Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 815
Filtrar
1.
Anal Chem ; 96(19): 7386-7393, 2024 May 14.
Artículo en Inglés | MEDLINE | ID: mdl-38698660

RESUMEN

Covalent labeling in combination with mass spectrometry is a powerful approach used in structural biology to study protein structures, interactions, and dynamics. Recently, the toolbox of covalent labeling techniques has been expanded with fast fluoroalkylation of proteins (FFAP). FFAP is a novel radical labeling method that utilizes fluoroalkyl radicals generated from hypervalent Togni reagents for targeting aromatic residues. This report further demonstrates the benefits of FFAP as a new method for structural characterization of therapeutic antibodies and interaction interfaces of antigen-antibody complexes. The results obtained from human trastuzumab and its complex with human epidermal growth factor receptor 2 (HER2) correlate well with previously published structural data and demonstrate the potential of FFAP in structural biology.


Asunto(s)
Mapeo Epitopo , Receptor ErbB-2 , Trastuzumab , Humanos , Mapeo Epitopo/métodos , Receptor ErbB-2/química , Receptor ErbB-2/inmunología , Trastuzumab/química , Alquilación , Anticuerpos Monoclonales/química , Anticuerpos Monoclonales/inmunología , Halogenación , Huella de Proteína/métodos , Complejo Antígeno-Anticuerpo/química
2.
J Am Chem Soc ; 146(19): 13455-13466, 2024 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-38703132

RESUMEN

The classical complement pathway is activated by antigen-bound IgG antibodies. Monomeric IgG must oligomerize to activate complement via the hexameric C1q complex, and hexamerizing mutants of IgG appear as promising therapeutic candidates. However, structural data have shown that it is not necessary to bind all six C1q arms to initiate complement, revealing a symmetry mismatch between C1 and the hexameric IgG complex that has not been adequately explained. Here, we use DNA nanotechnology to produce specific nanostructures to template antigens and thereby spatially control IgG valency. These DNA-nanotemplated IgG complexes can activate complement on cell-mimetic lipid membranes, which enabled us to determine the effect of IgG valency on complement activation without the requirement to mutate antibodies. We investigated this using biophysical assays together with 3D cryo-electron tomography. Our data revealed the importance of interantigen distance on antibody-mediated complement activation, and that the cleavage of complement component C4 by the C1 complex is proportional to the number of ideally spaced antigens. Increased IgG valency also translated to better terminal pathway activation and membrane attack complex formation. Together, these data provide insights into how nanopatterning antigen-antibody complexes influence the activation of the C1 complex and suggest routes to modulate complement activation by antibody engineering. Furthermore, to our knowledge, this is the first time DNA nanotechnology has been used to study the activation of the complement system.


Asunto(s)
Activación de Complemento , ADN , Inmunoglobulina G , Nanoestructuras , Nanoestructuras/química , Humanos , ADN/química , ADN/inmunología , Inmunoglobulina G/química , Inmunoglobulina G/inmunología , Complejo Antígeno-Anticuerpo/química , Complejo Antígeno-Anticuerpo/inmunología
3.
Int J Biol Macromol ; 268(Pt 2): 131697, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38688333

RESUMEN

Immobilization technology plays an important role in enhancing enzyme stability and environmental adaptability. Despite its rapid development, this technology still encounters many challenges such as enzyme leakage, difficulties in large-scale implementation, and limited reusability. Drawing inspiration from natural paired molecules, this study aimed to establish a method for immobilized α-glucosidase using artificial antibody-antigen interaction. The proposed method consists of three main parts: synthesis of artificial antibodies, synthesis of artificial antigens, and assembly of the artificial antibody-antigen complex. The critical step in this method involves selecting a pair of structurally similar compounds: catechol as a template for preparing artificial antibodies and protocatechualdehyde for modifying the enzyme to create the artificial antigens. By utilizing the same functional groups in these compounds, specific recognition of the antigen by the artificial antibody can be achieved, thereby immobilizing the enzymes. The results demonstrated that the immobilization amount, specific activity, and enzyme activity of the immobilized α-glucosidase were 25.09 ± 0.10 mg/g, 5.71 ± 0.17 U/mgprotein and 143.25 ± 1.71 U/gcarrier, respectively. The immobilized α-glucosidase not only exhibited excellent reusability but also demonstrated remarkable performance in catalyzing the hydrolysis of 4-methylumbelliferyl-α-D-glucopyranoside.


Asunto(s)
Enzimas Inmovilizadas , Himecromona , alfa-Glucosidasas , Enzimas Inmovilizadas/química , alfa-Glucosidasas/química , alfa-Glucosidasas/inmunología , Himecromona/química , Himecromona/análogos & derivados , Biocatálisis , Estabilidad de Enzimas , Hidrólisis , Biomimética/métodos , Cinética , Anticuerpos/química , Anticuerpos/inmunología , Materiales Biomiméticos/química , Complejo Antígeno-Anticuerpo/química , Concentración de Iones de Hidrógeno
4.
Bioinformatics ; 40(3)2024 Mar 04.
Artículo en Inglés | MEDLINE | ID: mdl-38449295

RESUMEN

MOTIVATION: Antibodies are proteins that the immune system produces in response to foreign pathogens. Designing antibodies that specifically bind to antigens is a key step in developing antibody therapeutics. The complementarity determining regions (CDRs) of the antibody are mainly responsible for binding to the target antigen, and therefore must be designed to recognize the antigen. RESULTS: We develop an antibody design model, AbFlex, that exhibits state-of-the-art performance in terms of structure prediction accuracy and amino acid recovery rate. Furthermore, >38% of newly designed antibody models are estimated to have better binding energies for their antigens than wild types. The effectiveness of the model is attributed to two different strategies that are developed to overcome the difficulty associated with the scarcity of antibody-antigen complex structure data. One strategy is to use an equivariant graph neural network model that is more data-efficient. More importantly, a new data augmentation strategy based on the flexible definition of CDRs significantly increases the performance of the CDR prediction model. AVAILABILITY AND IMPLEMENTATION: The source code and implementation are available at https://github.com/wsjeon92/AbFlex.


Asunto(s)
Complejo Antígeno-Anticuerpo , Regiones Determinantes de Complementariedad , Regiones Determinantes de Complementariedad/química , Regiones Determinantes de Complementariedad/metabolismo , Secuencia de Aminoácidos , Modelos Moleculares , Complejo Antígeno-Anticuerpo/química , Antígenos
5.
Database (Oxford) ; 20242024 Mar 19.
Artículo en Inglés | MEDLINE | ID: mdl-38502609

RESUMEN

Progression of various cancers and autoimmune diseases is associated with changes in systemic or local tissue temperatures, which may impact current therapies. The role of fever and acute inflammation-range temperatures on the stability and activity of antibodies relevant for cancers and autoimmunity is unknown. To produce molecular dynamics (MD) trajectories of immune complexes at relevant temperatures, we used the Research Collaboratory for Structural Bioinformatics (RCSB) database to identify 50 antibody:antigen complexes of interest, in addition to single antibodies and antigens, and deployed Groningen Machine for Chemical Simulations (GROMACS) to prepare and run the structures at different temperatures for 100-500 ns, in single or multiple random seeds. MD trajectories are freely available. Processed data include Protein Data Bank outputs for all files obtained every 50 ns, and free binding energy calculations for some of the immune complexes. Protocols for using the data are also available. Individual datasets contain unique DOIs. We created a web interface, ThermoPCD, as a platform to explore the data. The outputs of ThermoPCD allow the users to relate thermally-dependent changes in epitopes:paratopes interfaces to their free binding energies, or against own experimentally derived binding affinities. ThermoPCD is a free to use database of immune complexes' trajectories at different temperatures that does not require registration and allows for all the data to be available for download. Database URL: https://sites.google.com/view/thermopcd/home.


Asunto(s)
Simulación de Dinámica Molecular , Neoplasias , Humanos , Complejo Antígeno-Anticuerpo/química , Temperatura , Sitios de Unión de Anticuerpos
6.
MAbs ; 16(1): 2322533, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38477253

RESUMEN

Antibodies have increasingly been developed as drugs with over 100 now licensed in the US or EU. During development, it is often necessary to increase or reduce the affinity of an antibody and rational attempts to do so rely on having a structure of the antibody-antigen complex often obtained by modeling. The antigen-binding site consists primarily of six loops known as complementarity-determining regions (CDRs), and an open question has been whether these loops change their conformation when they bind to an antigen. Existing surveys of antibody-antigen complex structures have only examined CDR conformational change in case studies or small-scale surveys. With an increasing number of antibodies where both free and complexed structures have been deposited in the Protein Data Bank, a large-scale survey of CDR conformational change during binding is now possible. To this end, we built a dataset, AbAgDb, that currently includes 177 antibodies with high-quality CDRs, each of which has at least one bound and one unbound structure. We analyzed the conformational change of the Cα backbone of each CDR upon binding and found that, in most cases, the CDRs (other than CDR-H3) show minimal movement, while 70.6% and 87% of CDR-H3s showed global Cα RMSD ≤ 1.0Å and ≤ 2.0Å, respectively. We also compared bound CDR conformations with the conformational space of unbound CDRs and found most of the bound conformations are included in the unbound conformational space. In future, our results will contribute to developing insights into antibodies and new methods for modeling and docking.


Asunto(s)
Antígenos , Regiones Determinantes de Complementariedad , Secuencia de Aminoácidos , Modelos Moleculares , Conformación Proteica , Regiones Determinantes de Complementariedad/química , Complejo Antígeno-Anticuerpo/química , Sitios de Unión de Anticuerpos
7.
Protein Sci ; 33(1): e4824, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-37945533

RESUMEN

The atomic-resolution structural information that X-ray crystallography can provide on the binding interface between a Fab and its cognate antigen is highly valuable for understanding the mechanism of interaction. However, many Fab:antigen complexes are recalcitrant to crystallization, making the endeavor a considerable effort with no guarantee of success. Consequently, there have been significant steps taken to increase the likelihood of Fab:antigen complex crystallization by altering the Fab framework. In this investigation, we applied the surface entropy reduction strategy coupled with phage-display technology to identify a set of surface substitutions that improve the propensity of a human Fab framework to crystallize. In addition, we showed that combining these surface substitutions with previously reported Crystal Kappa and elbow substitutions results in an extraordinary improvement in Fab and Fab:antigen complex crystallizability, revealing a strong synergistic relationship between these sets of substitutions. Through comprehensive Fab and Fab:antigen complex crystallization screenings followed by structure determination and analysis, we defined the roles that each of these substitutions play in facilitating crystallization and how they complement each other in the process.


Asunto(s)
Complejo Antígeno-Anticuerpo , Fragmentos Fab de Inmunoglobulinas , Humanos , Cristalización/métodos , Fragmentos Fab de Inmunoglobulinas/genética , Fragmentos Fab de Inmunoglobulinas/química , Complejo Antígeno-Anticuerpo/química , Antígenos/química , Cristalografía por Rayos X , Conformación Proteica
8.
Protein Sci ; 32(9): e4745, 2023 09.
Artículo en Inglés | MEDLINE | ID: mdl-37550885

RESUMEN

Antibodies are used for many therapeutic and biotechnological purposes. Because the affinity of an antibody to the antigen is critical for clinical efficacy of pharmaceuticals, many affinity maturation strategies have been developed. Although we previously reported an affinity maturation strategy in which the association rate of the antibody toward its antigen is improved by introducing a cluster of arginine residues into the framework region of the antibody, the detailed molecular mechanism responsible for this improvement has been unknown. In this study, we introduced five arginine residues into an anti-hen egg white lysozyme antibody (HyHEL10) Fab fragment to create the R5-mutant and comprehensively characterized the interaction between antibody and antigen using thermodynamic analysis, X-ray crystallography, and molecular dynamics (MD) simulations. Our results indicate that introduction of charged residues strongly enhanced the association rate, as previously reported, and the antibody-antigen complex structure was almost the same for the R5-mutant and wild-type Fabs. The MD simulations indicate that the mutation increased conformational diversity in complementarity-determining region loops and thereby enhanced the association rate. These observations provide the molecular basis of affinity maturation by R5 mutation.


Asunto(s)
Complejo Antígeno-Anticuerpo , Antígenos , Conformación Proteica , Antígenos/química , Complejo Antígeno-Anticuerpo/química , Regiones Determinantes de Complementariedad/genética , Regiones Determinantes de Complementariedad/química , Fragmentos Fab de Inmunoglobulinas/genética , Fragmentos Fab de Inmunoglobulinas/química , Cristalografía por Rayos X
10.
Methods Mol Biol ; 2552: 267-282, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36346597

RESUMEN

In the recent years, therapeutic use of antibodies has seen a huge growth, "due to their inherent proprieties and technological advances in the methods used to study and characterize them. Effective design and engineering of antibodies for therapeutic purposes are heavily dependent on knowledge of the structural principles that regulate antibody-antigen interactions. Several experimental techniques such as X-ray crystallography, cryo-electron microscopy, NMR, or mutagenesis analysis can be applied, but these are usually expensive and time-consuming. Therefore computational approaches like molecular docking may offer a valuable alternative for the characterization of antibody-antigen complexes.Here we describe a protocol for the prediction of the 3D structure of antibody-antigen complexes using the integrative modelling platform HADDOCK. The protocol consists of (1) the identification of the antibody residues belonging to the hypervariable loops which are known to be crucial for the binding and can be used to guide the docking and (2) the detailed steps to perform docking with the HADDOCK 2.4 webserver following different strategies depending on the availability of information about epitope residues.


Asunto(s)
Complejo Antígeno-Anticuerpo , Simulación del Acoplamiento Molecular , Microscopía por Crioelectrón , Cristalografía por Rayos X , Espectroscopía de Resonancia Magnética , Complejo Antígeno-Anticuerpo/química , Unión Proteica
11.
J Mol Graph Model ; 118: 108364, 2023 01.
Artículo en Inglés | MEDLINE | ID: mdl-36356467

RESUMEN

Specific antibodies can bind to protein antigens with high affinity and specificity, and this property makes them one of the best protein-based therapeutics. Accurate prediction of antibody‒protein antigen binding affinity is crucial for designing effective antibodies. The current predictive methods for protein‒protein binding affinity usually fail to predict the binding affinity of an antibody‒protein antigen complex with a comparable level of accuracy. Here, new models specific for antibody‒antigen binding affinity prediction are developed according to the different types of interface and surface areas present in antibody‒antigen complex. The contacts-based descriptors are also employed to construct or train different models specific for antibody‒protein antigen binding affinity prediction. The results of this study show that (i) the area-based descriptors are slightly better than the contacts-based descriptors in terms of the predictive power; (ii) the new models specific for antibody‒protein antigen binding affinity prediction are superior to the previously-used general models for predicting the protein‒protein binding affinities; (iii) the performances of the best area-based and contacts-based models developed in this work are better than the performances of a recently-developed graph-based model (i.e., CSM-AB) specific for antibody‒protein antigen binding affinity prediction. The new models developed in this work would not only help understand the mechanisms underlying antibody‒protein antigen interactions, but would also be of some applicable utility in the design and virtual screening of antibody-based therapeutics.


Asunto(s)
Complejo Antígeno-Anticuerpo , Proteínas , Proteínas/química , Unión Proteica , Complejo Antígeno-Anticuerpo/química , Aprendizaje Automático , Antígenos/química
12.
Front Immunol ; 13: 910367, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35874680

RESUMEN

Antibody recognition of antigens is a critical element of adaptive immunity. One key class of antibody-antigen complexes is comprised of antibodies targeting linear epitopes of proteins, which in some cases are conserved elements of viruses and pathogens of relevance for vaccine design and immunotherapy. Here we report a detailed analysis of the structural and interface features of this class of complexes, based on a set of nearly 200 nonredundant high resolution antibody-peptide complex structures that were assembled from the Protein Data Bank. We found that antibody-bound peptides adopt a broad range of conformations, often displaying limited secondary structure, and that the same peptide sequence bound by different antibodies can in many cases exhibit varying conformations. Propensities of contacts with antibody loops and extent of antibody binding conformational changes were found to be broadly similar to those for antibodies in complex with larger protein antigens. However, antibody-peptide interfaces showed lower buried surface areas and fewer hydrogen bonds than antibody-protein antigen complexes, while calculated binding energy per buried interface area was found to be higher on average for antibody-peptide interfaces, likely due in part to a greater proportion of buried hydrophobic residues and higher shape complementarity. This dataset and these observations can be of use for future studies focused on this class of interactions, including predictive computational modeling efforts and the design of antibodies or epitope-based vaccine immunogens.


Asunto(s)
Complejo Antígeno-Anticuerpo , Vacunas , Complejo Antígeno-Anticuerpo/química , Antígenos , Sitios de Unión de Anticuerpos , Epítopos/química , Modelos Moleculares , Péptidos/química , Conformación Proteica
13.
Proc Natl Acad Sci U S A ; 119(28): e2123212119, 2022 07 12.
Artículo en Inglés | MEDLINE | ID: mdl-35867757

RESUMEN

Humans lack the capacity to produce the Galα1-3Galß1-4GlcNAc (α-gal) glycan, and produce anti-α-gal antibodies upon exposure to the carbohydrate on a diverse set of immunogens, including commensal gut bacteria, malaria parasites, cetuximab, and tick proteins. Here we use X-ray crystallographic analysis of antibodies from α-gal knockout mice and humans in complex with the glycan to reveal a common binding motif, centered on a germline-encoded tryptophan residue at Kabat position 33 (W33) of the complementarity-determining region of the variable heavy chain (CDRH1). Immunoglobulin sequencing of anti-α-gal B cells in healthy humans and tick-induced mammalian meat anaphylaxis patients revealed preferential use of heavy chain germline IGHV3-7, encoding W33, among an otherwise highly polyclonal antibody response. Antigen binding was critically dependent on the presence of the germline-encoded W33 residue for all of the analyzed antibodies; moreover, introduction of the W33 motif into naive IGHV3-23 antibody phage libraries enabled the rapid selection of α-gal binders. Our results outline structural and genetic factors that shape the human anti-α-galactosyl antibody response, and provide a framework for future therapeutics development.


Asunto(s)
Anafilaxia , Anticuerpos , Hipersensibilidad a los Alimentos , Cadenas Pesadas de Inmunoglobulina , Región Variable de Inmunoglobulina , Enfermedades por Picaduras de Garrapatas , Trisacáridos , Anafilaxia/inmunología , Animales , Anticuerpos/química , Anticuerpos/genética , Formación de Anticuerpos/genética , Complejo Antígeno-Anticuerpo/química , Cristalografía por Rayos X , Hipersensibilidad a los Alimentos/inmunología , Humanos , Cadenas Pesadas de Inmunoglobulina/química , Cadenas Pesadas de Inmunoglobulina/genética , Región Variable de Inmunoglobulina/química , Región Variable de Inmunoglobulina/inmunología , Ratones , Ratones Noqueados , Biblioteca de Péptidos , Conformación Proteica , Enfermedades por Picaduras de Garrapatas/inmunología , Trisacáridos/genética , Trisacáridos/inmunología
14.
Immunogenetics ; 74(5): 465-474, 2022 10.
Artículo en Inglés | MEDLINE | ID: mdl-35545703

RESUMEN

We herein analyzed all available protein-protein interfaces of the immune complexes from the Protein Data Bank whose antigens belong to pathogens or cancers that are modulated by fever in mammalian hosts. We also included, for comparison, protein interfaces from immune complexes that are not significantly modulated by the fever response. We highlight the distribution of amino acids at these viral, bacterial, protozoan and cancer epitopes, and at their corresponding paratopes that belong strictly to monoclonal antibodies. We identify the "hotspots", i.e. residues that are highly connected at such interfaces, and assess the structural, kinetic and thermodynamic parameters responsible for complex formation. We argue for an evolutionary pressure for the types of residues at these protein interfaces that may explain the role of fever as a selective force for optimizing antibody binding to antigens.


Asunto(s)
Anticuerpos Monoclonales , Complejo Antígeno-Anticuerpo , Animales , Anticuerpos Monoclonales/metabolismo , Complejo Antígeno-Anticuerpo/química , Sitios de Unión de Anticuerpos , Bases de Datos de Proteínas , Epítopos , Mamíferos
15.
J Virol ; 96(2): e0168921, 2022 01 26.
Artículo en Inglés | MEDLINE | ID: mdl-34730392

RESUMEN

The low abundance of envelope spikes and the inability of IgG to aggregate virions render HIV-1 an inadequate target for antibody-mediated clearance by phagocytes. In an attempt to improve the ability of antibody to mediate the internalization of HIV-1 virions, we generated multimers of the broadly neutralizing HIV-1-specific monoclonal antibody (MAb) VRC01 using site-directed mutagenesis of the Fc segment. We then measured virion internalization using primary human monocytes and neutrophils. We found that, in the absence of complement, immune complexes consisting of HIV-1 virions and VRC01 multimers were slightly more efficiently internalized than were complexes formed with monomeric VRC01. The presence of complement, however, greatly augmented internalization of immune complexes formed with the multimeric MAb but had little impact on monomeric MAb-mediated internalization. Multimerization and the presence of complement overcome the limited ability of monomeric antibody to mediate internalization of HIV-1 virions and may thus provide a therapeutic approach to clearing virus. IMPORTANCE Antibody-mediated internalization of HIV-1 by phagocytes, a potential mechanism for clearing virus, is very inefficient. In an effort to improve viral clearance, we produced a multimeric form of the broadly neutralizing monoclonal antibody VRC01. We found that VRC01 antibody multimers (primarily hexamers) were only slightly more efficient in mediating HIV-1 internalization than was monomeric VRC01. However, the addition of complement resulted in substantially greater internalization of multimer-opsonized virus. In contrast, complement had little if any impact on internalization of monomer-opsonized virus. Therefore, antibody multimerization in combination with complement may overcome the limited ability of monomeric antibody to mediate internalization of HIV-1 virions. Our findings may provide a therapeutic approach to clearing virus.


Asunto(s)
Proteínas del Sistema Complemento/inmunología , Anticuerpos Anti-VIH/inmunología , VIH-1/inmunología , Fagocitosis/inmunología , Virión/inmunología , Complejo Antígeno-Anticuerpo/química , Complejo Antígeno-Anticuerpo/genética , Complejo Antígeno-Anticuerpo/inmunología , Anticuerpos ampliamente neutralizantes/química , Anticuerpos ampliamente neutralizantes/genética , Anticuerpos ampliamente neutralizantes/inmunología , Anticuerpos Anti-VIH/química , Anticuerpos Anti-VIH/genética , Proteína gp41 de Envoltorio del VIH/inmunología , Humanos , Monocitos/inmunología , Mutación , Neutrófilos/inmunología , Multimerización de Proteína , Receptores Fc/genética , Receptores Fc/inmunología
16.
J Mol Biol ; 434(2): 167391, 2022 01 30.
Artículo en Inglés | MEDLINE | ID: mdl-34890647

RESUMEN

Previous reports present different models for the stabilization of the Fc-FcγRI immune complex. Although accord exists on the importance of L235 in IgG1 and some hydrophobic contacts for complex stabilization, discord exists regarding the existence of stabilizing glycoprotein contacts between glycans of IgG1 and a conserved FG-loop (171MGKHRY176) of FcγRIa. Complexes formed from the FcγRIa receptor and IgG1s containing biantennary glycans with N-acetylglucosamine, galactose, and α2,6-N-acetylneuraminic terminations were measured by hydrogen-deuterium exchange mass spectrometry (HDX-MS), classified for dissimilarity with Welch's ANOVA and Games-Howell post hoc procedures, and modeled with molecular dynamics (MD) simulations. For each glycoform of the IgG1-FcγRIa complex peptic peptides of Fab, Fc and FcγRIa report distinct H/D exchange rates. MD simulations corroborate the differences in the peptide deuterium content through calculation of the percent of time that transient glycan-peptide bonds exist. These results indicate that stability of IgG1-FcγRIa complexes correlate with the presence of intermolecular glycoprotein interactions between the IgG1 glycans and the 173KHR175 motif within the FG-loop of FcγRIa. The results also indicate that intramolecular glycan-protein bonds stabilize the Fc region in isolated and complexed IgG1. Moreover, HDX-MS data evince that the Fab domain has glycan-protein binding contacts within the IgG1-FcγRI complex.


Asunto(s)
Complejo Antígeno-Anticuerpo/química , Glicoproteínas/química , Espectrometría de Masas de Intercambio de Hidrógeno-Deuterio/métodos , Inmunoglobulina G/química , Simulación de Dinámica Molecular , Receptores de IgG/química , Anticuerpos Monoclonales/química , Complejo Antígeno-Anticuerpo/metabolismo , Galactosa , Glicoproteínas/metabolismo , Proteínas de la Membrana/química , Péptidos/química , Péptidos/metabolismo , Polisacáridos , Unión Proteica
17.
PLoS Comput Biol ; 17(12): e1009675, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34898603

RESUMEN

Identifying the epitope of an antibody is a key step in understanding its function and its potential as a therapeutic. Sequence-based clonal clustering can identify antibodies with similar epitope complementarity, however, antibodies from markedly different lineages but with similar structures can engage the same epitope. We describe a novel computational method for epitope profiling based on structural modelling and clustering. Using the method, we demonstrate that sequence dissimilar but functionally similar antibodies can be found across the Coronavirus Antibody Database, with high accuracy (92% of antibodies in multiple-occupancy structural clusters bind to consistent domains). Our approach functionally links antibodies with distinct genetic lineages, species origins, and coronavirus specificities. This indicates greater convergence exists in the immune responses to coronaviruses than is suggested by sequence-based approaches. Our results show that applying structural analytics to large class-specific antibody databases will enable high confidence structure-function relationships to be drawn, yielding new opportunities to identify functional convergence hitherto missed by sequence-only analysis.


Asunto(s)
Antígenos Virales/química , COVID-19/inmunología , COVID-19/virología , Epítopos de Linfocito B/química , SARS-CoV-2/química , SARS-CoV-2/inmunología , Secuencia de Aminoácidos , Animales , Anticuerpos Neutralizantes/química , Anticuerpos Neutralizantes/genética , Anticuerpos Antivirales/química , Anticuerpos Antivirales/genética , Anticuerpos Antivirales/metabolismo , Especificidad de Anticuerpos , Complejo Antígeno-Anticuerpo/química , Complejo Antígeno-Anticuerpo/genética , Reacciones Antígeno-Anticuerpo/genética , Reacciones Antígeno-Anticuerpo/inmunología , Biología Computacional , Coronavirus/química , Coronavirus/genética , Coronavirus/inmunología , Bases de Datos de Compuestos Químicos , Mapeo Epitopo , Epítopos de Linfocito B/genética , Humanos , Ratones , Modelos Moleculares , Pandemias , SARS-CoV-2/genética , Anticuerpos de Dominio Único/inmunología
18.
J Clin Invest ; 131(22)2021 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-34618682

RESUMEN

We used human monoclonal antibodies (humAbs) to study the mechanism of neuron intoxication by tetanus neurotoxin and to evaluate these antibodies as a safe preventive and therapeutic substitute for hyperimmune sera to treat tetanus in mice. By screening memory B cells from immune donors, we selected 2 tetanus neurotoxin-specific mAbs with exceptionally high neutralizing activities and extensively characterized them both structurally and functionally. We found that these antibodies interfered with the binding and translocation of the neurotoxin into neurons by interacting with 2 epitopes, whose identification pinpoints crucial events in the cellular pathogenesis of tetanus. Our observations explain the neutralization ability of these antibodies, which we found to be exceptionally potent in preventing experimental tetanus when injected into mice long before the toxin. Moreover, their Fab derivatives neutralized tetanus neurotoxin in post-exposure experiments, suggesting their potential for therapeutic use via intrathecal injection. As such, we believe these humAbs, as well as their Fab derivatives, meet the requirements to be considered for prophylactic and therapeutic use in human tetanus and are ready for clinical trials.


Asunto(s)
Anticuerpos Monoclonales/uso terapéutico , Metaloendopeptidasas/antagonistas & inhibidores , Toxina Tetánica/antagonistas & inhibidores , Tétanos/prevención & control , Adulto , Animales , Anticuerpos Monoclonales/química , Complejo Antígeno-Anticuerpo/química , Células HEK293 , Humanos , Fragmentos Fab de Inmunoglobulinas/química , Metaloendopeptidasas/química , Ratones , Conformación Proteica , Ratas , Tétanos/tratamiento farmacológico , Toxina Tetánica/química
19.
Clin Immunol ; 231: 108848, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34492381

RESUMEN

Antibodies to DNA (anti-DNA) are the serological hallmark of systemic lupus erythematosus. Previous studies have indicated that the phosphodiester backbone is the main antigenic target, with electrostatic interactions important for high avidity. To define further these interactions, the effects of ionic strength on anti-DNA binding of SLE plasmas were assessed in association and dissociation assays by ELISA. As these studies demonstrated, increasing ionic strength to a concentration of 1000 mM NaCl reduced antibody binding although the extent of the reduction varied among samples. In dissociation assays, differences among plasmas were also observed. For one of the plasmas, binding to DNA displayed resistance to dissociation by increasing ionic strength even though these concentrations limited binding in association assays. Time course studies showed a gradual change in binding interactions. These studies indicate that anti-DNA binding can involve both electrostatic and non-electrostatic interactions, with binding in some plasmas showing evidence of hysteresis.


Asunto(s)
Anticuerpos Antinucleares/inmunología , Afinidad de Anticuerpos/inmunología , Complejo Antígeno-Anticuerpo/inmunología , ADN/inmunología , Lupus Eritematoso Sistémico/inmunología , Anticuerpos Antinucleares/química , Complejo Antígeno-Anticuerpo/química , Autoantígenos/inmunología , Humanos , Concentración Osmolar
20.
Int J Mol Sci ; 22(13)2021 Jun 22.
Artículo en Inglés | MEDLINE | ID: mdl-34206395

RESUMEN

The innate immune system's natural killer (NK) cells exert their cytolytic function against a variety of pathological challenges, including tumors and virally infected cells. Their activation depends on net signaling mediated via inhibitory and activating receptors that interact with specific ligands displayed on the surfaces of target cells. The CD94/NKG2C heterodimer is one of the NK activating receptors and performs its function by interacting with the trimeric ligand comprised of the HLA-E/ß2m/nonameric peptide complex. Here, simulations of the all-atom multi-microsecond molecular dynamics in five immune complexes provide atomistic insights into the receptor-ligand molecular recognition, as well as the molecular events that facilitate the NK cell activation. We identify NKG2C, the HLA-Eα2 domain, and the nonameric peptide as the key elements involved in the molecular machinery of signal transduction via an intertwined hydrogen bond network. Overall, the study addresses the complex intricacies that are necessary to understand the mechanisms of the innate immune system.


Asunto(s)
Complejo Antígeno-Anticuerpo/química , Antígenos de Histocompatibilidad Clase I/química , Modelos Moleculares , Subfamília C de Receptores Similares a Lectina de Células NK/química , Subfamília D de Receptores Similares a Lectina de las Células NK/química , Péptidos/química , Secuencia de Aminoácidos , Complejo Antígeno-Anticuerpo/inmunología , Complejo Antígeno-Anticuerpo/metabolismo , Sitios de Unión , Antígenos de Histocompatibilidad Clase I/inmunología , Antígenos de Histocompatibilidad Clase I/metabolismo , Humanos , Enlace de Hidrógeno , Ligandos , Subfamília C de Receptores Similares a Lectina de Células NK/metabolismo , Subfamília D de Receptores Similares a Lectina de las Células NK/metabolismo , Péptidos/metabolismo , Unión Proteica , Conformación Proteica , Dominios y Motivos de Interacción de Proteínas , Transducción de Señal , Relación Estructura-Actividad , Antígenos HLA-E
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA