Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
1.
Ann N Y Acad Sci ; 1522(1): 60-73, 2023 04.
Artigo em Inglês | MEDLINE | ID: mdl-36722473

RESUMO

Respiratory viruses are a common cause of morbidity and mortality around the world. Viruses like influenza, RSV, and most recently SARS-CoV-2 can rapidly spread through a population, causing acute infection and, in vulnerable populations, severe or chronic disease. Developing effective treatment and prevention strategies often becomes a race against ever-evolving viruses that develop resistance, leaving therapy efficacy either short-lived or relevant for specific viral strains. On June 29 to July 2, 2022, researchers met for the Keystone symposium "Respiratory Viruses: New Frontiers." Researchers presented new insights into viral biology and virus-host interactions to understand the mechanisms of disease and identify novel treatment and prevention approaches that are effective, durable, and broad.


Assuntos
COVID-19 , Influenza Humana , Infecções por Vírus Respiratório Sincicial , Humanos , COVID-19/patologia , COVID-19/virologia , Interações entre Hospedeiro e Microrganismos , Influenza Humana/patologia , Influenza Humana/virologia , Infecções por Vírus Respiratório Sincicial/patologia , Infecções por Vírus Respiratório Sincicial/virologia , Vírus Sinciciais Respiratórios , SARS-CoV-2
2.
Semin Cell Dev Biol ; 146: 40-56, 2023 09 15.
Artigo em Inglês | MEDLINE | ID: mdl-36737258

RESUMO

Alternative RNA splicing is a co-transcriptional process that richly increases proteome diversity, and is dynamically regulated based on cell species, lineage, and activation state. Virus infection in vertebrate hosts results in rapid host transcriptome-wide changes, and regulation of alternative splicing can direct a combinatorial effect on the host transcriptome. There has been a recent increase in genome-wide studies evaluating host alternative splicing during viral infection, which integrates well with prior knowledge on viral interactions with host splicing proteins. A critical challenge remains in linking how these individual events direct global changes, and whether alternative splicing is an overall favorable pathway for fending off or supporting viral infection. Here, we introduce the process of alternative splicing, discuss how to analyze splice regulation, and detail studies on genome-wide and splice factor changes during viral infection. We seek to highlight where the field can focus on moving forward, and how incorporation of a virus-host co-evolutionary perspective can benefit this burgeoning subject.


Assuntos
Processamento Alternativo , Vírus , Processamento Alternativo/genética , Vírus/genética , Genoma , Transcriptoma , Proteoma/genética
3.
PLoS Biol ; 20(12): e3001934, 2022 12.
Artigo em Inglês | MEDLINE | ID: mdl-36542656

RESUMO

Viruses must balance their reliance on host cell machinery for replication while avoiding host defense. Influenza A viruses are zoonotic agents that frequently switch hosts, causing localized outbreaks with the potential for larger pandemics. The host range of influenza virus is limited by the need for successful interactions between the virus and cellular partners. Here we used immunocompetitive capture-mass spectrometry to identify cellular proteins that interact with human- and avian-style viral polymerases. We focused on the proviral activity of heterogenous nuclear ribonuclear protein U-like 1 (hnRNP UL1) and the antiviral activity of mitochondrial enoyl CoA-reductase (MECR). MECR is localized to mitochondria where it functions in mitochondrial fatty acid synthesis (mtFAS). While a small fraction of the polymerase subunit PB2 localizes to the mitochondria, PB2 did not interact with full-length MECR. By contrast, a minor splice variant produces cytoplasmic MECR (cMECR). Ectopic expression of cMECR shows that it binds the viral polymerase and suppresses viral replication by blocking assembly of viral ribonucleoprotein complexes (RNPs). MECR ablation through genome editing or drug treatment is detrimental for cell health, creating a generic block to virus replication. Using the yeast homolog Etr1 to supply the metabolic functions of MECR in MECR-null cells, we showed that specific antiviral activity is independent of mtFAS and is reconstituted by expressing cMECR. Thus, we propose a strategy where alternative splicing produces a cryptic antiviral protein that is embedded within a key metabolic enzyme.


Assuntos
Ácidos Graxos Dessaturases , Vírus da Influenza A , Humanos , Ácidos Graxos Dessaturases/metabolismo , Processamento Alternativo/genética , Mitocôndrias/metabolismo , Vírus da Influenza A/genética , Isoformas de Proteínas/metabolismo , Replicação Viral
4.
Cell Rep ; 29(8): 2175-2183.e4, 2019 11 19.
Artigo em Inglês | MEDLINE | ID: mdl-31747592

RESUMO

All viruses balance interactions between cellular machinery co-opted to support replication and host factors deployed to halt the infection. We use gene correlation analysis to perform an unbiased screen for host factors involved in influenza A virus (FLUAV) infection. Our screen identifies the cellular factor epidermal growth factor receptor pathway substrate 8 (EPS8) as the highest confidence pro-viral candidate. Knockout and overexpression of EPS8 confirm its importance in enhancing FLUAV infection and titers. Loss of EPS8 does not affect virion attachment, uptake, or fusion. Rather, our data show that EPS8 specifically functions during virion uncoating. EPS8 physically associates with incoming virion components, and subsequent nuclear import of released ribonucleoprotein complexes is significantly delayed in the absence of EPS8. Our study identifies EPS8 as a host factor important for uncoating, a crucial step of FLUAV infection during which the interface between the virus and host is still being discovered.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Vírus da Influenza A/patogenicidade , Proteínas Adaptadoras de Transdução de Sinal/genética , Humanos , Ribonucleoproteínas/genética , Ribonucleoproteínas/metabolismo , Transdução de Sinais/genética , Transdução de Sinais/fisiologia , Vírion/genética , Vírion/metabolismo
5.
Elife ; 82019 06 10.
Artigo em Inglês | MEDLINE | ID: mdl-31179971

RESUMO

The evolutionary history of ANP32 proteins impacts how influenza virus jumps from birds to mammals.


Assuntos
Vírus da Influenza A , Influenza Aviária , Orthomyxoviridae , Animais , Proteínas de Ligação a RNA , Proteínas Virais
6.
Cell Rep ; 24(10): 2581-2588.e4, 2018 09 04.
Artigo em Inglês | MEDLINE | ID: mdl-30184493

RESUMO

Adaptation of viruses to their hosts can result in specialization and a restricted host range. Species-specific polymorphisms in the influenza virus polymerase restrict its host range during transmission from birds to mammals. ANP32A was recently identified as a cellular co-factor affecting polymerase adaption and activity. Avian influenza polymerases require ANP32A containing an insertion resulting from an exon duplication uniquely encoded in birds. Here we find that natural splice variants surrounding this exon create avian ANP32A proteins with distinct effects on polymerase activity. We demonstrate species-independent direct interactions between all ANP32A variants and the PB2 polymerase subunit. This interaction is enhanced in the presence of viral genomic RNA. In contrast, only avian ANP32A restored ribonucleoprotein complex assembly for a restricted polymerase by enhancing RNA synthesis. Our data suggest that ANP32A splicing variation among birds differentially affects viral replication, polymerase adaption, and the potential of avian hosts to be reservoirs.


Assuntos
Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Orthomyxoviridae/genética , Orthomyxoviridae/patogenicidade , Splicing de RNA/genética , Proteínas Virais/metabolismo , Animais , Aves , Linhagem Celular , Éxons/genética , Imunofluorescência , Humanos , Imunoprecipitação , Peptídeos e Proteínas de Sinalização Intracelular/genética , Ligação Proteica/genética , Ligação Proteica/fisiologia , RNA Viral/genética , RNA Viral/metabolismo , Proteínas Virais/genética
7.
Elife ; 62017 07 31.
Artigo em Inglês | MEDLINE | ID: mdl-28758638

RESUMO

Influenza virus expresses transcripts early in infection and transitions towards genome replication at later time points. This process requires de novo assembly of the viral replication machinery, large ribonucleoprotein complexes (RNPs) composed of the viral polymerase, genomic RNA and oligomeric nucleoprotein (NP). Despite the central role of RNPs during infection, the factors dictating where and when they assemble are poorly understood. Here we demonstrate that human protein kinase C (PKC) family members regulate RNP assembly. Activated PKCδ interacts with the polymerase subunit PB2 and phospho-regulates NP oligomerization and RNP assembly during infection. Consistent with its role in regulating RNP assembly, knockout of PKCδ impairs virus infection by selectively disrupting genome replication. However, primary transcription from pre-formed RNPs deposited by infecting particles is unaffected. Thus, influenza virus exploits host PKCs to regulate RNP assembly, a step required for the transition from primary transcription to genome replication during the infectious cycle.


Assuntos
Interações Hospedeiro-Patógeno , Vírus da Influenza A Subtipo H1N1/genética , Proteína Quinase C-delta/genética , RNA Viral/genética , RNA Polimerase Dependente de RNA/genética , Ribonucleoproteínas/genética , Proteínas Virais/genética , Replicação Viral , Células A549 , Animais , Cães , Regulação da Expressão Gênica , Células HEK293 , Humanos , Vírus da Influenza A Subtipo H1N1/crescimento & desenvolvimento , Vírus da Influenza A Subtipo H1N1/metabolismo , Isoenzimas/genética , Isoenzimas/metabolismo , Células Madin Darby de Rim Canino , Modelos Moleculares , Mutação , Fosforilação , Ligação Proteica , Conformação Proteica , Proteína Quinase C-delta/metabolismo , RNA Viral/metabolismo , RNA Polimerase Dependente de RNA/metabolismo , Ribonucleoproteínas/metabolismo , Transdução de Sinais , Transcrição Gênica , Proteínas Virais/metabolismo
8.
Virus Res ; 225: 1-9, 2016 10 02.
Artigo em Inglês | MEDLINE | ID: mdl-27596738

RESUMO

Influenza virus neuraminidase (NA) plays a pivotal role during viral growth since its sialidase activity allows the efficient release of nascent virions from infected cells. Consequently, mutations in the NA catalytic site affecting sialic acid (SA) cleavage may influence the biological properties of influenza viruses. This study reports two amino acid substitutions (N386K and P431S) in the NA of the influenza A(H1N1)pdm09 virus that emerged in 2009 in Mexico. The NA sialidase activity to cleave SA-like substrates, and viral growth were examined and the mutant viruses had various deficiencies. NA mutations N386K and P431S together or separately, and in the presence or absence of H275Y were further evaluated using recombinant influenza A/California/04/2009 (pH1N1) viruses containing single, double, or triple mutations. Viral growth was reduced in the presence of mutation P431S alone or combined with N386K and/or H275Y. Substrates hydrolysis was reduced when recombinant pH1N1 viruses were analyzed by NA inhibitory assays. Moreover, elution assays with guinea pig red blood cells indicated an unbalanced hemagglutinin (HA):NA functionality. Altogether, our data underline the functional significance of mutations at highly conserved sites in influenza virus NA glycoprotein and the occurrence of permissive mutations to compensate virus viability in vitro.


Assuntos
Vírus da Influenza A Subtipo H1N1/enzimologia , Vírus da Influenza A Subtipo H1N1/genética , Mutação , Neuraminidase/genética , Neuraminidase/metabolismo , Proteínas Virais/genética , Proteínas Virais/metabolismo , Substituição de Aminoácidos , Animais , Antivirais/farmacologia , Linhagem Celular , Relação Dose-Resposta a Droga , Ativação Enzimática , Cobaias , Glicoproteínas de Hemaglutininação de Vírus da Influenza/metabolismo , Humanos , Vírus da Influenza A Subtipo H1N1/efeitos dos fármacos , Testes de Sensibilidade Microbiana , Replicação Viral
9.
PLoS One ; 11(6): e0156906, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27272307

RESUMO

Influenza A virus is a significant public health threat, but little is understood about the viral RNA structure and function. Current vaccines and therapeutic options to control influenza A virus infections are mostly protein-centric and of limited effectiveness. Here, we report using an ensemble defect approach to design mutations to misfold regions of conserved mRNA structures in influenza A virus segments 7 and 8. Influenza A mutant viruses inhibit pre-mRNA splicing and attenuate viral replication in cell culture, thus providing evidence for functions of the targeted regions. Targeting these influenza A viral RNA regions provides new possibilities for designing vaccines and therapeutics against this important human respiratory pathogen. The results also demonstrate that the ensemble defect approach is an efficient way to test for function of RNA sequences.


Assuntos
Vírus da Influenza A/fisiologia , Mutação , RNA Viral/química , RNA Viral/genética , Células A549 , Animais , Técnicas de Cultura de Células , Sequência Conservada , Cães , Células HEK293 , Humanos , Vírus da Influenza A/genética , Células Madin Darby de Rim Canino , Modelos Moleculares , Conformação de Ácido Nucleico , Dobramento de RNA , Splicing de RNA , Replicação Viral
10.
Viruses ; 8(7)2016 06 23.
Artigo em Inglês | MEDLINE | ID: mdl-27347991

RESUMO

Influenza A viruses (IAV) cause annual seasonal human respiratory disease epidemics. In addition, IAV have been implicated in occasional pandemics with inordinate health and economic consequences. Studying IAV, in vitro or in vivo, requires the use of laborious secondary methodologies to identify virus-infected cells. To circumvent this requirement, replication-competent IAV expressing an easily traceable reporter protein can be used. Here we discuss the development and applications of recombinant replication-competent IAV harboring diverse fluorescent or bioluminescent reporter genes in different locations of the viral genome. These viruses have been employed for in vitro and in vivo studies, such as the screening of neutralizing antibodies or antiviral compounds, the identification of host factors involved in viral replication, cell tropism, the development of vaccines, or the assessment of viral infection dynamics. In summary, reporter-expressing, replicating-competent IAV represent a powerful tool for the study of IAV both in vitro and in vivo.


Assuntos
Expressão Gênica , Genes Reporter , Vírus da Influenza A/genética , Vírus da Influenza A/fisiologia , Coloração e Rotulagem/métodos , Replicação Viral , Humanos , Proteínas Luminescentes/análise , Proteínas Luminescentes/genética , Recombinação Genética , Genética Reversa , Virologia/métodos
11.
PLoS One ; 11(1): e0147723, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26809059

RESUMO

Influenza A and B viruses (IAV and IBV, respectively) cause annual seasonal human respiratory disease epidemics. In addition, IAVs have been implicated in occasional pandemics with inordinate health and economic consequences. Studying influenza viruses in vitro or in vivo requires the use of laborious secondary methodologies to identify infected cells. To circumvent this requirement, replication-competent infectious influenza viruses expressing an easily traceable fluorescent reporter protein can be used. Timer is a fluorescent protein that undergoes a time-dependent color emission conversion from green to red. The rate of spectral change is independent of Timer protein concentration and can be used to chronologically measure the duration of its expression. Here, we describe the generation of replication-competent IAV and IBV where the viral non-structural protein 1 (NS1) was fused to the fluorescent dynamic Timer protein. Timer-expressing IAV and IBV displayed similar plaque phenotypes and growth kinetics to wild-type viruses in tissue culture. Within infected cells, Timer's spectral shift can be used to measure the rate and cell-to-cell spread of infection using fluorescent microscopy, plate readers, or flow cytometry. The progression of Timer-expressing IAV infection was also evaluated in a mouse model, demonstrating the feasibility to characterize IAV cell-to-cell infections in vivo. By providing the ability to chronologically track viral spread, Timer-expressing influenza viruses are an excellent option to evaluate the in vitro and in vivo dynamics of viral infection.


Assuntos
Vírus da Influenza A/fisiologia , Vírus da Influenza B/fisiologia , Influenza Humana/metabolismo , Proteínas Luminescentes/metabolismo , Animais , Cães , Células HEK293 , Humanos , Vírus da Influenza A/genética , Vírus da Influenza B/genética , Influenza Humana/genética , Proteínas Luminescentes/genética , Replicação Viral/genética , Replicação Viral/fisiologia
12.
J Biopharm Stat ; 26(3): 409-20, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26010892

RESUMO

Viruses that express reporter genes upon infection have been recently used to evaluate neutralizing antibody responses, where a lack of reporter expression indicates specific virus inhibition. The traditional model-based methods using standard outcome of percent neutralization could be applied to the data from the assays to estimate antibody titers. However, the data produced are sometimes irregular, which can yield meaningless outcomes of percent neutralization that do not fit the typical curves for immunoassays, making automated or semi-high throughput antibody titer estimation unreliable. We developed a type of new outcomes model, which is biologically meaningful and fits typical immunoassay curves well. Our simulation study indicates that the new response approach outperforms the traditional response approach regardless of the data variability. The proposed new response approach can be used in similar assays for other disease models.


Assuntos
Proteínas de Fluorescência Verde/química , Testes de Neutralização/métodos , Anticorpos Neutralizantes/análise , Anticorpos Antivirais/análise , Glicoproteínas de Hemaglutininação de Vírus da Influenza/análise , Modelos Estatísticos , Método de Monte Carlo
13.
Virus Res ; 216: 26-40, 2016 May 02.
Artigo em Inglês | MEDLINE | ID: mdl-26220478

RESUMO

The diverse host range, high transmissibility, and rapid evolution of influenza A viruses justify the importance of containing pathogenic viruses studied in the laboratory. Other than physically or mechanically changing influenza A virus containment procedures, modifying the virus to only replicate for a single round of infection similarly ensures safety and consequently decreases the level of biosafety containment required to study highly pathogenic members in the virus family. This biological containment is more ideal because it is less apt to computer, machine, or human error. With many necessary proteins that can be deleted, generation of single-cycle infectious influenza A viruses (sciIAV) can be achieved using a variety of approaches. Here, we review the recent burst in sciIAV generation and summarize the applications and findings on this important human pathogen using biocontained viral mimics.


Assuntos
Vírus da Influenza A/fisiologia , Influenza Humana/virologia , Animais , Humanos , Vírus da Influenza A/genética , Proteínas Virais/genética , Proteínas Virais/metabolismo , Replicação Viral
14.
Future Virol ; 10(6): 715-730, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-26213563

RESUMO

Vaccination represents the best option to protect humans against influenza virus. However, improving the effectiveness of current vaccines could better stifle the health burden caused by viral infection. Protein synthesis from individual genes can be downregulated by synthetically deoptimizing a gene's codon usage. With more rapid and affordable nucleotide synthesis, generating viruses that contain genes with deoptimized codons is now feasible. Attenuated, vaccine-candidate viruses can thus be engineered with hitherto uncharacterized properties. With eight gene segments, influenza A viruses with variably recoded genomes can produce a spectrum of attenuation that is contingent on the gene segment targeted and the number of codon changes. This review summarizes different targets and approaches to deoptimize influenza A virus codons for novel vaccine generation.

15.
Vaccine ; 33(30): 3562-70, 2015 Jul 09.
Artigo em Inglês | MEDLINE | ID: mdl-26044496

RESUMO

Avian-derived influenza A zoonoses are closely monitored and may be an indication of virus strains with pandemic potential. Both successful vaccination and convalescence of influenza A virus in humans typically results in the induction of antibodies that can neutralize viral infection. To improve long-standing and new-generation methodologies for detection of neutralizing antibodies, we have employed a novel reporter-based approach that allows for multiple antigenic testing within a single sample. Central to this approach is a single-cycle infectious influenza A virus (sciIAV), where a functional hemagglutinin (HA) gene was changed to encode either the green or the monomeric red fluorescent protein (GFP and mRFP, respectively) and HA is complemented in trans by stable HA-expressing cell lines. By using fluorescent proteins with non-overlapping emission spectra, this novel bivalent fluorescence-based microneutralization assay (BiFMA) can be used to detect neutralizing antibodies against two distinct influenza isolates in a single reaction, doubling the speed of experimentation while halving the amount of sera required. Moreover, this approach can be used for the rapid identification of influenza broadly neutralizing antibodies. Importantly, this novel BiFMA can be used for any given influenza HA-pseudotyped virus under BSL-2 facilities, including highly pathogenic influenza HA isolates.


Assuntos
Anticorpos Neutralizantes/sangue , Fluorometria/métodos , Vírus da Influenza A/imunologia , Testes de Neutralização/métodos , Glicoproteínas de Hemaglutininação de Vírus da Influenza/genética , Humanos , Proteínas Luminescentes/análise , Proteínas Luminescentes/genética
16.
J Infect Dis ; 212(8): 1270-8, 2015 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-25838266

RESUMO

Recent studies have shown that live attenuated influenza vaccines (LAIVs) expressing avian influenza virus hemagglutinins (HAs) prime for strong protective antibody responses to an inactivated influenza vaccine (IIV) containing the HA. To better understand this priming effect, we compared H7 HA head and stalk domain-specific B-cell responses in H7N7 LAIV-primed subjects and non-H7-primed controls after a single dose of H7N7 IIV. As previously reported, H7N7 LAIV-primed subjects but not control subjects generated strong hemagglutination-inhibiting and neutralizing antibody responses to the H7N7 IIV. Here, we found that the quantity, epitope diversity, and affinity of H7 head-specific antibodies increased rapidly in only H7N7 LAIV-primed subjects after receipt of the IIV. However, all cohorts generated a vigorous, high-affinity, stalk-specific antibody response. Consistent increases in circulating memory B-cell frequencies after receipt of the IIV reflected the specificity of high-affinity antibody production. Our findings emphasize the value of LAIVs as a vehicle for prepandemic vaccination.


Assuntos
Anticorpos Antivirais/sangue , Vírus da Influenza A Subtipo H7N7/imunologia , Vacinas contra Influenza/imunologia , Influenza Humana/imunologia , Anticorpos Neutralizantes/sangue , Linfócitos B/imunologia , Estudos de Coortes , Testes de Inibição da Hemaglutinação , Humanos , Influenza Humana/prevenção & controle , Influenza Humana/virologia , Testes de Neutralização , Vacinas Atenuadas/imunologia , Vacinas de Produtos Inativados/imunologia
17.
Virology ; 476: 206-216, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25553516

RESUMO

Like most animal viruses, studying influenza A in model systems requires secondary methodologies to identify infected cells. To circumvent this requirement, we describe the generation of replication-competent influenza A red fluorescent viruses. These influenza A viruses encode mCherry fused to the viral non-structural 1 (NS1) protein and display comparable growth kinetics to wild-type viruses in vitro. Infection of cells with influenza A mCherry viruses was neutralized with monoclonal antibodies and inhibited with antivirals to levels similar to wild-type virus. Influenza A mCherry viruses were also able to lethally infect mice, and strikingly, dose- and time-dependent kinetics of viral replication were monitored in whole excised mouse lungs using an in vivo imaging system (IVIS). By eliminating the need for secondary labeling of infected cells, influenza A mCherry viruses provide an ideal tool in the ongoing struggle to better characterize the virus and identify new therapeutics against influenza A viral infections.


Assuntos
Rastreamento de Células/métodos , Vírus da Influenza A Subtipo H1N1/fisiologia , Vírus da Influenza A/fisiologia , Influenza Humana/virologia , Proteínas Luminescentes/genética , Replicação Viral , Animais , Linhagem Celular , Feminino , Expressão Gênica , Humanos , Vírus da Influenza A Subtipo H1N1/genética , Vírus da Influenza A/genética , Proteínas Luminescentes/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Proteínas não Estruturais Virais/genética , Proteínas não Estruturais Virais/metabolismo , Proteína Vermelha Fluorescente
18.
J Virol ; 89(6): 3421-6, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25552727

RESUMO

The live attenuated influenza virus vaccine (LAIV) is preferentially recommended for use in persons 2 through 49 years of age but has not been approved for children under 2 or asthmatics due to safety concerns. Therefore, increasing safety is desirable. Here we describe a murine LAIV with reduced pathogenicity that retains lethality at high doses and further demonstrate that we can enhance safety in vivo through mutations within NS1. This model may permit preliminary safety analysis of improved LAIVs.


Assuntos
Modelos Animais de Doenças , Vírus da Influenza A/imunologia , Vacinas contra Influenza/administração & dosagem , Influenza Humana/prevenção & controle , Camundongos , Vacinas Atenuadas/administração & dosagem , Animais , Anticorpos Antivirais/imunologia , Feminino , Humanos , Vírus da Influenza A/classificação , Vírus da Influenza A/genética , Vacinas contra Influenza/efeitos adversos , Vacinas contra Influenza/imunologia , Influenza Humana/imunologia , Influenza Humana/virologia , Vacinas Atenuadas/efeitos adversos , Vacinas Atenuadas/genética , Vacinas Atenuadas/imunologia
19.
J Immunol ; 193(12): 6031-40, 2014 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-25392529

RESUMO

Influenza viruses remain a critical global health concern. More efficacious vaccines are needed to protect against influenza virus, yet few adjuvants are approved for routine use. Specialized proresolving mediators (SPMs) are powerful endogenous bioactive regulators of inflammation, with great clinical translational properties. In this study, we investigated the ability of the SPM 17-HDHA to enhance the adaptive immune response using an OVA immunization model and a preclinical influenza vaccination mouse model. Our findings revealed that mice immunized with OVA plus 17-HDHA or with H1N1-derived HA protein plus 17-HDHA increased Ag-specific Ab titers. 17-HDHA increased the number of Ab-secreting cells in vitro and the number of HA-specific Ab-secreting cells present in the bone marrow. Importantly, the 17-HDHA-mediated increased Ab production was more protective against live pH1N1 influenza infection in mice. To our knowledge, this is the first report on the biological effects of ω-3-derived SPMs on the humoral immune response. These findings illustrate a previously unknown biological link between proresolution signals and the adaptive immune system. Furthermore, this work has important implications for the understanding of B cell biology, as well as the development of new potential vaccine adjuvants.


Assuntos
Ácidos Docosa-Hexaenoicos/farmacologia , Imunidade Humoral/efeitos dos fármacos , Vírus da Influenza A Subtipo H1N1/imunologia , Infecções por Orthomyxoviridae/imunologia , Adjuvantes Imunológicos/farmacologia , Animais , Anticorpos Antivirais/imunologia , Formação de Anticorpos/efeitos dos fármacos , Formação de Anticorpos/imunologia , Subpopulações de Linfócitos B/citologia , Subpopulações de Linfócitos B/efeitos dos fármacos , Subpopulações de Linfócitos B/imunologia , Subpopulações de Linfócitos B/metabolismo , Diferenciação Celular/efeitos dos fármacos , Diferenciação Celular/imunologia , Glicoproteínas de Hemaglutininação de Vírus da Influenza/imunologia , Imunoglobulina G/imunologia , Imunoglobulina M/imunologia , Masculino , Camundongos , Infecções por Orthomyxoviridae/virologia , Plasmócitos/citologia , Plasmócitos/efeitos dos fármacos , Plasmócitos/imunologia , Plasmócitos/metabolismo
20.
J Virol ; 88(20): 12006-16, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25100831

RESUMO

The effector functions of specific CD8 T cells are crucial in mediating influenza heterologous protection. However, new approaches for influenza vaccines that can trigger effective CD8 T cell responses have not been extensively explored. We report here the generation of single-cycle infectious influenza virus that lacks a functional hemagglutinin (HA) gene on an X31 genetic background and demonstrate its potential for triggering protective CD8 T cell immunity against heterologous influenza virus challenge. In vitro, X31-sciIV can infect MDCK cells, but infectious virions are not produced unless HA is transcomplemented. In vivo, intranasal immunization with X31-sciIV does not cause any clinical symptoms in mice but generates influenza-specific CD8 T cells in lymphoid (mediastinal lymph nodes and spleen) and nonlymphoid tissues, including lung and bronchoalveolar lavage fluid, as measured by H2-Db NP366 and PA224 tetramer staining. In addition, a significant proportion of X31-sciIV-induced antigen-specific respiratory CD8 T cells expressed VLA-1, a marker that is associated with heterologous influenza protection. Further, these influenza-specific CD8 T cells produce antiviral cytokines when stimulated with NP366 and PA224 peptides, indicating that CD8 T cells triggered by X31-sciIV are functional. When challenged with a lethal dose of heterologous PR8 virus, X31-sciIV-primed mice were fully protected from death. However, when CD8 T cells were depleted after priming or before priming, mice could not effectively control virus replication or survive the lethal challenge, indicating that X31-sciIV-induced memory CD8 T cells mediate the heterologous protection. Thus, our results demonstrate the potential for sciIV as a CD8 T cell-inducing vaccine. Importance: One of the challenges for influenza prevention is the existence of multiple influenza virus subtypes and variants and the fact that new strains can emerge yearly. Numerous studies have indicated that the effector functions of specific CD8 T cells are crucial in mediating influenza heterologous protection. However, influenza vaccines that can trigger effective CD8 T cell responses for heterologous protection have not been developed. We report here the generation of an X31 (H3N2) virus-derived single-cycle infectious influenza virus, X31-sciIV. A one-dose immunization with X31-sciIV is capable of inducing functional influenza virus-specific CD8 T cells that can be recruited into respiratory tissues and provide protection against lethal heterologous challenge. Without these cells, protection against lethal challenge was essentially lost. Our data indicate that an influenza vaccine that primarily relies on CD8 T cells for protection could be developed.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Vírus da Influenza A Subtipo H1N1/imunologia , Vírus da Influenza A Subtipo H3N2/imunologia , Animais , Cães , Relação Dose-Resposta Imunológica , Ensaio de Imunoadsorção Enzimática , Feminino , Citometria de Fluxo , Células HEK293 , Glicoproteínas de Hemaglutininação de Vírus da Influenza/análise , Humanos , Depleção Linfocítica , Camundongos , Camundongos Endogâmicos C57BL
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA