Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
Virology ; 565: 13-21, 2022 01 02.
Article in English | MEDLINE | ID: mdl-34626907

ABSTRACT

Eastern equine encephalitis virus (EEEV), western equine encephalitis virus (WEEV) and Venezuelan equine encephalitis virus (VEEV) can cause fatal encephalitis in humans and equids. Some MAbs to the E1 glycoprotein are known to be cross-reactive, weakly neutralizing in vitro but can protect from disease in animal models. We investigated the mechanism of neutralization of VEEV infection by the broadly cross-reactive E1-specific MAb 1A4B-6. 1A4B-6 protected 3-week-old Swiss Webster mice prophylactically from lethal VEEV challenge. Likewise, 1A4B-6 inhibited virus growth in vitro at a pre-attachment step after virions were incubated at 37 °C and inhibited virus-mediated cell fusion. Amino acid residue N100 in the fusion loop of E1 protein was identified as critical for binding. The potential to elicit broadly cross-reactive MAbs with limited virus neutralizing activity in vitro but that can inhibit virus entry and protect animals from infection merits further exploration for vaccine and therapeutic developmental research.


Subject(s)
Antibodies, Viral/immunology , Encephalitis Virus, Venezuelan Equine/immunology , Encephalitis Virus, Venezuelan Equine/metabolism , Encephalomyelitis, Venezuelan Equine/immunology , Encephalomyelitis, Venezuelan Equine/virology , Viral Envelope Proteins/immunology , Virus Replication/drug effects , Alphavirus/immunology , Alphavirus Infections/immunology , Amino Acid Sequence , Animals , Antibodies, Monoclonal/immunology , Antibodies, Neutralizing/immunology , Cell Line , Chlorocebus aethiops , Cross Reactions , Encephalomyelitis, Venezuelan Equine/therapy , Glycoproteins/immunology , Immunotherapy , Mice , Protein Binding , Vero Cells , Viral Envelope Proteins/metabolism , Virion/immunology , Virion/metabolism
2.
Cell ; 184(17): 4414-4429.e19, 2021 08 19.
Article in English | MEDLINE | ID: mdl-34416146

ABSTRACT

Alphaviruses are emerging, mosquito-transmitted pathogens that cause musculoskeletal and neurological disease in humans. Although neutralizing antibodies that inhibit individual alphaviruses have been described, broadly reactive antibodies that protect against both arthritogenic and encephalitic alphaviruses have not been reported. Here, we identify DC2.112 and DC2.315, two pan-protective yet poorly neutralizing human monoclonal antibodies (mAbs) that avidly bind to viral antigen on the surface of cells infected with arthritogenic and encephalitic alphaviruses. These mAbs engage a conserved epitope in domain II of the E1 protein proximal to and within the fusion peptide. Treatment with DC2.112 or DC2.315 protects mice against infection by both arthritogenic (chikungunya and Mayaro) and encephalitic (Venezuelan, Eastern, and Western equine encephalitis) alphaviruses through multiple mechanisms, including inhibition of viral egress and monocyte-dependent Fc effector functions. These findings define a conserved epitope recognized by weakly neutralizing yet protective antibodies that could be targeted for pan-alphavirus immunotherapy and vaccine design.


Subject(s)
Alphavirus/immunology , Antibodies, Viral/immunology , Conserved Sequence/immunology , Epitopes/immunology , Viral Proteins/immunology , Alphavirus Infections/immunology , Alphavirus Infections/virology , Amino Acid Sequence , Animals , Antibodies, Monoclonal/immunology , Chikungunya Fever/immunology , Chikungunya Fever/virology , Chikungunya virus/immunology , Chlorocebus aethiops , Epitope Mapping , Epitopes/chemistry , Humans , Male , Mice, Inbred C57BL , Models, Biological , Monocytes/metabolism , Vero Cells , Viral Proteins/chemistry , Virus Release
3.
Nature ; 557(7706): 570-574, 2018 05.
Article in English | MEDLINE | ID: mdl-29769725

ABSTRACT

Arthritogenic alphaviruses comprise a group of enveloped RNA viruses that are transmitted to humans by mosquitoes and cause debilitating acute and chronic musculoskeletal disease 1 . The host factors required for alphavirus entry remain poorly characterized 2 . Here we use a genome-wide CRISPR-Cas9-based screen to identify the cell adhesion molecule Mxra8 as an entry mediator for multiple emerging arthritogenic alphaviruses, including chikungunya, Ross River, Mayaro and O'nyong nyong viruses. Gene editing of mouse Mxra8 or human MXRA8 resulted in reduced levels of viral infection of cells and, reciprocally, ectopic expression of these genes resulted in increased infection. Mxra8 bound directly to chikungunya virus particles and enhanced virus attachment and internalization into cells. Consistent with these findings, Mxra8-Fc fusion protein or anti-Mxra8 monoclonal antibodies blocked chikungunya virus infection in multiple cell types, including primary human synovial fibroblasts, osteoblasts, chondrocytes and skeletal muscle cells. Mutagenesis experiments suggest that Mxra8 binds to a surface-exposed region across the A and B domains of chikungunya virus E2 protein, which are a speculated site of attachment. Finally, administration of the Mxra8-Fc protein or anti-Mxra8 blocking antibodies to mice reduced chikungunya and O'nyong nyong virus infection as well as associated foot swelling. Pharmacological targeting of Mxra8 could form a strategy for mitigating infection and disease by multiple arthritogenic alphaviruses.


Subject(s)
Chikungunya virus/metabolism , Immunoglobulins/metabolism , Membrane Proteins/metabolism , O'nyong-nyong Virus/metabolism , Receptors, Virus/metabolism , 3T3 Cells , Animals , Antibodies, Blocking/immunology , CRISPR-Cas Systems/genetics , Chikungunya virus/pathogenicity , Chondrocytes/metabolism , Fibroblasts/metabolism , Humans , Immunoglobulins/immunology , Male , Membrane Proteins/antagonists & inhibitors , Membrane Proteins/immunology , Mice , Muscle, Skeletal/cytology , Muscle, Skeletal/metabolism , O'nyong-nyong Virus/pathogenicity , Osteoblasts/metabolism , Receptors, Fc/metabolism , Receptors, Virus/deficiency , Receptors, Virus/genetics
4.
PLoS Pathog ; 14(2): e1006837, 2018 02.
Article in English | MEDLINE | ID: mdl-29470533

ABSTRACT

Respiratory syncytial virus (RSV) is a major human pathogen that infects the majority of children by two years of age. The RSV fusion (F) protein is a primary target of human antibodies, and it has several antigenic regions capable of inducing neutralizing antibodies. Antigenic site IV is preserved in both the pre-fusion and post-fusion conformations of RSV F. Antibodies to antigenic site IV have been described that bind and neutralize both RSV and human metapneumovirus (hMPV). To explore the diversity of binding modes at antigenic site IV, we generated a panel of four new human monoclonal antibodies (mAbs) and competition-binding suggested the mAbs bind at antigenic site IV. Mutagenesis experiments revealed that binding and neutralization of two mAbs (3M3 and 6F18) depended on arginine (R) residue R429. We discovered two R429-independent mAbs (17E10 and 2N6) at this site that neutralized an RSV R429A mutant strain, and one of these mAbs (17E10) neutralized both RSV and hMPV. To determine the mechanism of cross-reactivity, we performed competition-binding, recombinant protein mutagenesis, peptide binding, and electron microscopy experiments. It was determined that the human cross-reactive mAb 17E10 binds to RSV F with a binding pose similar to 101F, which may be indicative of cross-reactivity with hMPV F. The data presented provide new concepts in RSV immune recognition and vaccine design, as we describe the novel idea that binding pose may influence mAb cross-reactivity between RSV and hMPV. Characterization of the site IV epitope bound by human antibodies may inform the design of a pan-Pneumovirus vaccine.


Subject(s)
Antibodies, Monoclonal/metabolism , Antibodies, Neutralizing/metabolism , Antibodies, Viral/metabolism , Epitopes/metabolism , Respiratory Syncytial Virus, Human/metabolism , Viral Fusion Proteins/metabolism , Amino Acid Sequence , Amino Acid Substitution , Antibodies, Monoclonal/chemistry , Antibodies, Neutralizing/chemistry , Antibodies, Viral/chemistry , Antibody Specificity , Binding Sites, Antibody , Binding, Competitive , Cross Reactions , Epitope Mapping , Humans , Kinetics , Metapneumovirus/immunology , Metapneumovirus/metabolism , Microscopy, Electron , Mutation , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Respiratory Syncytial Virus, Human/immunology , Viral Fusion Proteins/antagonists & inhibitors , Viral Fusion Proteins/genetics
5.
Proc Natl Acad Sci U S A ; 113(44): E6849-E6858, 2016 11 01.
Article in English | MEDLINE | ID: mdl-27791117

ABSTRACT

Palivizumab was the first antiviral monoclonal antibody (mAb) approved for therapeutic use in humans, and remains a prophylactic treatment for infants at risk for severe disease because of respiratory syncytial virus (RSV). Palivizumab is an engineered humanized version of a murine mAb targeting antigenic site II of the RSV fusion (F) protein, a key target in vaccine development. There are limited reported naturally occurring human mAbs to site II; therefore, the structural basis for human antibody recognition of this major antigenic site is poorly understood. Here, we describe a nonneutralizing class of site II-specific mAbs that competed for binding with palivizumab to postfusion RSV F protein. We also describe two classes of site II-specific neutralizing mAbs, one of which escaped competition with nonneutralizing mAbs. An X-ray crystal structure of the neutralizing mAb 14N4 in complex with F protein showed that the binding angle at which human neutralizing mAbs interact with antigenic site II determines whether or not nonneutralizing antibodies compete with their binding. Fine-mapping studies determined that nonneutralizing mAbs that interfere with binding of neutralizing mAbs recognize site II with a pose that facilitates binding to an epitope containing F surface residues on a neighboring protomer. Neutralizing antibodies, like motavizumab and a new mAb designated 3J20 that escape interference by the inhibiting mAbs, avoid such contact by binding at an angle that is shifted away from the nonneutralizing site. Furthermore, binding to rationally and computationally designed site II helix-loop-helix epitope-scaffold vaccines distinguished neutralizing from nonneutralizing site II antibodies.


Subject(s)
Antibodies, Neutralizing/immunology , Antibodies, Viral/immunology , Respiratory Syncytial Virus, Human/immunology , Viral Fusion Proteins/immunology , Animals , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/pharmacology , Antibodies, Monoclonal, Humanized/immunology , Antibodies, Neutralizing/chemistry , Antiviral Agents/pharmacology , Cell Line , Crystallography, X-Ray , Epitope Mapping , Epitopes/immunology , Humans , Mice , Mutagenesis , Palivizumab/pharmacology , Respiratory Syncytial Virus Vaccines/chemistry , Respiratory Syncytial Virus Vaccines/immunology , Respiratory Syncytial Virus, Human/drug effects
6.
MAbs ; 8(1): 129-40, 2016.
Article in English | MEDLINE | ID: mdl-26491897

ABSTRACT

Monitoring antigen-specific memory B cells and the antibodies they encode is important for understanding the specificity, breadth and duration of immune response to an infection or vaccination. The antibodies isolated could further help design vaccine antigens for raising relevant protective immune responses. However, developing assays to measure and isolate antigen-specific memory B cells is technically challenging due to the low frequencies of these cells that exist in the circulating blood. Here, we describe a flow cytometry method to identify and isolate dengue envelope-specific memory B cells using a labeled dengue envelope protein. We enumerated dengue-envelope specific memory B cells from a cohort of dengue seropositive donors using this direct flow cytometry assay. A more established and conventional assay, the cultured B ELISPOT, was used as a benchmark comparator. Furthermore, we were able to confirm the single-sorted memory B-cell specificity by culturing B cells and differentiating them into plasma cells using cell lines expressing CD40L. The culture supernatants were assayed for antigen binding and the ability of the antibodies to neutralize the cognate dengue virus. Moreover, we successfully isolated the heavy and light Ig sequences and expressed them as full-length recombinant antibodies to reproduce the activity seen in culture supernatants. Mapping of these antibodies revealed a novel epitope for dengue 2 virus serotype. In conclusion, we established a reproducible methodology to enumerate antigen-specific memory B cells and assay their encoded antibodies for functional characterization.


Subject(s)
Antibodies, Neutralizing/immunology , Antibodies, Viral/immunology , B-Lymphocytes/immunology , Dengue Virus/immunology , Immunologic Memory , Viral Envelope Proteins/immunology , B-Lymphocytes/cytology , Cell Culture Techniques , Female , Flow Cytometry , Humans , Male
7.
Cell Rep ; 13(11): 2553-2564, 2015 Dec 22.
Article in English | MEDLINE | ID: mdl-26686638

ABSTRACT

We evaluated the mechanism by which neutralizing human monoclonal antibodies inhibit chikungunya virus (CHIKV) infection. Potently neutralizing antibodies (NAbs) blocked infection at multiple steps of the virus life cycle, including entry and release. Cryo-electron microscopy structures of Fab fragments of two human NAbs and chikungunya virus-like particles showed a binding footprint that spanned independent domains on neighboring E2 subunits within one viral spike, suggesting a mechanism for inhibiting low-pH-dependent membrane fusion. Detailed epitope mapping identified amino acid E2-W64 as a critical interaction residue. An escape mutation (E2-W64G) at this residue rendered CHIKV attenuated in mice. Consistent with these data, CHIKV-E2-W64G failed to emerge in vivo under the selection pressure of one of the NAbs, IM-CKV063. As our study suggests that antibodies engaging the residue E2-W64 can potently inhibit CHIKV at multiple stages of infection, antibody-based therapies or immunogens that target this region might have protective value.


Subject(s)
Antibodies, Monoclonal/immunology , Antibodies, Neutralizing/immunology , Chikungunya virus/metabolism , Epitopes/immunology , Animals , Arthritis/metabolism , Arthritis/pathology , Chemokines/analysis , Chikungunya virus/genetics , Chikungunya virus/pathogenicity , Chlorocebus aethiops , Cytokines/analysis , Disease Models, Animal , Epitope Mapping , Genotype , Humans , Membrane Fusion , Mice , Mice, Inbred C57BL , Molecular Dynamics Simulation , Mutagenesis, Site-Directed , Protein Structure, Quaternary , Vero Cells , Viral Envelope Proteins/genetics , Viral Envelope Proteins/metabolism , Virus Internalization
8.
Cell ; 163(5): 1095-1107, 2015 Nov 19.
Article in English | MEDLINE | ID: mdl-26553503

ABSTRACT

We screened a panel of mouse and human monoclonal antibodies (MAbs) against chikungunya virus and identified several with inhibitory activity against multiple alphaviruses. Passive transfer of broadly neutralizing MAbs protected mice against infection by chikungunya, Mayaro, and O'nyong'nyong alphaviruses. Using alanine-scanning mutagenesis, loss-of-function recombinant proteins and viruses, and multiple functional assays, we determined that broadly neutralizing MAbs block multiple steps in the viral lifecycle, including entry and egress, and bind to a conserved epitope on the B domain of the E2 glycoprotein. A 16 Å resolution cryo-electron microscopy structure of a Fab fragment bound to CHIKV E2 B domain provided an explanation for its neutralizing activity. Binding to the B domain was associated with repositioning of the A domain of E2 that enabled cross-linking of neighboring spikes. Our results suggest that B domain antigenic determinants could be targeted for vaccine or antibody therapeutic development against multiple alphaviruses of global concern.


Subject(s)
Alphavirus Infections/immunology , Alphavirus/immunology , Antibodies, Monoclonal/immunology , Antibodies, Neutralizing/immunology , Antibodies, Viral/immunology , Epitopes , Viral Envelope Proteins/immunology , Alphavirus/classification , Alphavirus/metabolism , Alphavirus Infections/prevention & control , Alphavirus Infections/therapy , Amino Acid Sequence , Animals , Chikungunya virus/chemistry , Chikungunya virus/immunology , Cryoelectron Microscopy , Glycoproteins/chemistry , Glycoproteins/immunology , Humans , Immunoglobulin Fab Fragments/immunology , Immunoglobulin Fab Fragments/ultrastructure , Mice , Models, Molecular , Molecular Sequence Data , Protein Structure, Tertiary , Sequence Alignment , Viral Envelope Proteins/chemistry , Viral Vaccines/immunology , Virus Internalization
9.
Proc Natl Acad Sci U S A ; 112(45): 13898-903, 2015 Nov 10.
Article in English | MEDLINE | ID: mdl-26504196

ABSTRACT

Chikungunya virus (CHIKV) is a mosquito-transmitted alphavirus that causes severe acute and chronic disease in humans. Although highly inhibitory murine and human monoclonal antibodies (mAbs) have been generated, the structural basis of their neutralizing activity remains poorly characterized. Here, we determined the cryo-EM structures of chikungunya virus-like particles complexed with antibody fragments (Fab) of two highly protective human mAbs, 4J21 and 5M16, that block virus fusion with host membranes. Both mAbs bind primarily to sites within the A and B domains, as well as to the B domain's ß-ribbon connector of the viral glycoprotein E2. The footprints of these antibodies on the viral surface were consistent with results from loss-of-binding studies using an alanine scanning mutagenesis-based epitope mapping approach. The Fab fragments stabilized the position of the B domain relative to the virus, particularly for the complex with 5M16. This finding is consistent with a mechanism of neutralization in which anti-CHIKV mAbs that bridge the A and B domains impede movement of the B domain away from the underlying fusion loop on the E1 glycoprotein and therefore block the requisite pH-dependent fusion of viral and host membranes.


Subject(s)
Antibodies, Monoclonal/immunology , Antibodies, Neutralizing/immunology , Chikungunya Fever/therapy , Chikungunya virus/immunology , Cryoelectron Microscopy/methods , Antibodies, Monoclonal/chemistry , Antibodies, Monoclonal/therapeutic use , Antibodies, Neutralizing/chemistry , Antibodies, Neutralizing/therapeutic use , Humans , Protein Conformation
10.
J Virol ; 89(21): 10982-92, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26311869

ABSTRACT

UNLABELLED: Cocktails of monoclonal antibodies (MAbs) that target the surface glycoprotein (GP) of Ebola virus (EBOV) are effective in nonhuman primate models and have been used under emergency compassionate-treatment protocols in human patients. However, the amino acids that form the detailed binding epitopes for the MAbs in the ZMapp, ZMAb, and the related MB-003 cocktails have yet to be identified. Other binding properties that define how each MAb functionally interacts with GP­such as affinity, epitope conservation, and epitope accessibility­also remain largely unknown. To help define how each MAb interacts with GP, here we used comprehensive alanine-scanning mutagenesis (shotgun mutagenesis), neutralization escape, and whole virion binding to define each MAb's specific epitope, epitope accessibility, epitope conservation, and apparent affinity. Each of the six therapeutic MAbs binds nonidentical epitopes in the GP base, glycan cap, or mucin-like domain. Their apparent affinity, epitope complementarity, and epitope accessibility helps explain why MAbs 4G7 and 13C6 are more protective than 2G4 and 1H3. The mucin-like domain MAbs 6D8 and 13F6 bind with the strongest apparent affinity, helping to explain their effectiveness in vivo despite their inability to neutralize virus. IMPORTANCE: Ebola virus disease (EVD) can be caused by four different filovirus family members, including Ebola virus (EBOV), which infected 10 times more people in western Africa over the last year than all previous EVD outbreaks combined, with a number of cases distributed across the globe by travelers. Cocktails of inhibitory monoclonal antibodies (MAbs), such as ZMAb, MB-003, and in particular ZMapp, have demonstrated in animal models some of the most significant therapeutic potential for treating EVD, and in 2014, 15 patients were treated with ZMapp or ZMAb under compassionate-use protocols. Here, we have defined the epitope features for the most important therapeutic MAbs against EBOV developed to date. Defining the epitopes and binding characteristics for these MAbs, as well as the commonly used reference MAb KZ52, helps explain their breadth of reactivity against different ebolavirus species, predict viral evasion against these MAbs, and design new cocktails of MAbs with improved complementarity.


Subject(s)
Antibodies, Monoclonal/metabolism , Ebolavirus/metabolism , Viral Fusion Proteins/metabolism , Enzyme-Linked Immunosorbent Assay , Epitopes/genetics , Fluorescent Antibody Technique , Humans , Mutagenesis , Neutralization Tests , Protein Binding , Virion/metabolism
11.
Cell Host Microbe ; 18(1): 86-95, 2015 Jul 08.
Article in English | MEDLINE | ID: mdl-26159721

ABSTRACT

Chikungunya virus (CHIKV) is a mosquito-transmitted RNA virus that causes acute febrile infection associated with polyarthralgia in humans. Mechanisms of protective immunity against CHIKV are poorly understood, and no effective therapeutics or vaccines are available. We isolated and characterized human monoclonal antibodies (mAbs) that neutralize CHIKV infectivity. Among the 30 mAbs isolated, 13 had broad and ultrapotent neutralizing activity (IC50 < 10 ng/ml), and all of these mapped to domain A of the E2 envelope protein. Potent inhibitory mAbs blocked post-attachment steps required for CHIKV membrane fusion, and several were protective in a lethal challenge model in immunocompromised mice, even when administered at late time points after infection. These highly protective mAbs could be considered for prevention or treatment of CHIKV infection, and their epitope location in domain A of E2 could be targeted for rational structure-based vaccine development.


Subject(s)
Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/therapeutic use , Antibodies, Viral/immunology , Antibodies, Viral/therapeutic use , Chikungunya Fever/therapy , Chikungunya virus/immunology , Immunization, Passive/methods , Animals , Antibodies, Monoclonal/isolation & purification , Antibodies, Neutralizing/immunology , Antibodies, Neutralizing/isolation & purification , Antibodies, Neutralizing/therapeutic use , Antibodies, Viral/isolation & purification , Chemoprevention/methods , Chikungunya virus/physiology , Disease Models, Animal , Humans , Inhibitory Concentration 50 , Mice , Protein Binding , Survival Analysis , Treatment Outcome , Viral Envelope Proteins/immunology , Virus Internalization/drug effects
12.
J Virol ; 88(24): 14364-79, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25275138

ABSTRACT

UNLABELLED: Chikungunya virus (CHIKV) is a reemerging alphavirus that causes a debilitating arthritic disease and infects millions of people and for which no specific treatment is available. Like many alphaviruses, the structural targets on CHIKV that elicit a protective humoral immune response in humans are poorly defined. Here we used phage display against virus-like particles (VLPs) to isolate seven human monoclonal antibodies (MAbs) against the CHIKV envelope glycoproteins E2 and E1. One MAb, IM-CKV063, was highly neutralizing (50% inhibitory concentration, 7.4 ng/ml), demonstrated high-affinity binding (320 pM), and was capable of therapeutic and prophylactic protection in multiple animal models up to 24 h postexposure. Epitope mapping using a comprehensive shotgun mutagenesis library of 910 mutants with E2/E1 alanine mutations demonstrated that IM-CKV063 binds to an intersubunit conformational epitope on domain A, a functionally important region of E2. MAbs against the highly conserved fusion loop have not previously been reported but were also isolated in our studies. Fusion loop MAbs were broadly cross-reactive against diverse alphaviruses but were nonneutralizing. Fusion loop MAb reactivity was affected by temperature and reactivity conditions, suggesting that the fusion loop is hidden in infectious virions. Visualization of the binding sites of 15 different MAbs on the structure of E2/E1 revealed that all epitopes are located at the membrane-distal region of the E2/E1 spike. Interestingly, epitopes on the exposed topmost and outer surfaces of the E2/E1 trimer structure were neutralizing, whereas epitopes facing the interior of the trimer were not, providing a rationale for vaccine design and therapeutic MAb development using the intact CHIKV E2/E1 trimer. IMPORTANCE: CHIKV is the most important alphavirus affecting humans, resulting in a chronic arthritic condition that can persist for months or years. In recent years, millions of people have been infected globally, and the spread of CHIKV to the Americas is now beginning, with over 100,000 cases occurring in the Caribbean within 6 months of its arrival. Our study reports on seven human MAbs against the CHIKV envelope, including a highly protective MAb and rarely isolated fusion loop MAbs. Epitope mapping of these MAbs demonstrates how some E2/E1 epitopes are exposed or hidden from the human immune system and suggests a structural mechanism by which these MAbs protect (or fail to protect) against CHIKV infection. Our results suggest that the membrane-distal end of CHIKV E2/E1 is the primary target for the humoral immune response to CHIKV, and antibodies targeting the exposed topmost and outer surfaces of the E2/E1 trimer determine the neutralizing efficacy of this response.


Subject(s)
Antibodies, Neutralizing/immunology , Antibodies, Viral/immunology , Chikungunya virus/immunology , Epitopes/immunology , Viral Envelope Proteins/immunology , Animals , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/isolation & purification , Antibodies, Neutralizing/isolation & purification , Antibodies, Viral/isolation & purification , Binding Sites , Cell Surface Display Techniques , Chikungunya Fever/prevention & control , Disease Models, Animal , Epitope Mapping , Female , Humans , Immunization, Passive , Mice, Inbred C57BL , Models, Molecular , Protein Conformation , Survival Analysis
13.
PLoS Negl Trop Dis ; 7(9): e2423, 2013.
Article in English | MEDLINE | ID: mdl-24069479

ABSTRACT

The mosquito-borne alphavirus, chikungunya virus (CHIKV), has recently reemerged, producing the largest epidemic ever recorded for this virus, with up to 6.5 million cases of acute and chronic rheumatic disease. There are currently no licensed vaccines for CHIKV and current anti-inflammatory drug treatment is often inadequate. Here we describe the isolation and characterization of two human monoclonal antibodies, C9 and E8, from CHIKV infected and recovered individuals. C9 was determined to be a potent virus neutralizing antibody and a biosensor antibody binding study demonstrated it recognized residues on intact CHIKV VLPs. Shotgun mutagenesis alanine scanning of 98 percent of the residues in the E1 and E2 glycoproteins of CHIKV envelope showed that the epitope bound by C9 included amino-acid 162 in the acid-sensitive region (ASR) of the CHIKV E2 glycoprotein. The ASR is critical for the rearrangement of CHIKV E2 during fusion and viral entry into host cells, and we predict that C9 prevents these events from occurring. When used prophylactically in a CHIKV mouse model, C9 completely protected against CHIKV viremia and arthritis. We also observed that when administered therapeutically at 8 or 18 hours post-CHIKV challenge, C9 gave 100% protection in a pathogenic mouse model. Given that targeting this novel neutralizing epitope in E2 can potently protect both in vitro and in vivo, it is likely to be an important region both for future antibody and vaccine-based interventions against CHIKV.


Subject(s)
Alphavirus Infections/prevention & control , Antibodies, Monoclonal/immunology , Antibodies, Viral/immunology , Chikungunya virus/immunology , Viral Envelope Proteins/immunology , Alphavirus Infections/immunology , Animals , Antibodies, Monoclonal/administration & dosage , Antibodies, Monoclonal/isolation & purification , Antibodies, Viral/administration & dosage , Antibodies, Viral/isolation & purification , Chikungunya Fever , Disease Models, Animal , Epitope Mapping , Humans , Immunization, Passive , Mice , Mice, Inbred C57BL , Treatment Outcome
14.
J Virol ; 87(1): 52-66, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23077306

ABSTRACT

There are no available vaccines for dengue, the most important mosquito-transmitted viral disease. Mechanistic studies with anti-dengue virus (DENV) human monoclonal antibodies (hMAbs) provide a rational approach to identify and characterize neutralizing epitopes on DENV structural proteins that can serve to inform vaccine strategies. Here, we report a class of hMAbs that is likely to be an important determinant in the human humoral response to DENV infection. In this study, we identified and characterized three broadly neutralizing anti-DENV hMAbs: 4.8A, D11C, and 1.6D. These antibodies were isolated from three different convalescent patients with distinct histories of DENV infection yet demonstrated remarkable similarities. All three hMAbs recognized the E glycoprotein with high affinity, neutralized all four serotypes of DENV, and mediated antibody-dependent enhancement of infection in Fc receptor-bearing cells at subneutralizing concentrations. The neutralization activities of these hMAbs correlated with a strong inhibition of virus-liposome and intracellular fusion, not virus-cell binding. We mapped epitopes of these antibodies to the highly conserved fusion loop region of E domain II. Mutations at fusion loop residues W101, L107, and/or G109 significantly reduced the binding of the hMAbs to E protein. The results show that hMAbs directed against the highly conserved E protein fusion loop block viral entry downstream of virus-cell binding by inhibiting E protein-mediated fusion. Characterization of hMAbs targeting this region may provide new insights into DENV vaccine and therapeutic strategies.


Subject(s)
Antibodies, Monoclonal/immunology , Antibodies, Neutralizing/immunology , Antibodies, Viral/immunology , Dengue Virus/immunology , Viral Envelope Proteins/immunology , Amino Acid Substitution , Animals , Antibodies, Monoclonal/isolation & purification , Antibodies, Neutralizing/isolation & purification , Antibodies, Viral/isolation & purification , Antibody-Dependent Enhancement , Cell Line , Epitope Mapping , Epitopes, B-Lymphocyte/genetics , Epitopes, B-Lymphocyte/immunology , Humans , Macaca mulatta , Mutant Proteins/immunology , Neutralization Tests
SELECTION OF CITATIONS
SEARCH DETAIL
...