Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 4.154
Filter
1.
Acta Crystallogr F Struct Biol Commun ; 80(Pt 10): 234-235, 2024 Oct 01.
Article in English | MEDLINE | ID: mdl-39361148

ABSTRACT

The Section Editors welcome and introduce three new Co-editors to the journal and summarize their expertise in computational structural biology, experimental structural biology and cryo-electron microscopy.


Subject(s)
Cryoelectron Microscopy , Cryoelectron Microscopy/methods , Periodicals as Topic , Computational Biology/methods , Crystallography, X-Ray , Crystallography/methods
2.
J Appl Crystallogr ; 57(Pt 5): 1384-1391, 2024 Oct 01.
Article in English | MEDLINE | ID: mdl-39387090

ABSTRACT

Coherent diffractive imaging with X-ray free-electron lasers could enable structural studies of macromolecules at room temperature. This type of experiment could provide a means to study structural dynamics on the femtosecond timescale. However, the diffraction from a single protein is weak compared with the incoherent scattering from background sources, which negatively affects the reconstruction analysis. This work evaluates the effects of the presence of background on the analysis pipeline. Background measurements from the European X-ray Free-Electron Laser were combined with simulated diffraction patterns and treated by a standard reconstruction procedure, including orientation recovery with the expand, maximize and compress algorithm and 3D phase retrieval. Background scattering did have an adverse effect on the estimated resolution of the reconstructed density maps. Still, the reconstructions generally worked when the signal-to-background ratio was 0.6 or better, in the momentum transfer shell of the highest reconstructed resolution. The results also suggest that the signal-to-background requirement increases at higher resolution. This study gives an indication of what is possible at current setups at X-ray free-electron lasers with regards to expected background strength and establishes a target for experimental optimization of the background.

3.
Nature ; 2024 Oct 09.
Article in English | MEDLINE | ID: mdl-39384923
4.
Elife ; 132024 Oct 07.
Article in English | MEDLINE | ID: mdl-39373641

ABSTRACT

Polycystin-1 (PC1) is the protein product of the PKD1 gene whose mutation causes autosomal dominant Polycystic Kidney Disease (ADPKD). PC1 is an atypical G protein-coupled receptor (GPCR) with an autocatalytic GAIN domain that cleaves PC1 into extracellular N-terminal and membrane-embedded C-terminal (CTF) fragments. Recently, activation of PC1 CTF signaling was shown to be regulated by a stalk tethered agonist (TA), resembling the mechanism observed for adhesion GPCRs. Here, synthetic peptides of the first 9- (p9), 17- (p17), and 21-residues (p21) of the PC1 stalk TA were shown to re-activate signaling by a stalkless CTF mutant in human cell culture assays. Novel Peptide Gaussian accelerated molecular dynamics (Pep-GaMD) simulations elucidated binding conformations of p9, p17, and p21 and revealed multiple specific binding regions to the stalkless CTF. Peptide agonists binding to the TOP domain of PC1 induced close TOP-putative pore loop interactions, a characteristic feature of stalk TA-mediated PC1 CTF activation. Additional sequence coevolution analyses showed the peptide binding regions were consistent with covarying residue pairs identified between the TOP domain and the stalk TA. These insights into the structural dynamic mechanism of PC1 activation by TA peptide agonists provide an in-depth understanding that will facilitate the development of therapeutics targeting PC1 for ADPKD treatment.


Subject(s)
Molecular Dynamics Simulation , Peptides , Protein Binding , Signal Transduction , TRPP Cation Channels , Humans , TRPP Cation Channels/metabolism , TRPP Cation Channels/chemistry , TRPP Cation Channels/genetics , Peptides/metabolism , Peptides/chemistry , Peptides/pharmacology , Polycystic Kidney, Autosomal Dominant/metabolism , Polycystic Kidney, Autosomal Dominant/genetics , Polycystic Kidney, Autosomal Dominant/drug therapy , Protein Conformation
5.
Elife ; 132024 Oct 07.
Article in English | MEDLINE | ID: mdl-39374147

ABSTRACT

Lipopolysaccharides (LPS) confer resistance against harsh conditions, including antibiotics, in Gram-negative bacteria. The lipopolysaccharide transport (Lpt) complex, consisting of seven proteins (A-G), exports LPS across the cellular envelope. LptB2FG forms an ATP-binding cassette transporter that transfers LPS to LptC. How LptB2FG couples ATP binding and hydrolysis with LPS transport to LptC remains unclear. We observed the conformational heterogeneity of LptB2FG and LptB2FGC in micelles and/or proteoliposomes using pulsed dipolar electron spin resonance spectroscopy. Additionally, we monitored LPS binding and release using laser-induced liquid bead ion desorption mass spectrometry. The ß-jellyroll domain of LptF stably interacts with the LptG and LptC ß-jellyrolls in both the apo and vanadate-trapped states. ATP binding at the cytoplasmic side is allosterically coupled to the selective opening of the periplasmic LptF ß-jellyroll domain. In LptB2FG, ATP binding closes the nucleotide binding domains, causing a collapse of the first lateral gate as observed in structures. However, the second lateral gate, which forms the putative entry site for LPS, exhibits a heterogeneous conformation. LptC binding limits the flexibility of this gate to two conformations, likely representing the helix of LptC as either released from or inserted into the transmembrane domains. Our results reveal the regulation of the LPS entry gate through the dynamic behavior of the LptC transmembrane helix, while its ß-jellyroll domain is anchored in the periplasm. This, combined with long-range ATP-dependent allosteric gating of the LptF ß-jellyroll domain, may ensure efficient and unidirectional transport of LPS across the periplasm.


Subject(s)
ATP-Binding Cassette Transporters , Adenosine Triphosphate , Escherichia coli Proteins , Escherichia coli , Lipopolysaccharides , Lipopolysaccharides/metabolism , ATP-Binding Cassette Transporters/metabolism , ATP-Binding Cassette Transporters/chemistry , Escherichia coli Proteins/metabolism , Escherichia coli Proteins/chemistry , Escherichia coli Proteins/genetics , Adenosine Triphosphate/metabolism , Escherichia coli/metabolism , Escherichia coli/genetics , Biological Transport , Protein Binding , Protein Conformation , Membrane Proteins
6.
Br J Pharmacol ; 2024 Oct 02.
Article in English | MEDLINE | ID: mdl-39354889

ABSTRACT

This brief review highlights some of the structure-activity relationships of classic serotonergic psychedelics. In particular, we discuss structural features of three chemotypes: phenethylamines, ergolines and certain tryptamines, which possess psychedelic activity in humans. Where they are known, we point out the underlying molecular mechanisms utilized by each of the three chemotypes of psychedelic molecules. With a focus on the 5-HT2A receptor subtype, a G-protein coupled receptor known to be the primary target of psychedelics, we refer to several X-ray and cryoEM structures, with a variety of ligands bound, to illustrate the underlying atomistic basis for some of the known pharmacological observations of psychedelic drug actions.

8.
J Mol Biol ; : 168809, 2024 Oct 01.
Article in English | MEDLINE | ID: mdl-39362625

ABSTRACT

Membrane proteins are involved in numerous biological processes and represent more than half of all drug targets; thus, structural information on these proteins is invaluable. However, the low expression level of membrane proteins, as well as their poor stability in solution and tendency to precipitate and aggregate, are major bottlenecks in the preparation of purified membrane proteins for structural studies. Traditionally, the evaluation of membrane protein constructs for structural studies has been quite time consuming and expensive since it is necessary to express and purify the proteins on a large scale, particularly for X-ray crystallography. The emergence of fluorescence detection size exclusion chromatography (FSEC) has drastically changed this situation, as this method can be used to rapidly evaluate the expression and behavior of membrane proteins on a small scale without the need for purification. FSEC has become the most widely used method for the screening of expression conditions and sample evaluation for membrane proteins, leading to the successful determination of numerous structures. Even in the era of cryo-EM, FSEC and the new generation of FSEC derivative methods are being widely used in various manners to facilitate structural analysis. In addition, the application of FSEC is not limited to structural analysis; this method is also widely used for functional analysis of membrane proteins, including for analysis of oligomerization state, screening of antibodies and ligands, and affinity profiling. This review presents the latest advances and applications in membrane protein expression screening and sample evaluation, with a particular focus on FSEC methods.

9.
Int J Mol Sci ; 25(17)2024 Sep 08.
Article in English | MEDLINE | ID: mdl-39273672

ABSTRACT

Protein dynamics play a crucial role in biological function, encompassing motions ranging from atomic vibrations to large-scale conformational changes. Recent advancements in experimental techniques, computational methods, and artificial intelligence have revolutionized our understanding of protein dynamics. Nuclear magnetic resonance spectroscopy provides atomic-resolution insights, while molecular dynamics simulations offer detailed trajectories of protein motions. Computational methods applied to X-ray crystallography and cryo-electron microscopy (cryo-EM) have enabled the exploration of protein dynamics, capturing conformational ensembles that were previously unattainable. The integration of machine learning, exemplified by AlphaFold2, has accelerated structure prediction and dynamics analysis. These approaches have revealed the importance of protein dynamics in allosteric regulation, enzyme catalysis, and intrinsically disordered proteins. The shift towards ensemble representations of protein structures and the application of single-molecule techniques have further enhanced our ability to capture the dynamic nature of proteins. Understanding protein dynamics is essential for elucidating biological mechanisms, designing drugs, and developing novel biocatalysts, marking a significant paradigm shift in structural biology and drug discovery.


Subject(s)
Cryoelectron Microscopy , Machine Learning , Molecular Dynamics Simulation , Proteins , Cryoelectron Microscopy/methods , Proteins/chemistry , Proteins/metabolism , Protein Conformation , Humans , Nuclear Magnetic Resonance, Biomolecular/methods , Magnetic Resonance Spectroscopy/methods
10.
Cell ; 187(20): 5587-5603.e19, 2024 Oct 03.
Article in English | MEDLINE | ID: mdl-39293445

ABSTRACT

Filoviruses, including the Ebola and Marburg viruses, cause hemorrhagic fevers with up to 90% lethality. The viral nucleocapsid is assembled by polymerization of the nucleoprotein (NP) along the viral genome, together with the viral proteins VP24 and VP35. We employed cryo-electron tomography of cells transfected with viral proteins and infected with model Ebola virus to illuminate assembly intermediates, as well as a 9 Å map of the complete intracellular assembly. This structure reveals a previously unresolved third and outer layer of NP complexed with VP35. The intrinsically disordered region, together with the C-terminal domain of this outer layer of NP, provides the constant width between intracellular nucleocapsid bundles and likely functions as a flexible tether to the viral matrix protein in the virion. A comparison of intracellular nucleocapsids with prior in-virion nucleocapsid structures reveals that the nucleocapsid further condenses vertically in the virion. The interfaces responsible for nucleocapsid assembly are highly conserved and offer targets for broadly effective antivirals.


Subject(s)
Ebolavirus , Electron Microscope Tomography , Nucleocapsid , Virus Assembly , Ebolavirus/ultrastructure , Ebolavirus/chemistry , Ebolavirus/metabolism , Ebolavirus/physiology , Nucleocapsid/metabolism , Nucleocapsid/ultrastructure , Nucleocapsid/chemistry , Humans , Cryoelectron Microscopy/methods , Nucleocapsid Proteins/chemistry , Nucleocapsid Proteins/metabolism , Nucleocapsid Proteins/ultrastructure , Nucleoproteins/chemistry , Nucleoproteins/metabolism , Nucleoproteins/ultrastructure , Animals , Viral Proteins/metabolism , Viral Proteins/chemistry , Viral Proteins/ultrastructure , Models, Molecular , Virion/ultrastructure , Virion/metabolism , Hemorrhagic Fever, Ebola/virology , Chlorocebus aethiops
11.
Structure ; 2024 Sep 10.
Article in English | MEDLINE | ID: mdl-39305901

ABSTRACT

The dystrophin-glycoprotein-complex (DGC), anchored by the transmembrane protein dystroglycan, functions to mechanically link the extracellular matrix and actin cytoskeleton. Breaking this connection is associated with diseases such as muscular dystrophy, yet cleavage of dystroglycan by matrix-metalloproteinases (MMPs) remains an understudied mechanism to disrupt the DGC. We determined the crystal structure of the membrane-adjacent domain (amino acids 491-722) of E. coli expressed human dystroglycan to understand MMP cleavage regulation. The structural model includes tandem immunoglobulin-like (IGL) and sperm/enterokinase/agrin-like (SEAL) domains, which support proteolysis in diverse receptors to facilitate mechanotransduction, membrane protection, and viral entry. The structure reveals a C-terminal extension that buries the MMP site by packing into a hydrophobic pocket, a unique mechanism of MMP cleavage regulation. We further demonstrate structure-guided and disease-associated mutations disrupt proteolytic regulation using a cell-surface proteolysis assay. Thus disrupted proteolysis is a potentially relevant mechanism for "breaking" the DGC link to contribute to disease pathogenesis.

12.
Front Chem ; 12: 1426211, 2024.
Article in English | MEDLINE | ID: mdl-39246722

ABSTRACT

Understanding the functions of metal ions in biological systems is crucial for many aspects of research, including deciphering their roles in diseases and potential therapeutic use. Structural information about the molecular or atomic details of these interactions, generated by methods like X-ray crystallography, cryo-electron microscopy, or nucleic magnetic resonance, frequently provides details that no other method can. As with any experimental method, they have inherent limitations that sometimes lead to an erroneous interpretation. This manuscript highlights different aspects of structural data available for metal-protein complexes. We examine the quality of modeling metal ion binding sites across different structure determination methods, where different kinds of errors stem from, and how they can impact correct interpretations and conclusions.

13.
Toxins (Basel) ; 16(9)2024 Sep 19.
Article in English | MEDLINE | ID: mdl-39330864

ABSTRACT

ABC toxin complexes are a class of protein toxin translocases comprised of a multimeric assembly of protein subunits. Each subunit displays a unique composition, contributing to the formation of a syringe-like nano-machine with natural cargo carrying, targeting, and translocation capabilities. Many of these toxins are insecticidal, drawing increasing interest in agriculture for use as biological pesticides. The A subunit (TcA) is the largest subunit of the complex and contains domains associated with membrane permeation and targeting. The B and C subunits, TcB and TcC, respectively, package into a cocoon-like structure that contains a toxic peptide and are coupled to TcA to form a continuous channel upon final assembly. In this review, we outline the current understanding and gaps in the knowledge pertaining to ABC toxins, highlighting seven published structures of TcAs and how these structures have led to a better understanding of the mechanism of host tropism and toxin translocation. We also highlight similarities and differences between homologues that contribute to variations in host specificity and conformational change. Lastly, we review the biotechnological potential of ABC toxins as both pesticides and cargo-carrying shuttles that enable the transport of peptides into cells.


Subject(s)
ATP-Binding Cassette Transporters , Animals , Humans , ATP-Binding Cassette Transporters/chemistry , ATP-Binding Cassette Transporters/metabolism , Toxins, Biological/chemistry , Toxins, Biological/metabolism , Host Tropism
14.
J Bacteriol ; : e0014024, 2024 Sep 16.
Article in English | MEDLINE | ID: mdl-39283106

ABSTRACT

Powered by ion transport across the cell membrane, conserved ion-powered rotary motors (IRMs) drive bacterial motility by generating torque on the rotor of the bacterial flagellar motor. Homologous heteroheptameric IRMs have been structurally characterized in ion channels such as Tol/Ton/Exb/Gld, and most recently in phage defense systems such as Zor. Functional stator complexes synthesized from chimeras of PomB/MotB (PotB) have been used to study flagellar rotation at low ion-motive force achieved via reduced external sodium concentration. The function of such chimeras is highly sensitive to the location of the fusion site, and these hybrid proteins have thus far been arbitrarily designed. To date, no chimeras have been constructed using interchange of components from Tol/Ton/Exb/Gld and other ion-powered motors with more distant homology. Here, we synthesized chimeras of MotAB, PomAPotB, and ExbBD to assess their capacity for cross-compatibility. We generated motile strains powered by stator complexes with B-subunit chimeras. This motility was further optimized by directed evolution. Whole-genome sequencing of these strains revealed that motility-enhancing residue changes occurred in the A-subunit and at the peptidoglycan binding domain of the B-unit, which could improve motility. Overall, our work highlights the complexity of stator architecture and identifies the challenges associated with the rational design of chimeric IRMs. IMPORTANCE: Ion-powered rotary motors (IRMs) underpin the rotation of one of nature's oldest wheels, the flagellar motor. Recent structures show that this complex appears to be a fundamental molecular module with diverse biological utility where electrical energy is coupled to torque. Here, we attempted to rationally design chimeric IRMs to explore the cross-compatibility of these ancient motors. We succeeded in making one working chimera of a flagellar motor and a non-flagellar transport system protein. This had only a short hybrid stretch in the ion-conducting channel, and function was subsequently improved through additional substitutions at sites distant from this hybrid pore region. Our goal was to test the cross-compatibility of these homologous systems and highlight challenges arising when engineering new rotary motors.

15.
bioRxiv ; 2024 Sep 09.
Article in English | MEDLINE | ID: mdl-39314299

ABSTRACT

The structures of RNA:RNA complexes regulate many biological processes. Despite their importance, protein-free RNA:RNA complexes represent a tiny fraction of experimentally-determined structures. Here, we describe a joint small-angle X-ray and neutron scattering (SAXS/SANS) approach to structurally interrogate conformational changes in a model RNA:RNA complex. Using SAXS, we measured the solution structures of the individual RNAs in their free state and of the overall RNA:RNA complex. With SANS, we demonstrate, as a proof-of-principle, that isotope labeling and contrast matching (CM) can be combined to probe the bound state structure of an RNA within a selectively deuterated RNA:RNA complex. Furthermore, we show that experimental scattering data can validate and improve predicted AlphaFold 3 RNA:RNA complex structures to reflect its solution structure. Our work demonstrates that in silico modeling, SAXS, and CM-SANS can be used in concert to directly analyze conformational changes within RNAs when in complex, enhancing our understanding of RNA structure in functional assemblies.

16.
Elife ; 132024 Sep 02.
Article in English | MEDLINE | ID: mdl-39221775

ABSTRACT

BTB (bric-a-brack, Tramtrack, and broad complex) is a diverse group of protein-protein interaction domains found within metazoan proteins. Transcription factors contain a dimerizing BTB subtype with a characteristic N-terminal extension. The Tramtrack group (TTK) is a distinct type of BTB domain, which can multimerize. Single-particle cryo-EM microscopy revealed that the TTK-type BTB domains assemble into a hexameric structure consisting of three canonical BTB dimers connected through a previously uncharacterized interface. We demonstrated that the TTK-type BTB domains are found only in Arthropods and have undergone lineage-specific expansion in modern insects. The Drosophila genome encodes 24 transcription factors with TTK-type BTB domains, whereas only four have non-TTK-type BTB domains. Yeast two-hybrid analysis revealed that the TTK-type BTB domains have an unusually broad potential for heteromeric associations presumably through a dimer-dimer interaction interface. Thus, the TTK-type BTB domains are a structurally and functionally distinct group of protein domains specific to Arthropodan transcription factors.


Subject(s)
Arthropods , Protein Multimerization , Transcription Factors , Animals , Arthropods/metabolism , Arthropods/genetics , Cryoelectron Microscopy , Drosophila , Protein Domains , Transcription Factors/metabolism , Transcription Factors/chemistry , Transcription Factors/genetics , Two-Hybrid System Techniques
17.
Structure ; 2024 Sep 10.
Article in English | MEDLINE | ID: mdl-39293443

ABSTRACT

Recent studies have demonstrated BamA, the central component of the ß-barrel assembly machinery (BAM), as an important therapeutic target to combat infections caused by Acinetobacter baumannii and other Gram-negative pathogens. Homology modeling indicates BamA in A. baumannii consists of five polypeptide transport-associated (POTRA) domains and a ß-barrel membrane domain. We characterized the POTRA domains of BamA from A. baumannii in solution using size-exclusion chromatography small angle X-ray scattering (SEC-SAXS) analysis and determined crystal structures in two conformational states that are drastically different than those previously observed in BamA from other bacteria, indicating that the POTRA domains are even more conformationally dynamic than has been observed previously. Molecular dynamics simulations of the POTRA domains from A. baumannii and Escherichia coli allowed us to identify key structural features that contribute to the observed novel states. Together, these studies expand on our current understanding of the conformational plasticity within BamA across differing bacterial species.

18.
Enzymes ; 56: 55-83, 2024.
Article in English | MEDLINE | ID: mdl-39304291

ABSTRACT

Tyrosinase, a pivotal enzyme in melanin biosynthesis, orchestrates the pigmentation process in humans, affecting skin, hair, and eye color. This chapter examines the three-dimensional structure and functional aspects of tyrosinases from various sources, highlighting their di-metal ion coordination crucial for catalytic activity. I explore the biochemical pathwayscheme catalyzed by tyrosinase, specifically the oxidation of L-tyrosine to L-dopaquinone, a precursor in melanin synthesis. Detailed structural analyses, including 3D structures obtained from X-ray crystallography and computational modeling, reveal key insights into the enzyme's active site, variations among tyrosinases, and substrate binding mechanisms. Furthermore, the chapter investigates the role of human tyrosinase variants, their inhibitors, essential for developing therapeutic and cosmetic applications targeting hyperpigmentation disorders. Structural characterizations of tyrosinase-inhibitor complexes provide a foundation for designing effective inhibitors, with compounds like kojic acid, L-mimosine, and (S)-3-amino-tyrosine demonstrating significant inhibitory potential. This comprehensive examination of the structure, function, and inhibition mechanisms of tyrosinase offers avenues for innovative treatments in biotechnology, health, and beyond.


Subject(s)
Monophenol Monooxygenase , Humans , Catalytic Domain , Crystallography, X-Ray , Enzyme Inhibitors/pharmacology , Enzyme Inhibitors/chemistry , Melanins/metabolism , Melanins/biosynthesis , Melanins/chemistry , Models, Molecular , Monophenol Monooxygenase/antagonists & inhibitors , Monophenol Monooxygenase/metabolism , Monophenol Monooxygenase/chemistry , Protein Conformation , Tyrosine/chemistry , Tyrosine/metabolism
19.
J Clin Invest ; 134(18)2024 Sep 17.
Article in English | MEDLINE | ID: mdl-39286976

ABSTRACT

CD8+ T cells destroy insulin-producing pancreatic ß cells in type 1 diabetes through HLA class I-restricted presentation of self-antigens. Combinatorial peptide library screening was used to produce a preferred peptide recognition landscape for a patient-derived T cell receptor (TCR) that recognized the preproinsulin-derived (PPI-derived) peptide sequence LWMRLLPLL in the context of disease risk allele HLA A*24:02. Data were used to generate a strong superagonist peptide, enabling production of an autoimmune HLA A*24:02-peptide-TCR structure by crystal seeding. TCR binding to the PPI epitope was strongly focused on peptide residues Arg4 and Leu5, with more flexibility at other positions, allowing the TCR to strongly engage many peptides derived from pathogenic bacteria. We confirmed an epitope from Klebsiella that was recognized by PPI-reactive T cells from 3 of 3 HLA A*24:02+ patients. Remarkably, the same epitope selected T cells from 7 of 8 HLA A*24+ healthy donors that cross-reacted with PPI, leading to recognition and killing of HLA A*24:02+ cells expressing PPI. These data provide a mechanism by which molecular mimicry between pathogen and self-antigens could have resulted in the breaking of self-tolerance to initiate disease.


Subject(s)
Diabetes Mellitus, Type 1 , HLA-A24 Antigen , Insulin , Protein Precursors , Receptors, Antigen, T-Cell , Humans , Diabetes Mellitus, Type 1/immunology , Diabetes Mellitus, Type 1/genetics , Protein Precursors/immunology , Protein Precursors/genetics , Protein Precursors/metabolism , Insulin/immunology , Insulin/metabolism , HLA-A24 Antigen/immunology , HLA-A24 Antigen/genetics , Receptors, Antigen, T-Cell/immunology , Receptors, Antigen, T-Cell/genetics , Epitopes, T-Lymphocyte/immunology , Epitopes, T-Lymphocyte/genetics , CD8-Positive T-Lymphocytes/immunology , Female , Male
20.
Cell Rep ; 43(9): 114708, 2024 Sep 24.
Article in English | MEDLINE | ID: mdl-39243373

ABSTRACT

Lassa fever continues to be a major public health burden in West Africa, yet effective therapies or vaccines are lacking. The isolation of protective neutralizing antibodies against the Lassa virus glycoprotein complex (GPC) justifies the development of vaccines that can elicit strong neutralizing antibody responses. However, Lassa vaccine candidates have generally been unsuccessful at doing so, and the associated antibody responses to these vaccines remain poorly characterized. Here, we establish an electron microscopy-based epitope mapping workflow that enables high-resolution structural characterization of polyclonal antibodies to the GPC. By applying this method to rabbits vaccinated with a recombinant GPC vaccine and a GPC-derived virus-like particle, we reveal determinants of neutralization that involve epitopes of the GPC-A competition cluster. Furthermore, by identifying undescribed immunogenic off-target epitopes, we expose the challenges that recombinant GPC vaccines face. By enabling detailed polyclonal antibody characterization, our work ushers in a next generation of more rational Lassa vaccine design.


Subject(s)
Antibodies, Neutralizing , Antibodies, Viral , Lassa Fever , Lassa virus , Lassa virus/immunology , Animals , Antibodies, Neutralizing/immunology , Rabbits , Antibodies, Viral/immunology , Lassa Fever/immunology , Lassa Fever/prevention & control , Epitopes/immunology , Viral Vaccines/immunology , Humans , Epitope Mapping , Antibody Formation/immunology
SELECTION OF CITATIONS
SEARCH DETAIL