Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Nucleic Acids Res ; 49(22): 13019-13030, 2021 12 16.
Artigo em Inglês | MEDLINE | ID: mdl-34850141

RESUMO

SARS-CoV-2 is a positive-sense RNA virus responsible for the Coronavirus Disease 2019 (COVID-19) pandemic, which continues to cause significant morbidity, mortality and economic strain. SARS-CoV-2 can cause severe respiratory disease and death in humans, highlighting the need for effective antiviral therapies. The RNA synthesis machinery of SARS-CoV-2 is an ideal drug target and consists of non-structural protein 12 (nsp12), which is directly responsible for RNA synthesis, and numerous co-factors involved in RNA proofreading and 5' capping of viral RNAs. The formation of the 5' 7-methylguanosine (m7G) cap structure is known to require a guanylyltransferase (GTase) as well as a 5' triphosphatase and methyltransferases; however, the mechanism of SARS-CoV-2 RNA capping remains poorly understood. Here we find that SARS-CoV-2 nsp12 is involved in viral RNA capping as a GTase, carrying out the addition of a GTP nucleotide to the 5' end of viral RNA via a 5' to 5' triphosphate linkage. We further show that the nsp12 NiRAN (nidovirus RdRp-associated nucleotidyltransferase) domain performs this reaction, and can be inhibited by remdesivir triphosphate, the active form of the antiviral drug remdesivir. These findings improve understanding of coronavirus RNA synthesis and highlight a new target for novel or repurposed antiviral drugs against SARS-CoV-2.


Assuntos
Trifosfato de Adenosina/análogos & derivados , Antivirais/farmacologia , RNA-Polimerase RNA-Dependente de Coronavírus/metabolismo , Nucleotidiltransferases/antagonistas & inibidores , RNA Viral/biossíntese , SARS-CoV-2/enzimologia , Trifosfato de Adenosina/farmacologia , RNA-Polimerase RNA-Dependente de Coronavírus/antagonistas & inibidores , Genoma Viral/genética , Guanosina/análogos & derivados , Guanosina/metabolismo , Humanos , Nucleotidiltransferases/metabolismo , Capuzes de RNA/genética , SARS-CoV-2/genética , Vaccinia virus/enzimologia , Vaccinia virus/metabolismo , Tratamento Farmacológico da COVID-19
2.
Nat Microbiol ; 3(11): 1234-1242, 2018 11.
Artigo em Inglês | MEDLINE | ID: mdl-30224800

RESUMO

The molecular processes that determine the outcome of influenza virus infection in humans are multifactorial and involve a complex interplay between host, viral and bacterial factors1. However, it is generally accepted that a strong innate immune dysregulation known as 'cytokine storm' contributes to the pathology of infections with the 1918 H1N1 pandemic or the highly pathogenic avian influenza viruses of the H5N1 subtype2-4. The RNA sensor retinoic acid-inducible gene I (RIG-I) plays an important role in sensing viral infection and initiating a signalling cascade that leads to interferon expression5. Here, we show that short aberrant RNAs (mini viral RNAs (mvRNAs)), produced by the viral RNA polymerase during the replication of the viral RNA genome, bind to and activate RIG-I and lead to the expression of interferon-ß. We find that erroneous polymerase activity, dysregulation of viral RNA replication or the presence of avian-specific amino acids underlie mvRNA generation and cytokine expression in mammalian cells. By deep sequencing RNA samples from the lungs of ferrets infected with influenza viruses, we show that mvRNAs are generated during infection in vivo. We propose that mvRNAs act as the main agonists of RIG-I during influenza virus infection.


Assuntos
Proteína DEAD-box 58/metabolismo , Imunidade Inata/imunologia , Vírus da Influenza A/fisiologia , Infecções por Orthomyxoviridae/imunologia , RNA Viral/metabolismo , Animais , Linhagem Celular , Citocinas/metabolismo , Proteína DEAD-box 58/genética , Feminino , Furões , Vírus da Influenza A/genética , Interferon beta/genética , Interferon beta/metabolismo , Masculino , Mutação , Proteínas Virais/genética , Replicação Viral
3.
PLoS One ; 13(1): e0191226, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29338047

RESUMO

The influenza virus RNA genome is transcribed and replicated in the context of the viral ribonucleoprotein (vRNP) complex by the viral RNA polymerase. The nucleoprotein (NP) is the structural component of the vRNP providing a scaffold for the viral RNA. In the vRNP as well as during transcription and replication the viral polymerase interacts with NP but it is unclear which parts of the polymerase and NP mediate these interactions. Previously the C-terminal '627' domain (amino acids 538-693) of PB2 was shown to interact with NP. Here we report that a fragment encompassing amino acids 146-185 of NP is sufficient to mediate this interaction. Using NMR chemical shift perturbation assays we show that amino acid region 601 to 607 of the PB2 '627' domain interacts with this fragment of NP. Substitutions of these PB2 amino acids resulted in diminished RNP activity and surface plasmon resonance assays showed that amino acids D605 was essential for the interaction with NP and V606 may also play a partial role in the interaction. Collectively these results reveal a possible interaction surface between NP and the PB2 subunit of the RNA polymerase complex.


Assuntos
Virus da Influenza A Subtipo H5N1/química , Virus da Influenza A Subtipo H5N1/genética , Proteínas de Ligação a RNA/química , Proteínas de Ligação a RNA/genética , RNA Polimerase Dependente de RNA/química , RNA Polimerase Dependente de RNA/genética , Proteínas do Core Viral/química , Proteínas do Core Viral/genética , Proteínas Virais/química , Proteínas Virais/genética , Substituição de Aminoácidos , Ácido Aspártico/química , Genoma Viral , Células HEK293 , Humanos , Virus da Influenza A Subtipo H5N1/fisiologia , Influenza Humana/virologia , Modelos Moleculares , Ressonância Magnética Nuclear Biomolecular , Proteínas do Nucleocapsídeo , Domínios e Motivos de Interação entre Proteínas , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Ressonância de Plasmônio de Superfície , Valina/química , Replicação Viral
4.
J Virol ; 90(13): 6014-6021, 2016 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-27099314

RESUMO

UNLABELLED: Influenza viruses subvert the transcriptional machinery of their hosts to synthesize their own viral mRNA. Ongoing transcription by cellular RNA polymerase II (Pol II) is required for viral mRNA synthesis. By a process known as cap snatching, the virus steals short 5' capped RNA fragments from host capped RNAs and uses them to prime viral transcription. An interaction between the influenza A virus RNA polymerase and the C-terminal domain (CTD) of the large subunit of Pol II has been established, but the molecular details of this interaction remain unknown. We show here that the influenza virus ribonucleoprotein (vRNP) complex binds to the CTD of transcriptionally engaged Pol II. Furthermore, we provide evidence that the viral polymerase binds directly to the serine-5-phosphorylated form of the Pol II CTD, both in the presence and in the absence of viral RNA, and show that this interaction is conserved in evolutionarily distant influenza viruses. We propose a model in which direct binding of the viral RNA polymerase in the context of vRNPs to Pol II early in infection facilitates cap snatching, while we suggest that binding of free viral polymerase to Pol II late in infection may trigger Pol II degradation. IMPORTANCE: Influenza viruses cause yearly epidemics and occasional pandemics that pose a threat to human health, as well as represent a large economic burden to health care systems globally. Existing vaccines are not always effective, as they may not exactly match the circulating viruses. Furthermore, there are a limited number of antivirals available, and development of resistance to these is a concern. New measures to combat influenza are needed, but before they can be developed, it is necessary to better understand the molecular interactions between influenza viruses and their host cells. By providing further insights into the molecular details of how influenza viruses hijack the host transcriptional machinery, we aim to uncover novel targets for the development of antivirals.


Assuntos
Vírus da Influenza A/enzimologia , RNA Polimerase II/genética , RNA Polimerase II/metabolismo , RNA Polimerase Dependente de RNA/metabolismo , Ribonucleoproteínas/genética , Ribonucleoproteínas/metabolismo , Humanos , Vírus da Influenza A/genética , Vírus da Influenza A/metabolismo , Modelos Moleculares , Mimetismo Molecular , Peptídeos/metabolismo , Fosforilação , Ligação Proteica , Domínios Proteicos , RNA Polimerase II/química , RNA Viral/genética , RNA Viral/metabolismo , Ribonucleoproteínas/química , Serina , Replicação Viral
5.
Nature ; 527(7576): 114-7, 2015 Nov 05.
Artigo em Inglês | MEDLINE | ID: mdl-26503046

RESUMO

Negative-sense RNA viruses, such as influenza, encode large, multidomain RNA-dependent RNA polymerases that can both transcribe and replicate the viral RNA genome. In influenza virus, the polymerase (FluPol) is composed of three polypeptides: PB1, PB2 and PA/P3. PB1 houses the polymerase active site, whereas PB2 and PA/P3 contain, respectively, cap-binding and endonuclease domains required for transcription initiation by cap-snatching. Replication occurs through de novo initiation and involves a complementary RNA intermediate. Currently available structures of the influenza A and B virus polymerases include promoter RNA (the 5' and 3' termini of viral genome segments), showing FluPol in transcription pre-initiation states. Here we report the structure of apo-FluPol from an influenza C virus, solved by X-ray crystallography to 3.9 Å, revealing a new 'closed' conformation. The apo-FluPol forms a compact particle with PB1 at its centre, capped on one face by PB2 and clamped between the two globular domains of P3. Notably, this structure is radically different from those of promoter-bound FluPols. The endonuclease domain of P3 and the domains within the carboxy-terminal two-thirds of PB2 are completely rearranged. The cap-binding site is occluded by PB2, resulting in a conformation that is incompatible with transcription initiation. Thus, our structure captures FluPol in a closed, transcription pre-activation state. This reveals the conformation of newly made apo-FluPol in an infected cell, but may also apply to FluPol in the context of a non-transcribing ribonucleoprotein complex. Comparison of the apo-FluPol structure with those of promoter-bound FluPols allows us to propose a mechanism for FluPol activation. Our study demonstrates the remarkable flexibility of influenza virus RNA polymerase, and aids our understanding of the mechanisms controlling transcription and genome replication.


Assuntos
Gammainfluenzavirus/enzimologia , RNA Polimerase Dependente de RNA/química , Apoenzimas/química , Apoenzimas/metabolismo , Sítios de Ligação , Cristalografia por Raios X , Endonucleases/química , Endonucleases/metabolismo , Ativação Enzimática , Modelos Moleculares , Iniciação Traducional da Cadeia Peptídica , Regiões Promotoras Genéticas/genética , Ligação Proteica , Estrutura Terciária de Proteína , Subunidades Proteicas/química , Subunidades Proteicas/metabolismo , Capuzes de RNA/metabolismo , RNA Viral/biossíntese , RNA Viral/metabolismo , RNA Polimerase Dependente de RNA/metabolismo , Ribonucleoproteínas/química
6.
J Virol ; 89(2): 1452-5, 2015 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-25355893

RESUMO

In the influenza virus ribonucleoprotein complex, the oligomerization of the nucleoprotein is mediated by an interaction between the tail-loop of one molecule and the groove of the neighboring molecule. In this study, we show that phosphorylation of a serine residue (S165) within the groove of influenza A virus nucleoprotein inhibits oligomerization and, consequently, ribonucleoprotein activity and viral growth. We propose that nucleoprotein oligomerization in infected cells is regulated by reversible phosphorylation.


Assuntos
Vírus da Influenza A Subtipo H3N2/fisiologia , Multimerização Proteica , Proteínas de Ligação a RNA/metabolismo , Proteínas do Core Viral/metabolismo , Replicação Viral , Humanos , Proteínas do Nucleocapsídeo , Fosforilação
7.
Nat Commun ; 5: 4816, 2014 Sep 16.
Artigo em Inglês | MEDLINE | ID: mdl-25226414

RESUMO

Viruses use virions to spread between hosts, and virion composition is therefore the primary determinant of viral transmissibility and immunogenicity. However, the virions of many viruses are complex and pleomorphic, making them difficult to analyse in detail. Here we address this by identifying and quantifying virion proteins with mass spectrometry, producing a complete and quantified model of the hundreds of host-encoded and viral proteins that make up the pleomorphic virions of influenza viruses. We show that a conserved influenza virion architecture is maintained across diverse combinations of virus and host. This 'core' architecture, which includes substantial quantities of host proteins as well as the viral protein NS1, is elaborated with abundant host-dependent features. As a result, influenza virions produced by mammalian and avian hosts have distinct protein compositions. Finally, we note that influenza virions share an underlying protein composition with exosomes, suggesting that influenza virions form by subverting microvesicle production.


Assuntos
Especificidade de Hospedeiro/genética , Vírus da Influenza A Subtipo H1N1/ultraestrutura , Vírus da Influenza A Subtipo H3N2/ultraestrutura , Proteínas não Estruturais Virais/genética , Vírion/ultraestrutura , Sequência de Aminoácidos , Animais , Bovinos , Galinhas , Cães , Células Epiteliais/virologia , Expressão Gênica , Vírus da Influenza A Subtipo H1N1/genética , Vírus da Influenza A Subtipo H1N1/crescimento & desenvolvimento , Vírus da Influenza A Subtipo H3N2/genética , Vírus da Influenza A Subtipo H3N2/crescimento & desenvolvimento , Células Madin Darby de Rim Canino , Modelos Moleculares , Dados de Sequência Molecular , Óvulo/virologia , Alinhamento de Sequência , Carga Viral , Vírion/genética , Vírion/crescimento & desenvolvimento
8.
J Virol ; 88(22): 13284-99, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25187537

RESUMO

UNLABELLED: The negative-sense RNA genome of influenza A virus is transcribed and replicated by the viral RNA-dependent RNA polymerase (RdRP). The viral RdRP is an important host range determinant, indicating that its function is affected by interactions with cellular factors. However, the identities and the roles of most of these factors remain unknown. Here, we employed affinity purification followed by mass spectrometry to identify cellular proteins that interact with the influenza A virus RdRP in infected human cells. We purified RdRPs using a recombinant influenza virus in which the PB2 subunit of the RdRP is fused to a Strep-tag. When this tagged subunit was purified from infected cells, copurifying proteins included the other RdRP subunits (PB1 and PA) and the viral nucleoprotein and neuraminidase, as well as 171 cellular proteins. Label-free quantitative mass spectrometry revealed that the most abundant of these host proteins were chaperones, cytoskeletal proteins, importins, proteins involved in ubiquitination, kinases and phosphatases, and mitochondrial and ribosomal proteins. Among the phosphatases, we identified three subunits of the cellular serine/threonine protein phosphatase 6 (PP6), including the catalytic subunit PPP6C and regulatory subunits PPP6R1 and PPP6R3. PP6 was found to interact directly with the PB1 and PB2 subunits of the viral RdRP, and small interfering RNA (siRNA)-mediated knockdown of the catalytic subunit of PP6 in infected cells resulted in the reduction of viral RNA accumulation and the attenuation of virus growth. These results suggest that PP6 interacts with and positively regulates the activity of the influenza virus RdRP. IMPORTANCE: Influenza A viruses are serious clinical and veterinary pathogens, causing substantial health and economic impacts. In addition to annual seasonal epidemics, occasional global pandemics occur when viral strains adapt to humans from other species. To replicate efficiently and cause disease, influenza viruses must interact with a large number of host factors. The reliance of the viral RNA-dependent RNA polymerase (RdRP) on host factors makes it a major host range determinant. This study describes and quantifies host proteins that interact, directly or indirectly, with a subunit of the RdRP. It increases our understanding of the role of host proteins in viral replication and identifies a large number of potential barriers to pandemic emergence. Identifying host factors allows their importance for viral replication to be tested. Here, we demonstrate a role for the cellular phosphatase PP6 in promoting viral replication, contributing to our emerging knowledge of regulatory phosphorylation in influenza virus biology.


Assuntos
Interações Hospedeiro-Patógeno , Vírus da Influenza A/fisiologia , Fosfoproteínas Fosfatases/metabolismo , RNA Polimerase Dependente de RNA/metabolismo , Transcrição Gênica , Replicação Viral , Linhagem Celular , Células Epiteliais/virologia , Humanos , Espectrometria de Massas , Ligação Proteica , Mapeamento de Interação de Proteínas , RNA Polimerase Dependente de RNA/isolamento & purificação
9.
PLoS Pathog ; 10(3): e1003971, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24603687

RESUMO

Reassortment of influenza viral RNA (vRNA) segments in co-infected cells can lead to the emergence of viruses with pandemic potential. Replication of influenza vRNA occurs in the nucleus of infected cells, while progeny virions bud from the plasma membrane. However, the intracellular mechanics of vRNA assembly into progeny virions is not well understood. Here we used recent advances in microscopy to explore vRNA assembly and transport during a productive infection. We visualized four distinct vRNA segments within a single cell using fluorescent in situ hybridization (FISH) and observed that foci containing more than one vRNA segment were found at the external nuclear periphery, suggesting that vRNA segments are not exported to the cytoplasm individually. Although many cytoplasmic foci contain multiple vRNA segments, not all vRNA species are present in every focus, indicating that assembly of all eight vRNA segments does not occur prior to export from the nucleus. To extend the observations made in fixed cells, we used a virus that encodes GFP fused to the viral polymerase acidic (PA) protein (WSN PA-GFP) to explore the dynamics of vRNA assembly in live cells during a productive infection. Since WSN PA-GFP colocalizes with viral nucleoprotein and influenza vRNA segments, we used it as a surrogate for visualizing vRNA transport in 3D and at high speed by inverted selective-plane illumination microscopy. We observed cytoplasmic PA-GFP foci colocalizing and traveling together en route to the plasma membrane. Our data strongly support a model in which vRNA segments are exported from the nucleus as complexes that assemble en route to the plasma membrane through dynamic colocalization events in the cytoplasm.


Assuntos
Vírus da Influenza A/fisiologia , RNA Viral/metabolismo , Montagem de Vírus/fisiologia , Animais , Western Blotting , Linhagem Celular Tumoral , Citoplasma/metabolismo , Citoplasma/virologia , Imunofluorescência , Humanos , Hibridização in Situ Fluorescente , Microscopia Confocal , Proteínas Virais/metabolismo , Vírion/metabolismo
10.
J Virol ; 87(18): 10381-4, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-23864616

RESUMO

Transcription and replication of the influenza A virus RNA genome are mediated by the viral RNA polymerase from a promoter consisting of the partially base-paired 3' and 5' termini of viral genome segments. Here we show that transcription and replication can be uncoupled by mutation of an unpaired adenosine in the 5' strand of the promoter. This residue is important for transcription but not replication by being essential for the cap-binding activity of the RNA polymerase.


Assuntos
Vírus da Influenza A/fisiologia , Mutação Puntual , Regiões Promotoras Genéticas , RNA Viral/biossíntese , Transcrição Gênica , Replicação Viral , Adenosina/genética , Vírus da Influenza A/genética , Ligação Proteica , RNA Viral/genética , RNA Polimerase Dependente de RNA/metabolismo
11.
PLoS Pathog ; 8(11): e1002998, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23133386

RESUMO

Segment 7 of influenza A virus produces up to four mRNAs. Unspliced transcripts encode M1, spliced mRNA2 encodes the M2 ion channel, while protein products from spliced mRNAs 3 and 4 have not previously been identified. The M2 protein plays important roles in virus entry and assembly, and is a target for antiviral drugs and vaccination. Surprisingly, M2 is not essential for virus replication in a laboratory setting, although its loss attenuates the virus. To better understand how IAV might replicate without M2, we studied the reversion mechanism of an M2-null virus. Serial passage of a virus lacking the mRNA2 splice donor site identified a single nucleotide pseudoreverting mutation, which restored growth in cell culture and virulence in mice by upregulating mRNA4 synthesis rather than by reinstating mRNA2 production. We show that mRNA4 encodes a novel M2-related protein (designated M42) with an antigenically distinct ectodomain that can functionally replace M2 despite showing clear differences in intracellular localisation, being largely retained in the Golgi compartment. We also show that the expression of two distinct ion channel proteins is not unique to laboratory-adapted viruses but, most notably, was also a feature of the 1983 North American outbreak of H5N2 highly pathogenic avian influenza virus. In identifying a 14th influenza A polypeptide, our data reinforce the unexpectedly high coding capacity of the viral genome and have implications for virus evolution, as well as for understanding the role of M2 in the virus life cycle.


Assuntos
Processamento Alternativo , Vírus da Influenza A Subtipo H5N2/metabolismo , RNA Mensageiro/biossíntese , RNA Viral/biossíntese , Proteínas da Matriz Viral/biossíntese , Animais , Aves , Linhagem Celular Tumoral , Surtos de Doenças , Cães , Humanos , Vírus da Influenza A Subtipo H5N2/genética , Influenza Aviária/epidemiologia , Influenza Aviária/genética , Influenza Aviária/metabolismo , Influenza Humana/epidemiologia , Influenza Humana/genética , Influenza Humana/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , América do Norte/epidemiologia , RNA Mensageiro/genética , RNA Viral/genética , Proteínas da Matriz Viral/genética
12.
PLoS One ; 7(5): e36415, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22570712

RESUMO

The ribonucleoprotein (RNP) complex is the essential transcription-replication machinery of the influenza virus. It is composed of the trimeric polymerase (PA, PB1 and PB2), nucleoprotein (NP) and RNA. Elucidating the molecular mechanisms of RNP assembly is central to our understanding of the control of viral transcription and replication and the dependence of these processes on the host cell. In this report, we show, by RNP reconstitution assays and co-immunoprecipitation, that the interaction between NP and polymerase is crucial for the function of the RNP. The functional association of NP and polymerase involves the C-terminal '627' domain of PB2 and it requires NP arginine-150 and either lysine-627 or arginine-630 of PB2. Using surface plasmon resonance, we demonstrate that the interaction between NP and PB2 takes place without the involvement of RNA. At 33, 37 and 41°C in mammalian cells, more positive charges at aa. 627 and 630 of PB2 lead to stronger NP-polymerase interaction, which directly correlates with the higher RNP activity. In conclusion, our study provides new information on the NP-PB2 interaction and shows that the strength of NP-polymerase interaction and the resulting RNP activity are promoted by the positive charges at aa. 627 and 630 of PB2.


Assuntos
Orthomyxoviridae/metabolismo , Proteínas de Ligação a RNA/metabolismo , RNA Polimerase Dependente de RNA/metabolismo , Proteínas do Core Viral/metabolismo , Proteínas Virais/metabolismo , Sequência de Aminoácidos , Linhagem Celular , Ativação Enzimática/genética , Humanos , Cinética , Dados de Sequência Molecular , Complexos Multiproteicos/metabolismo , Mutação , Proteínas do Nucleocapsídeo , Orthomyxoviridae/enzimologia , Orthomyxoviridae/genética , Ligação Proteica , Domínios e Motivos de Interação entre Proteínas , RNA Viral/metabolismo , Proteínas de Ligação a RNA/genética , Ribonucleoproteínas/genética , Ribonucleoproteínas/metabolismo , Alinhamento de Sequência , Temperatura , Proteínas do Core Viral/genética , Proteínas Virais/química , Proteínas Virais/genética
13.
J Virol ; 85(22): 12073-8, 2011 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-21917965

RESUMO

The influenza virus nucleoprotein (NP) is believed to play a central role in directing a switch from RNA genome transcription to replication by the viral RNA polymerase. However, this role has recently been disputed with the proposal of alternative regulatory mechanisms. It has been suggested that the expression of viral polymerase and NP allows genome replication by stabilization of cRNA replication intermediates and complementary ribonucleoprotein (cRNP) assembly. Here, we demonstrate that the RNA-binding activity of NP is necessary for stabilization of cRNA, whereas, surprisingly, homo-oligomerization of NP is not essential. However, both RNA binding and homo-oligomerization activities are essential for genome replication.


Assuntos
Vírus da Influenza A/fisiologia , RNA Viral/metabolismo , Proteínas de Ligação a RNA/metabolismo , Proteínas do Core Viral/metabolismo , Replicação Viral , Linhagem Celular , Humanos , Influenza Humana , Proteínas do Nucleocapsídeo
14.
J Virol ; 85(10): 5228-31, 2011 May.
Artigo em Inglês | MEDLINE | ID: mdl-21411538

RESUMO

The influenza A virus genome consists of eight RNA segments that associate with the viral polymerase proteins (PB1, PB2, and PA) and nucleoprotein (NP) to form ribonucleoprotein complexes (RNPs). The viral NS1 protein was previously shown to associate with these complexes, although it was not clear which RNP component mediated the interaction. Using individual TAP (tandem affinity purification)-tagged PB1, PB2, PA, and NP, we demonstrated that the NS1 protein interacts specifically with NP and not the polymerase subunits. The region of NS1 that binds NP was mapped to the RNA-binding domain.


Assuntos
Mapeamento de Interação de Proteínas , Proteínas de Ligação a RNA/metabolismo , Proteínas do Core Viral/metabolismo , Proteínas não Estruturais Virais/metabolismo , Sequência de Aminoácidos , Sítios de Ligação , Linhagem Celular , Humanos , Dados de Sequência Molecular , Proteínas do Nucleocapsídeo , Proteínas não Estruturais Virais/genética
15.
J Virol ; 84(17): 8691-9, 2010 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-20573828

RESUMO

The RNA polymerase of influenza A virus is a host range determinant and virulence factor. In particular, the PB2 subunit of the RNA polymerase has been implicated as a crucial factor that affects cell tropism as well as virulence in animal models. These findings suggest that host factors associating with the PB2 protein may play an important role during viral replication. In order to identify host factors that associate with the PB2 protein, we purified recombinant PB2 from transiently transfected mammalian cells and identified copurifying host proteins by mass spectrometry. We found that the PB2 protein associates with the cytosolic chaperonin containing TCP-1 (CCT), stress-induced phosphoprotein 1 (STIP1), FK506 binding protein 5 (FKBP5), alpha- and beta-tubulin, Hsp60, and mitochondrial protein p32. Some of these binding partners associate with each other, suggesting that PB2 might interact with these proteins in multimeric complexes. More detailed analysis of the interaction of the PB2 protein with CCT revealed that PB2 associates with CCT as a monomer and that the CCT binding site is located in a central region of the PB2 protein. PB2 proteins from various influenza virus subtypes and origins can associate with CCT. Silencing of CCT resulted in reduced viral replication and reduced PB2 protein and viral RNA accumulation in a ribonucleoprotein reconstitution assay, suggesting an important function for CCT during the influenza virus life cycle. We propose that CCT might be acting as a chaperone for PB2 to aid its folding and possibly its incorporation into the trimeric RNA polymerase complex.


Assuntos
Chaperonina com TCP-1/metabolismo , Vírus da Influenza A/enzimologia , Influenza Humana/metabolismo , Proteínas Virais/metabolismo , Sítios de Ligação , Linhagem Celular , Chaperonina com TCP-1/química , Chaperonina com TCP-1/genética , Humanos , Vírus da Influenza A/genética , Vírus da Influenza A/fisiologia , Influenza Humana/genética , Ligação Proteica , Proteínas Virais/química , Proteínas Virais/genética
16.
J Virol ; 84(14): 7337-45, 2010 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-20463064

RESUMO

Homo-oligomerization of the nucleoprotein (NP) of influenza A virus is crucial for providing a major structural framework for the assembly of viral ribonucleoprotein (RNP) particles. The nucleoprotein is also essential for transcription and replication during the virus life cycle. In the H5N1 NP structure, the tail loop region is important for NP to form oligomers. Here, by an RNP reconstitution assay, we identified eight NP mutants that had different degrees of defects in forming functional RNPs, with the RNP activities of four mutants being totally abolished (E339A, V408S P410S, R416A, and L418S P419S mutants) and the RNP activities of the other four mutants being more than 50% decreased (R267A, I406S, R422A, and E449A mutants). Further characterization by static light scattering showed that the totally defective protein variants existed as monomers in vitro, deviating from the trimeric/oligomeric form of wild-type NP. The I406S, R422A, and E449A variants existed as a mixture of unstable oligomers, thus resulting in a reduction of RNP activity. Although the R267A variant existed as a monomer in vitro, it resumed an oligomeric form upon the addition of RNA and retained a certain degree of RNP activity. Our data suggest that there are three factors that govern the NP oligomerization event: (i) interaction between the tail loop and the insertion groove, (ii) maintenance of the tail loop conformation, and (iii) stabilization of the NP homo-oligomer. The work presented here provides information for the design of NP inhibitors for combating influenza virus infection.


Assuntos
Aminoácidos/metabolismo , Virus da Influenza A Subtipo H5N1/metabolismo , Proteínas de Ligação a RNA/química , Proteínas de Ligação a RNA/metabolismo , Proteínas do Core Viral/química , Proteínas do Core Viral/metabolismo , Aminoácidos/genética , Linhagem Celular , Humanos , Virus da Influenza A Subtipo H5N1/genética , Modelos Moleculares , Mutação , Proteínas do Nucleocapsídeo , Conformação Proteica , Multimerização Proteica , Proteínas de Ligação a RNA/genética , Proteínas do Core Viral/genética
17.
Cell ; 140(3): 397-408, 2010 Feb 05.
Artigo em Inglês | MEDLINE | ID: mdl-20144762

RESUMO

RIG-I is a key mediator of antiviral immunity, able to couple detection of infection by RNA viruses to the induction of interferons. Natural RIG-I stimulatory RNAs have variously been proposed to correspond to virus genomes, virus replication intermediates, viral transcripts, or self-RNA cleaved by RNase L. However, the relative contribution of each of these RNA species to RIG-I activation and interferon induction in virus-infected cells is not known. Here, we use three approaches to identify physiological RIG-I agonists in cells infected with influenza A virus or Sendai virus. We show that RIG-I agonists are exclusively generated by the process of virus replication and correspond to full-length virus genomes. Therefore, nongenomic viral transcripts, short replication intermediates, and cleaved self-RNA do not contribute substantially to interferon induction in cells infected with these negative strand RNA viruses. Rather, single-stranded RNA viral genomes bearing 5'-triphosphates constitute the natural RIG-I agonists that trigger cell-intrinsic innate immune responses during infection.


Assuntos
RNA Helicases DEAD-box/imunologia , Proteínas de Membrana/imunologia , Proteínas do Tecido Nervoso/imunologia , Infecções por Vírus de RNA/imunologia , RNA Viral/imunologia , Animais , Linhagem Celular , Proteína DEAD-box 58 , Cães , Humanos , Interferons/imunologia , Camundongos , Vírus de RNA/fisiologia , Receptores de Superfície Celular , Receptores Imunológicos , Replicação Viral
18.
Virology ; 394(1): 154-63, 2009 Nov 10.
Artigo em Inglês | MEDLINE | ID: mdl-19744689

RESUMO

The negative sense RNA genome of influenza A virus is transcribed and replicated in the nuclei of infected cells by the viral RNA polymerase. Only four viral polypeptides are required but multiple cellular components are potentially involved. We used fluorescence recovery after photobleaching (FRAP) to characterise the dynamics of GFP-tagged viral ribonucleoprotein (RNP) components in living cells. The nucleoprotein (NP) displayed very slow mobility that significantly increased on formation of transcriptionally active RNPs. Conversely, single or dimeric polymerase subunits showed fast nuclear dynamics that decreased upon formation of heterotrimers, suggesting increased interaction of the full polymerase complex with a relatively immobile cellular component(s). Treatment with inhibitors of cellular transcription indicated that in part, this reflected an interaction with cellular RNA polymerase II. Analysis of mutated influenza virus polymerase complexes further suggested that this was through an interaction between PB2 and RNA Pol II separate from PB2 cap-binding activity.


Assuntos
Vírus da Influenza A/fisiologia , Proteínas de Ligação a RNA/metabolismo , Proteínas do Core Viral/metabolismo , Replicação Viral , Linhagem Celular , Núcleo Celular/química , Humanos , Proteínas do Nucleocapsídeo , Ligação Proteica , RNA Polimerase II/metabolismo , RNA Polimerase Dependente de RNA/metabolismo , Proteínas Virais/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA