Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 384
Filtrar
Mais filtros

Bases de dados
Tipo de documento
Intervalo de ano de publicação
1.
Cell ; 185(1): 113-130.e15, 2022 01 06.
Artigo em Inglês | MEDLINE | ID: mdl-34921774

RESUMO

mRNA-1273 vaccine efficacy against SARS-CoV-2 Delta wanes over time; however, there are limited data on the impact of durability of immune responses on protection. Here, we immunized rhesus macaques and assessed immune responses over 1 year in blood and upper and lower airways. Serum neutralizing titers to Delta were 280 and 34 reciprocal ID50 at weeks 6 (peak) and 48 (challenge), respectively. Antibody-binding titers also decreased in bronchoalveolar lavage (BAL). Four days after Delta challenge, the virus was unculturable in BAL, and subgenomic RNA declined by ∼3-log10 compared with control animals. In nasal swabs, sgRNA was reduced by 1-log10, and the virus remained culturable. Anamnestic antibodies (590-fold increased titer) but not T cell responses were detected in BAL by day 4 post-challenge. mRNA-1273-mediated protection in the lungs is durable but delayed and potentially dependent on anamnestic antibody responses. Rapid and sustained protection in upper and lower airways may eventually require a boost.

2.
Cell ; 181(5): 1004-1015.e15, 2020 05 28.
Artigo em Inglês | MEDLINE | ID: mdl-32375025

RESUMO

Coronaviruses make use of a large envelope protein called spike (S) to engage host cell receptors and catalyze membrane fusion. Because of the vital role that these S proteins play, they represent a vulnerable target for the development of therapeutics. Here, we describe the isolation of single-domain antibodies (VHHs) from a llama immunized with prefusion-stabilized coronavirus spikes. These VHHs neutralize MERS-CoV or SARS-CoV-1 S pseudotyped viruses, respectively. Crystal structures of these VHHs bound to their respective viral targets reveal two distinct epitopes, but both VHHs interfere with receptor binding. We also show cross-reactivity between the SARS-CoV-1 S-directed VHH and SARS-CoV-2 S and demonstrate that this cross-reactive VHH neutralizes SARS-CoV-2 S pseudotyped viruses as a bivalent human IgG Fc-fusion. These data provide a molecular basis for the neutralization of pathogenic betacoronaviruses by VHHs and suggest that these molecules may serve as useful therapeutics during coronavirus outbreaks.


Assuntos
Anticorpos Neutralizantes/isolamento & purificação , Betacoronavirus/imunologia , Anticorpos de Domínio Único/isolamento & purificação , Animais , Anticorpos Neutralizantes/química , Anticorpos Neutralizantes/imunologia , COVID-19 , Camelídeos Americanos/imunologia , Infecções por Coronavirus/terapia , Reações Cruzadas , Imunoglobulina G/química , Imunoglobulina G/imunologia , Modelos Moleculares , Pandemias , Pneumonia Viral/terapia , Domínios Proteicos , Receptores Virais/química , SARS-CoV-2 , Anticorpos de Domínio Único/química , Anticorpos de Domínio Único/imunologia , Glicoproteína da Espícula de Coronavírus/química , Glicoproteína da Espícula de Coronavírus/imunologia
3.
Cell ; 179(7): 1636-1646.e15, 2019 12 12.
Artigo em Inglês | MEDLINE | ID: mdl-31787378

RESUMO

B cell receptor (BCR) sequencing is a powerful tool for interrogating immune responses to infection and vaccination, but it provides limited information about the antigen specificity of the sequenced BCRs. Here, we present LIBRA-seq (linking B cell receptor to antigen specificity through sequencing), a technology for high-throughput mapping of paired heavy- and light-chain BCR sequences to their cognate antigen specificities. B cells are mixed with a panel of DNA-barcoded antigens so that both the antigen barcode(s) and BCR sequence are recovered via single-cell next-generation sequencing. Using LIBRA-seq, we mapped the antigen specificity of thousands of B cells from two HIV-infected subjects. The predicted specificities were confirmed for a number of HIV- and influenza-specific antibodies, including known and novel broadly neutralizing antibodies. LIBRA-seq will be an integral tool for antibody discovery and vaccine development efforts against a wide range of antigen targets.


Assuntos
Mapeamento de Epitopos/métodos , Epitopos/química , Receptores de Antígenos de Linfócitos B/química , Análise de Sequência de DNA/métodos , Análise de Célula Única/métodos , Anticorpos Neutralizantes/química , Anticorpos Neutralizantes/imunologia , Antígenos/química , Antígenos/imunologia , Células Cultivadas , Epitopos/imunologia , Células HEK293 , Anticorpos Anti-HIV/química , Anticorpos Anti-HIV/imunologia , Sequenciamento de Nucleotídeos em Larga Escala/métodos , Ensaios de Triagem em Larga Escala/métodos , Humanos , Receptores de Antígenos de Linfócitos B/imunologia , Células THP-1
4.
Cell ; 178(1): 202-215.e14, 2019 06 27.
Artigo em Inglês | MEDLINE | ID: mdl-31204102

RESUMO

Despite the worldwide success of vaccination, newborns remain vulnerable to infections. While neonatal vaccination has been hampered by maternal antibody-mediated dampening of immune responses, enhanced regulatory and tolerogenic mechanisms, and immune system immaturity, maternal pre-natal immunization aims to boost neonatal immunity via antibody transfer to the fetus. However, emerging data suggest that antibodies are not transferred equally across the placenta. To understand this, we used systems serology to define Fc features associated with antibody transfer. The Fc-profile of neonatal and maternal antibodies differed, skewed toward natural killer (NK) cell-activating antibodies. This selective transfer was linked to digalactosylated Fc-glycans that selectively bind FcRn and FCGR3A, resulting in transfer of antibodies able to efficiently leverage innate immune cells present at birth. Given emerging data that vaccination may direct antibody glycosylation, our study provides insights for the development of next-generation maternal vaccines designed to elicit antibodies that will most effectively aid neonates.


Assuntos
Antígenos de Histocompatibilidade Classe I/imunologia , Antígenos de Histocompatibilidade Classe I/metabolismo , Imunoglobulina G/metabolismo , Placenta/metabolismo , Polissacarídeos/metabolismo , Receptores Fc/imunologia , Receptores Fc/metabolismo , Adolescente , Adulto , Bélgica , Degranulação Celular , Estudos de Coortes , Feminino , Glicosilação , Humanos , Recém-Nascido , Células Matadoras Naturais/imunologia , Ativação Linfocitária/imunologia , Masculino , Gravidez , Receptores de IgG/metabolismo , Células THP-1 , Estados Unidos , Vacinação , Adulto Jovem
5.
Immunity ; 57(3): 574-586.e7, 2024 Mar 12.
Artigo em Inglês | MEDLINE | ID: mdl-38430907

RESUMO

Continuously evolving influenza viruses cause seasonal epidemics and pose global pandemic threats. Although viral neuraminidase (NA) is an effective drug and vaccine target, our understanding of the NA antigenic landscape still remains incomplete. Here, we describe NA-specific human antibodies that target the underside of the NA globular head domain, inhibit viral propagation of a wide range of human H3N2, swine-origin variant H3N2, and H2N2 viruses, and confer both pre- and post-exposure protection against lethal H3N2 infection in mice. Cryo-EM structures of two such antibodies in complex with NA reveal non-overlapping epitopes covering the underside of the NA head. These sites are highly conserved among N2 NAs yet inaccessible unless the NA head tilts or dissociates. Our findings help guide the development of effective countermeasures against ever-changing influenza viruses by identifying hidden conserved sites of vulnerability on the NA underside.


Assuntos
Vacinas contra Influenza , Influenza Humana , Infecções por Orthomyxoviridae , Humanos , Animais , Camundongos , Suínos , Proteínas Virais/genética , Neuraminidase , Vírus da Influenza A Subtipo H3N2 , Anticorpos Monoclonais , Anticorpos Antivirais
6.
Immunity ; 56(10): 2425-2441.e14, 2023 Oct 10.
Artigo em Inglês | MEDLINE | ID: mdl-37689061

RESUMO

Nanoparticles for multivalent display and delivery of vaccine antigens have emerged as a promising avenue for enhancing B cell responses to protein subunit vaccines. Here, we evaluated B cell responses in rhesus macaques immunized with prefusion-stabilized respiratory syncytial virus (RSV) F glycoprotein trimer compared with nanoparticles displaying 10 or 20 copies of the same antigen. We show that multivalent display skews antibody specificities and drives epitope-focusing of responding B cells. Antibody cloning and repertoire sequencing revealed that focusing was driven by the expansion of clonally distinct B cells through recruitment of diverse precursors. We identified two antibody lineages that developed either ultrapotent neutralization or pneumovirus cross-neutralization from precursor B cells with low initial affinity for the RSV-F immunogen. This suggests that increased avidity by multivalent display facilitates the activation and recruitment of these cells. Diversification of the B cell response by multivalent nanoparticle immunogens has broad implications for vaccine design.

8.
Nat Immunol ; 20(3): 362-372, 2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-30742080

RESUMO

The present vaccine against influenza virus has the inevitable risk of antigenic discordance between the vaccine and the circulating strains, which diminishes vaccine efficacy. This necessitates new approaches that provide broader protection against influenza. Here we designed a vaccine using the hypervariable receptor-binding domain (RBD) of viral hemagglutinin displayed on a nanoparticle (np) able to elicit antibody responses that neutralize H1N1 influenza viruses spanning over 90 years. Co-display of RBDs from multiple strains across time, so that the adjacent RBDs are heterotypic, provides an avidity advantage to cross-reactive B cells. Immunization with the mosaic RBD-np elicited broader antibody responses than those induced by an admixture of nanoparticles encompassing the same set of RBDs as separate homotypic arrays. Furthermore, we identified a broadly neutralizing monoclonal antibody in a mouse immunized with mosaic RBD-np. The mosaic antigen array signifies a unique approach that subverts monotypic immunodominance and allows otherwise subdominant cross-reactive B cell responses to emerge.


Assuntos
Glicoproteínas de Hemaglutininação de Vírus da Influenza/imunologia , Vírus da Influenza A Subtipo H1N1/imunologia , Vacinas contra Influenza/imunologia , Influenza Humana/imunologia , Nanopartículas/química , Infecções por Orthomyxoviridae/imunologia , Animais , Anticorpos Neutralizantes/administração & dosagem , Anticorpos Neutralizantes/imunologia , Anticorpos Antivirais/imunologia , Linfócitos B/efeitos dos fármacos , Linfócitos B/imunologia , Linfócitos B/virologia , Reações Cruzadas/efeitos dos fármacos , Reações Cruzadas/imunologia , Feminino , Glicoproteínas de Hemaglutininação de Vírus da Influenza/química , Humanos , Imunização , Vírus da Influenza A Subtipo H1N1/metabolismo , Vírus da Influenza A Subtipo H1N1/fisiologia , Vacinas contra Influenza/administração & dosagem , Vacinas contra Influenza/química , Influenza Humana/prevenção & controle , Influenza Humana/virologia , Camundongos Endogâmicos BALB C , Infecções por Orthomyxoviridae/prevenção & controle , Infecções por Orthomyxoviridae/virologia
9.
Immunity ; 55(12): 2405-2418.e7, 2022 12 13.
Artigo em Inglês | MEDLINE | ID: mdl-36356572

RESUMO

Current influenza vaccines predominantly induce immunity to the hypervariable hemagglutinin (HA) head, requiring frequent vaccine reformulation. Conversely, the immunosubdominant yet conserved HA stem harbors a supersite that is targeted by broadly neutralizing antibodies (bnAbs), representing a prime target for universal vaccines. Here, we showed that the co-immunization of two HA stem immunogens derived from group 1 and 2 influenza A viruses elicits cross-group protective immunity and neutralizing antibody responses in mice, ferrets, and nonhuman primates (NHPs). Immunized mice were protected from multiple group 1 and 2 viruses, and all animal models showed broad serum-neutralizing activity. A bnAb isolated from an immunized NHP broadly neutralized and protected against diverse viruses, including H5N1 and H7N9. Genetic and structural analyses revealed strong homology between macaque and human bnAbs, illustrating common biophysical constraints for acquiring cross-group specificity. Vaccine elicitation of stem-directed cross-group-protective immunity represents a step toward the development of broadly protective influenza vaccines.


Assuntos
Virus da Influenza A Subtipo H5N1 , Subtipo H7N9 do Vírus da Influenza A , Vacinas contra Influenza , Influenza Humana , Infecções por Orthomyxoviridae , Animais , Camundongos , Humanos , Hemaglutininas , Anticorpos Amplamente Neutralizantes , Glicoproteínas de Hemaglutininação de Vírus da Influenza , Anticorpos Antivirais , Furões , Anticorpos Neutralizantes , Imunização
10.
Cell ; 167(3): 625-631, 2016 Oct 20.
Artigo em Inglês | MEDLINE | ID: mdl-27693357

RESUMO

The emergence of Zika virus in the Americas and Caribbean created an urgent need for vaccines to reduce transmission and prevent disease, particularly the devastating neurodevelopmental defects that occur in utero. Rapid advances in Zika immunity and the development of vaccine candidates provide cautious optimism that preventive measures are possible.


Assuntos
Vacinas Virais/imunologia , Infecção por Zika virus/imunologia , Infecção por Zika virus/prevenção & controle , Zika virus/imunologia , Anticorpos Antivirais/sangue , Anticorpos Antivirais/imunologia , Humanos , Imunidade Inata , Linfócitos T/imunologia , Infecção por Zika virus/sangue , Infecção por Zika virus/transmissão
11.
Cell ; 167(3): 684-694.e9, 2016 Oct 20.
Artigo em Inglês | MEDLINE | ID: mdl-27768891

RESUMO

Monkeypox (MPXV) and cowpox (CPXV) are emerging agents that cause severe human infections on an intermittent basis, and variola virus (VARV) has potential for use as an agent of bioterror. Vaccinia immune globulin (VIG) has been used therapeutically to treat severe orthopoxvirus infections but is in short supply. We generated a large panel of orthopoxvirus-specific human monoclonal antibodies (Abs) from immune subjects to investigate the molecular basis of broadly neutralizing antibody responses for diverse orthopoxviruses. Detailed analysis revealed the principal neutralizing antibody specificities that are cross-reactive for VACV, CPXV, MPXV, and VARV and that are determinants of protection in murine challenge models. Optimal protection following respiratory or systemic infection required a mixture of Abs that targeted several membrane proteins, including proteins on enveloped and mature virion forms of virus. This work reveals orthopoxvirus targets for human Abs that mediate cross-protective immunity and identifies new candidate Ab therapeutic mixtures to replace VIG.


Assuntos
Anticorpos Monoclonais/imunologia , Anticorpos Neutralizantes/imunologia , Anticorpos Antivirais/imunologia , Especificidade de Anticorpos , Infecções por Poxviridae/imunologia , Varíola Bovina/imunologia , Vírus da Varíola Bovina/imunologia , Reações Cruzadas , Humanos , Leucócitos Mononucleares/imunologia , Mpox/imunologia , Monkeypox virus/imunologia , Varíola/imunologia , Vacínia/imunologia , Vaccinia virus/imunologia , Vírus da Varíola/imunologia
12.
Cell ; 166(3): 609-623, 2016 Jul 28.
Artigo em Inglês | MEDLINE | ID: mdl-27453470

RESUMO

Antibodies capable of neutralizing divergent influenza A viruses could form the basis of a universal vaccine. Here, from subjects enrolled in an H5N1 DNA/MIV-prime-boost influenza vaccine trial, we sorted hemagglutinin cross-reactive memory B cells and identified three antibody classes, each capable of neutralizing diverse subtypes of group 1 and group 2 influenza A viruses. Co-crystal structures with hemagglutinin revealed that each class utilized characteristic germline genes and convergent sequence motifs to recognize overlapping epitopes in the hemagglutinin stem. All six analyzed subjects had sequences from at least one multidonor class, and-in half the subjects-multidonor-class sequences were recovered from >40% of cross-reactive B cells. By contrast, these multidonor-class sequences were rare in published antibody datasets. Vaccination with a divergent hemagglutinin can thus increase the frequency of B cells encoding broad influenza A-neutralizing antibodies. We propose the sequence signature-quantified prevalence of these B cells as a metric to guide universal influenza A immunization strategies.


Assuntos
Anticorpos Neutralizantes/imunologia , Anticorpos Antivirais/imunologia , Vírus da Influenza A/imunologia , Vacinas contra Influenza/imunologia , Adulto , Sequência de Aminoácidos , Anticorpos Neutralizantes/química , Anticorpos Neutralizantes/genética , Anticorpos Antivirais/química , Anticorpos Antivirais/genética , Linfócitos B/imunologia , Epitopos de Linfócito B , Feminino , Rearranjo Gênico de Cadeia Pesada de Linfócito B , Humanos , Memória Imunológica , Virus da Influenza A Subtipo H5N1/imunologia , Masculino , Pessoa de Meia-Idade , Modelos Moleculares , Estrutura Terciária de Proteína , Relação Estrutura-Atividade , Adulto Jovem
13.
Nat Immunol ; 19(1): 20-28, 2018 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-29199281

RESUMO

Emerging infectious diseases will continue to threaten public health and are sustained by global commerce, travel and disruption of ecological systems. Most pandemic threats are caused by viruses from either zoonotic sources or vector-borne sources. Developing better ways to anticipate and manage the ongoing microbial challenge will be critical for achieving the United Nations Sustainable Development Goals and, conversely, each such goal will affect the ability to control infectious diseases. Here we discuss how technology can be applied effectively to better prepare for and respond to new viral diseases with a focus on new paradigms for vaccine development.


Assuntos
Doenças Transmissíveis Emergentes/imunologia , Vacinação/métodos , Vacinas Virais/imunologia , Viroses/imunologia , Animais , Doenças Transmissíveis Emergentes/prevenção & controle , Doenças Transmissíveis Emergentes/virologia , Transmissão de Doença Infecciosa/prevenção & controle , Humanos , Pandemias/prevenção & controle , Saúde Pública/métodos , Vacinas Virais/uso terapêutico , Viroses/epidemiologia , Viroses/prevenção & controle
14.
Immunity ; 54(8): 1636-1651, 2021 08 10.
Artigo em Inglês | MEDLINE | ID: mdl-34348117

RESUMO

The development of effective vaccines to combat infectious diseases is a complex multi-year and multi-stakeholder process. To accelerate the development of vaccines for coronavirus disease 2019 (COVID-19), a novel pathogen emerging in late 2019 and spreading globally by early 2020, the United States government (USG) mounted an operation bridging public and private sector expertise and infrastructure. The success of the endeavor can be seen in the rapid advanced development of multiple vaccine candidates, with several demonstrating efficacy and now being administered around the globe. Here, we review the milestones enabling the USG-led effort, the methods utilized, and ensuing outcomes. We discuss the current status of COVID-19 vaccine development and provide a perspective for how partnership and preparedness can be better utilized in response to future public-health pandemic emergencies.


Assuntos
Vacinas contra COVID-19/imunologia , COVID-19/epidemiologia , COVID-19/prevenção & controle , Pesquisa , SARS-CoV-2/imunologia , Bioengenharia , Biotecnologia , Vacinas contra COVID-19/administração & dosagem , Humanos , Modelos Moleculares , Avaliação de Resultados em Cuidados de Saúde , Vigilância em Saúde Pública , Pesquisa/estatística & dados numéricos , Pesquisa/tendências , Estados Unidos/epidemiologia , Cobertura Vacinal/estatística & dados numéricos , Vacinologia
15.
Immunity ; 54(8): 1869-1882.e6, 2021 08 10.
Artigo em Inglês | MEDLINE | ID: mdl-34270939

RESUMO

Vaccine-associated enhanced respiratory disease (VAERD) was previously observed in some preclinical models of severe acute respiratory syndrome (SARS) and MERS coronavirus vaccines. We used the SARS coronavirus 2 (SARS-CoV-2) mouse-adapted, passage 10, lethal challenge virus (MA10) mouse model of acute lung injury to evaluate the immune response and potential for immunopathology in animals vaccinated with research-grade mRNA-1273. Whole-inactivated virus or heat-denatured spike protein subunit vaccines with alum designed to elicit low-potency antibodies and Th2-skewed CD4+ T cells resulted in reduced viral titers and weight loss post challenge but more severe pathological changes in the lung compared to saline-immunized animals. In contrast, a protective dose of mRNA-1273 induced favorable humoral and cellular immune responses that protected from viral replication in the upper and lower respiratory tract upon challenge. A subprotective dose of mRNA-1273 reduced viral replication and limited histopathological manifestations compared to animals given saline. Overall, our findings demonstrate an immunological signature associated with antiviral protection without disease enhancement following vaccination with mRNA-1273.


Assuntos
Vacinas contra COVID-19/imunologia , COVID-19/imunologia , COVID-19/prevenção & controle , Interações Hospedeiro-Patógeno/imunologia , SARS-CoV-2/imunologia , Vacinas Sintéticas/imunologia , Animais , Anticorpos Neutralizantes/imunologia , Anticorpos Antivirais/imunologia , Biópsia , Vacinas contra COVID-19/administração & dosagem , Modelos Animais de Doenças , Humanos , Imunoglobulina G , Imuno-Histoquímica , Camundongos , Avaliação de Resultados em Cuidados de Saúde , RNA Mensageiro , Glicoproteína da Espícula de Coronavírus/imunologia , Subpopulações de Linfócitos T/imunologia , Subpopulações de Linfócitos T/metabolismo , Vacinas Sintéticas/administração & dosagem , Vacinas de mRNA
16.
Immunity ; 54(4): 769-780.e6, 2021 04 13.
Artigo em Inglês | MEDLINE | ID: mdl-33823129

RESUMO

An effective vaccine for respiratory syncytial virus (RSV) is an unrealized public health goal. A single dose of the prefusion-stabilized fusion (F) glycoprotein subunit vaccine (DS-Cav1) substantially increases serum-neutralizing activity in healthy adults. We sought to determine whether DS-Cav1 vaccination induces a repertoire mirroring the pre-existing diversity from natural infection or whether antibody lineages targeting specific epitopes predominate. We evaluated RSV F-specific B cell responses before and after vaccination in six participants using complementary B cell sequencing methodologies and identified 555 clonal lineages. DS-Cav1-induced lineages recognized the prefusion conformation of F (pre-F) and were genetically diverse. Expressed antibodies recognized all six antigenic sites on the pre-F trimer. We identified 34 public clonotypes, and structural analysis of two antibodies from a predominant clonotype revealed a common mode of recognition. Thus, vaccination with DS-Cav1 generates a diverse polyclonal response targeting the antigenic sites on pre-F, supporting the development and advanced testing of pre-F-based vaccines against RSV.


Assuntos
Anticorpos Antivirais/imunologia , Formação de Anticorpos/imunologia , Infecções por Vírus Respiratório Sincicial/imunologia , Vacinas contra Vírus Sincicial Respiratório/imunologia , Vírus Sincicial Respiratório Humano/imunologia , Adolescente , Adulto , Idoso , Idoso de 80 Anos ou mais , Anticorpos Neutralizantes/imunologia , Linhagem Celular , Linhagem Celular Tumoral , Criança , Pré-Escolar , Estudos de Coortes , Epitopos/imunologia , Feminino , Células HEK293 , Humanos , Lactente , Recém-Nascido , Masculino , Pessoa de Meia-Idade , Vacinação/métodos , Proteínas Virais de Fusão/imunologia , Adulto Jovem
18.
Cell ; 162(5): 1090-100, 2015 Aug 27.
Artigo em Inglês | MEDLINE | ID: mdl-26279189

RESUMO

Epstein-Barr virus (EBV) represents a major global health problem. Though it is associated with infectious mononucleosis and ∼200,000 cancers annually worldwide, a vaccine is not available. The major target of immunity is EBV glycoprotein 350/220 (gp350) that mediates attachment to B cells through complement receptor 2 (CR2/CD21). Here, we created self-assembling nanoparticles that displayed different domains of gp350 in a symmetric array. By focusing presentation of the CR2-binding domain on nanoparticles, potent neutralizing antibodies were elicited in mice and non-human primates. The structurally designed nanoparticle vaccine increased neutralization 10- to 100-fold compared to soluble gp350 by targeting a functionally conserved site of vulnerability, improving vaccine-induced protection in a mouse model. This rational approach to EBV vaccine design elicited potent neutralizing antibody responses by arrayed presentation of a conserved viral entry domain, a strategy that can be applied to other viruses.


Assuntos
Vacinas contra Herpesvirus/química , Vacinas contra Herpesvirus/imunologia , Animais , Anticorpos Neutralizantes/imunologia , Cristalografia por Raios X , Desenho de Fármacos , Feminino , Herpesvirus Humano 4 , Vacinas contra Herpesvirus/genética , Vacinas contra Herpesvirus/isolamento & purificação , Macaca fascicularis , Camundongos , Camundongos Endogâmicos BALB C , Nanopartículas/química , Nanopartículas/ultraestrutura , Receptores de Complemento 3d/química , Receptores de Complemento 3d/imunologia , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/imunologia , Proteínas Recombinantes/isolamento & purificação
19.
Immunity ; 51(3): 429-442, 2019 09 17.
Artigo em Inglês | MEDLINE | ID: mdl-31533056

RESUMO

Respiratory syncytial virus (RSV) has eluded active vaccination efforts for more than five decades and continues to cause substantial morbidity and mortality in infants, the immunocompromised, and older adults. Although newer approaches of passive antibody-mediated protection show promise, vaccines aimed at eliciting fusion protein (F)-targeting antibodies have repeatedly failed to meet pre-established, modest-efficacy goals. Newer candidates, including protein-based vaccines, live-attenuated viruses, and gene-based delivery platforms, incorporate structurally defined and stabilized versions of the prefusion form of the F glycoprotein and are advancing rapidly into critical efficacy studies in susceptible target populations. This review discusses the storied history of RSV vaccine development, immunological lessons learned along the way, and critical findings about protein structure that remodeled our understanding of protective immunity to this important pathogen.


Assuntos
Infecções por Vírus Respiratório Sincicial/imunologia , Vacinas contra Vírus Sincicial Respiratório/imunologia , Vírus Sinciciais Respiratórios/imunologia , Animais , Anticorpos Antivirais/imunologia , Humanos , Proteínas Virais de Fusão/imunologia
20.
Immunity ; 51(4): 735-749.e8, 2019 10 15.
Artigo em Inglês | MEDLINE | ID: mdl-31563464

RESUMO

Antibody paratopes are formed by hypervariable complementarity-determining regions (CDRH3s) and variable gene-encoded CDRs. The latter show biased usage in human broadly neutralizing antibodies (bnAbs) against both HIV and influenza virus, suggesting the existence of gene-endowed targeting solutions that may be amenable to pathway amplification. To test this, we generated transgenic mice with human CDRH3 diversity but simultaneously constrained to individual user-defined human immunoglobulin variable heavy-chain (VH) genes, including IGHV1-69, which shows biased usage in human bnAbs targeting the hemagglutinin stalk of group 1 influenza A viruses. Sequential immunization with a stalk-only hemagglutinin nanoparticle elicited group 1 bnAbs, but only in IGHV1-69 mice. This VH-endowed response required minimal affinity maturation, was elicited alongside pre-existing influenza immunity, and when IGHV1-69 B cells were diluted to match the frequency measured in humans. These results indicate that the human repertoire could, in principle, support germline-encoded bnAb elicitation using a single recombinant hemagglutinin immunogen.


Assuntos
Anticorpos Antivirais/metabolismo , Linfócitos B/imunologia , Anticorpos Amplamente Neutralizantes/metabolismo , Vírus da Influenza A/fisiologia , Vacinas contra Influenza/imunologia , Influenza Humana/imunologia , Receptores de Antígenos de Linfócitos B/genética , Animais , Anticorpos Antivirais/genética , Afinidade de Anticorpos , Anticorpos Amplamente Neutralizantes/genética , Regiões Determinantes de Complementaridade/genética , Mutação em Linhagem Germinativa/genética , Glicoproteínas de Hemaglutininação de Vírus da Influenza/imunologia , Humanos , Imunidade Humoral , Imunização Secundária , Cadeias Pesadas de Imunoglobulinas/genética , Camundongos , Camundongos Transgênicos , Nanopartículas , Engenharia de Proteínas
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA