Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 49
Filter
Add more filters










Publication year range
1.
Viruses ; 16(3)2024 03 09.
Article in English | MEDLINE | ID: mdl-38543786

ABSTRACT

Influenza A viruses (IAVs) possess a segmented genome consisting of eight viral RNAs (vRNAs) associated with multiple copies of viral nucleoprotein (NP) and a viral polymerase complex. Despite the crucial role of RNA structure in IAV replication, the impact of NP binding on vRNA structure is not well understood. In this study, we employed SHAPE chemical probing to compare the structure of NS and M vRNAs of WSN IAV in various states: before the addition of NP, in complex with NP, and after the removal of NP. Comparison of the RNA structures before the addition of NP and after its removal reveals that NP, while introducing limited changes, remodels local structures in both vRNAs and long-range interactions in the NS vRNA, suggesting a potentially biologically relevant RNA chaperone activity. In contrast, NP significantly alters the structure of vRNAs in vRNA/NP complexes, though incorporating experimental data into RNA secondary structure prediction proved challenging. Finally, our results suggest that NP not only binds single-stranded RNA but also helices with interruptions, such as bulges or small internal loops, with a preference for G-poor and C/U-rich regions.


Subject(s)
Influenza A virus , Nucleocapsid Proteins , Influenza A virus/genetics , Influenza A virus/metabolism , Nucleoproteins/metabolism , RNA, Viral/metabolism , Genomics
2.
Sci Adv ; 9(50): eadj9974, 2023 Dec 15.
Article in English | MEDLINE | ID: mdl-38100595

ABSTRACT

Influenza virus genome encapsidation is essential for the formation of a helical viral ribonucleoprotein (vRNP) complex composed of nucleoproteins (NP), the trimeric polymerase, and the viral genome. Although low-resolution vRNP structures are available, it remains unclear how the viral RNA is encapsidated and how NPs assemble into the helical filament specific of influenza vRNPs. In this study, we established a biological tool, the RNP-like particles assembled from recombinant influenza A virus NP and synthetic RNA, and we present the first subnanometric cryo-electron microscopy structure of the helical NP-RNA complex (8.7 to 5.3 Å). The helical RNP-like structure reveals a parallel double-stranded conformation, allowing the visualization of NP-NP and NP-RNA interactions. The RNA, located at the interface of neighboring NP protomers, interacts with conserved residues previously described as essential for the NP-RNA interaction. The NP undergoes conformational changes to enable RNA binding and helix formation. Together, our findings provide relevant insights for understanding the mechanism for influenza genome encapsidation.


Subject(s)
Influenza, Human , Nucleoproteins , Humans , Nucleoproteins/chemistry , Cryoelectron Microscopy , Ribonucleoproteins/genetics , RNA, Viral/metabolism , Nucleocapsid/metabolism
3.
J Am Chem Soc ; 145(38): 20985-21001, 2023 09 27.
Article in English | MEDLINE | ID: mdl-37707433

ABSTRACT

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.


Subject(s)
Influenza in Birds , Animals , Humans , Nucleotidyltransferases , Amino Acids , Mutation , Probability , Mammals , Nuclear Proteins , RNA-Binding Proteins/genetics
4.
Viruses ; 15(6)2023 06 02.
Article in English | MEDLINE | ID: mdl-37376615

ABSTRACT

In recent years, cryo-electron microscopy (cryo-EM) has emerged as an important standalone technique within structural biology [...].


Subject(s)
Molecular Biology , Cryoelectron Microscopy/methods
5.
Viruses ; 14(11)2022 10 26.
Article in English | MEDLINE | ID: mdl-36366462

ABSTRACT

Determining the structural organisation of viral replication complexes and unravelling the impact of infection on cellular homeostasis represent important challenges in virology. This may prove particularly useful when confronted with viruses that pose a significant threat to human health, that appear unique within their family, or for which knowledge is scarce. Among Mononegavirales, bornaviruses (family Bornaviridae) stand out due to their compact genomes and their nuclear localisation for replication. The recent recognition of the zoonotic potential of several orthobornaviruses has sparked a surge of interest in improving our knowledge on this viral family. In this work, we provide a complete analysis of the structural organisation of Borna disease virus 1 (BoDV-1) phosphoprotein (P), an important cofactor for polymerase activity. Using X-ray diffusion and diffraction experiments, we revealed that BoDV-1 P adopts a long coiled-coil α-helical structure split into two parts by an original ß-strand twist motif, which is highly conserved across the members of whole Orthobornavirus genus and may regulate viral replication. In parallel, we used BioID to determine the proximal interactome of P in living cells. We confirmed previously known interactors and identified novel proteins linked to several biological processes such as DNA repair or mRNA metabolism. Altogether, our study provides important structure/function cues, which may improve our understanding of BoDV-1 pathogenesis.


Subject(s)
Borna disease virus , Bornaviridae , Animals , Humans , Borna disease virus/genetics , Phosphoproteins/genetics , Bornaviridae/genetics , DNA Repair , DNA , RNA, Messenger/genetics
6.
EMBO J ; 41(12): e108306, 2022 06 14.
Article in English | MEDLINE | ID: mdl-35506364

ABSTRACT

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.


Subject(s)
Influenza A Virus, H1N1 Subtype , Influenza, Human , Orthomyxoviridae Infections , Pneumonia , Animals , Antiviral Agents/pharmacology , Humans , Influenza A Virus, H3N2 Subtype/metabolism , Mice , Nucleocapsid Proteins , Nucleoproteins/metabolism , Succinic Acid/metabolism , Succinic Acid/pharmacology , Succinic Acid/therapeutic use , Virus Replication
7.
Viruses ; 13(12)2021 12 09.
Article in English | MEDLINE | ID: mdl-34960735

ABSTRACT

Vesicular stomatitis virus (VSV), the founding member of the mononegavirus order (Mononegavirales), was found to be a negative strand RNA virus in the 1960s, and since then the number of such viruses has continually increased with no end in sight. Sendai virus (SeV) was noted soon afterwards due to an outbreak of newborn pneumonitis in Japan whose putative agent was passed in mice, and nowadays this mouse virus is mainly the bane of animal houses and immunologists. However, SeV was important in the study of this class of viruses because, like flu, it grows to high titers in embryonated chicken eggs, facilitating the biochemical characterization of its infection and that of its nucleocapsid, which is very close to that of measles virus (MeV). This review and opinion piece follow SeV as more is known about how various mononegaviruses express their genetic information and carry out their RNA synthesis, and proposes a unified model based on what all MNV have in common.


Subject(s)
Mononegavirales Infections/virology , Mononegavirales/genetics , RNA, Viral/genetics , Sendai virus/genetics , Animals , Genome, Viral , Humans , Mononegavirales/metabolism , RNA, Viral/metabolism , Respirovirus Infections/virology , Sendai virus/metabolism
8.
PLoS Pathog ; 17(12): e1010106, 2021 12.
Article in English | MEDLINE | ID: mdl-34969061

ABSTRACT

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.


Subject(s)
COVID-19/virology , Influenza, Human/virology , Orthomyxoviridae/physiology , SARS-CoV-2/physiology , Animals , Antiviral Agents , COVID-19/therapy , COVID-19/transmission , Drug Development , Evolution, Molecular , Humans , Influenza, Human/therapy , Influenza, Human/transmission , Orthomyxoviridae/immunology , SARS-CoV-2/immunology , Selection, Genetic , Viral Load , Viral Vaccines
9.
Viruses ; 12(8)2020 07 30.
Article in English | MEDLINE | ID: mdl-32751671

ABSTRACT

Influenza viruses are negative single-stranded RNA viruses with nuclear transcription and replication. They enter the nucleus by using the cellular importin-α/-ß nuclear import machinery. Influenza nucleoproteins from influenza A, B, C and D viruses possess a nuclear localization signal (NLS) localized on an intrinsically disordered extremity (NPTAIL). In this paper, using size exclusion chromatography (SEC), SEC-multi-angle laser light scattering (SEC-MALLS) analysis, surface plasmon resonance (SPR) and fluorescence anisotropy, we provide the first comparative study designed to dissect the interaction between the four NPTAILs and four importins-α identified as partners. All interactions between NPTAILs and importins-α have high association and dissociation rates and present a distinct and specific behaviour. D/NPTAIL interacts strongly with all importins-α while B/NPTAIL shows weak affinity for importins-α. A/NPTAIL and C/NPTAIL present preferential importin-α partners. Mutations in B/NPTAIL and D/NPTAIL show a loss of importin-α binding, confirming key NLS residues. Taken together, our results provide essential highlights of this complex translocation mechanism.


Subject(s)
Host Microbial Interactions , Nucleocapsid Proteins/metabolism , Orthomyxoviridae/metabolism , alpha Karyopherins/metabolism , Active Transport, Cell Nucleus , Cell Nucleus/metabolism , Fluorescence Polarization , Humans , Mutation , Nuclear Localization Signals , Nucleocapsid Proteins/genetics , Orthomyxoviridae/genetics , Protein Binding , Surface Plasmon Resonance
10.
J Mol Biol ; 432(10): 3353-3359, 2020 05 01.
Article in English | MEDLINE | ID: mdl-32222384

ABSTRACT

Here, we describe the crystal structures of two distinct isoforms of ligand-free human karyopherin RanBP5 and investigate its global propensity to interact with influenza A virus polymerase. Our results confirm the general architecture and mechanism of the IMB3 karyopherin-ß subfamily whilst also highlighting differences with the yeast orthologue Kap121p. Moreover, our results provide insight into the structural flexibility of ß-importins in the unbound state. Based on docking of a nuclear localisation sequence, point mutations were designed, which suppress influenza PA-PB1 subcomplex binding to RanBP5 in a binary protein complementation assay.


Subject(s)
Cell Nucleus/metabolism , Influenza A virus/enzymology , Point Mutation , RNA-Dependent RNA Polymerase/metabolism , Viral Proteins/metabolism , beta Karyopherins/chemistry , Binding Sites , Crystallography, X-Ray , Humans , Models, Molecular , Molecular Docking Simulation , Protein Binding , Protein Conformation , Protein Transport , beta Karyopherins/genetics
11.
Proc Natl Acad Sci U S A ; 116(22): 10968-10977, 2019 05 28.
Article in English | MEDLINE | ID: mdl-31076555

ABSTRACT

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.


Subject(s)
Antiviral Agents/pharmacology , Chromosomal Proteins, Non-Histone/metabolism , Cytokines/metabolism , Orthomyxoviridae/drug effects , RNA Splicing Factors/metabolism , A549 Cells , Chromosomal Proteins, Non-Histone/chemistry , Chromosomal Proteins, Non-Histone/genetics , Cytokines/chemistry , Cytokines/genetics , HEK293 Cells , Host-Pathogen Interactions/drug effects , Humans , Molecular Docking Simulation , Orthomyxoviridae/pathogenicity , Protein Binding/drug effects , Protein Stability/drug effects , RNA Splicing , RNA Splicing Factors/chemistry , RNA Splicing Factors/genetics , Spliceosomes/drug effects
12.
Sci Rep ; 9(1): 600, 2019 01 24.
Article in English | MEDLINE | ID: mdl-30679709

ABSTRACT

This paper focuses on the nucleoprotein (NP) of the newly identified member of the Orthomyxoviridae family, Influenza D virus. To date several X-ray structures of NP of Influenza A (A/NP) and B (B/NP) viruses and of infectious salmon anemia (ISA/NP) virus have been solved. Here we purified, characterized and solved the X-ray structure of the tetrameric D/NP at 2.4 Å resolution. The crystal structure of its core is similar to NP of other Influenza viruses. However, unlike A/NP and B/NP which possess a flexible amino-terminal tail containing nuclear localization signals (NLS) for their nuclear import, D/NP possesses a carboxy-terminal tail (D/NPTAIL). We show that D/NPTAIL harbors a bipartite NLS and designed C-terminal truncated mutants to demonstrate the role of D/NPTAIL for nuclear transport.


Subject(s)
Cell Nucleus/metabolism , Nucleoproteins/chemistry , Orthomyxoviridae/metabolism , Thogotovirus/metabolism , Active Transport, Cell Nucleus , Amino Acid Sequence , Crystallography, X-Ray , HEK293 Cells , Humans , Nucleoproteins/genetics , Nucleoproteins/metabolism , Protein Binding , Protein Structure, Quaternary , Recombinant Proteins/biosynthesis , Recombinant Proteins/chemistry , Recombinant Proteins/isolation & purification , Sequence Alignment , alpha Karyopherins/metabolism
13.
Sci Rep ; 7(1): 17164, 2017 12 07.
Article in English | MEDLINE | ID: mdl-29215074

ABSTRACT

Influenza viruses are negative strand RNA viruses that replicate in the nucleus of the cell. The viral nucleoprotein (NP) is the major component of the viral ribonucleoprotein. In this paper we show that the NP of influenza B has a long N-terminal tail of 70 residues with intrinsic flexibility. This tail contains the Nuclear Location Signal (NLS). The nuclear trafficking of the viral components mobilizes cellular import factors at different stages, making these host-pathogen interactions promising targets for new therapeutics. NP is imported into the nucleus by the importin-α/ß pathway, through a direct interaction with importin-α isoforms. Here we provide a combined nuclear magnetic resonance and small-angle X-ray scattering (NMR/SAXS) analysis to describe the dynamics of the interaction between influenza B NP and the human importin-α. The NP of influenza B does not have a single NLS nor a bipartite NLS but our results suggest that the tail harbors several adjacent NLS sequences, located between residues 30 and 71.


Subject(s)
Protein Conformation , RNA-Binding Proteins/chemistry , RNA-Binding Proteins/metabolism , Viral Core Proteins/chemistry , Viral Core Proteins/metabolism , alpha Karyopherins/chemistry , alpha Karyopherins/metabolism , Amino Acid Sequence , Humans , Magnetic Resonance Spectroscopy , Models, Molecular , Nucleocapsid Proteins , Protein Binding , RNA-Binding Proteins/genetics , Scattering, Small Angle , Viral Core Proteins/genetics , alpha Karyopherins/genetics
14.
Viruses ; 8(9)2016 09 13.
Article in English | MEDLINE | ID: mdl-27649229

ABSTRACT

This paper describes a biochemical study for making complexes between the nucleoprotein of influenza viruses A and B (A/NP and B/NP) and small RNAs (polyUC RNAs from 5 to 24 nucleotides (nt)), starting from monomeric proteins. We used negative stain electron microscopy, size exclusion chromatography-multi-angle laser light scattering (SEC-MALLS) analysis, and fluorescence anisotropy measurements to show how the NP-RNA complexes evolve. Both proteins make small oligomers with 24-nt RNAs, trimers for A/NP, and dimers, tetramers, and larger complexes for B/NP. With shorter RNAs, the affinities of NP are all in the same range at 50 mM NaCl, showing that the RNAs bind on the same site. The affinity of B/NP for a 24-nt RNA does not change with salt. However, the affinity of A/NP for a 24-nt RNA is lower at 150 and 300 mM NaCl, suggesting that the RNA binds to another site, either on the same protomer or on a neighbour protomer. For our fluorescence anisotropy experiments, we used 6-fluorescein amidite (FAM)-labelled RNAs. By using a (UC)6-FAM(3') RNA with 150 mM NaCl, we observed an interesting phenomenon that gives macromolecular complexes similar to the ribonucleoprotein particles purified from the viruses.


Subject(s)
Orthomyxoviridae/physiology , RNA-Binding Proteins/metabolism , RNA/metabolism , Viral Core Proteins/metabolism , Binding Sites , Nucleocapsid Proteins , Protein Binding , Virus Assembly
15.
Sci Rep ; 6: 24727, 2016 Apr 20.
Article in English | MEDLINE | ID: mdl-27095520

ABSTRACT

The genome of influenza A virus (IAV) comprises eight RNA segments (vRNA) which are transcribed and replicated by the heterotrimeric IAV RNA-dependent RNA-polymerase (RdRp). RdRp consists of three subunits (PA, PB1 and PB2) and binds both the highly conserved 3'- and 5'-ends of the vRNA segment. The IAV RdRp is an important antiviral target, but its structural mechanism has remained largely elusive to date. By applying a polyprotein strategy, we produced RdRp complexes and define a minimal human IAV RdRp core complex. We show that PA-PB1 forms a stable heterodimeric submodule that can strongly interact with 5'-vRNA. In contrast, 3'-vRNA recognition critically depends on the PB2 N-terminal domain. Moreover, we demonstrate that PA-PB1 forms a stable and stoichiometric complex with host nuclear import factor RanBP5 that can be modelled using SAXS and we show that the PA-PB1-RanPB5 complex is no longer capable of 5'-vRNA binding. Our results provide further evidence for a step-wise assembly of IAV structural components, regulated by nuclear transport mechanisms and host factor binding.


Subject(s)
Influenza A virus/genetics , Influenza A virus/metabolism , Influenza, Human/metabolism , Influenza, Human/virology , Protein Subunits/metabolism , RNA-Dependent RNA Polymerase/genetics , RNA-Dependent RNA Polymerase/metabolism , beta Karyopherins/metabolism , Gene Expression Regulation, Viral , Humans , Promoter Regions, Genetic , Protein Binding , Protein Multimerization , RNA, Viral/genetics , RNA-Dependent RNA Polymerase/chemistry , Viral Proteins/genetics , Viral Proteins/metabolism
16.
Methods Mol Biol ; 1320: 37-57, 2016.
Article in English | MEDLINE | ID: mdl-26227036

ABSTRACT

tRNAs occupy a central role in the cellular life, and they are involved in a broad range of biological processes that relies on their interaction with proteins and RNA. Crystallization and structure resolution of tRNA or/and tRNA/partner complexes can yield in valuable information on structural organizations of key elements of cellular machinery. However, crystallization of RNA, is often challenging. Here we review two methods to produce and purify tRNA in quantity and quality to perform X-ray studies.


Subject(s)
Crystallography, X-Ray/methods , RNA, Transfer/chemistry , Base Sequence , Chromatography , Crystallization , DNA-Directed RNA Polymerases/chemistry , Escherichia coli/enzymology , Hydrogen-Ion Concentration , Magnetic Resonance Spectroscopy , Molecular Sequence Data , Nucleic Acid Conformation , Plasmids , Reproducibility of Results , Ribonucleases/chemistry , Transcription, Genetic , Viral Proteins/chemistry , X-Rays
17.
Curr Opin Struct Biol ; 32: 139-46, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25996897

ABSTRACT

Polyproteins are chains of covalently conjoined smaller proteins that occur in nature as versatile means to organize the proteome of viruses including HIV. During maturation, viral polyproteins are typically cleaved into the constituent proteins with different biological functions by highly specific proteases, and structural analyses at defined stages of this maturation process can provide clues for antiviral intervention strategies. Recombinant polyproteins that use similar mechanisms are emerging as powerful tools for producing hitherto inaccessible protein targets such as the influenza polymerase, for high-resolution structure determination by X-ray crystallography. Conversely, covalent linking of individual protein subunits into single polypeptide chains are exploited to overcome sample preparation bottlenecks. Moreover, synthetic polyproteins provide a promising tool to dissect dynamic folding of polypeptide chains into three-dimensional architectures in single-molecule structure analysis by atomic force microscopy (AFM). The recent use of natural and synthetic polyproteins in structural biology and major achievements are highlighted in this contribution.


Subject(s)
Polyproteins/chemistry , Amino Acid Sequence , Animals , Crystallography, X-Ray/methods , Humans , Microscopy, Atomic Force/methods , Models, Molecular , Molecular Sequence Data , Protein Conformation , Protein Folding , Recombinant Proteins/chemistry , Viral Proteins/chemistry , Viruses/chemistry
18.
Expert Opin Drug Discov ; 10(4): 345-71, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25792362

ABSTRACT

INTRODUCTION: Influenza viruses are a threat to human health. There are presently only two methods for treating influenza: vaccines, which require yearly updates, and two classes of antivirals that suffer with the problem of resistance by current human influenza viruses; this is especially the case with amantadine and rimantadine. Consequently, there is an urgent need for the development of new antivirals with new mechanisms of action. AREAS COVERED: In this review, the authors focus on viral protein domains, their associated activity and their inhibition by small molecules defined by a structure-based design with a special emphasis on the ribonucleoprotein complex and its inhibitors. Several new classes of antiviral candidates targeting viral replication through individual domains of the polymerase and the nucleoprotein (NP) have been developed through structure-based design. EXPERT OPINION: To date, the antivirals targeting neuraminidase are by far the most developed and potent. Antiviral candidates targeting the NP and polymerase domains are in the pipeline but their pharmacokinetics needs further studies. The recently published structures of the polymerase expand the possibilities for development of new antivirals. Combination therapies targeting conserved viral targets and new cellular proteins or exploiting drug promiscuity hold promises to fight against the emergence of resistance.


Subject(s)
Antiviral Agents/therapeutic use , Drug Design , Influenza, Human/drug therapy , Animals , Antiviral Agents/pharmacokinetics , Antiviral Agents/pharmacology , Drug Resistance, Viral , Humans , Influenza, Human/virology , Ribonucleoproteins/antagonists & inhibitors , Ribonucleoproteins/metabolism , Structure-Activity Relationship , Virus Replication/drug effects
19.
Nature ; 516(7531): 361-6, 2014 Dec 18.
Article in English | MEDLINE | ID: mdl-25409151

ABSTRACT

Influenza virus polymerase uses a capped primer, derived by 'cap-snatching' from host pre-messenger RNA, to transcribe its RNA genome into mRNA and a stuttering mechanism to generate the poly(A) tail. By contrast, genome replication is unprimed and generates exact full-length copies of the template. Here we use crystal structures of bat influenza A and human influenza B polymerases (FluA and FluB), bound to the viral RNA promoter, to give mechanistic insight into these distinct processes. In the FluA structure, a loop analogous to the priming loop of flavivirus polymerases suggests that influenza could initiate unprimed template replication by a similar mechanism. Comparing the FluA and FluB structures suggests that cap-snatching involves in situ rotation of the PB2 cap-binding domain to direct the capped primer first towards the endonuclease and then into the polymerase active site. The polymerase probably undergoes considerable conformational changes to convert the observed pre-initiation state into the active initiation and elongation states.


Subject(s)
DNA-Directed RNA Polymerases/metabolism , Influenza A virus/enzymology , Influenza B virus/enzymology , Models, Molecular , RNA Caps , RNA, Viral/biosynthesis , RNA, Viral/chemistry , Catalytic Domain , Crystallization , DNA-Directed RNA Polymerases/chemistry , Gene Expression Regulation, Viral , Influenza A virus/chemistry , Influenza B virus/chemistry , Promoter Regions, Genetic , Protein Binding , Protein Structure, Tertiary , RNA Caps/chemistry , RNA Caps/metabolism , Virus Replication
20.
Nucleic Acids Res ; 42(20): 12939-48, 2014 Nov 10.
Article in English | MEDLINE | ID: mdl-25326326

ABSTRACT

Eukaryotic elongation factor eEF1A transits between the GTP- and GDP-bound conformations during the ribosomal polypeptide chain elongation. eEF1A*GTP establishes a complex with the aminoacyl-tRNA in the A site of the 80S ribosome. Correct codon-anticodon recognition triggers GTP hydrolysis, with subsequent dissociation of eEF1A*GDP from the ribosome. The structures of both the 'GTP'- and 'GDP'-bound conformations of eEF1A are unknown. Thus, the eEF1A-related ribosomal mechanisms were anticipated only by analogy with the bacterial homolog EF-Tu. Here, we report the first crystal structure of the mammalian eEF1A2*GDP complex which indicates major differences in the organization of the nucleotide-binding domain and intramolecular movements of eEF1A compared to EF-Tu. Our results explain the nucleotide exchange mechanism in the mammalian eEF1A and suggest that the first step of eEF1A*GDP dissociation from the 80S ribosome is the rotation of the nucleotide-binding domain observed after GTP hydrolysis.


Subject(s)
Guanosine Diphosphate/chemistry , Guanosine Triphosphate/chemistry , Peptide Elongation Factor 1/chemistry , Animals , Crystallography, X-Ray , Guanosine Diphosphate/metabolism , Guanosine Triphosphate/metabolism , Magnesium/chemistry , Models, Molecular , Peptide Elongation Factor 1/metabolism , Protein Binding , Protein Conformation , Protein Isoforms/chemistry , Protein Isoforms/metabolism , Rabbits
SELECTION OF CITATIONS
SEARCH DETAIL
...