Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 118
Filtrar
Mais filtros

Intervalo de ano de publicação
1.
Annu Rev Immunol ; 34: 395-420, 2016 05 20.
Artigo em Inglês | MEDLINE | ID: mdl-26907212

RESUMO

Systemic autoimmune diseases are characterized by specific targeting of a limited group of ubiquitously expressed autoantigens by the immune system. This review examines the mechanisms underlying their selection as immune targets. Initiation of autoimmune responses likely reflects the presentation of antigens with a distinct structure not previously encountered by the immune system, in a proimmune context (injury, malignancy, or infection). Causes of modified structure include somatic mutation and posttranslational modifications (including citrullination and proteolysis). Many autoantigens are components of multimolecular complexes, and some of the other components may provide adjuvant activity. Propagation of autoimmune responses appears to reflect a bidirectional interaction between the immune response and the target tissues in a mutually reinforcing cycle: Immune effector pathways generate additional autoantigen, which feeds further immune response. We propose that this resonance may be a critical principle underlying disease propagation, with specific autoantigens functioning as the hubs around which amplification occurs.


Assuntos
Autoantígenos/imunologia , Doenças Autoimunes/imunologia , Epitopos Imunodominantes/imunologia , Complexos Multiproteicos/imunologia , Doenças Reumáticas/imunologia , Adjuvantes Imunológicos , Animais , Autoimunidade , Retroalimentação Fisiológica , Humanos , Tolerância Imunológica , Relação Estrutura-Atividade
2.
Cell ; 184(19): 4996-5014.e26, 2021 09 16.
Artigo em Inglês | MEDLINE | ID: mdl-34534464

RESUMO

CD8 T cell responses against different tumor neoantigens occur simultaneously, yet little is known about the interplay between responses and its impact on T cell function and tumor control. In mouse lung adenocarcinoma, we found that immunodominance is established in tumors, wherein CD8 T cell expansion is predominantly driven by the antigen that most stably binds MHC. T cells responding to subdominant antigens were enriched for a TCF1+ progenitor phenotype correlated with response to immune checkpoint blockade (ICB) therapy. However, the subdominant T cell response did not preferentially benefit from ICB due to a dysfunctional subset of TCF1+ cells marked by CCR6 and Tc17 differentiation. Analysis of human samples and sequencing datasets revealed that CCR6+ TCF1+ cells exist across human cancers and are not correlated with ICB response. Vaccination eliminated CCR6+ TCF1+ cells and dramatically improved the subdominant response, highlighting a strategy to optimally engage concurrent neoantigen responses against tumors.


Assuntos
Adenocarcinoma de Pulmão/imunologia , Antígenos de Neoplasias/imunologia , Linfócitos T CD8-Positivos/imunologia , Fator 1-alfa Nuclear de Hepatócito/metabolismo , Neoplasias Pulmonares/imunologia , Células-Tronco/imunologia , Sequência de Aminoácidos , Animais , Antígeno CTLA-4/metabolismo , Epitopos , Feminino , Humanos , Inibidores de Checkpoint Imunológico/farmacologia , Neoplasias Pulmonares/patologia , Camundongos , Peptídeos/química , Fenótipo , Receptor de Morte Celular Programada 1/metabolismo , RNA-Seq , Receptores de Antígenos de Linfócitos T/metabolismo , Receptores CCR6/metabolismo , Análise de Célula Única , Vacinação
3.
Immunity ; 55(10): 1909-1923.e6, 2022 10 11.
Artigo em Inglês | MEDLINE | ID: mdl-36115338

RESUMO

Reciprocal interactions between host T helper cells and gut microbiota enforce local immunological tolerance and modulate extra-intestinal immunity. However, our understanding of antigen-specific tolerance to the microbiome is limited. Here, we developed a systematic approach to predict HLA class-II-specific epitopes using the humanized bacteria-originated T cell antigen (hBOTA) algorithm. We identified a diverse set of microbiome epitopes spanning all major taxa that are compatible with presentation by multiple HLA-II alleles. In particular, we uncovered an immunodominant epitope from the TonB-dependent receptor SusC that was universally recognized and ubiquitous among Bacteroidales. In healthy human subjects, SusC-reactive T cell responses were characterized by IL-10-dominant cytokine profiles, whereas in patients with active Crohn's disease, responses were associated with elevated IL-17A. Our results highlight the potential of targeted antigen discovery within the microbiome to reveal principles of tolerance and functional transitions during inflammation.


Assuntos
Doença de Crohn , Epitopos Imunodominantes , Linfócitos T CD4-Positivos , Epitopos de Linfócito T , Humanos , Interleucina-10 , Interleucina-17
4.
Immunity ; 54(6): 1290-1303.e7, 2021 06 08.
Artigo em Inglês | MEDLINE | ID: mdl-34022127

RESUMO

Dissecting the evolution of memory B cells (MBCs) against SARS-CoV-2 is critical for understanding antibody recall upon secondary exposure. Here, we used single-cell sequencing to profile SARS-CoV-2-reactive B cells in 38 COVID-19 patients. Using oligo-tagged antigen baits, we isolated B cells specific to the SARS-CoV-2 spike, nucleoprotein (NP), open reading frame 8 (ORF8), and endemic human coronavirus (HCoV) spike proteins. SARS-CoV-2 spike-specific cells were enriched in the memory compartment of acutely infected and convalescent patients several months post symptom onset. With severe acute infection, substantial populations of endemic HCoV-reactive antibody-secreting cells were identified and possessed highly mutated variable genes, signifying preexisting immunity. Finally, MBCs exhibited pronounced maturation to NP and ORF8 over time, especially in older patients. Monoclonal antibodies against these targets were non-neutralizing and non-protective in vivo. These findings reveal antibody adaptation to non-neutralizing intracellular antigens during infection, emphasizing the importance of vaccination for inducing neutralizing spike-specific MBCs.


Assuntos
Anticorpos Antivirais/imunologia , Formação de Anticorpos/imunologia , Linfócitos B/imunologia , COVID-19/imunologia , Interações Hospedeiro-Patógeno/imunologia , Epitopos Imunodominantes/imunologia , SARS-CoV-2/imunologia , Anticorpos Neutralizantes/imunologia , Formação de Anticorpos/genética , Linfócitos B/metabolismo , Biologia Computacional/métodos , Reações Cruzadas/imunologia , Mapeamento de Epitopos , Feminino , Perfilação da Expressão Gênica , Sequenciamento de Nucleotídeos em Larga Escala , Interações Hospedeiro-Patógeno/genética , Humanos , Epitopos Imunodominantes/genética , Memória Imunológica , Masculino , Testes de Neutralização , Análise de Célula Única/métodos , Glicoproteína da Espícula de Coronavírus/imunologia , Transcriptoma
5.
Immunol Rev ; 307(1): 116-133, 2022 05.
Artigo em Inglês | MEDLINE | ID: mdl-35174510

RESUMO

Random VDJ recombination early in T and B cell development enables the adaptive immune system to recognize a vast array of evolving pathogens via antigen receptors. However, the potential of such randomly generated TCRs and BCRs to recognize and respond to self-antigens requires layers of tolerance mechanisms to mitigate the risk of life-threatening autoimmunity. Since they were originally cloned more than three decades ago, the NR4A family of nuclear hormone receptors have been implicated in many critical aspects of immune tolerance, including negative selection of thymocytes, peripheral T cell tolerance, regulatory T cells (Treg), and most recently in peripheral B cell tolerance. In this review, we discuss important insights from many laboratories as well as our own group into the function and mechanisms by which this small class of primary response genes promotes self-tolerance and immune homeostasis to balance the need for host defense against the inherent risks posed by the adaptive immune system.


Assuntos
Tolerância Imunológica , Membro 1 do Grupo A da Subfamília 4 de Receptores Nucleares , Linfócitos B , Humanos , Membro 1 do Grupo A da Subfamília 4 de Receptores Nucleares/genética , Tolerância a Antígenos Próprios , Linfócitos T Reguladores
6.
Eur J Immunol ; : e2451200, 2024 Aug 13.
Artigo em Inglês | MEDLINE | ID: mdl-39138621

RESUMO

This study aims to understand the impact of early antiretroviral therapy (ART) on HIV-specific T-cell responses measured after treatment interruption may inform strategies to deliver ART-free immune-mediated viral suppression. HIV-specific T-cell immunity was analysed using gamma interferon enzyme-linked immunospot assays in two studies. SPARTAC included individuals with primary HIV infection randomised to 48 weeks of ART (n = 24) or no immediate therapy (n = 37). The PITCH (n = 7) cohort started antiretroviral therapy in primary infection for at least one year, followed by TI. In SPARTAC, participants treated in PHI for 48 weeks followed by TI for 12 weeks, and those who remained untreated for 60 weeks made similar HIV Gag-directed responses (both magnitude and breadth) at week 60. However, the treated group made a greater proportion of novel HIV Gag-directed responses by Week 60, suggestive of a greater reserve to produce new potentially protective responses. In the more intensively followed PITCH study, 6/7 participants showed dominant Gag and/or Pol-specific responses post-TI compared with pre-TI. Although early ART in PHI was not associated with major differences in HIV-specific immunity following TI compared with untreated participants, the potential to make more new Gag-directed responses warrants further investigation as this may inform strategies to achieve ART-free control.

7.
Immunol Rev ; 296(1): 120-131, 2020 07.
Artigo em Inglês | MEDLINE | ID: mdl-32483855

RESUMO

The majority of all vaccines work by inducing protective antibody responses. The mechanisms by which the B cells responsible for producing protective antibodies are elicited to respond are not well understood. Interclonal B cell competition to complex antigens, particularly in germinal centers, has emerged as an important hurdle in designing effective vaccines. This review will focus on recent advances in understanding the roles of B cell precursor frequency, B cell receptor affinity for antigen, antigen avidity, and other factors that can substantially alter the outcomes of B cell responses to complex antigens. Understanding the interdependence of these fundamental factors that affect B cell responses can inform current vaccine design efforts for pathogens with complex proteins as candidate immunogens such as HIV, influenza, and coronaviruses.


Assuntos
Linfócitos B/imunologia , Linfócitos B/metabolismo , Competição entre as Células/imunologia , Epitopos Imunodominantes/imunologia , Imunomodulação , Animais , Antígenos/metabolismo , Linfócitos B/citologia , Competição entre as Células/genética , Diferenciação Celular/imunologia , Centro Germinativo/imunologia , Centro Germinativo/metabolismo , Humanos , Receptores de Antígenos de Linfócitos B/metabolismo
8.
Immunol Rev ; 296(1): 191-204, 2020 07.
Artigo em Inglês | MEDLINE | ID: mdl-32666572

RESUMO

Natural influenza virus infections and seasonal vaccinations often do not confer broadly neutralizing immunity across diverse influenza strains. In addition, the virus is capable of rapid antigenic drift in order to evade pre-existing immunity. The surface glycoproteins, hemagglutinin, and neuraminidase can easily mutate their immunodominant epitopes without impacting fitness. Skewing human antibody repertoires to target more conserved epitopes is thus an expanding area of research: Many groups are attempting to produce universal influenza vaccines that can protect across a wide variety of strains. Achieving this goal will require a detailed understanding of how infection history impacts humoral responses. It will also require the ability to manipulate or enhance B cell selection in order to expand clones that can recognize subdominant but protective epitopes. In this review, we will discuss what immune imprinting means to immunologists and describe efforts to overcome or silence imprinting in order to improve vaccination efficiency.


Assuntos
Interações Hospedeiro-Patógeno/imunologia , Epitopos Imunodominantes/imunologia , Influenza Humana/imunologia , Orthomyxoviridae/imunologia , Animais , Antígenos Virais/imunologia , Seleção Clonal Mediada por Antígeno , Humanos , Imunidade Celular , Imunidade Humoral , Vacinas contra Influenza/administração & dosagem , Vacinas contra Influenza/imunologia , Influenza Humana/prevenção & controle , Influenza Humana/virologia , Vacinação
9.
Br J Haematol ; 202(4): 874-878, 2023 08.
Artigo em Inglês | MEDLINE | ID: mdl-37323051

RESUMO

Respiratory syncytial virus (RSV)-associated viral infections are a major public health problem affecting the immunologically naïve/compromised populations. Given the RSV-associated morbidity and the limited treatment options, we sought to characterize the cellular immune response to RSV to develop a targeted T cell therapy for off-the-shelf administration to immunocompromised individuals. Here we report on the immunological profiling, as well as manufacturing, characterization and antiviral properties of these RSV-targeted T cells. A randomized, phase 1/2 clinical trial evaluating their safety and activity in haematopoietic stem cell transplant recipients as an off-the-shelf multi-respiratory virus-directed product is currently underway (NCT04933968, https://clinicaltrials.gov).


Assuntos
Infecções por Vírus Respiratório Sincicial , Vírus Sincicial Respiratório Humano , Humanos , Antivirais/uso terapêutico , Imunoterapia , Infecções por Vírus Respiratório Sincicial/tratamento farmacológico , Linfócitos T
10.
Proc Natl Acad Sci U S A ; 117(37): 22920-22931, 2020 09 15.
Artigo em Inglês | MEDLINE | ID: mdl-32873644

RESUMO

Animal models of human antigen-specific B cell receptors (BCRs) generally depend on "inferred germline" sequences, and thus their relationship to authentic naive human B cell BCR sequences and affinities is unclear. Here, BCR sequences from authentic naive human VRC01-class B cells from healthy human donors were selected for the generation of three BCR knockin mice. The BCRs span the physiological range of affinities found in humans, and use three different light chains (VK3-20, VK1-5, and VK1-33) found among subclasses of naive human VRC01-class B cells and HIV broadly neutralizing antibodies (bnAbs). The germline-targeting HIV immunogen eOD-GT8 60mer is currently in clinical trial as a candidate bnAb vaccine priming immunogen. To attempt to model human immune responses to the eOD-GT8 60mer, we tested each authentic naive human VRC01-class BCR mouse model under rare human physiological B cell precursor frequency conditions. B cells with high (HuGL18HL) or medium (HuGL17HL) affinity BCRs were primed, recruited to germinal centers, and they affinity matured, and formed memory B cells. Precursor frequency and affinity interdependently influenced responses. Taken together, these experiments utilizing authentic naive human VRC01-class BCRs validate a central tenet of germline-targeting vaccine design and extend the overall concept of the reverse vaccinology approach to vaccine development.


Assuntos
Anticorpos Monoclonais/imunologia , Anticorpos Amplamente Neutralizantes/imunologia , Anticorpos Anti-HIV/imunologia , Receptores de Antígenos de Linfócitos B/imunologia , Vacinas contra a AIDS/imunologia , Sequência de Aminoácidos/genética , Animais , Anticorpos Neutralizantes/imunologia , Linfócitos B/imunologia , Anticorpos Amplamente Neutralizantes/farmacologia , Antígenos CD4/imunologia , Técnicas de Introdução de Genes/métodos , Centro Germinativo/imunologia , Antígenos HIV , Infecções por HIV/imunologia , HIV-1/imunologia , Humanos , Camundongos , Camundongos Endogâmicos , Camundongos Transgênicos , Células Precursoras de Linfócitos B/imunologia , Vacinação/métodos
11.
Biochem Biophys Res Commun ; 607: 124-130, 2022 06 04.
Artigo em Inglês | MEDLINE | ID: mdl-35367824

RESUMO

CD8+ T-cell responses exert strong suppressive pressure on viral replication and select for viral escape mutations in HIV infection. Multiple viral epitopes restricted by major histocompatibility complex class I (MHC-I) are targeted by CD8+ T cells. Sequential selection of viral escape mutations in individual epitope-coding regions could result in failure in CD8+ T cell-based viral control leading to disease progression. However, how this sequential selection of epitope mutations occurs has not fully been determined. Here, we examined sequential selection of viral mutations in seven CD8+ T-cell epitope-coding regions in a macaque AIDS model of simian immunodeficiency virus mac239 (SIVmac239) infection. In seven SIVmac239-infected Burmese rhesus macaques possessing MHC-I haplotype 90-120-Ia, selection of viral mutations was observed in five to seven of the seven 90-120-Ia-associated CD8+ T-cell epitope-coding regions in a year post-infection. Of the seven CD8+ T-cell epitopes, viral mutation selection was detected first at two epitopes, Gag206-216 and Nef9-19, but was found finally at Vif114-124 epitope in most animals. Viral loads in 6 months were significantly associated with the number of mutated CD8+ T-cell epitope-coding regions 1 year post-infection. Tetramer analysis revealed early induction of Gag241-249 specific CD8+ T-cell responses, which did not always result in early selection of viral mutations in the Gag241-249 epitope, suggesting that the order of epitope mutation selection may not be determined only by immunodominance. This SIV infection model using 90-120-Ia-positive macaques would be useful for analysis of the determinants for sequential epitope mutation selection, contributing to our understanding of virus-host CD8+ T-cell interaction in HIV infection.


Assuntos
Infecções por HIV , Síndromes de Imunodeficiência , Síndrome de Imunodeficiência Adquirida dos Símios , Vírus da Imunodeficiência Símia , Animais , Linfócitos T CD8-Positivos , Epitopos de Linfócito T/genética , Antígenos de Histocompatibilidade Classe I/genética , Macaca mulatta , Mutação , Síndrome de Imunodeficiência Adquirida dos Símios/genética , Vírus da Imunodeficiência Símia/genética
12.
Proc Natl Acad Sci U S A ; 116(27): 13474-13479, 2019 07 02.
Artigo em Inglês | MEDLINE | ID: mdl-31213541

RESUMO

A major obstacle to vaccination against antigenically variable viruses is skewing of antibody responses to variable immunodominant epitopes. For influenza virus hemagglutinin (HA), the immunodominance of the variable head impairs responses to the highly conserved stem. Here, we show that head immunodominance depends on the physical attachment of head to stem. Stem immunogenicity is enhanced by immunizing with stem-only constructs or by increasing local HA concentration in the draining lymph node. Surprisingly, coimmunization of full-length HA and stem alters stem-antibody class switching. Our findings delineate strategies for overcoming immunodominance, with important implications for human vaccination.


Assuntos
Anticorpos Neutralizantes/imunologia , Epitopos/imunologia , Hemaglutininas/imunologia , Epitopos Imunodominantes/imunologia , Vírus da Influenza A/imunologia , Vacinas contra Influenza/imunologia , Animais , Anticorpos Antivirais/imunologia , Linfócitos B/imunologia , Feminino , Camundongos , Camundongos Endogâmicos C57BL , Infecções por Orthomyxoviridae/imunologia , Infecções por Orthomyxoviridae/prevenção & controle , Células-Tronco/imunologia
13.
Immunol Rev ; 283(1): 113-128, 2018 05.
Artigo em Inglês | MEDLINE | ID: mdl-29664573

RESUMO

During infections and cancer, the composition of the T-cell receptor (TCR) repertoire of antigen-specific CD8+ T cells changes over time. TCR avidity is thought to be a major driver of this process, thereby interacting with several additional regulators of T-cell responses to form a composite immune response architecture. Infections with latent viruses, such as cytomegalovirus (CMV), can lead to large T-cell responses characterized by an oligoclonal TCR repertoire. Here, we review the current status of experimental studies and theoretical models of TCR repertoire evolution during CMV infection. We will particularly discuss the degree to which this process may be determined through structural TCR avidity. As engineered TCR-redirected T cells have moved into the spotlight for providing more effective immunotherapies, it is essential to understand how the key features of a given TCR influence T-cell expansion and maintenance in settings of infection or malignancy. Deeper insights into these mechanisms will improve our basic understanding of T-cell immunology and help to identify optimal TCRs for immunotherapy.


Assuntos
Infecções por Citomegalovirus/genética , Infecções por Citomegalovirus/imunologia , Citomegalovirus/imunologia , Receptores de Antígenos de Linfócitos T/genética , Especificidade do Receptor de Antígeno de Linfócitos T/imunologia , Linfócitos T/imunologia , Linfócitos T/metabolismo , Animais , Infecções por Citomegalovirus/metabolismo , Epitopos de Linfócito T/imunologia , Variação Genética , Humanos , Imunoterapia , Ativação Linfocitária/imunologia , Receptores de Antígenos de Linfócitos T/metabolismo , Especificidade do Receptor de Antígeno de Linfócitos T/genética , Vacinas/imunologia
14.
Immunol Rev ; 284(1): 91-105, 2018 07.
Artigo em Inglês | MEDLINE | ID: mdl-29944766

RESUMO

CD4 T cells convey a number of discrete functions to protective immunity to influenza, a complexity that distinguishes this arm of adaptive immunity from B cells and CD8 T cells. Although the most well recognized function of CD4 T cells is provision of help for antibody production, CD4 T cells are important in many aspects of protective immunity. Our studies have revealed that viral antigen specificity is a key determinant of CD4 T cell function, as illustrated both by mouse models of infection and human vaccine responses, a factor whose importance is due at least in part to events in viral antigen handling. We discuss research that has provided insight into the diverse viral epitope specificity of CD4 T cells elicited after infection, how this primary response is modified as CD4 T cells home to the lung, establish memory, and after challenge with a secondary and distinct influenza virus strain. Our studies in human subjects point out the challenges facing vaccine efforts to facilitate responses to novel and avian strains of influenza, as well as strategies that enhance the ability of CD4 T cells to promote protective antibody responses to both seasonal and potentially pandemic strains of influenza.


Assuntos
Antígenos Virais/imunologia , Linfócitos T CD4-Positivos/imunologia , Glicoproteínas de Hemaglutininação de Vírus da Influenza/imunologia , Memória Imunológica/imunologia , Vírus da Influenza A Subtipo H1N1/imunologia , Influenza Humana/imunologia , Animais , Anticorpos Antivirais/imunologia , Linfócitos B , Modelos Animais de Doenças , Humanos , Camundongos
15.
J Virol ; 94(5)2020 02 14.
Artigo em Inglês | MEDLINE | ID: mdl-31826989

RESUMO

Reactivation of herpes simplex virus 1 (HSV-1) from neurons in sensory ganglia such as the trigeminal ganglia (TG) is influenced by virus-specific CD8+ T cells that infiltrate the ganglia at the onset of latency and contract to a stable activated tissue-resident memory population. In C57BL/6 mice, half of HSV-specific CD8+ T cells (gB-CD8s) recognize one dominant epitope (residues 498 to 505) on glycoprotein B (gB498-505), while the remainder (non-gB-CD8s) recognize 19 subdominant epitopes from 12 viral proteins. To address how expression by HSV-1 influences the formation and ganglionic retention of CD8+ T cell populations, we developed recombinant HSV-1 with the native immunodominant gB epitope disrupted but then expressed ectopically from different viral promoters. In mice, the epitope expressed from the gB promoter restored full gB-CD8 immunodominance to 50%. Intriguingly, earlier expression from constitutive, immediate-early, and early promoters did not significantly increase immunodominance, indicating that these promoters cannot elicit more than half of the CD8 compartment. Epitope expressed from candidate viral promoters of "true late" HSV-1 genes either delayed or reduced the priming efficiency of gB-CD8s and their levels in the TG at early times. HSV expressing the epitope from the full latency-associated transcript promoter did not efficiently prime gB-CD8s; however, gB-CD8s primed by a concurrent wild-type flank infection infiltrated the TG and were retained long term, suggesting that latent epitope expression is sufficient to retain gB-CD8s. Taken together, the data indicate that viral promoters shape latent HSV-1-specific CD8+ T cell populations and should be an important consideration in future vaccine design.IMPORTANCE Latency of HSV-1 in host neurons enables long-term persistence from which reactivation may occur to cause recurrent diseases, such as blinding herpetic stromal keratitis. Latency is not antigenically silent, and viral proteins are sporadically expressed at low levels without full virion production. This protein expression is recognized by ganglion-resident HSV-1-specific CD8+ T cells that maintain a protective resident population. Since these T cells can influence lytic/latent decisions in reactivating neurons, we argue that improving their ganglionic retention and function may offer a strategy in vaccine design to reduce reactivation and recurrent disease. To understand factors driving the infiltration and retention of ganglionic CD8s, we examined several HSV recombinants that have different viral promoters driving expression of the immunodominant gB epitope. We show that the selection of epitope promoter influences CD8+ T cell population hierarchies and their function.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Gânglios/imunologia , Herpes Simples/imunologia , Herpesvirus Humano 1/imunologia , Epitopos Imunodominantes/imunologia , Animais , Chlorocebus aethiops , Modelos Animais de Doenças , Feminino , Gânglios Sensitivos/imunologia , Herpes Simples/virologia , Herpesvirus Humano 1/genética , Ceratite Herpética/imunologia , Cinética , Camundongos , Camundongos Endogâmicos C57BL , Gânglio Trigeminal/virologia , Células Vero , Proteínas do Envelope Viral/genética
16.
J Virol ; 94(16)2020 07 30.
Artigo em Inglês | MEDLINE | ID: mdl-32493826

RESUMO

Humoral immune protection against influenza virus infection is mediated largely by antibodies against hemagglutinin (HA) and neuraminidase (NA), the two major glycoproteins on the virus surface. While influenza virus vaccination efforts have focused mainly on HA, NA-based immunity has been shown to reduce disease severity and provide heterologous protection. Current seasonal vaccines do not elicit strong anti-NA responses-in part due to the immunodominance of the HA protein. Here, we demonstrate that by swapping the 5' and 3' terminal packaging signals of the HA and NA genomic segments, which contain the RNA promoters, we are able to rescue influenza viruses that express more NA and less HA. Vaccination with formalin-inactivated "rewired" viruses significantly enhances the anti-NA antibody response compared to vaccination with unmodified viruses. Passive transfer of sera from mice immunized with rewired virus vaccines shows better protection against influenza virus challenge. Our results provide evidence that the immunodominance of HA stems in part from its abundance on the viral surface, and that rewiring viral packaging signals-thereby increasing the NA content on viral particles-is a viable strategy for improving the immunogenicity of NA in an influenza virus vaccine.IMPORTANCE Influenza virus infections are a major source of morbidity and mortality worldwide. Increasing evidence highlights neuraminidase as a potential vaccination target. This report demonstrates the efficacy of rewiring influenza virus packaging signals for creating vaccines with more neuraminidase content which provide better neuraminidase (NA)-based protection.


Assuntos
Vírus da Influenza A/genética , Neuraminidase/genética , Neuraminidase/imunologia , Animais , Anticorpos Antivirais/imunologia , Proteção Cruzada , Reações Cruzadas , Feminino , Expressão Gênica/genética , Regulação Viral da Expressão Gênica/genética , Células HEK293 , Glicoproteínas de Hemaglutininação de Vírus da Influenza/genética , Hemaglutininas/imunologia , Humanos , Vírus da Influenza A Subtipo H1N1/genética , Vírus da Influenza A Subtipo H3N2/genética , Virus da Influenza A Subtipo H5N1/genética , Vacinas contra Influenza/imunologia , Influenza Humana/virologia , Camundongos , Camundongos Endogâmicos BALB C , Infecções por Orthomyxoviridae/virologia , RNA/genética , Vacinação/métodos
17.
Immunol Invest ; 50(7): 780-801, 2021 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-33779464

RESUMO

As one of the most important weapons against infectious diseases, vaccines have saved countless lives since their first use in the late eighteenth century. Antibodies produced by effector B cells upon vaccination play a critical role in mediating protection. The past several decades of research have led to a revolution in our understanding of B cell response to vaccination. Vaccines against SARS-CoV-2 coronavirus were developed at an unprecedented speed to power our global fight against COVID-19 pandemic. Nevertheless, we still face many challenges in the development of vaccines against many other deadly viruses, such as human immunodeficiency virus (HIV) and influenza virus. In this review, we summarize the latest findings on B cell response to vaccination and pathogen infection. We also discuss the current challenges in the field and the potential strategies targeting B cell response to improve vaccine efficacy.Key abbreviations box: BCR: B cell receptor; bNAb: broadly neutralizing antibody; DC: dendritic cells; DZ: dark zone; EF response: extrafollicular response; FDC: follicular dendritic cell; GC: germinal center; HIV: human immunodeficiency virus; IC: immune complex; LLPC: long-lived plasma cell; LZ: light zone; MBC: memory B cell; SLPB: short-lived plasmablast; TFH: T follicular helper cells; TLR: Toll-like receptor.


Assuntos
Linfócitos B/imunologia , Vacinas contra COVID-19/imunologia , COVID-19/imunologia , Pandemias/prevenção & controle , Animais , Humanos , SARS-CoV-2/imunologia , Vacinação/métodos
18.
Immunogenetics ; 72(1-2): 109-118, 2020 02.
Artigo em Inglês | MEDLINE | ID: mdl-31811313

RESUMO

Upon recognition of peptide-MHC complexes by T cell receptors (TCR), the cognate T cells expand and differentiate into effector T cells to generate protective immunity. Despite the fact that any immune response generates a diverse set of TCR clones against a particular epitope, only a few clones are highly expanded in any immune response. Previous studies observed that the highest frequency clones usually control viral infections better than subdominant clones, but the reasons for this dominance among T cell clones are still unclear. Here, we used publicly available TCR amino acid sequences to study which factors determine whether a response becomes immunodominance (ID) per donor; we classified the largest T cell clone as the epitope-specific dominant clone and all the other clones as subdominant responses (SD). We observed a distinctively hydrophobic CDR3 in ID responses against a dominant epitope from influenza A virus, compared to the SD responses. The common V-J combinations were shared between ID and SD responses, suggesting that the biased V-J recombination events are restricted by epitope specificity; thus, the immunodominance is not directly determined by a bias combination of V and J genetic segments. Our findings reveal a close similarity of global sequence properties between dominant and subdominant clones of epitope-specific responses but detectable distinctive amino acid enrichments in ID. Taken together, we believe this first comparative study of immunodominant and subdominant TCR sequences can guide further studies to resolve factors determining the immunodominance of antiviral as well as tumor-specific T cell responses.


Assuntos
Regiões Determinantes de Complementaridade/genética , Epitopos Imunodominantes/genética , Receptores de Antígenos de Linfócitos T/genética , Sequência de Aminoácidos , Linfócitos T CD8-Positivos/imunologia , Regiões Determinantes de Complementaridade/metabolismo , Bases de Dados Factuais , Epitopos/genética , Epitopos/imunologia , Epitopos de Linfócito T/imunologia , Humanos , Imunidade Celular , Epitopos Imunodominantes/imunologia , Ativação Linfocitária , Dados de Sequência Molecular , Receptores de Antígenos de Linfócitos T/imunologia , Recombinação V(D)J/genética
19.
J Virol ; 93(18)2019 09 15.
Artigo em Inglês | MEDLINE | ID: mdl-31375573

RESUMO

Influenza viruses express two surface glycoproteins, the hemagglutinin (HA) and the neuraminidase (NA). Anti-NA antibodies protect from lethal influenza virus challenge in the mouse model and correlate inversely with virus shedding and symptoms in humans. Consequently, the NA is a promising target for influenza virus vaccine design. Current seasonal vaccines, however, poorly induce anti-NA antibodies, partly because of the immunodominance of the HA over the NA when the two glycoproteins are closely associated. To address this issue, here we investigated whether extending the stalk domain of the NA could render it more immunogenic on virus particles. Two recombinant influenza viruses based on the H1N1 strain A/Puerto Rico/8/1934 (PR8) were rescued with NA stalk domains extended by 15 or 30 amino acids. Formalin-inactivated viruses expressing wild-type NA or the stalk-extended NA variants were used to vaccinate mice. The virus with the 30-amino-acid stalk extension induced significantly higher anti-NA IgG responses (characterized by increased in vitro antibody-dependent cellular cytotoxicity [ADCC] activity) than the wild-type PR8 virus, while anti-HA IgG levels were unaffected. Similarly, extending the stalk domain of the NA of a recent H3N2 virus enhanced the induction of anti-NA IgGs in mice. On the basis of these results, we hypothesize that the subdominance of the NA can be modulated if the protein is modified such that its height surpasses that of the HA on the viral membrane. Extending the stalk domain of NA may help to enhance its immunogenicity in influenza virus vaccines without compromising antibody responses to HA.IMPORTANCE The efficacy of influenza virus vaccines could be improved by enhancing the immunogenicity of the NA protein. One of the reasons for its poor immunogenicity is the immunodominance of the HA over the NA in many seasonal influenza virus vaccines. Here we demonstrate that, in the mouse model, extending the stalk domain of the NA protein can enhance its immunogenicity on virus particles and overcome the immunodominance of the HA without affecting antibody responses to the HA. The antibody repertoire is broadened by the extended NA and includes additional ADCC-active antibodies. Our findings may assist in the efforts toward more effective influenza virus vaccines.


Assuntos
Neuraminidase/imunologia , Orthomyxoviridae/imunologia , Orthomyxoviridae/metabolismo , Animais , Anticorpos Antivirais/imunologia , Reações Cruzadas , Cães , Feminino , Células HEK293 , Glicoproteínas de Hemaglutininação de Vírus da Influenza/imunologia , Hemaglutininas/imunologia , Humanos , Vírus da Influenza A Subtipo H1N1/imunologia , Vírus da Influenza A Subtipo H3N2/imunologia , Virus da Influenza A Subtipo H5N1/imunologia , Vacinas contra Influenza/imunologia , Influenza Humana/virologia , Células Madin Darby de Rim Canino , Camundongos Endogâmicos BALB C , Neuraminidase/genética , Neuraminidase/metabolismo , Infecções por Orthomyxoviridae/virologia , Vacinação
20.
J Virol ; 93(2)2019 01 15.
Artigo em Inglês | MEDLINE | ID: mdl-30381487

RESUMO

The influenza B virus hemagglutinin contains four major antigenic sites (the 120 loop, the 150 loop, the 160 loop, and the 190 helix) within the head domain. These immunodominant antigenic sites are the main targets of neutralizing antibodies and are subject to antigenic drift. Yet little is known about the specific antibody responses toward each site in terms of antibody prevalence and hemagglutination inhibition activity. In this study, we used modified hemagglutinins of influenza B virus which display only one or none of the major antigenic sites to measure antibody responses toward the classical as well as the noncanonical epitopes in mice, ferrets, and humans. With our novel reagents, we found that both hemagglutination inhibition antibodies and total IgGs were mostly induced by the major antigenic sites. However, in human adults, we observed high hemagglutination inhibition antibody responses toward the noncanonical epitopes. By stratifying the human samples into age groups, we found that the noncanonical antibody responses appeared to increase with age.IMPORTANCE This study dissected the specific antibody responses toward the major antigenic sites and the noncanonical epitopes of influenza B virus hemagglutinin in animals and humans using novel reagents. These findings will guide the design of the next generation of influenza virus vaccines.


Assuntos
Anticorpos Neutralizantes/metabolismo , Glicoproteínas de Hemaglutininação de Vírus da Influenza/química , Vírus da Influenza B/imunologia , Influenza Humana/imunologia , Adulto , Fatores Etários , Idoso , Animais , Anticorpos Antivirais/metabolismo , Pré-Escolar , Cães , Furões , Deriva Genética , Células HEK293 , Glicoproteínas de Hemaglutininação de Vírus da Influenza/genética , Glicoproteínas de Hemaglutininação de Vírus da Influenza/imunologia , Humanos , Epitopos Imunodominantes/imunologia , Lactente , Vírus da Influenza B/genética , Vacinas contra Influenza/imunologia , Influenza Humana/virologia , Células Madin Darby de Rim Canino , Camundongos , Pessoa de Meia-Idade , Especificidade da Espécie
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA