Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 189
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Cell ; 185(6): 995-1007.e18, 2022 03 17.
Article in English | MEDLINE | ID: mdl-35303429

ABSTRACT

Several ebolaviruses cause outbreaks of severe disease. Vaccines and monoclonal antibody cocktails are available to treat Ebola virus (EBOV) infections, but not Sudan virus (SUDV) or other ebolaviruses. Current cocktails contain antibodies that cross-react with the secreted soluble glycoprotein (sGP) that absorbs virus-neutralizing antibodies. By sorting memory B cells from EBOV infection survivors, we isolated two broadly reactive anti-GP monoclonal antibodies, 1C3 and 1C11, that potently neutralize, protect rodents from disease, and lack sGP cross-reactivity. Both antibodies recognize quaternary epitopes in trimeric ebolavirus GP. 1C11 bridges adjacent protomers via the fusion loop. 1C3 has a tripartite epitope in the center of the trimer apex. One 1C3 antigen-binding fragment anchors simultaneously to the three receptor-binding sites in the GP trimer, and separate 1C3 paratope regions interact differently with identical residues on the three protomers. A cocktail of both antibodies completely protected nonhuman primates from EBOV and SUDV infections, indicating their potential clinical value.


Subject(s)
Antibodies, Neutralizing , Antibodies, Viral , Ebolavirus , Hemorrhagic Fever, Ebola , Animals , Epitopes , Glycoproteins/chemistry , Protein Subunits
2.
Nat Immunol ; 24(7): 1161-1172, 2023 07.
Article in English | MEDLINE | ID: mdl-37322179

ABSTRACT

Despite the success of COVID-19 vaccines, severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) variants of concern have emerged that can cause breakthrough infections. Although protection against severe disease has been largely preserved, the immunological mediators of protection in humans remain undefined. We performed a substudy on the ChAdOx1 nCoV-19 (AZD1222) vaccinees enrolled in a South African clinical trial. At peak immunogenicity, before infection, no differences were observed in immunoglobulin (Ig)G1-binding antibody titers; however, the vaccine induced different Fc-receptor-binding antibodies across groups. Vaccinees who resisted COVID-19 exclusively mounted FcγR3B-binding antibodies. In contrast, enhanced IgA and IgG3, linked to enriched FcγR2B binding, was observed in individuals who experienced breakthrough. Antibodies unable to bind to FcγR3B led to immune complex clearance and resulted in inflammatory cascades. Differential antibody binding to FcγR3B was linked to Fc-glycosylation differences in SARS-CoV-2-specific antibodies. These data potentially point to specific FcγR3B-mediated antibody functional profiles as critical markers of immunity against COVID-19.


Subject(s)
COVID-19 , Vaccines , Humans , ChAdOx1 nCoV-19 , COVID-19 Vaccines/adverse effects , COVID-19/prevention & control , SARS-CoV-2 , Antibodies, Viral , Immunoglobulin G , Receptors, Fc/genetics , Antibodies, Neutralizing , Vaccination
3.
Cell ; 181(1): 6, 2020 04 02.
Article in English | MEDLINE | ID: mdl-32243796

ABSTRACT

Ervebo is the first licensed vaccine for prevention of Ebola virus disease. The vaccine, originally developed by the Public Health Agency of Canada, is delivered in a single 1 mL dose and has been delivered to >200,000 people in an ongoing 2018-2020 outbreak of disease. To view this Bench to Bedside, open or download the PDF.


Subject(s)
Antibodies, Viral/immunology , Disease Outbreaks/prevention & control , Ebola Vaccines/immunology , Hemorrhagic Fever, Ebola/prevention & control , Mass Vaccination , Drug Approval , Ebolavirus , Humans , Viral Envelope Proteins/immunology
4.
Cell ; 183(1): 1-3, 2020 10 01.
Article in English | MEDLINE | ID: mdl-33007260

ABSTRACT

The pandemic has impacted every scientist differently. Many negative impacts are frequently discussed. Here we highlight unexpected positives that we have found and hope will persist: improved access to experts; deeper and broader human engagement among colleagues, collaborators, and competitors; and significant democratization of research.


Subject(s)
COVID-19/psychology , Pandemics/ethics , Humans , Optimism/psychology , SARS-CoV-2/pathogenicity
5.
Cell ; 182(3): 786-786.e1, 2020 08 06.
Article in English | MEDLINE | ID: mdl-32763187

ABSTRACT

In order to initiate successful infection, viruses have to transmit and deliver their genome from one host cell or organism to another. To achieve this, enveloped viruses must first fuse their membrane with those of the target host cell. Here, we describe the sequence of events leading to the entry of representative enveloped viruses, highlighting the strategies they use to gain access to the host cell cytosol.


Subject(s)
Endocytosis , Endosomes/virology , Membrane Fusion , Virus Internalization , Viruses/metabolism , Animals , Endosomes/metabolism , Humans , Virus Diseases/enzymology , Virus Diseases/metabolism , Viruses/genetics
6.
Cell ; 182(4): 812-827.e19, 2020 08 20.
Article in English | MEDLINE | ID: mdl-32697968

ABSTRACT

A SARS-CoV-2 variant carrying the Spike protein amino acid change D614G has become the most prevalent form in the global pandemic. Dynamic tracking of variant frequencies revealed a recurrent pattern of G614 increase at multiple geographic levels: national, regional, and municipal. The shift occurred even in local epidemics where the original D614 form was well established prior to introduction of the G614 variant. The consistency of this pattern was highly statistically significant, suggesting that the G614 variant may have a fitness advantage. We found that the G614 variant grows to a higher titer as pseudotyped virions. In infected individuals, G614 is associated with lower RT-PCR cycle thresholds, suggestive of higher upper respiratory tract viral loads, but not with increased disease severity. These findings illuminate changes important for a mechanistic understanding of the virus and support continuing surveillance of Spike mutations to aid with development of immunological interventions.


Subject(s)
Betacoronavirus/genetics , Betacoronavirus/pathogenicity , Coronavirus Infections/virology , Pneumonia, Viral/virology , Spike Glycoprotein, Coronavirus/genetics , COVID-19 , Coronavirus Infections/epidemiology , Coronavirus Infections/physiopathology , Epidemiological Monitoring , Genetic Fitness , Genetic Variation , Geographic Information Systems , Hospitalization , Humans , Pandemics , Phylogeny , Pneumonia, Viral/epidemiology , Pneumonia, Viral/physiopathology , Respiratory System/virology , SARS-CoV-2 , Severity of Illness Index , Viral Load
7.
Cell ; 183(4): 996-1012.e19, 2020 11 12.
Article in English | MEDLINE | ID: mdl-33010815

ABSTRACT

Limited knowledge is available on the relationship between antigen-specific immune responses and COVID-19 disease severity. We completed a combined examination of all three branches of adaptive immunity at the level of SARS-CoV-2-specific CD4+ and CD8+ T cell and neutralizing antibody responses in acute and convalescent subjects. SARS-CoV-2-specific CD4+ and CD8+ T cells were each associated with milder disease. Coordinated SARS-CoV-2-specific adaptive immune responses were associated with milder disease, suggesting roles for both CD4+ and CD8+ T cells in protective immunity in COVID-19. Notably, coordination of SARS-CoV-2 antigen-specific responses was disrupted in individuals ≥ 65 years old. Scarcity of naive T cells was also associated with aging and poor disease outcomes. A parsimonious explanation is that coordinated CD4+ T cell, CD8+ T cell, and antibody responses are protective, but uncoordinated responses frequently fail to control disease, with a connection between aging and impaired adaptive immune responses to SARS-CoV-2.


Subject(s)
Adaptive Immunity , Antigens, Viral/immunology , Coronavirus Infections/pathology , Pneumonia, Viral/pathology , Acute Disease , Adult , Aged , Aged, 80 and over , Antibodies, Neutralizing/blood , Antibodies, Viral/blood , Betacoronavirus/immunology , Betacoronavirus/isolation & purification , Betacoronavirus/metabolism , CD4-Positive T-Lymphocytes/cytology , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , COVID-19 , Coronavirus Infections/immunology , Coronavirus Infections/virology , Cytokines/blood , Female , Humans , Male , Middle Aged , Pandemics , Pneumonia, Viral/immunology , Pneumonia, Viral/virology , SARS-CoV-2 , Severity of Illness Index , Young Adult
8.
Cell ; 178(4): 1004-1015.e14, 2019 08 08.
Article in English | MEDLINE | ID: mdl-31398326

ABSTRACT

Lassa virus (LASV) causes hemorrhagic fever and is endemic in West Africa. Protective antibody responses primarily target the LASV surface glycoprotein (GPC), and GPC-B competition group antibodies often show potent neutralizing activity in humans. However, which features confer potent and broadly neutralizing antibody responses is unclear. Here, we compared three crystal structures of LASV GPC complexed with GPC-B antibodies of varying neutralization potency. Each GPC-B antibody recognized an overlapping epitope involved in binding of two adjacent GPC monomers and preserved the prefusion trimeric conformation. Differences among GPC-antibody interactions highlighted specific residues that enhance neutralization. Using structure-guided amino acid substitutions, we increased the neutralization potency and breadth of these antibodies to include all major LASV lineages. The ability to define antibody residues that allow potent and broad neutralizing activity, together with findings from analyses of inferred germline precursors, is critical to develop potent therapeutics and for vaccine design and assessment.


Subject(s)
Antibodies, Neutralizing/immunology , Antibodies, Viral/immunology , Germ Cells/immunology , Lassa Fever/immunology , Lassa virus/immunology , Membrane Glycoproteins/chemistry , Viral Envelope Proteins/chemistry , Animals , Antigens, Viral/immunology , Chlorocebus aethiops , Drosophila/cytology , Epitopes/chemistry , Epitopes/immunology , HEK293 Cells , Humans , Lassa Fever/virology , Membrane Glycoproteins/immunology , Protein Structure, Secondary , Vero Cells , Viral Envelope Proteins/immunology , Viral Vaccines/immunology
9.
Cell ; 177(6): 1566-1582.e17, 2019 05 30.
Article in English | MEDLINE | ID: mdl-31104840

ABSTRACT

Ebola virus (EBOV) remains a public health threat. We performed a longitudinal study of B cell responses to EBOV in four survivors of the 2014 West African outbreak. Infection induced lasting EBOV-specific immunoglobulin G (IgG) antibodies, but their subclass composition changed over time, with IgG1 persisting, IgG3 rapidly declining, and IgG4 appearing late. Striking changes occurred in the immunoglobulin repertoire, with massive recruitment of naive B cells that subsequently underwent hypermutation. We characterized a large panel of EBOV glycoprotein-specific monoclonal antibodies (mAbs). Only a small subset of mAbs that bound glycoprotein by ELISA recognized cell-surface glycoprotein. However, this subset contained all neutralizing mAbs. Several mAbs protected against EBOV disease in animals, including one mAb that targeted an epitope under evolutionary selection during the 2014 outbreak. Convergent antibody evolution was seen across multiple donors, particularly among VH3-13 neutralizing antibodies specific for the GP1 core. Our study provides a benchmark for assessing EBOV vaccine-induced immunity.


Subject(s)
Antibodies, Monoclonal/immunology , B-Lymphocytes/physiology , Hemorrhagic Fever, Ebola/immunology , Adult , Amino Acid Sequence/genetics , Animals , Antibodies, Monoclonal/isolation & purification , Antibodies, Neutralizing/immunology , Antibodies, Viral/immunology , B-Lymphocytes/metabolism , Chlorocebus aethiops , Ebola Vaccines/immunology , Ebolavirus/genetics , Ebolavirus/metabolism , Ebolavirus/pathogenicity , Epitopes/blood , Female , Glycoproteins/genetics , Hemorrhagic Fever, Ebola/metabolism , Hemorrhagic Fever, Ebola/virology , Humans , Immunoglobulin G/immunology , Jurkat Cells , Longitudinal Studies , Male , Mice , Mice, Inbred BALB C , Survivors , Vero Cells , Viral Envelope Proteins/genetics
10.
Cell ; 174(4): 938-952.e13, 2018 08 09.
Article in English | MEDLINE | ID: mdl-30096313

ABSTRACT

Antibodies are promising post-exposure therapies against emerging viruses, but which antibody features and in vitro assays best forecast protection are unclear. Our international consortium systematically evaluated antibodies against Ebola virus (EBOV) using multidisciplinary assays. For each antibody, we evaluated epitopes recognized on the viral surface glycoprotein (GP) and secreted glycoprotein (sGP), readouts of multiple neutralization assays, fraction of virions left un-neutralized, glycan structures, phagocytic and natural killer cell functions elicited, and in vivo protection in a mouse challenge model. Neutralization and induction of multiple immune effector functions (IEFs) correlated most strongly with protection. Neutralization predominantly occurred via epitopes maintained on endosomally cleaved GP, whereas maximal IEF mapped to epitopes farthest from the viral membrane. Unexpectedly, sGP cross-reactivity did not significantly influence in vivo protection. This comprehensive dataset provides a rubric to evaluate novel antibodies and vaccine responses and a roadmap for therapeutic development for EBOV and related viruses.


Subject(s)
Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/isolation & purification , Ebolavirus/immunology , Epitopes/immunology , Hemorrhagic Fever, Ebola/prevention & control , Membrane Glycoproteins/immunology , Animals , Antibodies, Monoclonal/administration & dosage , Female , Hemorrhagic Fever, Ebola/immunology , Hemorrhagic Fever, Ebola/virology , Immunization , Mice , Mice, Inbred BALB C , Treatment Outcome
11.
Immunity ; 55(5): 738-748, 2022 05 10.
Article in English | MEDLINE | ID: mdl-35545026

ABSTRACT

The brutal toll of another viral pandemic can be blunted by investing now in research that uncovers mechanisms of broad-based immunity so we may have vaccines and therapeutics at the ready. We do not know exactly what pathogen may trigger the next wave or next pandemic. We do know, however, that the human immune system must respond and must be bolstered with effective vaccines and other therapeutics to preserve lives and livelihoods. These countermeasures must focus on features conserved among families of pathogens in order to be responsive against something yet to emerge. Here, we focus on immunological approaches to mitigate the impact of the next emerging virus pandemic by developing vaccines that elicit both broadly protective antibodies and T cells. Identifying human immune mechanisms of broad protection against virus families with pandemic potential will be our best defense for humanity in the future.


Subject(s)
COVID-19 , Vaccines , Antibodies, Viral , Humans , Pandemics/prevention & control , SARS-CoV-2 , T-Lymphocytes
12.
Immunity ; 55(2): 355-365.e4, 2022 02 08.
Article in English | MEDLINE | ID: mdl-35090580

ABSTRACT

SARS-CoV-2 mRNA vaccines confer robust protection against COVID-19, but the emergence of variants has generated concerns regarding the protective efficacy of the currently approved vaccines, which lose neutralizing potency against some variants. Emerging data suggest that antibody functions beyond neutralization may contribute to protection from the disease, but little is known about SARS-CoV-2 antibody effector functions. Here, we profiled the binding and functional capacity of convalescent antibodies and Moderna mRNA-1273 COVID-19 vaccine-induced antibodies across SARS-CoV-2 variants of concern (VOCs). Although the neutralizing responses to VOCs decreased in both groups, the Fc-mediated responses were distinct. In convalescent individuals, although antibodies exhibited robust binding to VOCs, they showed compromised interactions with Fc-receptors. Conversely, vaccine-induced antibodies also bound robustly to VOCs but continued to interact with Fc-receptors and mediate antibody effector functions. These data point to a resilience in the mRNA-vaccine-induced humoral immune response that may continue to offer protection from SARS-CoV-2 VOCs independent of neutralization.


Subject(s)
2019-nCoV Vaccine mRNA-1273/immunology , Antibodies, Viral/immunology , COVID-19/metabolism , COVID-19/prevention & control , Receptors, Fc/metabolism , SARS-CoV-2/immunology , 2019-nCoV Vaccine mRNA-1273/administration & dosage , Adult , Antibodies, Neutralizing/immunology , Cross Reactions/immunology , Female , Host-Pathogen Interactions , Humans , Male , Middle Aged , Neutralization Tests , Protein Binding , Spike Glycoprotein, Coronavirus/immunology , Vaccination , Young Adult
13.
Cell ; 164(3): 392-405, 2016 Jan 28.
Article in English | MEDLINE | ID: mdl-26806128

ABSTRACT

Recent studies have suggested that antibody-mediated protection against the Ebolaviruses may be achievable, but little is known about whether or not antibodies can confer cross-reactive protection against viruses belonging to diverse Ebolavirus species, such as Ebola virus (EBOV), Sudan virus (SUDV), and Bundibugyo virus (BDBV). We isolated a large panel of human monoclonal antibodies (mAbs) against BDBV glycoprotein (GP) using peripheral blood B cells from survivors of the 2007 BDBV outbreak in Uganda. We determined that a large proportion of mAbs with potent neutralizing activity against BDBV bind to the glycan cap and recognize diverse epitopes within this major antigenic site. We identified several glycan cap-specific mAbs that neutralized multiple ebolaviruses, including SUDV, and a cross-reactive mAb that completely protected guinea pigs from the lethal challenge with heterologous EBOV. Our results provide a roadmap to develop a single antibody-based treatment effective against multiple Ebolavirus infections.


Subject(s)
Antibodies, Monoclonal/immunology , Antibodies, Neutralizing/immunology , Ebolavirus/immunology , Hemorrhagic Fever, Ebola/immunology , Survivors , Animals , Cross Reactions , Disease Models, Animal , Epitope Mapping , Guinea Pigs , Humans , Mice , Mice, Inbred BALB C , Microscopy, Electron , Models, Molecular , Mutagenesis , Uganda
14.
Nat Immunol ; 19(11): 1169-1178, 2018 11.
Article in English | MEDLINE | ID: mdl-30333617

ABSTRACT

Recent Ebola virus disease epidemics have highlighted the need for effective vaccines and therapeutics to prevent future outbreaks. Antibodies are clearly critical for control of this deadly disease; however, the specific mechanisms of action of protective antibodies have yet to be defined. In this Perspective we discuss the antibody features that correlate with in vivo protection during infection with Ebola virus, based on the results of a systematic and comprehensive study of antibodies directed against this virus. Although neutralization activity mediated by the Fab domains of the antibody is strongly correlated with protection, recruitment of immune effector functions by the Fc domain has also emerged as a complementary, and sometimes alternative, route to protection. For a subset of antibodies, Fc-mediated clearance and killing of infected cells seems to be the main driver of protection after exposure and mirrors observations in vaccination studies. Continued analysis of antibodies that achieve protection partially or wholly through Fc-mediated functions, the precise functions required, the intersection with specificity and the importance of these functions in different animal models is needed to identify and begin to capitalize on Fc-mediated protection in vaccines and therapeutics alike.


Subject(s)
Antibodies, Viral/immunology , Ebolavirus/immunology , Hemorrhagic Fever, Ebola/immunology , Animals , Antibodies, Neutralizing/immunology , Humans
15.
Immunity ; 54(4): 815-828.e5, 2021 04 13.
Article in English | MEDLINE | ID: mdl-33852832

ABSTRACT

Protective Ebola virus (EBOV) antibodies have neutralizing activity and induction of antibody constant domain (Fc)-mediated innate immune effector functions. Efforts to enhance Fc effector functionality often focus on maximizing antibody-dependent cellular cytotoxicity, yet distinct combinations of functions could be critical for antibody-mediated protection. As neutralizing antibodies have been cloned from EBOV disease survivors, we sought to identify survivor Fc effector profiles to help guide Fc optimization strategies. Survivors developed a range of functional antibody responses, and we therefore applied a rapid, high-throughput Fc engineering platform to define the most protective profiles. We generated a library of Fc variants with identical antigen-binding fragments (Fabs) from an EBOV neutralizing antibody. Fc variants with antibody-mediated complement deposition and moderate natural killer (NK) cell activity demonstrated complete protective activity in a stringent in vivo mouse model. Our findings highlight the importance of specific effector functions in antibody-mediated protection, and the experimental platform presents a generalizable resource for identifying correlates of immunity to guide therapeutic antibody design.


Subject(s)
Ebolavirus/immunology , Hemorrhagic Fever, Ebola/immunology , Immunoglobulin Fab Fragments/immunology , Immunoglobulin Fc Fragments/immunology , Animals , Antibodies, Neutralizing/immunology , Antibodies, Viral/immunology , Antibody Formation/immunology , Antibody-Dependent Cell Cytotoxicity/immunology , Female , HEK293 Cells , Hemorrhagic Fever, Ebola/virology , Humans , Immunoglobulin G/immunology , Mice, Inbred BALB C , Receptors, Fc/immunology
16.
Cell ; 163(5): 1053-1054, 2015 Nov 19.
Article in English | MEDLINE | ID: mdl-26590413

ABSTRACT

Chikungunya virus recently caused large outbreaks world-wide. In this issue of Cell, Fox et al. describe several potently neutralizing antibodies against multiple alphaviruses. The structure of the virus in complex with one of the antibodies reveals the antibody-induced rearrangement and crosslinking of the viral surface proteins that result in neutralization.


Subject(s)
Alphavirus Infections/immunology , Alphavirus/immunology , Antibodies, Monoclonal/immunology , Antibodies, Neutralizing/immunology , Antibodies, Viral/immunology , Epitopes , Viral Envelope Proteins/immunology , Animals , Humans
17.
Cell ; 160(5): 904-912, 2015 Feb 26.
Article in English | MEDLINE | ID: mdl-25723165

ABSTRACT

The filoviruses, including Marburg and Ebola, express a single glycoprotein on their surface, termed GP, which is responsible for attachment and entry of target cells. Filovirus GPs differ by up to 70% in protein sequence, and no antibodies are yet described that cross-react among them. Here, we present the 3.6 Å crystal structure of Marburg virus GP in complex with a cross-reactive antibody from a human survivor, and a lower resolution structure of the antibody bound to Ebola virus GP. The antibody, MR78, recognizes a GP1 epitope conserved across the filovirus family, which likely represents the binding site of their NPC1 receptor. Indeed, MR78 blocks binding of the essential NPC1 domain C. These structures and additional small-angle X-ray scattering of mucin-containing MARV and EBOV GPs suggest why such antibodies were not previously elicited in studies of Ebola virus, and provide critical templates for development of immunotherapeutics and inhibitors of entry.


Subject(s)
Antibodies, Neutralizing/chemistry , Antibodies, Neutralizing/immunology , Marburgvirus/chemistry , Viral Envelope Proteins/chemistry , Amino Acid Sequence , Animals , Antibodies, Monoclonal/chemistry , Antibodies, Monoclonal/metabolism , Antibodies, Neutralizing/isolation & purification , Antibodies, Neutralizing/metabolism , Antibodies, Viral/chemistry , Antibodies, Viral/immunology , Antibodies, Viral/metabolism , Antigen-Antibody Complex/chemistry , Cell Line , Cross Reactions , Crystallography, X-Ray , Drosophila , Ebolavirus/chemistry , Humans , Immunoglobulin Fab Fragments/chemistry , Immunoglobulin Fab Fragments/metabolism , Marburg Virus Disease/immunology , Marburgvirus/genetics , Marburgvirus/immunology , Models, Molecular , Molecular Sequence Data , Mucins/chemistry , Sequence Alignment , Viral Envelope Proteins/metabolism
18.
Cell ; 160(5): 893-903, 2015 Feb 26.
Article in English | MEDLINE | ID: mdl-25723164

ABSTRACT

The mechanisms by which neutralizing antibodies inhibit Marburg virus (MARV) are not known. We isolated a panel of neutralizing antibodies from a human MARV survivor that bind to MARV glycoprotein (GP) and compete for binding to a single major antigenic site. Remarkably, several of the antibodies also bind to Ebola virus (EBOV) GP. Single-particle EM structures of antibody-GP complexes reveal that all of the neutralizing antibodies bind to MARV GP at or near the predicted region of the receptor-binding site. The presence of the glycan cap or mucin-like domain blocks binding of neutralizing antibodies to EBOV GP, but not to MARV GP. The data suggest that MARV-neutralizing antibodies inhibit virus by binding to infectious virions at the exposed MARV receptor-binding site, revealing a mechanism of filovirus inhibition.


Subject(s)
Antibodies, Neutralizing/chemistry , Antibodies, Neutralizing/immunology , Antigen-Antibody Complex/ultrastructure , Marburg Virus Disease/immunology , Marburgvirus/chemistry , Viral Envelope Proteins/chemistry , Adult , Animals , Antibodies, Monoclonal/chemistry , Antibodies, Monoclonal/metabolism , Antibodies, Neutralizing/isolation & purification , Antibodies, Neutralizing/metabolism , Antibodies, Viral/chemistry , Antibodies, Viral/immunology , Antibodies, Viral/metabolism , B-Lymphocytes/immunology , Female , Humans , Immunoglobulin Fab Fragments/chemistry , Immunoglobulin Fab Fragments/metabolism , Marburgvirus/genetics , Marburgvirus/immunology , Models, Molecular , Mutation , Protein Structure, Tertiary , Viral Envelope Proteins/metabolism
19.
Immunity ; 52(2): 388-403.e12, 2020 02 18.
Article in English | MEDLINE | ID: mdl-32023489

ABSTRACT

Structural principles underlying the composition of protective antiviral monoclonal antibody (mAb) cocktails are poorly defined. Here, we exploited antibody cooperativity to develop a therapeutic mAb cocktail against Ebola virus. We systematically analyzed the antibody repertoire in human survivors and identified a pair of potently neutralizing mAbs that cooperatively bound to the ebolavirus glycoprotein (GP). High-resolution structures revealed that in a two-antibody cocktail, molecular mimicry was a major feature of mAb-GP interactions. Broadly neutralizing mAb rEBOV-520 targeted a conserved epitope on the GP base region. mAb rEBOV-548 bound to a glycan cap epitope, possessed neutralizing and Fc-mediated effector function activities, and potentiated neutralization by rEBOV-520. Remodeling of the glycan cap structures by the cocktail enabled enhanced GP binding and virus neutralization. The cocktail demonstrated resistance to virus escape and protected non-human primates (NHPs) against Ebola virus disease. These data illuminate structural principles of antibody cooperativity with implications for development of antiviral immunotherapeutics.


Subject(s)
Antibodies, Monoclonal/immunology , Antibodies, Neutralizing/immunology , Antibodies, Viral/immunology , Ebolavirus/immunology , Glycoproteins/immunology , Hemorrhagic Fever, Ebola/immunology , Animals , Antibodies, Monoclonal/therapeutic use , Antibodies, Neutralizing/therapeutic use , Antibodies, Viral/therapeutic use , Cell Line , Disease Models, Animal , Drug Therapy, Combination , Epitopes , Female , Glycoproteins/chemistry , Hemorrhagic Fever, Ebola/prevention & control , Humans , Immunoglobulin Fab Fragments/immunology , Macaca mulatta , Male , Mice , Mice, Inbred BALB C , Molecular Mimicry , Protein Conformation
20.
Cell ; 154(4): 763-74, 2013 Aug 15.
Article in English | MEDLINE | ID: mdl-23953110

ABSTRACT

Proteins, particularly viral proteins, can be multifunctional, but the mechanisms behind multifunctionality are not fully understood. Here, we illustrate through multiple crystal structures, biochemistry, and cellular microscopy that VP40 rearranges into different structures, each with a distinct function required for the ebolavirus life cycle. A butterfly-shaped VP40 dimer traffics to the cellular membrane. Once there, electrostatic interactions trigger rearrangement of the polypeptide into a linear hexamer. These hexamers construct a multilayered, filamentous matrix structure that is critical for budding and resembles tomograms of authentic virions. A third structure of VP40, formed by a different rearrangement, is not involved in virus assembly but instead uniquely binds RNA to regulate viral transcription inside infected cells. These results provide a functional model for ebolavirus matrix assembly and the other roles of VP40 in the virus life cycle and demonstrate how a single wild-type, unmodified polypeptide can assemble into different structures for different functions.


Subject(s)
Ebolavirus/metabolism , Viral Matrix Proteins/chemistry , Viral Matrix Proteins/metabolism , Crystallography, X-Ray , Dimerization , Ebolavirus/chemistry , Ebolavirus/classification , Ebolavirus/genetics , Models, Molecular , Mutagenesis , Point Mutation , Viral Matrix Proteins/genetics , Virus Assembly , Virus Release
SELECTION OF CITATIONS
SEARCH DETAIL