forces collectively contribute to the strong binding. Attempts to engineer the TD peptide show that the proline residue is crucial for the formation of the 310 helix in order to fit into the binding cleft. Isothermal calorimetry (ITC) reported a dissociation constant KD of 12 ± 2.8 nm, indicating a strong interaction between the TD peptide and GD-26 Fab. High specificity of GD-26 IgG to the TD peptide was demonstrated by western blotting, ELISA and immunofluorescence as only TD-tagged proteins were detected, suggesting the effectiveness of the GD-26/TD peptide tagging system. In addition to already-existing epitope tags such as the FLAG tag and the ALFA tag adopting either extended or α-helix conformations, the unique 310 helix conformation of the TD peptide together with the corresponding monoclonal antibody GD-26 offers a novel tagging option for research.


Subject(s)
Antibodies, Monoclonal/immunology , Bacteriorhodopsins/immunology , Epitopes/immunology , Peptides/immunology , Antibodies, Monoclonal/genetics , Antibodies, Monoclonal/ultrastructure , Antibody Specificity/genetics , Bacteriorhodopsins/genetics , Bacteriorhodopsins/ultrastructure , Crystallography, X-Ray , Enzyme-Linked Immunosorbent Assay , Epitopes/genetics , Epitopes/ultrastructure , Haloarcula marismortui/immunology , Immunoglobulin Fab Fragments/genetics , Immunoglobulin Fab Fragments/immunology , Immunoglobulin Fab Fragments/ultrastructure , Peptides/genetics
2.
Sci Rep ; 11(1): 15374, 2021 07 28.
Article in English | MEDLINE | ID: mdl-34321560

ABSTRACT

Mayaro virus (MAYV), which causes mayaro fever, is endemic to limited regions of South America that may expand due to the possible involvement of Aedes spp. mosquitoes in its transmission. Its effective control will require the accurate identification of infected individuals, which has been restricted to nucleic acid-based tests due to similarities with other emerging members of the Alphavirus genus of the Togaviridae family; both in structure and clinical symptoms. Serological tests have a more significant potential to expand testing at a reasonable cost, and their performance primarily reflects that of the antigen utilized to capture pathogen-specific antibodies. Here, we describe the assembly of a synthetic gene encoding multiple copies of antigenic determinants mapped from the nsP1, nsP2, E1, and E2 proteins of MAYV that readily expressed as a stable chimeric protein in bacteria. Its serological performance as the target in ELISAs revealed a high accuracy for detecting anti-MAYV IgM antibodies. No cross-reactivity was observed with serum from seropositive individuals for dengue, chikungunya, yellow fever, Zika, and other infectious diseases as well as healthy individuals. Our data suggest that this bioengineered antigen could be used to develop high-performance serological tests for MAYV infections.


Subject(s)
Alphavirus Infections/diagnosis , Alphavirus/immunology , Epitopes/immunology , Togaviridae Infections/diagnosis , Aedes/virology , Alphavirus/pathogenicity , Alphavirus Infections/immunology , Alphavirus Infections/transmission , Alphavirus Infections/virology , Animals , Enzyme-Linked Immunosorbent Assay , Epitopes/genetics , Epitopes/ultrastructure , Female , Genes, Synthetic/genetics , Genes, Synthetic/immunology , Humans , Immunoglobulin M/immunology , Male , Serologic Tests , South America/epidemiology , Togaviridae/isolation & purification , Togaviridae/pathogenicity , Togaviridae Infections/immunology , Togaviridae Infections/transmission , Togaviridae Infections/virology
3.
Nat Commun ; 11(1): 4421, 2020 09 04.
Article in English | MEDLINE | ID: mdl-32887891

ABSTRACT

Receptor usage that determines cell tropism and drives viral classification closely correlates with the virus structure. Enterovirus B (EV-B) consists of several subgroups according to receptor usage, among which echovirus 30 (E30), a leading causative agent for human aseptic meningitis, utilizes FcRn as an uncoating receptor. However, receptors for many EVs remain unknown. Here we analyzed the atomic structures of E30 mature virion, empty- and A-particles, which reveals serotype-specific epitopes and striking conformational differences between the subgroups within EV-Bs. Of these, the VP1 BC loop markedly distinguishes E30 from other EV-Bs, indicative of a role as a structural marker for EV-B. By obtaining cryo-electron microscopy structures of E30 in complex with its receptor FcRn and CD55 and comparing its homologs, we deciphered the underlying molecular basis for receptor recognition. Together with experimentally derived viral receptor identifications, we developed a structure-based in silico algorithm to inform a rational prediction for EV receptor usage.


Subject(s)
Antigen-Antibody Complex/ultrastructure , Enterovirus B, Human/ultrastructure , Antigens, Viral/ultrastructure , CD55 Antigens/immunology , Cryoelectron Microscopy , Enterovirus B, Human/immunology , Epitopes/ultrastructure , Humans , Receptors, Fc/immunology , Virion/ultrastructure
4.
Nat Commun ; 11(1): 4419, 2020 09 04.
Article in English | MEDLINE | ID: mdl-32887892

ABSTRACT

Echovirus 30 (E30), a serotype of Enterovirus B (EV-B), recently emerged as a major causative agent of aseptic meningitis worldwide. E30 is particularly devastating in the neonatal population and currently no vaccine or antiviral therapy is available. Here we characterize two highly potent E30-specific monoclonal antibodies, 6C5 and 4B10, which efficiently block binding of the virus to its attachment receptor CD55 and uncoating receptor FcRn. Combinations of 6C5 and 4B10 augment the sum of their individual anti-viral activities. High-resolution structures of E30-6C5-Fab and E30-4B10-Fab define the location and nature of epitopes targeted by the antibodies. 6C5 and 4B10 engage the capsid loci at the north rim of the canyon and in-canyon, respectively. Notably, these regions exhibit antigenic variability across EV-Bs, highlighting challenges in development of broad-spectrum antibodies. Our structures of these neutralizing antibodies of E30 are instructive for development of vaccines and therapeutics against EV-B infections.


Subject(s)
Antibodies, Neutralizing/ultrastructure , Antigen-Antibody Complex/ultrastructure , Capsid Proteins/immunology , Enterovirus B, Human/immunology , Animals , Antibodies, Monoclonal/ultrastructure , Antigens, Viral , CD55 Antigens/immunology , Cryoelectron Microscopy , Epitopes/ultrastructure , Humans , Meningitis, Aseptic/virology , Mice , Receptors, Fc/immunology , Serogroup
5.
Nat Commun ; 11(1): 3114, 2020 06 19.
Article in English | MEDLINE | ID: mdl-32561744

ABSTRACT

Revealing antibody-antigen interactions at the single-molecule level will deepen our understanding of immunology. However, structural determination under crystal or cryogenic conditions does not provide temporal resolution for resolving transient, physiologically or pathologically relevant functional antibody-antigen complexes. Here, we develop a triangular DNA origami framework with site-specifically anchored and spatially organized artificial epitopes to capture transient conformations of immunoglobulin Gs (IgGs) at room temperature. The DNA origami epitopes (DOEs) allows programmed spatial distribution of epitope spikes, which enables direct imaging of functional complexes with atomic force microscopy (AFM). We establish the critical dependence of the IgG avidity on the lateral distance of epitopes within 3-20 nm at the single-molecule level. High-speed AFM imaging of transient conformations further provides structural and dynamic evidence for the IgG avidity from monovalent to bivalent in a single event, which sheds light on various applications including virus neutralization, diagnostic detection and cancer immunotherapy.


Subject(s)
Antibody Affinity , Epitopes/ultrastructure , Immunoglobulin G/ultrastructure , Molecular Probes/ultrastructure , Single Molecule Imaging/methods , Antigen-Antibody Complex/ultrastructure , DNA, Single-Stranded/immunology , DNA, Single-Stranded/metabolism , DNA, Single-Stranded/ultrastructure , Epitopes/immunology , Epitopes/metabolism , Fluorescence Resonance Energy Transfer/methods , Humans , Immunoglobulin G/immunology , Immunoglobulin G/metabolism , Microscopy, Atomic Force/methods , Molecular Dynamics Simulation , Molecular Probes/immunology , Molecular Probes/metabolism , Nanotechnology , Structure-Activity Relationship
6.
J Struct Biol ; 211(1): 107512, 2020 07 01.
Article in English | MEDLINE | ID: mdl-32325220

ABSTRACT

Dipeptidase 3 (DPEP3) is one of three glycosylphosphatidylinositol-anchored metallopeptidases potentially involved in the hydrolytic metabolism of dipeptides. While its exact biological function is not clear, DPEP3 expression is normally limited to testis, but can be elevated in ovarian cancer. Antibody drug conjugates targeting DPEP3 have shown efficacy in preclinical models with a pyrrolobenzodiazepine conjugate, SC-003, dosed in a phase I clinical trial (NCT02539719). Here we reveal the novel atomic structure of DPEP3 alone and in complex with the SC-003 Fab fragment at 1.8 and 2.8 Å, respectively. The structure of DPEP3/SC-003 Fab complex reveals an eighteen-residue epitope across the DPEP3 dimerization interface distinct from the enzymatic active site. DPEP1 and DPEP3 extracellular domains share a conserved, dimeric TIM (ß/α)8-barrel fold, consistent with 49% sequence identity. However, DPEP3 diverges from DPEP1 and DPEP2 in key positions of its active site: a histidine to tyrosine variation at position 269 reduces affinity for the ß zinc and may cause substrate steric hindrance, whereas an aspartate to asparagine change at position 359 abolishes activation of the nucleophilic water/hydroxide, resulting in no in vitro activity against a variety of dipeptides and biological substrates (imipenem, leukotriene D4 and cystinyl-bis-glycine). Hence DPEP3, unlike DPEP1 and DPEP2, may require an activating co-factor in vivo or may remain an inactive, degenerate enzyme. This report sheds light on the structural discriminants between active and inactive membrane dipeptidases and provides a benchmark to characterize current and future DPEP3-targeted therapeutic approaches.


Subject(s)
Dipeptidases/ultrastructure , Epitopes/ultrastructure , Immunoconjugates/ultrastructure , Antibodies/chemistry , Antibodies/immunology , Antibodies/ultrastructure , Dipeptidases/chemistry , Dipeptidases/genetics , Dipeptidases/immunology , Epitopes/genetics , Epitopes/immunology , Humans , Immunoconjugates/genetics , Immunoconjugates/immunology , Immunoglobulin Fab Fragments/genetics , Immunoglobulin Fab Fragments/immunology , Immunoglobulin Fab Fragments/ultrastructure , Membrane Proteins/immunology , Membrane Proteins/ultrastructure , Proteolysis
7.
Virol Sin ; 35(1): 1-13, 2020 Feb.
Article in English | MEDLINE | ID: mdl-31916022

ABSTRACT

Antibodies play critical roles in neutralizing viral infections and are increasingly used as therapeutic drugs and diagnostic tools. Structural studies on virus-antibody immune complexes are important for better understanding the molecular mechanisms of antibody-mediated neutralization and also provide valuable information for structure-based vaccine design. Cryo-electron microscopy (cryo-EM) has recently matured as a powerful structural technique for studying bio-macromolecular complexes. When combined with X-ray crystallography, cryo-EM provides a routine approach for structurally characterizing the immune complexes formed between icosahedral viruses and their antibodies. In this review, recent advances in the structural understanding of virus-antibody interactions are outlined for whole virions with icosahedral T = pseudo 3 (picornaviruses) and T = 3 (flaviviruses) architectures, focusing on the dynamic nature of viral shells in different functional states. Glycoprotein complexes from pleomorphic enveloped viruses are also discussed as immune complex antigens. Improving our understanding of viral epitope structures using virus-based platforms would provide a fundamental road map for future vaccine development.


Subject(s)
Antibodies, Viral/ultrastructure , Antigen-Antibody Complex/ultrastructure , Cryoelectron Microscopy , Virion/ultrastructure , Animals , Antibodies, Viral/immunology , Epitopes/immunology , Epitopes/ultrastructure , Flavivirus/immunology , Flavivirus/ultrastructure , Humans , Picornaviridae/immunology , Picornaviridae/ultrastructure , Protein Binding , Protein Conformation , Virion/immunology
8.
Drug Discov Today Technol ; 35-36: 23-33, 2020 Dec.
Article in English | MEDLINE | ID: mdl-33388125

ABSTRACT

Microbial surface polysaccharides are important virulence factors and targets for vaccine development. Glycoconjugate vaccines, obtained by covalently linking carbohydrates and proteins, are well established tools for prevention of bacterial infections. Elucidation of the minimal portion involved in the interactions with functional antibodies is of utmost importance for the understanding of their mechanism of induction of protective immune responses and the design of synthetic glycan based vaccines. Typically, this is achieved by combination of different techniques, which include ELISA, glycoarray, Surface Plasmon Resonance in conjunction with approaches for mapping at atomic level the position involved in binding, such as Saturation Transfer NMR and X-ray crystallography. This review provides an overview of the structural studies performed to map glycan epitopes (glycotopes), with focus on the highly complex structure of Group B Streptococcus type III (GBSIII) capsular polysaccharide. Furthermore, it describes the rational process followed to translate the obtained information into the design of a protective glycoconjugate vaccine based on a well-defined synthetic glycan epitope.


Subject(s)
Polysaccharides, Bacterial/administration & dosage , Streptococcal Infections/prevention & control , Streptococcal Vaccines/administration & dosage , Streptococcus agalactiae/immunology , Animals , Crystallography, X-Ray , Disease Models, Animal , Drug Design , Epitopes/administration & dosage , Epitopes/immunology , Epitopes/ultrastructure , Glycoconjugates/administration & dosage , Glycoconjugates/chemistry , Glycoconjugates/immunology , Humans , Immunogenicity, Vaccine , Polysaccharides, Bacterial/immunology , Polysaccharides, Bacterial/ultrastructure , Streptococcal Infections/immunology , Streptococcal Infections/microbiology , Streptococcal Vaccines/chemical synthesis , Streptococcal Vaccines/immunology , Structure-Activity Relationship , Vaccines, Synthetic/administration & dosage , Vaccines, Synthetic/chemistry , Vaccines, Synthetic/immunology
9.
Drug Discov Today Technol ; 35-36: 45-56, 2020 Dec.
Article in English | MEDLINE | ID: mdl-33388127

ABSTRACT

An effective prophylactic HIV-1 vaccine is essential in order to contain the HIV/AIDS global pandemic. The discovery of different broadly neutralizing antibodies (bnAbs) in the last decades has enabled the characterization of several minimal epitopes on the HIV envelope (Env) spike, including glycan-dependent fragments. Herein, we provide a brief overview of the progress made on the development of synthetic carbohydrate-based epitope mimics for the elicitation of bnAbs directed to certain regions on Env gp120 protein: the outer domain high-mannose cluster and the variable loops V1V2 and V3. We focus on the design, synthesis and biological evaluation of minimal immunogens and discuss key aspects towards the development of a successful protective vaccine against HIV-1.


Subject(s)
AIDS Vaccines/administration & dosage , Antibodies, Viral/immunology , Broadly Neutralizing Antibodies/immunology , HIV Infections/prevention & control , HIV-1/immunology , AIDS Vaccines/chemical synthesis , AIDS Vaccines/immunology , Animals , Antibodies, Viral/metabolism , Broadly Neutralizing Antibodies/metabolism , Disease Models, Animal , Drug Design , Epitopes/immunology , Epitopes/metabolism , Epitopes/ultrastructure , HIV Envelope Protein gp120/immunology , HIV Envelope Protein gp120/metabolism , HIV Envelope Protein gp120/ultrastructure , HIV Infections/immunology , HIV Infections/virology , HIV-1/ultrastructure , Humans , Immunogenicity, Vaccine , Macaca , Mannose/chemistry , Mannose/immunology , Protein Domains/immunology , Vaccines, Synthetic/administration & dosage , Vaccines, Synthetic/chemistry , Vaccines, Synthetic/immunology
10.
Cell Host Microbe ; 26(6): 729-738.e4, 2019 12 11.
Article in English | MEDLINE | ID: mdl-31757767

ABSTRACT

Influenza virus neuraminidase (NA) is a major target for small-molecule antiviral drugs. Antibodies targeting the NA surface antigen could also inhibit virus entry and egress to provide host protection. However, our understanding of the nature and range of target epitopes is limited because of a lack of human antibody structures with influenza neuraminidase. Here, we describe crystal and cryogenic electron microscopy (cryo-EM) structures of NAs from human-infecting avian H7N9 viruses in complex with five human anti-N9 antibodies, systematically defining several antigenic sites and antibody epitope footprints. These antibodies either fully or partially block the NA active site or bind to epitopes distant from the active site while still showing neuraminidase inhibition. The inhibition of antibodies to NAs was further analyzed by glycan array and solution-based NA activity assays. Together, these structural studies provide insights into protection by anti-NA antibodies and templates for the development of NA-based influenza virus vaccines and therapeutics.


Subject(s)
Antibodies, Neutralizing , Antibodies, Viral , Epitopes/ultrastructure , Neuraminidase , Orthomyxoviridae Infections/drug therapy , Animals , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/ultrastructure , Antibodies, Neutralizing/immunology , Antibodies, Neutralizing/ultrastructure , Antibodies, Viral/immunology , Antibodies, Viral/ultrastructure , Antiviral Agents/immunology , Cryoelectron Microscopy , Epitopes/immunology , Epitopes/metabolism , Humans , Influenza A Virus, H7N9 Subtype/immunology , Influenza Vaccines , Neuraminidase/chemistry , Neuraminidase/ultrastructure , Orthomyxoviridae Infections/prevention & control , Viral Proteins/chemistry , Viral Proteins/ultrastructure
11.
IUBMB Life ; 71(10): 1522-1536, 2019 10.
Article in English | MEDLINE | ID: mdl-31185142

ABSTRACT

Acetaldehyde is a metabolite of ethanol, an important constituent of tobacco pyrolysis and the aldehydic product of lipid peroxidation. Acetaldehyde induced toxicity is mainly due to its binding to cellular macromolecules resulting in the formation of stable adducts accompanied by oxidative stress. The aim of this study was to characterize structural and immunological alterations in human immunoglobulin G (IgG) modified with acetaldehyde in the presence of sodium borohydride, a reducing agent. The IgG modifications were studied by various physicochemical techniques such as fluorescence and CD spectroscopy, free amino group estimation, 2,2-azobis 2-amidinopropane (AAPH) induced red blood cell hemolysis as well as transmission electron microscopy. Molecular docking was also employed to predict the preferential binding of acetaldehyde to IgG. The immunogenicity of native and acetaldehyde-modified IgG was investigated by immunizing female New Zealand white rabbits using native and modified IgG as antigens. Binding specificity and cross reactivity of rabbit antibodies was screened by competitive inhibition ELISA and band shift assays. The modification of human IgG with acetaldehyde results in quenching of the fluorescence of tyrosine residues, decrease in free amino group content, a change in the antioxidant property as well as formation of cross-linked structures in human IgG. Molecular docking reveals strong binding of IgG to acetaldehyde. Moreover, acetaldehyde modified IgG induced high titer antibodies (>1:12800) in the experimental animals. The antibodies exhibited high specificity in competitive binding assay toward acetaldehyde modified human IgG. The results indicate that acetaldehyde induces alterations in secondary and tertiary structure of IgG molecule that leads to formation of neo-epitopes on IgG that enhances its immunogenicity.


Subject(s)
Acetaldehyde/chemistry , Epitopes/ultrastructure , Immunoglobulin G/ultrastructure , Protein Conformation , Animals , Binding Sites/immunology , Enzyme-Linked Immunosorbent Assay , Epitopes/immunology , Erythrocytes/immunology , Female , Hemolysis/immunology , Humans , Immunoglobulin G/chemistry , Immunoglobulin G/immunology , Microscopy, Electron, Transmission , Molecular Docking Simulation , Oxidative Stress/immunology , Protein Binding/immunology , Rabbits , Tyrosine/immunology
12.
Sci Rep ; 9(1): 4933, 2019 03 20.
Article in English | MEDLINE | ID: mdl-30894561

ABSTRACT

Group 21 and 5 allergens are homologous house dust mite proteins known as mid-tier allergens. To reveal the biological function of group 21 allergens and to understand better the allergenicity of the rDer f 21 allergen, we determined the 1.5 Å crystal structure of rDer f 21 allergen from Dermatophagoides farinae. The rDer f 21 protein consists of a three helical bundle, similar to available structures of group 21 and homologous group 5 allergens. The rDer f 21 dimer forms a hydrophobic binding pocket similar to the one in the Der p 5 allergen, which indicates that both of the homologous groups could share a similar function. By performing structure-guided mutagenesis, we mutated all 38 surface-exposed polar residues of the rDer f 21 allergen and carried out immuno-dot blot assays using 24 atopic sera. Six residues, K10, K26, K42, E43, K46, and K48, which are located in the region between the N-terminus and the loop 1 of rDer f 21 were identified as the major IgE epitopes of rDer f 21. Epitope mapping of all potential IgE epitopes on the surface of the rDer f 21 crystal structure revealed heterogeneity in the sIgE recognition of the allergen epitopes in atopic individuals. The higher the allergen-sIgE level of an individual, the higher the number of epitope residues that are found in the allergen. The results illustrate the clear correlation between the number of specific major epitope residues in an allergen and the sIgE level of the atopic population.


Subject(s)
Antigens, Dermatophagoides/immunology , Arthropod Proteins/immunology , Dermatophagoides farinae/immunology , Epitopes/ultrastructure , Hypersensitivity, Immediate/immunology , Immunoglobulin E/blood , Animals , Antigens, Dermatophagoides/metabolism , Antigens, Dermatophagoides/ultrastructure , Arthropod Proteins/metabolism , Arthropod Proteins/ultrastructure , Crystallography, X-Ray , Epitope Mapping , Epitopes/immunology , Epitopes/metabolism , Humans , Hypersensitivity, Immediate/blood , Immunoglobulin E/immunology
13.
J Struct Biol ; 203(3): 236-241, 2018 09.
Article in English | MEDLINE | ID: mdl-29775653

ABSTRACT

AAV2.5 represents the first structure-guided in-silico designed Adeno-associated virus (AAV) gene delivery vector. This engineered vector combined the receptor attachment properties of AAV serotype 2 (AAV2) with the muscle tropic properties of AAV1, and exhibited an antibody escape phenotype because of a modified antigenic epitope. To confirm the design, the structure of the vector was determined to a resolution of 2.78 Šusing cryo-electron microscopy and image reconstruction. The structure of the major viral protein (VP), VP3, was ordered from residue 219 to 736, as reported for other AAV structures, and the five AAV2.5 residues exchanged from AAV2 to AAV1, Q263A, T265 (insertion), N706A, V709A, and T717N, were readily interpretable. Significantly, the surface loops containing these residues adopt the AAV1 conformation indicating the importance of amino acid residues in dictating VP structure.


Subject(s)
Cryoelectron Microscopy/methods , Gene Transfer Techniques , Genetic Vectors/ultrastructure , Parvovirinae/ultrastructure , Capsid/chemistry , Capsid/ultrastructure , Capsid Proteins/chemistry , Capsid Proteins/ultrastructure , Dependovirus , Epitopes/chemistry , Epitopes/ultrastructure , Genetic Therapy , Genetic Vectors/chemistry , Genetic Vectors/genetics , Humans , Parvovirinae/chemistry , Parvovirinae/genetics , Protein Binding
14.
J Struct Biol ; 194(2): 191-8, 2016 May.
Article in English | MEDLINE | ID: mdl-26876146

ABSTRACT

The cholesteryl ester transfer protein (CETP) enables the transfer of cholesteryl ester (CE) from high-density lipoproteins (HDL) to low-density lipoproteins (LDL) in the plasma compartment. CETP inhibition raises plasma levels of HDL cholesterol; a ternary tunnel complex with CETP bridging HDL and LDL was suggested as a mechanism. Here, we test whether the inhibition of CETP tunnel complex formation is a promising approach to suppress CE transfer from HDL to LDL, for potential treatment of cardio-vascular disease (CVD). Three monoclonal antibodies against different epitopes of CETP are assayed for their potential to interfere with CE transfer between HDL and/or LDL. Surprisingly, antibodies that target the tips of the elongated CETP molecule, interaction sites sterically required to form the suggested transfer complexes, do not interfere with CETP activity, but an antibody binding to the central region does. We show that CETP interacts with HDL, but not with LDL. Our findings demonstrate that a ternary tunnel complex is not the mechanistic prerequisite to transfer CE among lipoproteins.


Subject(s)
Cholesterol Ester Transfer Proteins/metabolism , Cholesterol Esters/metabolism , Epitopes/chemistry , Lipoproteins, HDL/metabolism , Lipoproteins, LDL/metabolism , Antibodies, Monoclonal/chemistry , Antibodies, Monoclonal/isolation & purification , Biological Transport , Cell Line , Cholesterol Ester Transfer Proteins/genetics , Cholesterol Ester Transfer Proteins/ultrastructure , Epitopes/ultrastructure , Gene Expression , Humans , Lipoproteins, HDL/ultrastructure , Lipoproteins, LDL/ultrastructure , Microscopy, Electron, Transmission , Protein Binding , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Recombinant Proteins/ultrastructure
15.
Plant Biotechnol J ; 14(1): 231-9, 2016 Jan.
Article in English | MEDLINE | ID: mdl-25879277

ABSTRACT

We describe a novel strategy to produce vaccine antigens using a plant cell-suspension culture system in lieu of the conventional bacterial or animal cell-culture systems. We generated transgenic cell-suspension cultures from Nicotiana benthamiana leaves carrying wild-type or chimeric Bamboo mosaic virus (BaMV) expression constructs encoding the viral protein 1 (VP1) epitope of foot-and-mouth disease virus (FMDV). Antigens accumulated to high levels in BdT38 and BdT19 transgenic cell lines co-expressing silencing suppressor protein P38 or P19. BaMV chimeric virus particles (CVPs) were subsequently purified from the respective cell lines (1.5 and 2.1 mg CVPs/20 g fresh weight of suspended biomass, respectively), and the resulting CVPs displayed VP1 epitope on the surfaces. Guinea pigs vaccinated with purified CVPs produced humoral antibodies. This study represents an important advance in the large-scale production of immunopeptide vaccines in a cost-effective manner using a plant cell-suspension culture system.


Subject(s)
Chimera/metabolism , Epitopes/metabolism , Nicotiana/genetics , Plant Cells/metabolism , Potexvirus/physiology , Virion/metabolism , Animals , Antibody Specificity/immunology , Epitopes/immunology , Epitopes/ultrastructure , Guinea Pigs , Immunization , Plants, Genetically Modified , Recombination, Genetic/genetics , Suspensions , Nicotiana/cytology , Nicotiana/virology , Virion/ultrastructure
16.
Virology ; 485: 199-204, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26295280

ABSTRACT

Human norovirus is a dominant cause of acute gastroenteritis around the world. Several norovirus disinfectants label citric acid as an active ingredient. In this study, we showed that norovirus virus-like particles (VLPs) treated with citrate buffer caused the particles to alter their morphology, including increased diameters associated with a new ring-like structure. We also found that epitopes on the protruding (P) domain on these particles were more readily accessible to antibodies after the citrate treatment. These results suggested that citrate had a direct effect on the norovirus particles. Using X-ray crystallography, we showed that the P domain bound citrate from lemon juice and a disinfectant containing citric acid. Importantly, citrate binds at the histo-blood group antigen binding pocket, which are attachment factors for norovirus infections. Taken together, these new findings suggested that it might be possible to treat/reduce norovirus infections with citrate, although further studies are needed.


Subject(s)
Antibodies, Viral/chemistry , Capsid Proteins/chemistry , Citric Acid/chemistry , Epitopes/chemistry , Norovirus/chemistry , Virion/chemistry , Baculoviridae/genetics , Blood Group Antigens/chemistry , Capsid Proteins/ultrastructure , Crystallography, X-Ray , Disinfectants/chemistry , Epitopes/ultrastructure , Gene Expression , Microscopy, Electron , Models, Molecular , Norovirus/ultrastructure , Protein Binding , Protein Structure, Tertiary , Recombinant Proteins/chemistry , Recombinant Proteins/ultrastructure , Single-Domain Antibodies/chemistry , Virion/ultrastructure
17.
J Mol Biol ; 427(16): 2617-28, 2015 Aug 14.
Article in English | MEDLINE | ID: mdl-26135247

ABSTRACT

Hepatitis C virus (HCV) is a positive-strand RNA virus within the Flaviviridae family. The viral "spike" of HCV is formed by two envelope glycoproteins, E1 and E2, which together mediate viral entry by engaging host receptors and undergoing conformational changes to facilitate membrane fusion. While E2 can be readily produced in the absence of E1, E1 cannot be expressed without E2 and few reagents, including monoclonal antibodies (mAbs), are available for study of this essential HCV glycoprotein. A human mAb to E1, IGH526, was previously reported to cross-neutralize different HCV isolates, and therefore, we sought to further characterize the IGH526 neutralizing epitope to obtain information for vaccine design. We found that mAb IGH526 bound to a discontinuous epitope, but with a major component corresponding to E1 residues 314-324. The crystal structure of IGH526 Fab with this E1 glycopeptide at 1.75Å resolution revealed that the antibody binds to one face of an α-helical peptide. Single mutations on the helix substantially lowered IGH526 binding but did not affect neutralization, indicating either that multiple mutations are required or that additional regions are recognized by the antibody in the context of the membrane-associated envelope oligomer. Molecular dynamics simulations indicate that the free peptide is flexible in solution, suggesting that it requires stabilization for use as a candidate vaccine immunogen.


Subject(s)
Epitopes/ultrastructure , Hepatitis C Antibodies/immunology , Immunoglobulin Fab Fragments/immunology , Viral Envelope Proteins/immunology , Viral Envelope Proteins/ultrastructure , Antibodies, Monoclonal/immunology , Antibodies, Neutralizing/immunology , Binding Sites, Antibody , Cell Line , Crystallography, X-Ray , Epitope Mapping , Epitopes/immunology , HEK293 Cells , Hepacivirus/immunology , Humans , Molecular Dynamics Simulation
18.
Nat Nanotechnol ; 10(5): 472-9, 2015 May.
Article in English | MEDLINE | ID: mdl-25822932

ABSTRACT

Nanoparticles in a biological milieu are known to form a sufficiently long-lived and well-organized 'corona' of biomolecules to confer a biological identity to the particle. Because this nanoparticle-biomolecule complex interacts with cells and biological barriers, potentially engaging with different biological pathways, it is important to clarify the presentation of functional biomolecular motifs at its interface. Here, we demonstrate that by using antibody-labelled gold nanoparticles, differential centrifugal sedimentation and various imaging techniques it is possible to identify the spatial location of proteins, their functional motifs and their binding sites. We show that for transferrin-coated polystyrene nanoparticles only a minority of adsorbed proteins exhibit functional motifs and the spatial organization appears random, which is consistent, overall, with a stochastic and irreversible adsorption process. Our methods are applicable to a wide array of nanoparticles and can offer a microscopic molecular description of the biological identity of nanoparticles.


Subject(s)
Binding Sites/physiology , Epitopes/ultrastructure , Metal Nanoparticles/chemistry , Proteins/ultrastructure , Epitopes/chemistry , Gold/chemistry , Gold/metabolism , Humans , Immunohistochemistry , Nanotechnology , Polystyrenes/chemistry , Protein Binding , Proteins/chemistry , Receptors, Transferrin , Transferrin
19.
Mol Immunol ; 59(2): 200-7, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24667070

ABSTRACT

Bla g 1 is a major allergen from Blatella germanica and one of the primary allergens used to assess cockroach allergen exposure. The epitope of an anti-Bla g 1 scFv was mapped in order to better understand cross reactivity with other group 1 cockroach allergens and patient IgE epitopes. X-ray crystallography was used to determine the structure of the scFv. The scFv epitope on Bla g 1 was located by alanine scanning site-directed mutagenesis and ELISA. Twenty-six rBla g 1-GST alanine mutants were evaluated for variations in binding to the scFv compared to the wild type allergen. Six mutants showed a significant difference in scFv binding affinity. These mutations clustered to form a discontinuous epitope mainly comprising two helices of Bla g 1. The allergen-scFv complex was modeled based on the results, and the epitope region was found to have low sequence similarity with Per a 1, especially among the residues identified as functionally important for the scFv binding to Bla g 1. Indeed, the scFv failed to bind Per a 1 in American cockroach extract. The scFv was unable to inhibit the binding of IgE antibodies from a highly cockroach allergic patient to Bla g 1. Based on the surface area of Bla g 1 occluded by the scFv, putative regions of patient IgE-Bla g 1 interactions can be inferred. This scFv could be best utilized as a capture antibody in an IgE detection ELISA, or to differentiate Bla g 1 from Per a 1 in environmental exposure assays.


Subject(s)
Allergens/immunology , Cockroaches/immunology , Single-Chain Antibodies/immunology , Single-Chain Antibodies/ultrastructure , Allergens/genetics , Animals , Binding Sites, Antibody/immunology , Cross Reactions/immunology , Crystallography, X-Ray , Epitope Mapping , Epitopes/immunology , Epitopes/ultrastructure , Humans , Immunoglobulin E/immunology , Models, Molecular , Mutation
SELECTION OF CITATIONS
SEARCH DETAIL
...