Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
Curr Opin Virol ; 11: 98-102, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25837466

ABSTRACT

The development of accurate tools for predicting B-cell epitopes is important but difficult. Traditional methods have examined which regions in an antigen are likely binding sites of an antibody. However, it is becoming increasingly clear that most antigen surface residues will be able to bind one or more of the myriad of possible antibodies. In recent years, new approaches have emerged for predicting an epitope for a specific antibody, utilizing information encoded in antibody sequence or structure. Applying such antibody-specific predictions to groups of antibodies in combination with easily obtainable experimental data improves the performance of epitope predictions. We expect that further advances of such tools will be possible with the integration of immunoglobulin repertoire sequencing data.


Subject(s)
Antibodies/immunology , Computational Biology/methods , Epitopes, B-Lymphocyte/immunology , Immunoglobulins/genetics , Animals , Humans , Immunoglobulins/chemistry , Sequence Analysis, DNA
2.
Bioinformatics ; 31(8): 1313-5, 2015 Apr 15.
Article in English | MEDLINE | ID: mdl-25432167

ABSTRACT

UNLABELLED: Antibody epitope mapping is a key step in understanding antibody-antigen recognition and is of particular interest for drug development, diagnostics and vaccine design. Most computational methods for epitope prediction are based on properties of the antigen sequence and/or structure, not taking into account the antibody for which the epitope is predicted. Here, we introduce PEASE, a web server predicting antibody-specific epitopes, utilizing the sequence of the antibody. The predictions are provided both at the residue level and as patches on the antigen structure. The tradeoff between recall and precision can be tuned by the user, by changing the default parameters. The results are provided as text and HTML files as well as a graph, and can be viewed on the antigen 3D structure. AVAILABILITY AND IMPLEMENTATION: PEASE is freely available on the web at www.ofranlab.org/PEASE. CONTACT: yanay@ofranlab.org.


Subject(s)
Algorithms , Antibodies/chemistry , Antigens/chemistry , Epitope Mapping/methods , Epitopes, B-Lymphocyte/chemistry , Internet , Antibodies/metabolism , Artificial Intelligence , Complementarity Determining Regions/genetics , Epitopes, B-Lymphocyte/metabolism , Humans , Protein Conformation
3.
J Virol ; 88(19): 11339-55, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25031354

ABSTRACT

UNLABELLED: Vaccinia virus (VACV) L1 is an important target for viral neutralization and has been included in multicomponent DNA or protein vaccines against orthopoxviruses. To further understand the protective mechanism of the anti-L1 antibodies, we generated five murine anti-L1 monoclonal antibodies (MAbs), which clustered into 3 distinct epitope groups. While two groups of anti-L1 failed to neutralize, one group of 3 MAbs potently neutralized VACV in an isotype- and complement-independent manner. This is in contrast to neutralizing antibodies against major VACV envelope proteins, such as H3, D8, or A27, which failed to completely neutralize VACV unless the antibodies are of complement-fixing isotypes and complement is present. Compared to nonneutralizing anti-L1 MAbs, the neutralization antibodies bound to the recombinant L1 protein with a significantly higher affinity and also could bind to virions. By using a variety of techniques, including the isolation of neutralization escape mutants, hydrogen/deuterium exchange mass spectrometry, and X-ray crystallography, the epitope of the neutralizing antibodies was mapped to a conformational epitope with Asp35 as the key residue. This epitope is similar to the epitope of 7D11, a previously described potent VACV neutralizing antibody. The epitope was recognized mainly by CDR1 and CDR2 of the heavy chain, which are highly conserved among antibodies recognizing the epitope. These antibodies, however, had divergent light-chain and heavy-chain CDR3 sequences. Our study demonstrates that the conformational L1 epitope with Asp35 is a common site of vulnerability for potent neutralization by a divergent group of antibodies. IMPORTANCE: Vaccinia virus, the live vaccine for smallpox, is one of the most successful vaccines in human history, but it presents a level of risk that has become unacceptable for the current population. Studying the immune protection mechanism of smallpox vaccine is important for understanding the basic principle of successful vaccines and the development of next-generation, safer vaccines for highly pathogenic orthopoxviruses. We studied antibody targets in smallpox vaccine by developing potent neutralizing antibodies against vaccinia virus and comprehensively characterizing their epitopes. We found a site in vaccinia virus L1 protein as the target of a group of highly potent murine neutralizing antibodies. The analysis of antibody-antigen complex structure and the sequences of the antibody genes shed light on how these potent neutralizing antibodies are elicited from immunized mice.


Subject(s)
Antibodies, Monoclonal/chemistry , Antibodies, Neutralizing/chemistry , Antibodies, Viral/chemistry , Vaccinia virus/immunology , Vaccinia/immunology , Viral Envelope Proteins/chemistry , Amino Acid Sequence , Animals , Antigens, Viral , Epitopes/chemistry , Epitopes/immunology , Female , Mice , Mice, Inbred BALB C , Models, Molecular , Molecular Sequence Data , Neutralization Tests , Protein Interaction Domains and Motifs , Survival Analysis , Vaccination , Vaccinia/mortality , Vaccinia/prevention & control , Vaccinia/virology , Vaccinia virus/chemistry , Viral Envelope Proteins/administration & dosage , Viral Envelope Proteins/immunology , Virion/chemistry , Virion/immunology
4.
Structure ; 22(4): 646-57, 2014 Apr 08.
Article in English | MEDLINE | ID: mdl-24631463

ABSTRACT

Antibody epitope mapping is crucial for understanding B cell-mediated immunity and required for characterizing therapeutic antibodies. In contrast to T cell epitope mapping, no computational tools are in widespread use for prediction of B cell epitopes. Here, we show that, utilizing the sequence of an antibody, it is possible to identify discontinuous epitopes on its cognate antigen. The predictions are based on residue-pairing preferences and other interface characteristics. We combined these antibody-specific predictions with results of cross-blocking experiments that identify groups of antibodies with overlapping epitopes to improve the predictions. We validate the high performance of this approach by mapping the epitopes of a set of antibodies against the previously uncharacterized D8 antigen, using complementary techniques to reduce method-specific biases (X-ray crystallography, peptide ELISA, deuterium exchange, and site-directed mutagenesis). These results suggest that antibody-specific computational predictions and simple cross-blocking experiments allow for accurate prediction of residues in conformational B cell epitopes.


Subject(s)
Antibodies, Monoclonal/chemistry , Antigen-Antibody Complex/chemistry , Antigens, Viral/chemistry , Epitopes, B-Lymphocyte/chemistry , Peptides/chemistry , Amino Acid Sequence , Animals , Antibodies, Monoclonal/immunology , Antigen-Antibody Complex/immunology , Antigens, Viral/immunology , Crystallography, X-Ray , Deuterium Exchange Measurement , Enzyme-Linked Immunosorbent Assay , Epitope Mapping , Epitopes, B-Lymphocyte/immunology , Humans , Mice , Models, Molecular , Molecular Sequence Data , Mutagenesis, Site-Directed , Peptides/immunology , Vaccinia virus/chemistry
5.
FEBS J ; 281(1): 306-19, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24279419

ABSTRACT

The principles of affinity maturation of antibodies (Abs), which underlies B cell-mediated immunity, are still under debate. It is unclear whether the antigen (Ag) binding site is a preferred target for mutations, and what the role of activation-induced deaminase (AID) hotspots is in this process. Here we report a structural analysis of 3495 residues that have been replaced through somatic hypermutations (SHMs) in 196 Abs. We show that there is no correlation between the propensity of an amino acid to be in AID hotspot and the probability that it is replaced during the SHM process. Although AID hotspots may be necessary to enable SHMs, they are not a major driving force in determining which residues are mutated. We identified Ab positions that are highly mutated and significantly affect binding. The effect of mutation on binding energy is a major factor in determining which structural regions of the Ab are mutated. There is a clear preference for mutations at the Ag-binding site. However, positions outside this region that also affect binding are often preferred targets for SHMs. As for amino acid preferences, a general trend during SHM is to make Ab-Ag interfaces more similar to protein-protein interfaces in general. In different regions of the Ab, there are different sets of preferences for amino acid substitution. This mapping improves our understanding of Ab affinity maturation and may assist in Ab engineering.


Subject(s)
Amino Acids/metabolism , Antibodies/genetics , Cytidine Deaminase/metabolism , Germ-Line Mutation/genetics , Somatic Hypermutation, Immunoglobulin/genetics , Amino Acid Sequence , Amino Acid Substitution , Animals , Antibody Affinity , Humans , Immunoglobulin Heavy Chains/genetics , Immunoglobulin Heavy Chains/metabolism , Immunoglobulin Light Chains/genetics , Immunoglobulin Light Chains/metabolism , Immunoglobulin Variable Region/genetics , Immunoglobulin Variable Region/metabolism , Mice , Molecular Sequence Data , Sequence Homology, Amino Acid
6.
Front Immunol ; 4: 302, 2013 Oct 08.
Article in English | MEDLINE | ID: mdl-24115948

ABSTRACT

The function of antibodies (Abs) involves specific binding to antigens (Ags) and activation of other components of the immune system to fight pathogens. The six hypervariable loops within the variable domains of Abs, commonly termed complementarity determining regions (CDRs), are widely assumed to be responsible for Ag recognition, while the constant domains are believed to mediate effector activation. Recent studies and analyses of the growing number of available Ab structures, indicate that this clear functional separation between the two regions may be an oversimplification. Some positions within the CDRs have been shown to never participate in Ag binding and some off-CDRs residues often contribute critically to the interaction with the Ag. Moreover, there is now growing evidence for non-local and even allosteric effects in Ab-Ag interaction in which Ag binding affects the constant region and vice versa. This review summarizes and discusses the structural basis of Ag recognition, elaborating on the contribution of different structural determinants of the Ab to Ag binding and recognition. We discuss the CDRs, the different approaches for their identification and their relationship to the Ag interface. We also review what is currently known about the contribution of non-CDRs regions to Ag recognition, namely the framework regions (FRs) and the constant domains. The suggested mechanisms by which these regions contribute to Ag binding are discussed. On the Ag side of the interaction, we discuss attempts to predict B-cell epitopes and the suggested idea to incorporate Ab information into B-cell epitope prediction schemes. Beyond improving the understanding of immunity, characterization of the functional role of different parts of the Ab molecule may help in Ab engineering, design of CDR-derived peptides, and epitope prediction.

7.
J Immunol ; 190(5): 2327-34, 2013 Mar 01.
Article in English | MEDLINE | ID: mdl-23359499

ABSTRACT

Determining which parts of the Ab are essential for Ag recognition and binding is crucial for understanding B cell-mediated immunity. Identification of fragments of Abs that maintain specificity to the Ag will also allow for the development of improved Ab-based therapy and diagnostics. In this article, we show that structural analysis of Ab-Ag complexes reveals which fragments of the Ab may bind the Ag on their own. In particular, it is possible to predict whether a given CDR is likely to bind the Ag as a peptide by analyzing the energetic contribution of each CDR to Ag binding and by assessing to what extent the interaction between that CDR and the Ag depends on other CDRs. To demonstrate this, we analyzed five Ab-Ag complexes and predicted for each of them which of the CDRs may bind the Ag on its own as a peptide. We then show that these predictions are in agreement with our experimental analysis and with previously published experimental results. These findings promote our understanding of the modular nature of Ab-Ag interactions and lay the foundation for the rational design of active CDR-derived peptides.


Subject(s)
Antigen-Antibody Complex/chemistry , Antigens/chemistry , Complementarity Determining Regions/chemistry , Immunoglobulin Heavy Chains/chemistry , Peptides/chemistry , Single-Chain Antibodies/chemistry , Animals , Antibody Affinity , Antibody Specificity , Antigen-Antibody Complex/genetics , Antigen-Antibody Complex/immunology , Antigens/genetics , Antigens/immunology , Binding Sites, Antibody , Chickens , Cloning, Molecular , Complementarity Determining Regions/genetics , Complementarity Determining Regions/immunology , Databases, Protein , Escherichia coli/genetics , Humans , Immunoglobulin Heavy Chains/genetics , Immunoglobulin Heavy Chains/immunology , Models, Molecular , Peptides/genetics , Peptides/immunology , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/immunology , Single-Chain Antibodies/genetics , Single-Chain Antibodies/immunology , Thermodynamics
8.
J Immunol ; 189(10): 4890-9, 2012 Nov 15.
Article in English | MEDLINE | ID: mdl-23066154

ABSTRACT

To study structural changes that occur in Abs upon Ag binding, we systematically compared free and bound structures of all 141 crystal structures of the 49 Abs that were solved in these two forms. We found that many structural changes occur far from the Ag binding site. Some of them may constitute a mechanism for the recently suggested allosteric effects in Abs. Within the binding site itself, CDR-H3 is the only element that shows significant binding-related conformational changes; however, this occurs in only one third of the Abs. Beyond the binding site, Ag binding is associated with changes in the relative orientation of the H and L chains in both the variable and constant domains. An even larger change occurs in the elbow angle between the variable and the constant domains, and it is significantly larger for binding of big Ags than for binding of small ones. The most consistent and substantial conformational changes occur in a loop in the H chain constant domain. This loop is implicated in the interaction between the H and L chains, is often intrinsically disordered, and is involved in complement binding. Hence, we suggest that it may have a role in Ab function. These findings provide structural insight into the recently proposed allosteric effects in Abs.


Subject(s)
Antigens/chemistry , Binding Sites, Antibody , Databases, Protein , Immunoglobulin Heavy Chains/chemistry , Immunoglobulin Light Chains/chemistry , Animals , Antigens/genetics , Antigens/immunology , Humans , Immunoglobulin Heavy Chains/genetics , Immunoglobulin Heavy Chains/immunology , Immunoglobulin Light Chains/genetics , Immunoglobulin Light Chains/immunology , Protein Structure, Quaternary , Protein Structure, Tertiary
9.
J Biol Chem ; 287(34): 28480-94, 2012 Aug 17.
Article in English | MEDLINE | ID: mdl-22722932

ABSTRACT

Deletion of Phe-508 (F508del) in the first nucleotide binding domain (NBD1) of the cystic fibrosis transmembrane conductance regulator (CFTR) leads to defects in folding and channel gating. NMR data on human F508del NBD1 indicate that an H620Q mutant, shown to increase channel open probability, and the dual corrector/potentiator CFFT-001 similarly disrupt interactions between ß-strands S3, S9, and S10 and the C-terminal helices H8 and H9, shifting a preexisting conformational equilibrium from helix to coil. CFFT-001 appears to interact with ß-strands S3/S9/S10, consistent with docking simulations. Decreases in T(m) from differential scanning calorimetry with H620Q or CFFT-001 suggest direct compound binding to a less thermostable state of NBD1. We hypothesize that, in full-length CFTR, shifting the conformational equilibrium to reduce H8/H9 interactions with the uniquely conserved strands S9/S10 facilitates release of the regulatory region from the NBD dimerization interface to promote dimerization and thereby increase channel open probability. These studies enabled by our NMR assignments for F508del NBD1 provide a window into the conformational fluctuations within CFTR that may regulate function and contribute to folding energetics.


Subject(s)
Cystic Fibrosis Transmembrane Conductance Regulator/chemistry , Protein Folding , Protein Multimerization , Amino Acid Sequence , Amino Acid Substitution , Cystic Fibrosis Transmembrane Conductance Regulator/genetics , Cystic Fibrosis Transmembrane Conductance Regulator/metabolism , Humans , Mutation, Missense , Protein Structure, Secondary , Protein Structure, Tertiary , Sequence Deletion
SELECTION OF CITATIONS
SEARCH DETAIL
...