Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Nature ; 573(7773): 287-290, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31485076

RESUMEN

Influenza A viruses are responsible for seasonal epidemics, and pandemics can arise from the transmission of novel zoonotic influenza A viruses to humans1,2. Influenza A viruses contain a segmented negative-sense RNA genome, which is transcribed and replicated by the viral-RNA-dependent RNA polymerase (FluPolA) composed of PB1, PB2 and PA subunits3-5. Although the high-resolution crystal structure of FluPolA of bat influenza A virus has previously been reported6, there are no complete structures available for human and avian FluPolA. Furthermore, the molecular mechanisms of genomic viral RNA (vRNA) replication-which proceeds through a complementary RNA (cRNA) replicative intermediate, and requires oligomerization of the polymerase7-10-remain largely unknown. Here, using crystallography and cryo-electron microscopy, we determine the structures of FluPolA from human influenza A/NT/60/1968 (H3N2) and avian influenza A/duck/Fujian/01/2002 (H5N1) viruses at a resolution of 3.0-4.3 Å, in the presence or absence of a cRNA or vRNA template. In solution, FluPolA forms dimers of heterotrimers through the C-terminal domain of the PA subunit, the thumb subdomain of PB1 and the N1 subdomain of PB2. The cryo-electron microscopy structure of monomeric FluPolA bound to the cRNA template reveals a binding site for the 3' cRNA at the dimer interface. We use a combination of cell-based and in vitro assays to show that the interface of the FluPolA dimer is required for vRNA synthesis during replication of the viral genome. We also show that a nanobody (a single-domain antibody) that interferes with FluPolA dimerization inhibits the synthesis of vRNA and, consequently, inhibits virus replication in infected cells. Our study provides high-resolution structures of medically relevant FluPolA, as well as insights into the replication mechanisms of the viral RNA genome. In addition, our work identifies sites in FluPolA that could be targeted in the development of antiviral drugs.


Asunto(s)
Genoma Viral/genética , Subtipo H3N2 del Virus de la Influenza A/enzimología , Subtipo H5N1 del Virus de la Influenza A/enzimología , Modelos Moleculares , ARN Polimerasa Dependiente del ARN/química , Microscopía por Crioelectrón , Cristalización , Estructura Terciaria de Proteína , Anticuerpos de Dominio Único/metabolismo , Replicación Viral
3.
J Virol ; 92(16)2018 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-29875249

RESUMEN

The heterotrimeric influenza A virus RNA-dependent RNA polymerase complex, composed of PB1, PB2, and PA subunits, is responsible for transcribing and replicating the viral RNA genome. The N-terminal endonuclease domain of the PA subunit performs endonucleolytic cleavage of capped host RNAs to generate capped RNA primers for viral transcription. A surface-exposed flexible loop (PA51-72-loop) in the PA endonuclease domain has been shown to be dispensable for endonuclease activity. Interestingly, the PA51-72-loop was found to form different intramolecular interactions depending on the conformational arrangement of the polymerase. In this study, we show that a PA subunit lacking the PA51-72-loop assembles into a heterotrimeric polymerase with PB1 and PB2. We demonstrate that in a cellular context, the PA51-72-loop is required for RNA replication but not transcription by the viral polymerase. In agreement, recombinant viral polymerase lacking the PA51-72-loop is able to carry out cap-dependent transcription but is inhibited in de novo replication initiation in vitro Furthermore, viral RNA (vRNA) synthesis is also restricted during ApG-primed extension, indicating that the PA51-72-loop is required not only for replication initiation but also for elongation on a cRNA template. We propose that the PA51-72-loop plays a role in the stabilization of the replicase conformation of the polymerase. Together, these results further our understanding of influenza virus RNA genome replication in general and highlight a role of the PA endonuclease domain in polymerase function in particular.IMPORTANCE Influenza A viruses are a major global health threat, not only causing significant morbidity and mortality every year but also having the potential to cause severe pandemic outbreaks like the 1918 influenza pandemic. The viral polymerase is a protein complex which is responsible for transcription and replication of the viral genome and therefore is an attractive target for antiviral drug development. For that purpose it is important to understand the mechanisms of how the virus replicates its genome and how the viral polymerase works on a molecular level. In this report, we characterize the role of the flexible surface-exposed PA51-72-loop in polymerase function and offer new insights into the replication mechanism of influenza A viruses.


Asunto(s)
Virus de la Influenza A/enzimología , Virus de la Influenza A/fisiología , Proteínas Mutantes/metabolismo , ARN Polimerasa Dependiente del ARN/metabolismo , Proteínas Virales/metabolismo , Replicación Viral , Proteínas Mutantes/genética , Unión Proteica , Multimerización de Proteína , ARN Viral/biosíntesis , ARN Polimerasa Dependiente del ARN/genética , Eliminación de Secuencia , Proteínas Virales/genética
4.
Mol Cell ; 70(6): 1101-1110.e4, 2018 06 21.
Artículo en Inglés | MEDLINE | ID: mdl-29910112

RESUMEN

Influenza virus RNA polymerase (FluPol), a heterotrimer composed of PB1, PB2, and PA subunits (P3 in influenza C), performs both transcription and replication of the viral RNA genome. For transcription, FluPol interacts with the C-terminal domain (CTD) of RNA polymerase II (Pol II), which enables FluPol to snatch capped RNA primers from nascent host RNAs. Here, we describe the co-crystal structure of influenza C virus polymerase (FluPolC) bound to a Ser5-phosphorylated CTD (pS5-CTD) peptide. The position of the CTD-binding site at the interface of PB1, P3, and the flexible PB2 C-terminal domains suggests that CTD binding stabilizes the transcription-competent conformation of FluPol. In agreement, both cap snatching and capped primer-dependent transcription initiation by FluPolC are enhanced in the presence of pS5-CTD. Mutations of amino acids in the CTD-binding site reduce viral mRNA synthesis. We propose a model for the activation of the influenza virus transcriptase through its association with pS5-CTD of Pol II.


Asunto(s)
ARN Polimerasas Dirigidas por ADN/genética , Gammainfluenzavirus/genética , Gammainfluenzavirus/ultraestructura , ARN Polimerasas Dirigidas por ADN/fisiología , Humanos , Unión Proteica , Dominios Proteicos/fisiología , Caperuzas de ARN/genética , ARN Polimerasa II/metabolismo , ARN Polimerasa II/fisiología , ARN Viral/metabolismo , ARN Polimerasa Dependiente del ARN/genética , Transcripción Genética , Proteínas Virales/genética , Replicación Viral
5.
J Virol ; 90(13): 6014-6021, 2016 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-27099314

RESUMEN

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.


Asunto(s)
Virus de la Influenza A/enzimología , ARN Polimerasa II/genética , ARN Polimerasa II/metabolismo , ARN Polimerasa Dependiente del ARN/metabolismo , Ribonucleoproteínas/genética , Ribonucleoproteínas/metabolismo , Humanos , Virus de la Influenza A/genética , Virus de la Influenza A/metabolismo , Modelos Moleculares , Imitación Molecular , Péptidos/metabolismo , Fosforilación , Unión Proteica , Dominios Proteicos , ARN Polimerasa II/química , ARN Viral/genética , ARN Viral/metabolismo , Ribonucleoproteínas/química , Serina , Replicación Viral
6.
Mol Cell ; 61(1): 125-37, 2016 Jan 07.
Artículo en Inglés | MEDLINE | ID: mdl-26711008

RESUMEN

Influenza virus polymerase transcribes or replicates the segmented RNA genome (vRNA) into respectively viral mRNA or full-length copies and initiates RNA synthesis by binding the conserved 3' and 5' vRNA ends (the promoter). In recent structures of promoter-bound polymerase, the cap-binding and endonuclease domains are configured for cap snatching, which generates capped transcription primers. Here, we present a FluB polymerase structure with a bound complementary cRNA 5' end that exhibits a major rearrangement of the subdomains within the C-terminal two-thirds of PB2 (PB2-C). Notably, the PB2 nuclear localization signal (NLS)-containing domain translocates ∼90 Što bind to the endonuclease domain. FluA PB2-C alone and RNA-free FluC polymerase are similarly arranged. Biophysical and cap-dependent endonuclease assays show that in solution the polymerase explores different conformational distributions depending on which RNA is bound. The inherent flexibility of the polymerase allows it to adopt alternative conformations that are likely important during polymerase maturation into active progeny RNPs.


Asunto(s)
Gammainfluenzavirus/enzimología , Subtipo H5N1 del Virus de la Influenza A/enzimología , Virus de la Influenza B/enzimología , ARN Polimerasa Dependiente del ARN/metabolismo , Proteínas Virales/metabolismo , Secuencia de Aminoácidos , Cristalografía por Rayos X , Humanos , Subtipo H5N1 del Virus de la Influenza A/genética , Virus de la Influenza B/genética , Gammainfluenzavirus/genética , Rayos Láser , Espectrometría de Masas , Modelos Moleculares , Datos de Secuencia Molecular , Señales de Localización Nuclear/metabolismo , Dominios y Motivos de Interacción de Proteínas , ARN Viral/metabolismo , ARN Polimerasa Dependiente del ARN/química , ARN Polimerasa Dependiente del ARN/genética , Ribonucleoproteínas/metabolismo , Dispersión del Ángulo Pequeño , Relación Estructura-Actividad , Proteínas Virales/química , Proteínas Virales/genética
7.
Nature ; 527(7576): 114-7, 2015 Nov 05.
Artículo en Inglés | MEDLINE | ID: mdl-26503046

RESUMEN

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.


Asunto(s)
Gammainfluenzavirus/enzimología , ARN Polimerasa Dependiente del ARN/química , Apoenzimas/química , Apoenzimas/metabolismo , Sitios de Unión , Cristalografía por Rayos X , Endonucleasas/química , Endonucleasas/metabolismo , Activación Enzimática , Modelos Moleculares , Iniciación de la Cadena Peptídica Traduccional , Regiones Promotoras Genéticas/genética , Unión Proteica , Estructura Terciaria de Proteína , Subunidades de Proteína/química , Subunidades de Proteína/metabolismo , Caperuzas de ARN/metabolismo , ARN Viral/biosíntesis , ARN Viral/metabolismo , ARN Polimerasa Dependiente del ARN/metabolismo , Ribonucleoproteínas/química
8.
Proc Natl Acad Sci U S A ; 111(32): E3335-42, 2014 Aug 12.
Artículo en Inglés | MEDLINE | ID: mdl-25071209

RESUMEN

The influenza virus is a major human and animal pathogen responsible for seasonal epidemics and occasional pandemics. The genome of the influenza A virus comprises eight segments of single-stranded, negative-sense RNA with highly conserved 5' and 3' termini. These termini interact to form a double-stranded promoter structure that is recognized and bound by the viral RNA-dependent RNA polymerase (RNAP); however, no 3D structural information for the influenza polymerase-bound promoter exists. Functional studies have led to the proposal of several 2D models for the secondary structure of the bound promoter, including a corkscrew model in which the 5' and 3' termini form short hairpins. We have taken advantage of an insect-cell system to prepare large amounts of active recombinant influenza virus RNAP, and used this to develop a highly sensitive single-molecule FRET assay to measure distances between fluorescent dyes located on the promoter and map its structure both with and without the polymerase bound. These advances enabled the direct analysis of the influenza promoter structure in complex with the viral RNAP, and provided 3D structural information that is in agreement with the corkscrew model for the influenza virus promoter RNA. Our data provide insights into the mechanisms of promoter binding by the influenza RNAP and have implications for the understanding of the regulatory mechanisms involved in the transcription of viral genes and replication of the viral RNA genome. In addition, the simplicity of this system should translate readily to the study of any virus polymerase-promoter interaction.


Asunto(s)
Subtipo H3N2 del Virus de la Influenza A/química , Subtipo H3N2 del Virus de la Influenza A/genética , Regiones Promotoras Genéticas , ARN Viral/química , ARN Viral/genética , ARN Polimerasa Dependiente del ARN/metabolismo , Proteínas Virales/metabolismo , Animales , Sitios de Unión , Transferencia Resonante de Energía de Fluorescencia , Humanos , Modelos Moleculares , Conformación de Ácido Nucleico , ARN Polimerasa Dependiente del ARN/química , Proteínas Virales/química
9.
Biochem J ; 458(2): 387-94, 2014 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-24313835

RESUMEN

A structure-guided fragment-based approach was used to target the lipophilic allosteric binding site of Mycobacterium tuberculosis EthR. This elongated channel has many hydrophobic residues lining the binding site, with few opportunities for hydrogen bonding. We demonstrate that a fragment-based approach involving the inclusion of flexible fragments in the library leads to an efficient exploration of chemical space, that fragment binding can lead to an extension of the cavity, and that fragments are able to identify hydrogen-bonding opportunities in this hydrophobic environment that are not exploited in Nature. In the present paper, we report the identification of a 1 µM affinity ligand obtained by structure-guided fragment linking.


Asunto(s)
Sitio Alostérico/fisiología , Sistemas de Liberación de Medicamentos/métodos , Descubrimiento de Drogas/métodos , Fragmentos de Péptidos/fisiología , Proteínas Represoras/antagonistas & inhibidores , Proteínas Represoras/química , Factores de Transcripción/antagonistas & inhibidores , Factores de Transcripción/química , Sitios de Unión/fisiología , Cristalografía por Rayos X , Lípidos , Mycobacterium tuberculosis , Fragmentos de Péptidos/antagonistas & inhibidores , Estructura Secundaria de Proteína , Estructura Terciaria de Proteína , Proteínas Represoras/metabolismo , Relación Estructura-Actividad , Factores de Transcripción/metabolismo
10.
Proc Natl Acad Sci U S A ; 110(45): E4238-45, 2013 Nov 05.
Artículo en Inglés | MEDLINE | ID: mdl-24145413

RESUMEN

Negative-strand RNA viruses represent a significant class of important pathogens that cause substantial morbidity and mortality in human and animal hosts worldwide. A defining feature of these viruses is that their single-stranded RNA genomes are of opposite polarity to messenger RNA and are replicated through a positive-sense intermediate. The replicative intermediate is thought to exist as a complementary ribonucleoprotein (cRNP) complex. However, isolation of such complexes from infected cells has never been accomplished. Here we report the development of an RNA-based affinity-purification strategy for the isolation of cRNPs of influenza A virus from infected cells. This technological advance enabled the structural and functional characterization of this elusive but essential component of the viral RNA replication machine. The cRNP exhibits a filamentous double-helical organization with defined termini, containing the viral RNA-dependent RNA polymerase (RdRp) at one end and a loop structure at the other end. In vitro characterization of cRNP activity yielded mechanistic insights into the workings of this RNA synthesis machine. In particular, we found that cRNPs show activity in vitro only in the presence of added RdRp. Intriguingly, a replication-inactive RdRp mutant was also able to activate cRNP-templated viral RNA synthesis. We propose a model of influenza virus genome replication that relies on the trans-activation of the cRNP-associated RdRp. The described purification strategy should be applicable to other negative-strand RNA viruses and will promote studies into their replication mechanisms.


Asunto(s)
Genoma Viral/genética , Virus de la Influenza A/genética , Modelos Genéticos , ARN Complementario/genética , ARN Viral/biosíntesis , Replicación Viral/genética , Animales , Western Blotting , Bovinos , Células HEK293 , Humanos , Virus de la Influenza A/ultraestructura , Microscopía Electrónica de Transmisión , Oligonucleótidos/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...