Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 63
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Annu Rev Biochem ; 90: 321-348, 2021 06 20.
Artigo em Inglês | MEDLINE | ID: mdl-33770447

RESUMO

Influenza virus RNA-dependent RNA polymerase (FluPol) transcribes the viral RNA genome in the infected cell nucleus. In the 1970s, researchers showed that viral transcription depends on host RNA polymerase II (RNAP II) activity and subsequently that FluPol snatches capped oligomers from nascent RNAP II transcripts to prime its own transcription. Exactly how this occurs remains elusive. Here, we review recent advances in the mechanistic understanding of FluPol transcription and early events in RNAP II transcription that are relevant to cap-snatching. We describe the known direct interactions between FluPol and the RNAP II C-terminal domain and summarize the transcription-related host factors that have been found to interact with FluPol. We also discuss open questions regarding how FluPol may be targeted to actively transcribing RNAP II and the exact context and timing of cap-snatching, which is presumed to occur after cap completion but before the cap is sequestered by the nuclear cap-binding complex.


Assuntos
Interações Hospedeiro-Patógeno/fisiologia , Orthomyxoviridae/enzimologia , RNA Polimerase Dependente de RNA/metabolismo , Transcrição Gênica , Proteínas Virais/metabolismo , Humanos , Orthomyxoviridae/patogenicidade , Proteínas de Ligação ao Cap de RNA/genética , Proteínas de Ligação ao Cap de RNA/metabolismo , RNA Polimerase II/química , RNA Polimerase II/metabolismo , RNA Polimerase Dependente de RNA/genética , Proteínas Virais/genética
2.
Cell ; 167(3): 643-656.e17, 2016 Oct 20.
Artigo em Inglês | MEDLINE | ID: mdl-27768888

RESUMO

Humans differ in the outcome that follows exposure to life-threatening pathogens, yet the extent of population differences in immune responses and their genetic and evolutionary determinants remain undefined. Here, we characterized, using RNA sequencing, the transcriptional response of primary monocytes from Africans and Europeans to bacterial and viral stimuli-ligands activating Toll-like receptor pathways (TLR1/2, TLR4, and TLR7/8) and influenza virus-and mapped expression quantitative trait loci (eQTLs). We identify numerous cis-eQTLs that contribute to the marked differences in immune responses detected within and between populations and a strong trans-eQTL hotspot at TLR1 that decreases expression of pro-inflammatory genes in Europeans only. We find that immune-responsive regulatory variants are enriched in population-specific signals of natural selection and show that admixture with Neandertals introduced regulatory variants into European genomes, affecting preferentially responses to viral challenges. Together, our study uncovers evolutionarily important determinants of differences in host immune responsiveness between human populations.


Assuntos
Adaptação Fisiológica/genética , Adaptação Fisiológica/imunologia , Imunidade Adaptativa , Homem de Neandertal/genética , Homem de Neandertal/imunologia , Imunidade Adaptativa/genética , Alelos , Animais , Infecções Bacterianas/genética , Infecções Bacterianas/imunologia , Sequência de Bases , Evolução Biológica , População Negra/genética , Regulação da Expressão Gênica , Variação Genética , Humanos , Sistema Imunitário , Locos de Características Quantitativas , RNA/genética , Seleção Genética , Análise de Sequência de RNA , Receptores Toll-Like/genética , Transcrição Gênica , Viroses/genética , Viroses/imunologia , População Branca/genética
3.
EMBO J ; 41(12): e108306, 2022 06 14.
Artigo em Inglês | MEDLINE | ID: mdl-35506364

RESUMO

Influenza virus infection causes considerable morbidity and mortality, but current therapies have limited efficacy. We hypothesized that investigating the metabolic signaling during infection may help to design innovative antiviral approaches. Using bronchoalveolar lavages of infected mice, we here demonstrate that influenza virus induces a major reprogramming of lung metabolism. We focused on mitochondria-derived succinate that accumulated both in the respiratory fluids of virus-challenged mice and of patients with influenza pneumonia. Notably, succinate displays a potent antiviral activity in vitro as it inhibits the multiplication of influenza A/H1N1 and A/H3N2 strains and strongly decreases virus-triggered metabolic perturbations and inflammatory responses. Moreover, mice receiving succinate intranasally showed reduced viral loads in lungs and increased survival compared to control animals. The antiviral mechanism involves a succinate-dependent posttranslational modification, that is, succinylation, of the viral nucleoprotein at the highly conserved K87 residue. Succinylation of viral nucleoprotein altered its electrostatic interactions with viral RNA and further impaired the trafficking of viral ribonucleoprotein complexes. The finding that succinate efficiently disrupts the influenza replication cycle opens up new avenues for improved treatment of influenza pneumonia.


Assuntos
Vírus da Influenza A Subtipo H1N1 , Influenza Humana , Infecções por Orthomyxoviridae , Pneumonia , Animais , Antivirais/farmacologia , Humanos , Vírus da Influenza A Subtipo H3N2/metabolismo , Camundongos , Proteínas do Nucleocapsídeo , Nucleoproteínas/metabolismo , Ácido Succínico/metabolismo , Ácido Succínico/farmacologia , Ácido Succínico/uso terapêutico , Replicação Viral
4.
Nucleic Acids Res ; 52(12): 7188-7210, 2024 Jul 08.
Artigo em Inglês | MEDLINE | ID: mdl-38686810

RESUMO

Genome-wide approaches have significantly advanced our knowledge of the repertoire of RNA-binding proteins (RBPs) that associate with cellular polyadenylated mRNAs within eukaryotic cells. Recent studies focusing on the RBP interactomes of viral mRNAs, notably SARS-Cov-2, have revealed both similarities and differences between the RBP profiles of viral and cellular mRNAs. However, the RBPome of influenza virus mRNAs remains unexplored. Herein, we identify RBPs that associate with the viral mRNA encoding the nucleoprotein (NP) of an influenza A virus. Focusing on TDP-43, we show that it binds several influenza mRNAs beyond the NP-mRNA, and that its depletion results in lower levels of viral mRNAs and proteins within infected cells, and a decreased yield of infectious viral particles. We provide evidence that the viral polymerase recruits TDP-43 onto viral mRNAs through a direct interaction with the disordered C-terminal domain of TDP-43. Notably, other RBPs found to be associated with influenza virus mRNAs also interact with the viral polymerase, which points to a role of the polymerase in orchestrating the assembly of viral messenger ribonucleoproteins.


Assuntos
Proteínas de Ligação a DNA , Vírus da Influenza A , RNA Mensageiro , RNA Viral , Proteínas de Ligação a RNA , Replicação Viral , Humanos , Replicação Viral/genética , RNA Viral/metabolismo , RNA Viral/genética , Proteínas de Ligação a DNA/metabolismo , Proteínas de Ligação a DNA/genética , RNA Mensageiro/metabolismo , RNA Mensageiro/genética , Proteínas de Ligação a RNA/metabolismo , Proteínas de Ligação a RNA/genética , Vírus da Influenza A/genética , Vírus da Influenza A/fisiologia , Vírus da Influenza A/metabolismo , Proteínas do Nucleocapsídeo/metabolismo , Proteínas do Nucleocapsídeo/genética , Células HEK293 , Proteínas do Core Viral/metabolismo , Proteínas do Core Viral/genética , Ligação Proteica , Animais
5.
Proc Natl Acad Sci U S A ; 120(6): e2211098120, 2023 02 07.
Artigo em Inglês | MEDLINE | ID: mdl-36730204

RESUMO

The segmented RNA genome of influenza A viruses (IAVs) enables viral evolution through genetic reassortment after multiple IAVs coinfect the same cell, leading to viruses harboring combinations of eight genomic segments from distinct parental viruses. Existing data indicate that reassortant genotypes are not equiprobable; however, the low throughput of available virology techniques does not allow quantitative analysis. Here, we have developed a high-throughput single-cell droplet microfluidic system allowing encapsulation of IAV-infected cells, each cell being infected by a single progeny virion resulting from a coinfection process. Customized barcoded primers for targeted viral RNA sequencing enabled the analysis of 18,422 viral genotypes resulting from coinfection with two circulating human H1N1pdm09 and H3N2 IAVs. Results were highly reproducible, confirmed that genetic reassortment is far from random, and allowed accurate quantification of reassortants including rare events. In total, 159 out of the 254 possible reassortant genotypes were observed but with widely varied prevalence (from 0.038 to 8.45%). In cells where eight segments were detected, all 112 possible pairwise combinations of segments were observed. The inclusion of data from single cells where less than eight segments were detected allowed analysis of pairwise cosegregation between segments with very high confidence. Direct coupling analysis accurately predicted the fraction of pairwise segments and full genotypes. Overall, our results indicate that a large proportion of reassortant genotypes can emerge upon coinfection and be detected over a wide range of frequencies, highlighting the power of our tool for systematic and exhaustive monitoring of the reassortment potential of IAVs.


Assuntos
Coinfecção , Vírus da Influenza A , Influenza Humana , Humanos , Vírus da Influenza A/genética , Vírus da Influenza A Subtipo H3N2/genética , Infecções por Orthomyxoviridae , Vírus Reordenados/genética , RNA Viral/genética , Análise de Sequência de RNA
7.
PLoS Pathog ; 18(5): e1010328, 2022 05.
Artigo em Inglês | MEDLINE | ID: mdl-35605026

RESUMO

During annual influenza epidemics, influenza B viruses (IBVs) co-circulate with influenza A viruses (IAVs), can become predominant and cause severe morbidity and mortality. Phylogenetic analyses suggest that IAVs (primarily avian viruses) and IBVs (primarily human viruses) have diverged over long time scales. Identifying their common and distinctive features is an effective approach to increase knowledge about the molecular details of influenza infection. The virus-encoded RNA-dependent RNA polymerases (FluPolB and FluPolA) are PB1-PB2-PA heterotrimers that perform transcription and replication of the viral genome in the nucleus of infected cells. Initiation of viral mRNA synthesis requires a direct association of FluPol with the host RNA polymerase II (RNAP II), in particular the repetitive C-terminal domain (CTD) of the major RNAP II subunit, to enable "cap-snatching" whereby 5'-capped oligomers derived from nascent RNAP II transcripts are pirated to prime viral transcription. Here, we present the first high-resolution co-crystal structure of FluPolB bound to a CTD mimicking peptide at a binding site crossing from PA to PB2. By performing structure-based mutagenesis of FluPolB and FluPolA followed by a systematic investigation of FluPol-CTD binding, FluPol activity and viral phenotype, we demonstrate that IBVs and IAVs have evolved distinct binding interfaces to recruit the RNAP II CTD, despite the CTD sequence being highly conserved across host species. We find that the PB2 627 subdomain, a major determinant of FluPol-host cell interactions and IAV host-range, is involved in CTD-binding for IBVs but not for IAVs, and we show that FluPolB and FluPolA bind to the host RNAP II independently of the CTD. Altogether, our results suggest that the CTD-binding modes of IAV and IBV may represent avian- and human-optimized binding modes, respectively, and that their divergent evolution was shaped by the broader interaction network between the FluPol and the host transcriptional machinery.


Assuntos
Vírus da Influenza A , Influenza Humana , Humanos , Vírus da Influenza A/genética , Vírus da Influenza B/metabolismo , Filogenia , RNA Polimerase II/genética , RNA Polimerase II/metabolismo , RNA Polimerase Dependente de RNA/genética , Replicação Viral/genética
8.
J Am Chem Soc ; 145(38): 20985-21001, 2023 09 27.
Artigo em Inglês | MEDLINE | ID: mdl-37707433

RESUMO

Adaptation of avian influenza RNA polymerase (FluPol) to human cells requires mutations on the 627-NLS domains of the PB2 subunit. The E627K adaptive mutation compensates a 33-amino-acid deletion in the acidic intrinsically disordered domain of the host transcription regulator ANP32A, a deletion that restricts FluPol activity in mammalian cells. The function of ANP32A in the replication transcription complex and in particular its role in host restriction remains poorly understood. Here we characterize ternary complexes formed between ANP32A, FluPol, and the viral nucleoprotein, NP, supporting the putative role of ANP32A in shuttling NP to the replicase complex. We demonstrate that while FluPol and NP can simultaneously bind distinct linear motifs on avian ANP32A, the deletion in the shorter human ANP32A blocks this mode of colocalization. NMR reveals that NP and human-adapted FluPol, containing the E627 K mutation, simultaneously bind the identical extended linear motif on human ANP32A in an electrostatically driven, highly dynamic and multivalent ternary complex. This study reveals a probable molecular mechanism underlying host adaptation, whereby E627K, which enhances the basic surface of the 627 domain, is selected to confer the necessary multivalent properties to allow ANP32A to colocalize NP and FluPol in human cells.


Assuntos
Influenza Aviária , Animais , Humanos , Nucleotidiltransferases , Aminoácidos , Mutação , Probabilidade , Mamíferos , Proteínas Nucleares , Proteínas de Ligação a RNA/genética
9.
PLoS Pathog ; 17(12): e1010106, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34969061

RESUMO

The development of safe and effective vaccines in a record time after the emergence of the Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) is a remarkable achievement, partly based on the experience gained from multiple viral outbreaks in the past decades. However, the Coronavirus Disease 2019 (COVID-19) crisis also revealed weaknesses in the global pandemic response and large gaps that remain in our knowledge of the biology of coronaviruses (CoVs) and influenza viruses, the 2 major respiratory viruses with pandemic potential. Here, we review current knowns and unknowns of influenza viruses and CoVs, and we highlight common research challenges they pose in 3 areas: the mechanisms of viral emergence and adaptation to humans, the physiological and molecular determinants of disease severity, and the development of control strategies. We outline multidisciplinary approaches and technological innovations that need to be harnessed in order to improve preparedeness to the next pandemic.


Assuntos
COVID-19/virologia , Influenza Humana/virologia , Orthomyxoviridae/fisiologia , SARS-CoV-2/fisiologia , Animais , Antivirais , COVID-19/terapia , COVID-19/transmissão , Desenvolvimento de Medicamentos , Evolução Molecular , Humanos , Influenza Humana/terapia , Influenza Humana/transmissão , Orthomyxoviridae/imunologia , SARS-CoV-2/imunologia , Seleção Genética , Carga Viral , Vacinas Virais
10.
Nature ; 541(7635): 117-121, 2017 01 05.
Artigo em Inglês | MEDLINE | ID: mdl-28002402

RESUMO

The heterotrimeric influenza polymerase (FluPol), comprising subunits PA, PB1 and PB2, binds to the conserved 5' and 3' termini (the 'promoter') of each of the eight single-stranded viral RNA (vRNA) genome segments and performs both transcription and replication of vRNA in the infected cell nucleus. To transcribe viral mRNAs, FluPol associates with cellular RNA polymerase II (Pol II), which enables it to take 5'-capped primers from nascent Pol II transcripts. Here we present a co-crystal structure of bat influenza A polymerase bound to a Pol II C-terminal domain (CTD) peptide mimic, which shows two distinct phosphoserine-5 (SeP5)-binding sites in the polymerase PA subunit, accommodating four CTD heptad repeats overall. Mutagenesis of the SeP5-contacting basic residues (PA K289, R454, K635 and R638) weakens CTD repeat binding in vitro without affecting the intrinsic cap-primed (transcription) or unprimed (replication) RNA synthesis activity of recombinant polymerase, whereas in cell-based minigenome assays the same mutations substantially reduce overall polymerase activity. Only recombinant viruses with a single mutation in one of the SeP5-binding sites can be rescued, but these viruses are severely attenuated and genetically unstable. Several previously described mutants that modulate virulence can be rationalized by our results, including a second site mutation (PA(C453R)) that enables the highly attenuated mutant virus (PA(R638A)) to revert to near wild-type infectivity. We conclude that direct binding of FluPol to the SeP5 Pol II CTD is fine-tuned to allow efficient viral transcription and propose that the CTD-binding site on FluPol could be targeted for antiviral drug development.


Assuntos
Quirópteros/virologia , Orthomyxoviridae/enzimologia , RNA Polimerase II/química , RNA Polimerase II/metabolismo , RNA Polimerase Dependente de RNA/química , RNA Polimerase Dependente de RNA/metabolismo , Sequência de Aminoácidos , Animais , Antivirais/farmacologia , Sítios de Ligação/efeitos dos fármacos , Sítios de Ligação/genética , Cristalografia por Raios X , Vírus da Influenza A/enzimologia , Vírus da Influenza B/enzimologia , Modelos Moleculares , Terapia de Alvo Molecular , Mutação , Orthomyxoviridae/genética , Orthomyxoviridae/crescimento & desenvolvimento , Orthomyxoviridae/patogenicidade , Infecções por Orthomyxoviridae/tratamento farmacológico , Infecções por Orthomyxoviridae/enzimologia , Infecções por Orthomyxoviridae/virologia , Fragmentos de Peptídeos/química , Fragmentos de Peptídeos/metabolismo , Fosfosserina/metabolismo , Ligação Proteica/efeitos dos fármacos , Domínios Proteicos , Subunidades Proteicas , RNA Polimerase Dependente de RNA/genética , Virulência/genética , Replicação Viral
11.
RNA ; 26(12): 1905-1918, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-32929001

RESUMO

Like most RNA viruses, influenza viruses generate defective viral genomes (DVGs) with large internal deletions during replication. There is accumulating evidence supporting a biological relevance of such DVGs. However, further understanding of the molecular mechanisms that underlie the production and biological activity of DVGs is conditioned upon the sensitivity and accuracy of detection methods, that is, next-generation sequencing (NGS) technologies and related bioinformatics algorithms. Although many algorithms were developed, their sensitivity and reproducibility were mostly assessed on simulated data. Here, we introduce DG-seq, a time-efficient pipeline for DVG detection and quantification, and a set of biological controls to assess the performance of not only our bioinformatics algorithm but also the upstream NGS steps. Using these tools, we provide the first rigorous comparison of the two commonly used sample processing methods for RNA-seq, with or without a PCR preamplification step. Our data show that preamplification confers a limited advantage in terms of sensitivity and introduces size- but also sequence-dependent biases in DVG quantification, thereby providing a strong rationale to favor preamplification-free methods. We further examine the features of DVGs produced by wild-type and transcription-defective (PA-K635A or PA-R638A) influenza viruses, and show an increased diversity and frequency of DVGs produced by the PA mutants compared to the wild-type virus. Finally, we demonstrate a significant enrichment in DVGs showing direct, A/T-rich sequence repeats at the deletion breakpoint sites. Our findings provide novel insights into the mechanisms of influenza virus DVG production.


Assuntos
Vírus Defeituosos/genética , Genoma Viral , Influenza Humana/genética , Orthomyxoviridae/genética , RNA Viral/genética , RNA-Seq/métodos , Humanos , Influenza Humana/virologia , Replicação Viral
12.
Proc Natl Acad Sci U S A ; 116(22): 10968-10977, 2019 05 28.
Artigo em Inglês | MEDLINE | ID: mdl-31076555

RESUMO

New therapeutic strategies targeting influenza are actively sought due to limitations in current drugs available. Host-directed therapy is an emerging concept to target host functions involved in pathogen life cycles and/or pathogenesis, rather than pathogen components themselves. From this perspective, we focused on an essential host partner of influenza viruses, the RED-SMU1 splicing complex. Here, we identified two synthetic molecules targeting an α-helix/groove interface essential for RED-SMU1 complex assembly. We solved the structure of the SMU1 N-terminal domain in complex with RED or bound to one of the molecules identified to disrupt this complex. We show that these compounds inhibiting RED-SMU1 interaction also decrease endogenous RED-SMU1 levels and inhibit viral mRNA splicing and viral multiplication, while preserving cell viability. Overall, our data demonstrate the potential of RED-SMU1 destabilizing molecules as an antiviral therapy that could be active against a wide range of influenza viruses and be less prone to drug resistance.


Assuntos
Antivirais/farmacologia , Proteínas Cromossômicas não Histona/metabolismo , Citocinas/metabolismo , Orthomyxoviridae/efeitos dos fármacos , Fatores de Processamento de RNA/metabolismo , Células A549 , Proteínas Cromossômicas não Histona/química , Proteínas Cromossômicas não Histona/genética , Citocinas/química , Citocinas/genética , Células HEK293 , Interações Hospedeiro-Patógeno/efeitos dos fármacos , Humanos , Simulação de Acoplamento Molecular , Orthomyxoviridae/patogenicidade , Ligação Proteica/efeitos dos fármacos , Estabilidade Proteica/efeitos dos fármacos , Splicing de RNA , Fatores de Processamento de RNA/química , Fatores de Processamento de RNA/genética , Spliceossomos/efeitos dos fármacos
13.
PLoS Pathog ; 15(10): e1008034, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-31581279

RESUMO

The influenza A virus RNA-dependent RNA polymerase complex consists in three subunits, PB2, PB1 and PA, that perform transcription and replication of the viral genome through very distinct mechanisms. Biochemical and structural studies have revealed that the polymerase can adopt multiple conformations and form oligomers. However so far it remained unclear whether the available oligomeric crystal structures represent a functional state of the polymerase. Here we gained new insights into this question, by investigating the incompatibility between non-cognate subunits of influenza polymerase brought together through genetic reassortment. We observed that a 7:1 reassortant virus whose PB2 segment derives from the A/WSN/33 (WSN) virus in an otherwise A/PR/8/34 (PR8) backbone is attenuated, despite a 97% identity between the PR8-PB2 and WSN-PB2 proteins. Independent serial passages led to the selection of phenotypic revertants bearing distinct second-site mutations on PA, PB1 and/or PB2. The constellation of mutations present on one revertant virus was studied extensively using reverse genetics and cell-based reconstitution of the viral polymerase. The PA-E349K mutation appeared to play a major role in correcting the initial defect in replication (cRNA -> vRNA) of the PR8xWSN-PB2 reassortant. Strikingly the PA-E349K mutation, and also the PB2-G74R and PB1-K577G mutations present on other revertants, are located at a dimerization interface of the polymerase. All three restore wild-type-like polymerase activity in a minigenome assay while decreasing the level of polymerase dimerization. Overall, our data show that the polymerase subunits co-evolve to ensure not only optimal inter-subunit interactions within the heterotrimer, but also proper levels of dimerization of the heterotrimer which appears to be essential for efficient viral RNA replication. Our findings point to influenza polymerase dimerization as a feature that is controlled by a complex interplay of genetic determinants, can restrict genetic reassortment, and could become a target for antiviral drug development.


Assuntos
Vírus da Influenza A/enzimologia , Influenza Humana/virologia , Mutação , Multimerização Proteica , RNA Polimerase Dependente de RNA/química , RNA Polimerase Dependente de RNA/genética , Vírus Reordenados/genética , Células A549 , Células HEK293 , Humanos , Influenza Humana/genética , Conformação Proteica , Subunidades Proteicas , RNA Polimerase Dependente de RNA/metabolismo , Proteínas Virais/química , Proteínas Virais/genética , Proteínas Virais/metabolismo , Replicação Viral
14.
Am J Physiol Lung Cell Mol Physiol ; 310(7): L689-99, 2016 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-26747784

RESUMO

Influenza A viruses (IAV) trigger contagious acute respiratory diseases. A better understanding of the molecular mechanisms of IAV pathogenesis and host immune responses is required for the development of more efficient treatments of severe influenza. Calpains are intracellular proteases that participate in diverse cellular responses, including inflammation. Here, we used in vitro and in vivo approaches to investigate the role of calpain signaling in IAV pathogenesis. Calpain expression and activity were found altered in IAV-infected bronchial epithelial cells. With the use of small-interfering RNA (siRNA) gene silencing, specific synthetic inhibitors of calpains, and mice overexpressing calpastatin, we found that calpain inhibition dampens IAV replication and IAV-triggered secretion of proinflammatory mediators and leukocyte infiltration. Remarkably, calpain inhibition has a protective impact in IAV infection, since it significantly reduced mortality of mice challenged not only by seasonal H3N2- but also by hypervirulent H5N1 IAV strains. Hence, our study suggests that calpains are promising therapeutic targets for treating IAV acute pneumonia.


Assuntos
Calpaína/fisiologia , Virus da Influenza A Subtipo H5N1/imunologia , Influenza Humana/enzimologia , Animais , Citocinas/metabolismo , Cães , Células Epiteliais/imunologia , Células Epiteliais/metabolismo , Regulação Enzimológica da Expressão Gênica , Regulação Viral da Expressão Gênica , Humanos , Influenza Humana/imunologia , Influenza Humana/virologia , Células Madin Darby de Rim Canino , Camundongos Endogâmicos C57BL , Receptores de Reconhecimento de Padrão/metabolismo , Transdução de Sinais , Replicação Viral
15.
J Virol ; 89(12): 6376-90, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25855727

RESUMO

UNLABELLED: The influenza virus RNA-dependent RNA polymerase catalyzes genome replication and transcription within the cell nucleus. Efficient nuclear import and assembly of the polymerase subunits PB1, PB2, and PA are critical steps in the virus life cycle. We investigated the structure and function of the PA linker (residues 197 to 256), located between its N-terminal endonuclease domain and its C-terminal structured domain that binds PB1, the polymerase core. Circular dichroism experiments revealed that the PA linker by itself is structurally disordered. A large series of PA linker mutants exhibited a temperature-sensitive (ts) phenotype (reduced viral growth at 39.5°C versus 37°C/33°C), suggesting an alteration of folding kinetic parameters. The ts phenotype was associated with a reduced efficiency of replication/transcription of a pseudoviral reporter RNA in a minireplicon assay. Using a fluorescent-tagged PB1, we observed that ts and lethal PA mutants did not efficiently recruit PB1 to reach the nucleus at 39.5°C. A protein complementation assay using PA mutants, PB1, and ß-importin IPO5 tagged with fragments of the Gaussia princeps luciferase showed that increasing the temperature negatively modulated the PA-PB1 and the PA-PB1-IPO5 interactions or complex stability. The selection of revertant viruses allowed the identification of different types of compensatory mutations located in one or the other of the three polymerase subunits. Two ts mutants were shown to be attenuated and able to induce antibodies in mice. Taken together, our results identify a PA domain critical for PB1-PA nuclear import and that is a "hot spot" to engineer ts mutants that could be used to design novel attenuated vaccines. IMPORTANCE: By targeting a discrete domain of the PA polymerase subunit of influenza virus, we were able to identify a series of 9 amino acid positions that are appropriate to engineer temperature-sensitive (ts) mutants. This is the first time that a large number of ts mutations were engineered in such a short domain, demonstrating that rational design of ts mutants can be achieved. We were able to associate this phenotype with a defect of transport of the PA-PB1 complex into the nucleus. Reversion substitutions restored the ability of the complex to move to the nucleus. Two of these ts mutants were shown to be attenuated and able to produce antibodies in mice. These results are of high interest for the design of novel attenuated vaccines and to develop new antiviral drugs.


Assuntos
Vírus da Influenza A Subtipo H1N1/fisiologia , Vírus da Influenza A Subtipo H1N1/efeitos da radiação , Mutação , RNA Polimerase Dependente de RNA/genética , RNA Polimerase Dependente de RNA/metabolismo , Proteínas Virais/genética , Proteínas Virais/metabolismo , Replicação Viral/efeitos da radiação , Transporte Ativo do Núcleo Celular , Animais , Dicroísmo Circular , Feminino , Teste de Complementação Genética , Humanos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Proteínas Mutantes/genética , Proteínas Mutantes/metabolismo , Ligação Proteica/efeitos da radiação , Domínios e Motivos de Interação entre Proteínas , Estrutura Secundária de Proteína , RNA Polimerase Dependente de RNA/química , Temperatura , Proteínas Virais/química
16.
PLoS Pathog ; 10(6): e1004164, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24945353

RESUMO

Influenza A viruses are major pathogens in humans and in animals, whose genome consists of eight single-stranded RNA segments of negative polarity. Viral mRNAs are synthesized by the viral RNA-dependent RNA polymerase in the nucleus of infected cells, in close association with the cellular transcriptional machinery. Two proteins essential for viral multiplication, the exportin NS2/NEP and the ion channel protein M2, are produced by splicing of the NS1 and M1 mRNAs, respectively. Here we identify two human spliceosomal factors, RED and SMU1, that control the expression of NS2/NEP and are required for efficient viral multiplication. We provide several lines of evidence that in infected cells, the hetero-trimeric viral polymerase recruits a complex formed by RED and SMU1 through interaction with its PB2 and PB1 subunits. We demonstrate that the splicing of the NS1 viral mRNA is specifically affected in cells depleted of RED or SMU1, leading to a decreased production of the spliced mRNA species NS2, and to a reduced NS2/NS1 protein ratio. In agreement with the exportin function of NS2, these defects impair the transport of newly synthesized viral ribonucleoproteins from the nucleus to the cytoplasm, and strongly reduce the production of infectious influenza virions. Overall, our results unravel a new mechanism of viral subversion of the cellular splicing machinery, by establishing that the human splicing factors RED and SMU1 act jointly as key regulators of influenza virus gene expression. In addition, our data point to a central role of the viral RNA polymerase in coupling transcription and alternative splicing of the viral mRNAs.


Assuntos
Processamento Alternativo , Proteínas Cromossômicas não Histona/metabolismo , Citocinas/metabolismo , Vírus da Influenza A/fisiologia , RNA Mensageiro/metabolismo , RNA Viral/metabolismo , RNA Polimerase Dependente de RNA/metabolismo , Proteínas Virais/metabolismo , Transporte Ativo do Núcleo Celular , Linhagem Celular , Proteínas Cromossômicas não Histona/antagonistas & inibidores , Proteínas Cromossômicas não Histona/química , Proteínas Cromossômicas não Histona/genética , Citocinas/antagonistas & inibidores , Citocinas/química , Citocinas/genética , Inativação Gênica , Interações Hospedeiro-Patógeno , Humanos , Vírus da Influenza A/enzimologia , Carioferinas/genética , Carioferinas/metabolismo , Domínios e Motivos de Interação entre Proteínas , Estrutura Quaternária de Proteína , RNA Polimerase Dependente de RNA/genética , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/metabolismo , Spliceossomos/enzimologia , Spliceossomos/metabolismo , Técnicas do Sistema de Duplo-Híbrido , Proteínas Virais/genética , Replicação Viral
17.
Mol Cell Proteomics ; 12(10): 2845-55, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23816991

RESUMO

A precise mapping of pathogen-host interactions is essential for comprehensive understanding of the processes of infection and pathogenesis. The most frequently used techniques for interactomics are the yeast two-hybrid binary methodologies, which do not recapitulate the pathogen life cycle, and the tandem affinity purification mass spectrometry co-complex methodologies, which cannot distinguish direct from indirect interactions. New technologies are thus needed to improve the mapping of pathogen-host interactions. In the current study, we detected binary interactions between influenza A virus polymerase and host proteins during the course of an actual viral infection, using a new strategy based on trans-complementation of the Gluc1 and Gluc2 fragments of Gaussia princeps luciferase. Infectious recombinant influenza viruses that encode a Gluc1-tagged polymerase subunit were engineered to infect cultured cells transiently expressing a selected set of Gluc2-tagged cellular proteins involved in nucleocytoplasmic trafficking pathways. A random set and a literature-curated set of Gluc2-tagged cellular proteins were tested in parallel. Our assay allowed the sensitive and accurate recovery of previously described interactions, and it revealed 30% of positive, novel viral-host protein-protein interactions within the exploratory set. In addition to cellular proteins involved in the nuclear import pathway, components of the nuclear pore complex such as NUP62 and mRNA export factors such as NXF1, RMB15B, and DDX19B were identified for the first time as interactors of the viral polymerase. Gene silencing experiments further showed that NUP62 is required for efficient viral replication. Our findings give new insights regarding the subversion of host nucleocytoplasmic trafficking pathways by influenza A viruses. They also demonstrate the potential of our infectious protein complementation assay for high-throughput exploration of influenza virus interactomics in infected cells. With more infectious reverse genetics systems becoming available, this strategy should be widely applicable to numerous pathogens.


Assuntos
Vírus da Influenza A/metabolismo , Luciferases/genética , Mapeamento de Interação de Proteínas/métodos , Amantadina/farmacologia , Animais , Antivirais/farmacologia , Linhagem Celular , Linhagem Celular Tumoral , Copépodes/enzimologia , Cricetinae , DNA Polimerase Dirigida por DNA/genética , Cães , Interações Hospedeiro-Patógeno , Humanos , Vírus da Influenza A/genética , Vírus da Influenza A/patogenicidade , Luciferases/metabolismo , Células Madin Darby de Rim Canino , Genética Reversa , Ribavirina/farmacologia , Proteínas Virais de Fusão , Proteínas Virais/metabolismo , Replicação Viral
18.
J Gen Virol ; 95(Pt 6): 1233-1243, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24694396

RESUMO

Currently circulating H5N1 influenza viruses have undergone a complex evolution since the appearance of their progenitor A/Goose/Guangdong/1/96 in 1996. After the eradication of the H5N1 viruses that emerged in Hong Kong in 1997 (HK/97 viruses), new genotypes of H5N1 viruses emerged in the same region in 2000 that were more pathogenic for both chickens and mice than HK/97 viruses. These, as well as virtually all highly pathogenic H5N1 viruses since 2000, harbour a deletion of aa 80-84 in the unstructured region of the non-structural (NS) protein NS1 linking its RNA-binding domain to its effector domain. NS segments harbouring this mutation have since been found in non-H5N1 viruses and we asked whether this 5 aa deletion could have a general effect not limited to the NS1 of H5N1 viruses. We genetically engineered this deletion in the NS segment of a duck-origin avian H1N1 virus, and compared the in vivo and in vitro properties of the WT and NSdel8084 viruses. In experimentally infected chickens, the NSdel8084 virus showed both an increased replication potential and an increased pathogenicity. This in vivo phenotype was correlated with a higher replicative efficiency in vitro, both in embryonated eggs and in a chicken lung epithelial cell line. Our data demonstrated that the increased replicative potential conferred by this small deletion was a general feature not restricted to NS1 from H5N1 viruses and suggested that viruses acquiring this mutation may be selected positively in the future.


Assuntos
Vírus da Influenza A Subtipo H1N1/fisiologia , Vírus da Influenza A Subtipo H1N1/patogenicidade , Proteínas não Estruturais Virais/química , Proteínas não Estruturais Virais/fisiologia , Sequência de Aminoácidos , Animais , Sequência de Bases , Linhagem Celular , Embrião de Galinha , Galinhas , Citocinas/genética , DNA Viral/genética , Vírus da Influenza A Subtipo H1N1/genética , Virus da Influenza A Subtipo H5N1/química , Virus da Influenza A Subtipo H5N1/genética , Virus da Influenza A Subtipo H5N1/fisiologia , Influenza Aviária/imunologia , Influenza Aviária/patologia , Influenza Aviária/virologia , Interferon Tipo I/biossíntese , Pulmão/patologia , Pulmão/virologia , Camundongos , Dados de Sequência Molecular , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Deleção de Sequência , Especificidade da Espécie , Carga Viral , Proteínas não Estruturais Virais/genética , Virulência/genética , Virulência/fisiologia , Replicação Viral/genética , Replicação Viral/fisiologia
19.
Nat Commun ; 15(1): 1064, 2024 Feb 05.
Artigo em Inglês | MEDLINE | ID: mdl-38316757

RESUMO

The current model is that the influenza virus polymerase (FluPol) binds either to host RNA polymerase II (RNAP II) or to the acidic nuclear phosphoprotein 32 (ANP32), which drives its conformation and activity towards transcription or replication of the viral genome, respectively. Here, we provide evidence that the FluPol-RNAP II binding interface, beyond its well-acknowledged function in cap-snatching during transcription initiation, has also a pivotal role in replication of the viral genome. Using a combination of cell-based and in vitro approaches, we show that the RNAP II C-terminal-domain, jointly with ANP32, enhances FluPol replication activity. We observe successive conformational changes to switch from a transcriptase to a replicase conformation in the presence of the bound RNPAII C-terminal domain and propose a model in which the host RNAP II is the anchor for transcription and replication of the viral genome. Our data open new perspectives on the spatial coupling of viral transcription and replication and the coordinated balance between these two activities.


Assuntos
Orthomyxoviridae , RNA Polimerase II , RNA Polimerase II/metabolismo , RNA Polimerase Dependente de RNA/metabolismo , RNA Viral/genética , Orthomyxoviridae/genética , RNA Polimerases Dirigidas por DNA , Replicação Viral/genética
20.
PLoS One ; 19(1): e0292977, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38271396

RESUMO

The life cycle of influenza A viruses (IAV), and notably intracellular trafficking of the viral genome, depends on multiple interactions with the cellular cytoskeleton and endomembrane system. A limitation of the conventional cellular models used for mechanistic study and subcellular imaging of IAV infection is that they are cultured in two dimensions (2D) under non-polarizing conditions, and therefore they do not recapitulate the intracellular organization of the polarized respiratory epithelial cells naturally targeted by IAVs. To overcome this limitation, we developed an IAV-infection assay in a 3D cell culture system which allows imaging along the baso-lateral axis of polarized cells, with subcellular resolution. Here we describe a protocol to grow polarized monolayers of Caco2-TC7 cells on static Cytodex-3 microcarrier beads, infect them with IAV, and subsequently perform immunostaining and confocal imaging, or electron microscopy, on polarized IAV-infected cells. This method can be extended to other pathogens that infect human polarized epithelial cells.


Assuntos
Vírus da Influenza A , Influenza Humana , Infecções por Orthomyxoviridae , Humanos , Células CACO-2 , Células Epiteliais/metabolismo , Replicação Viral
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa