Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 34
Filter
Add more filters










Publication year range
1.
Nat Commun ; 15(1): 4189, 2024 May 17.
Article in English | MEDLINE | ID: mdl-38760379

ABSTRACT

The viral polymerase complex, comprising the large protein (L) and phosphoprotein (P), is crucial for both genome replication and transcription in non-segmented negative-strand RNA viruses (nsNSVs), while structures corresponding to these activities remain obscure. Here, we resolved two L-P complex conformations from the mumps virus (MuV), a typical member of nsNSVs, via cryogenic-electron microscopy. One conformation presents all five domains of L forming a continuous RNA tunnel to the methyltransferase domain (MTase), preferably as a transcription state. The other conformation has the appendage averaged out, which is inaccessible to MTase. In both conformations, parallel P tetramers are revealed around MuV L, which, together with structures of other nsNSVs, demonstrates the diverse origins of the L-binding X domain of P. Our study links varying structures of nsNSV polymerase complexes with genome replication and transcription and points to a sliding model for polymerase complexes to advance along the RNA templates.


Subject(s)
Cryoelectron Microscopy , Mumps virus , Viral Proteins , Mumps virus/genetics , Mumps virus/ultrastructure , Mumps virus/metabolism , Viral Proteins/metabolism , Viral Proteins/ultrastructure , Viral Proteins/chemistry , Viral Proteins/genetics , Models, Molecular , RNA, Viral/metabolism , RNA, Viral/ultrastructure , RNA, Viral/genetics , DNA-Directed RNA Polymerases/metabolism , DNA-Directed RNA Polymerases/ultrastructure , DNA-Directed RNA Polymerases/chemistry , DNA-Directed RNA Polymerases/genetics , Protein Domains , Phosphoproteins/metabolism , Phosphoproteins/chemistry , Phosphoproteins/ultrastructure , RNA-Dependent RNA Polymerase/metabolism , RNA-Dependent RNA Polymerase/ultrastructure , RNA-Dependent RNA Polymerase/chemistry , RNA-Dependent RNA Polymerase/genetics , Virus Replication , Transcription, Genetic , Protein Conformation
2.
Nature ; 625(7995): 611-617, 2024 Jan.
Article in English | MEDLINE | ID: mdl-38123676

ABSTRACT

The respiratory syncytial virus (RSV) polymerase is a multifunctional RNA-dependent RNA polymerase composed of the large (L) protein and the phosphoprotein (P). It transcribes the RNA genome into ten viral mRNAs and replicates full-length viral genomic and antigenomic RNAs1. The RSV polymerase initiates RNA synthesis by binding to the conserved 3'-terminal RNA promoters of the genome or antigenome2. However, the lack of a structure of the RSV polymerase bound to the RNA promoter has impeded the mechanistic understanding of RSV RNA synthesis. Here we report cryogenic electron microscopy structures of the RSV polymerase bound to its genomic and antigenomic viral RNA promoters, representing two of the first structures of an RNA-dependent RNA polymerase in complex with its RNA promoters in non-segmented negative-sense RNA viruses. The overall structures of the promoter-bound RSV polymerases are similar to that of the unbound (apo) polymerase. Our structures illustrate the interactions between the RSV polymerase and the RNA promoters and provide the structural basis for the initiation of RNA synthesis at positions 1 and 3 of the RSV promoters. These structures offer a deeper understanding of the pre-initiation state of the RSV polymerase and could aid in antiviral research against RSV.


Subject(s)
Promoter Regions, Genetic , RNA-Dependent RNA Polymerase , Respiratory Syncytial Virus, Human , Promoter Regions, Genetic/genetics , Respiratory Syncytial Virus, Human/enzymology , Respiratory Syncytial Virus, Human/genetics , RNA, Viral/biosynthesis , RNA, Viral/genetics , RNA, Viral/metabolism , RNA-Dependent RNA Polymerase/chemistry , RNA-Dependent RNA Polymerase/metabolism , RNA-Dependent RNA Polymerase/ultrastructure , Virus Replication/genetics , Cryoelectron Microscopy , Subgenomic RNA/biosynthesis , Subgenomic RNA/genetics , Subgenomic RNA/metabolism
3.
Hepatology ; 75(1): 170-181, 2022 01.
Article in English | MEDLINE | ID: mdl-34387882

ABSTRACT

BACKGROUND AND AIMS: Hepatitis E virus (HEV) is one of the most common causes of acute hepatitis worldwide. Its positive-strand RNA genome encodes three open reading frames (ORF). ORF1 is translated into a large protein composed of multiple domains and is known as the viral replicase. The RNA-dependent RNA polymerase (RDRP) domain is responsible for the synthesis of viral RNA. APPROACH AND RESULTS: Here, we identified a highly conserved α-helix located in the RDRP thumb subdomain. Nuclear magnetic resonance demonstrated an amphipathic α-helix extending from amino acids 1628 to 1644 of the ORF1 protein. Functional analyses revealed a dual role of this helix in HEV RNA replication and virus production, including assembly and release. Mutations on the hydrophobic side of the amphipathic α-helix impaired RNA replication and resulted in the selection of a second-site compensatory change in the RDRP palm subdomain. Other mutations enhanced RNA replication but impaired virus assembly and/or release. CONCLUSIONS: Structure-function analyses identified a conserved amphipathic α-helix in the thumb subdomain of the HEV RDRP with a dual role in viral RNA replication and infectious particle production. This study provides structural insights into a key segment of the ORF1 protein and describes the successful use of reverse genetics in HEV, revealing functional interactions between the RDRP thumb and palm subdomains. On a broader scale, it demonstrates that the HEV replicase, similar to those of other positive-strand RNA viruses, is also involved in virus production.


Subject(s)
Hepatitis E virus/pathogenicity , Hepatitis E/virology , RNA-Dependent RNA Polymerase/metabolism , Virus Replication/genetics , Hep G2 Cells , Hepatitis E virus/genetics , Humans , Mutation , Protein Conformation, alpha-Helical/genetics , RNA, Viral/metabolism , RNA-Dependent RNA Polymerase/genetics , RNA-Dependent RNA Polymerase/ultrastructure , Structure-Activity Relationship
4.
Nature ; 587(7835): 638-643, 2020 11.
Article in English | MEDLINE | ID: mdl-33208942

ABSTRACT

Aquatic birds represent a vast reservoir from which new pandemic influenza A viruses can emerge1. Influenza viruses contain a negative-sense segmented RNA genome that is transcribed and replicated by the viral heterotrimeric RNA polymerase (FluPol) in the context of viral ribonucleoprotein complexes2,3. RNA polymerases of avian influenza A viruses (FluPolA) replicate viral RNA inefficiently in human cells because of species-specific differences in acidic nuclear phosphoprotein 32 (ANP32), a family of essential host proteins for FluPol activity4. Host-adaptive mutations, particularly a glutamic-acid-to-lysine mutation at amino acid residue 627 (E627K) in the 627 domain of the PB2 subunit, enable avian FluPolA to overcome this restriction and efficiently replicate viral RNA in the presence of human ANP32 proteins. However, the molecular mechanisms of genome replication and the interplay with ANP32 proteins remain largely unknown. Here we report cryo-electron microscopy structures of influenza C virus polymerase (FluPolC) in complex with human and chicken ANP32A. In both structures, two FluPolC molecules form an asymmetric dimer bridged by the N-terminal leucine-rich repeat domain of ANP32A. The C-terminal low-complexity acidic region of ANP32A inserts between the two juxtaposed PB2 627 domains of the asymmetric FluPolA dimer, suggesting a mechanism for how the adaptive PB2(E627K) mutation enables the replication of viral RNA in mammalian hosts. We propose that this complex represents a replication platform for the viral RNA genome, in which one of the FluPol molecules acts as a replicase while the other initiates the assembly of the nascent replication product into a viral ribonucleoprotein complex.


Subject(s)
Cryoelectron Microscopy , Gammainfluenzavirus/enzymology , Host-Pathogen Interactions , Nuclear Proteins/chemistry , Nuclear Proteins/metabolism , RNA-Binding Proteins/chemistry , RNA-Binding Proteins/metabolism , RNA-Dependent RNA Polymerase/chemistry , RNA-Dependent RNA Polymerase/metabolism , Animals , Chickens/virology , Genome, Viral/genetics , HEK293 Cells , Humans , Gammainfluenzavirus/genetics , Models, Molecular , Nuclear Proteins/ultrastructure , Orthomyxoviridae Infections/genetics , Orthomyxoviridae Infections/metabolism , Orthomyxoviridae Infections/virology , Protein Multimerization , RNA, Viral/biosynthesis , RNA, Viral/genetics , RNA-Binding Proteins/ultrastructure , RNA-Dependent RNA Polymerase/ultrastructure , Sf9 Cells
5.
Nat Commun ; 11(1): 3656, 2020 07 21.
Article in English | MEDLINE | ID: mdl-32694517

ABSTRACT

Avian influenza polymerase undergoes host adaptation in order to efficiently replicate in human cells. Adaptive mutants are localised on the C-terminal (627-NLS) domains of the PB2 subunit. In particular, mutation of PB2 residue 627 from E to K rescues polymerase activity in mammalian cells. A host transcription regulator ANP32A, comprising a long C-terminal intrinsically disordered domain (IDD), is responsible for this adaptation. Human ANP32A IDD lacks a 33 residue insertion compared to avian ANP32A, and this deletion restricts avian influenza polymerase activity. We used NMR to determine conformational ensembles of E627 and K627 forms of 627-NLS of PB2 in complex with avian and human ANP32A. Human ANP32A IDD transiently binds to the 627 domain, exploiting multivalency to maximise affinity. E627 interrupts the polyvalency of the interaction, an effect compensated by an avian-unique motif in the IDD. The observed binding mode is maintained in the context of heterotrimeric influenza polymerase, placing ANP32A in the immediate vicinity of known host-adaptive PB2 mutants.


Subject(s)
Avian Proteins/ultrastructure , Influenza A Virus, H5N1 Subtype/pathogenicity , Nuclear Proteins/ultrastructure , Protein Domains/genetics , RNA-Binding Proteins/ultrastructure , RNA-Dependent RNA Polymerase/ultrastructure , Viral Proteins/ultrastructure , Animals , Avian Proteins/metabolism , Birds/virology , Humans , Influenza A Virus, H5N1 Subtype/genetics , Influenza A Virus, H5N1 Subtype/metabolism , Influenza in Birds/virology , Influenza, Human/virology , Mutation , Nuclear Magnetic Resonance, Biomolecular , Nuclear Proteins/metabolism , Protein Binding/genetics , RNA-Binding Proteins/metabolism , RNA-Dependent RNA Polymerase/genetics , RNA-Dependent RNA Polymerase/metabolism , Species Specificity , Viral Proteins/genetics , Viral Proteins/metabolism , Virus Replication
6.
J Struct Biol ; 211(1): 107510, 2020 07 01.
Article in English | MEDLINE | ID: mdl-32353513

ABSTRACT

Enterovirus D68 (EV-D68) is an emerging viral pathogen belonging to the Enterovirus genus of the Picornaviridae family, which is a serious threat to human health and has resulted in significant economic losses. The EV-D68 genome encodes an RNA-dependent RNA polymerase (RdRp) 3Dpol, which is central for viral genome replication and considered as a promising target for specific antiviral therapeutics. In this study, we report the crystal structures of human EV-D68 RdRp in the apo state and in complex with the inhibitor NADPH, which was selected by using a structure-based virtual screening approach. The EV-D68-RdRp-NADPH complex is the first RdRp-inhibitor structure identified in the species Enterovirus D. The inhibitor NADPH occupies the RNA template binding channel of EV-D68 RdRp with a novel binding pocket. Additionally, residues involved in the NADPH binding pocket of EV-D68 RdRp are highly conserved in RdRps of enteroviruses. Therefore, the enzyme activity of three RdRps from EV-D68, poliovirus, and enterovirus A71 is shown to decrease when titrated with NADPH separately in vitro. Furthermore, we identified that NADPH plays a pivotal role as an RdRp inhibitor instead of a chain terminator during restriction of RNA-dependent RNA replication. In the future, derivatives of NADPH may pave the way for novel inhibitors of RdRp through compound modification, providing potential antiviral agents for treating enteroviral infection and related diseases.


Subject(s)
Enterovirus D, Human/ultrastructure , Enterovirus Infections/virology , NADP/ultrastructure , RNA-Dependent RNA Polymerase/ultrastructure , Binding Sites/genetics , Enterovirus D, Human/genetics , Enterovirus D, Human/pathogenicity , Enterovirus Infections/genetics , Genome, Viral/genetics , Humans , NADP/chemistry , RNA/genetics , RNA/ultrastructure , RNA-Dependent RNA Polymerase/chemistry , RNA-Dependent RNA Polymerase/genetics , Virus Replication/genetics
7.
Nature ; 584(7819): 154-156, 2020 08.
Article in English | MEDLINE | ID: mdl-32438371

ABSTRACT

The new coronavirus severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) uses an RNA-dependent RNA polymerase (RdRp) for the replication of its genome and the transcription of its genes1-3. Here we present a cryo-electron microscopy structure of the SARS-CoV-2 RdRp in an active form that mimics the replicating enzyme. The structure comprises the viral proteins non-structural protein 12 (nsp12), nsp8 and nsp7, and more than two turns of RNA template-product duplex. The active-site cleft of nsp12 binds to the first turn of RNA and mediates RdRp activity with conserved residues. Two copies of nsp8 bind to opposite sides of the cleft and position the second turn of RNA. Long helical extensions in nsp8 protrude along exiting RNA, forming positively charged 'sliding poles'. These sliding poles can account for the known processivity of RdRp that is required for replicating the long genome of coronaviruses3. Our results enable a detailed analysis of the inhibitory mechanisms that underlie the antiviral activity of substances such as remdesivir, a drug for the treatment of coronavirus disease 2019 (COVID-19)4.


Subject(s)
Betacoronavirus/enzymology , Cryoelectron Microscopy , RNA, Viral/biosynthesis , RNA-Dependent RNA Polymerase/chemistry , RNA-Dependent RNA Polymerase/metabolism , Viral Nonstructural Proteins/chemistry , Viral Nonstructural Proteins/metabolism , Adenosine Monophosphate/analogs & derivatives , Adenosine Monophosphate/pharmacology , Alanine/analogs & derivatives , Alanine/pharmacology , Betacoronavirus/drug effects , Betacoronavirus/genetics , Betacoronavirus/ultrastructure , Coronavirus RNA-Dependent RNA Polymerase , Models, Molecular , Protein Conformation , RNA, Viral/chemistry , RNA, Viral/metabolism , RNA-Dependent RNA Polymerase/genetics , RNA-Dependent RNA Polymerase/ultrastructure , SARS-CoV-2 , Viral Nonstructural Proteins/genetics , Viral Nonstructural Proteins/ultrastructure
8.
Science ; 368(6492): 779-782, 2020 05 15.
Article in English | MEDLINE | ID: mdl-32277040

ABSTRACT

A novel coronavirus [severe acute respiratory syndrome-coronavirus 2 (SARS-CoV-2)] outbreak has caused a global coronavirus disease 2019 (COVID-19) pandemic, resulting in tens of thousands of infections and thousands of deaths worldwide. The RNA-dependent RNA polymerase [(RdRp), also named nsp12] is the central component of coronaviral replication and transcription machinery, and it appears to be a primary target for the antiviral drug remdesivir. We report the cryo-electron microscopy structure of COVID-19 virus full-length nsp12 in complex with cofactors nsp7 and nsp8 at 2.9-angstrom resolution. In addition to the conserved architecture of the polymerase core of the viral polymerase family, nsp12 possesses a newly identified ß-hairpin domain at its N terminus. A comparative analysis model shows how remdesivir binds to this polymerase. The structure provides a basis for the design of new antiviral therapeutics that target viral RdRp.


Subject(s)
Betacoronavirus/enzymology , RNA-Dependent RNA Polymerase/chemistry , RNA-Dependent RNA Polymerase/ultrastructure , Viral Nonstructural Proteins/chemistry , Viral Nonstructural Proteins/ultrastructure , Adenosine Monophosphate/analogs & derivatives , Adenosine Monophosphate/metabolism , Adenosine Monophosphate/pharmacology , Alanine/analogs & derivatives , Alanine/metabolism , Alanine/pharmacology , Antiviral Agents/metabolism , Antiviral Agents/pharmacology , Catalytic Domain , Coronavirus RNA-Dependent RNA Polymerase , Cryoelectron Microscopy , Drug Design , Models, Molecular , Multiprotein Complexes/chemistry , Multiprotein Complexes/metabolism , Multiprotein Complexes/ultrastructure , Protein Conformation, beta-Strand , Protein Domains , RNA-Dependent RNA Polymerase/antagonists & inhibitors , RNA-Dependent RNA Polymerase/metabolism , SARS-CoV-2 , Viral Nonstructural Proteins/antagonists & inhibitors , Viral Nonstructural Proteins/metabolism
9.
Nature ; 579(7800): 615-619, 2020 03.
Article in English | MEDLINE | ID: mdl-32214249

ABSTRACT

Arenaviruses can cause severe haemorrhagic fever and neurological diseases in humans and other animals, exemplified by Lassa mammarenavirus, Machupo mammarenavirus and lymphocytic choriomeningitis virus, posing great threats to public health1-4. These viruses encode a large multi-domain RNA-dependent RNA polymerase for transcription and replication of the viral genome5. Viral polymerases are one of the leading antiviral therapeutic targets. However, the structure of arenavirus polymerase is not yet known. Here we report the near-atomic resolution structures of Lassa and Machupo virus polymerases in both apo and promoter-bound forms. These structures display a similar overall architecture to influenza virus and bunyavirus polymerases but possess unique local features, including an arenavirus-specific insertion domain that regulates the polymerase activity. Notably, the ordered active site of arenavirus polymerase is inherently switched on, without the requirement for allosteric activation by 5'-viral RNA, which is a necessity for both influenza virus and bunyavirus polymerases6,7. Moreover, dimerization could facilitate the polymerase activity. These findings advance our understanding of the mechanism of arenavirus replication and provide an important basis for developing antiviral therapeutics.


Subject(s)
Arenaviruses, New World/enzymology , Cryoelectron Microscopy , Lassa virus/enzymology , RNA-Dependent RNA Polymerase/chemistry , RNA-Dependent RNA Polymerase/ultrastructure , Virus Replication , Apoenzymes/chemistry , Apoenzymes/metabolism , Apoenzymes/ultrastructure , Arenaviruses, New World/ultrastructure , Catalytic Domain , Lassa virus/ultrastructure , Lymphocytic choriomeningitis virus/enzymology , Lymphocytic choriomeningitis virus/ultrastructure , Models, Molecular , Promoter Regions, Genetic/genetics , RNA-Dependent RNA Polymerase/metabolism
10.
Cell Rep ; 30(1): 53-60.e5, 2020 01 07.
Article in English | MEDLINE | ID: mdl-31914397

ABSTRACT

The large (L) proteins of non-segmented, negative-strand RNA viruses are multifunctional enzymes that produce capped, methylated, and polyadenylated mRNA and replicate the viral genome. A phosphoprotein (P), required for efficient RNA-dependent RNA polymerization from the viral ribonucleoprotein (RNP) template, regulates the function and conformation of the L protein. We report the structure of vesicular stomatitis virus L in complex with its P cofactor determined by electron cryomicroscopy at 3.0 Å resolution, enabling us to visualize bound segments of P. The contacts of three P segments with multiple L domains show how P induces a closed, compact, initiation-competent conformation. Binding of P to L positions its N-terminal domain adjacent to a putative RNA exit channel for efficient encapsidation of newly synthesized genomes with the nucleoprotein and orients its C-terminal domain to interact with an RNP template. The model shows that a conserved tryptophan in the priming loop can support the initiating 5' nucleotide.


Subject(s)
Coenzymes/metabolism , Phosphoproteins/metabolism , RNA-Dependent RNA Polymerase/chemistry , RNA-Dependent RNA Polymerase/metabolism , Viral Proteins/chemistry , Viral Proteins/metabolism , Amino Acid Sequence , Animals , Cell Line , Humans , Models, Molecular , Phosphoproteins/chemistry , Phosphoproteins/ultrastructure , Protein Binding , Protein Subunits/chemistry , Protein Subunits/metabolism , RNA-Dependent RNA Polymerase/ultrastructure , Viral Proteins/ultrastructure
11.
J Mol Biol ; 432(2): 358-366, 2020 01 17.
Article in English | MEDLINE | ID: mdl-31629769

ABSTRACT

RNA-dependent RNA polymerases (RdRps) catalyze RNA synthesis of RNA viruses. During initiation of RNA synthesis, the RdRp catalyzes the formation of the first dinucleotide, acting as primer for subsequent processive RNA elongation. Here, we present the structure of the RdRp complexes in the dinucleotide primed state in situ within a transcribing cypovirus under near physiological conditions using cryo-electron microscopy. The 3' end of RNA templates, paired RNA dinucleotide primer, incoming nucleotide, and catalytic divalent cations in the RdRp were resolved at 3.8 Å resolution. The end of the RNA template and the dinucleotide is buttressed by the aromatic tyrosine in a loop from the RdRp bracelet domain. Our structure reveals the interactions between the nucleotide substrates and the conserved residues during the RdRp initiation, and the coordinated structural changes preceding the elongation stage. In addition, it provides the direct evidence for existence of the slow step of the dinucleotide primed state in the viral RdRp transcription.


Subject(s)
RNA Viruses/ultrastructure , RNA-Dependent RNA Polymerase/ultrastructure , RNA/biosynthesis , Reoviridae/ultrastructure , Cryoelectron Microscopy , Multiprotein Complexes , Protein Conformation , RNA/chemistry , RNA/genetics , RNA Viruses/enzymology , RNA-Dependent RNA Polymerase/chemistry , RNA-Dependent RNA Polymerase/genetics , Reoviridae/chemistry , Reoviridae/genetics , Transcription, Genetic
12.
Nucleic Acids Res ; 48(3): 1392-1405, 2020 02 20.
Article in English | MEDLINE | ID: mdl-31863580

ABSTRACT

The enterovirus 71 (EV71) 3Dpol is an RNA-dependent RNA polymerase (RdRP) that plays the central role in the viral genome replication, and is an important target in antiviral studies. Here, we report a crystal structure of EV71 3Dpol elongation complex (EC) at 1.8 Å resolution. The structure reveals that the 5'-end guanosine of the downstream RNA template interacts with a fingers domain pocket, with the base sandwiched by H44 and R277 side chains through hydrophobic stacking interactions, and these interactions are still maintained after one in-crystal translocation event induced by nucleotide incorporation, implying that the pocket could regulate the functional properties of the polymerase by interacting with RNA. When mutated, residue R277 showed an impact on virus proliferation in virological studies with residue H44 having a synergistic effect. In vitro biochemical data further suggest that mutations at these two sites affect RNA binding, EC stability, but not polymerase catalytic rate (kcat) and apparent NTP affinity (KM,NTP). We propose that, although rarely captured by crystallography, similar surface pocket interaction with nucleobase may commonly exist in nucleic acid motor enzymes to facilitate their processivity. Potential applications in antiviral drug and vaccine development are also discussed.


Subject(s)
Enterovirus A, Human/ultrastructure , Multiprotein Complexes/ultrastructure , Protein Conformation , RNA-Dependent RNA Polymerase/ultrastructure , Antiviral Agents/chemistry , Binding Sites , Crystallography, X-Ray , Enterovirus A, Human/chemistry , Enterovirus A, Human/genetics , Genome, Viral , Humans , Models, Molecular , Multiprotein Complexes/chemistry , Nucleotides/chemistry , RNA, Viral/chemistry , RNA, Viral/ultrastructure , RNA-Dependent RNA Polymerase/chemistry , Virus Replication/genetics
13.
Nat Struct Mol Biol ; 26(11): 1023-1034, 2019 11.
Article in English | MEDLINE | ID: mdl-31695188

ABSTRACT

Endogenous RNA transcription characterizes double-stranded RNA (dsRNA) viruses in the Reoviridae, a family that is exemplified by its simple, single-shelled member cytoplasmic polyhedrosis virus (CPV). Because of the lack of in situ structures of the intermediate stages of RNA-dependent RNA polymerase (RdRp) during transcription, it is poorly understood how RdRp detects environmental cues and internal transcriptional states to initiate and coordinate repeated cycles of transcript production inside the capsid. Here, we captured five high-resolution (2.8-3.5 Å) RdRp-RNA in situ structures-representing quiescent, initiation, early elongation, elongation and abortive states-under seven experimental conditions of CPV. We observed the 'Y'-form initial RNA fork in the initiation state and the complete transcription bubble in the elongation state. These structures reveal that de novo RNA transcription involves three major conformational changes during state transitions. Our results support an ouroboros model for endogenous conservative transcription in dsRNA viruses.


Subject(s)
RNA, Double-Stranded/genetics , RNA, Viral/genetics , Reoviridae/genetics , Transcription, Genetic , Cryoelectron Microscopy , Humans , Models, Molecular , RNA, Double-Stranded/chemistry , RNA, Double-Stranded/ultrastructure , RNA, Viral/chemistry , RNA, Viral/ultrastructure , RNA-Dependent RNA Polymerase/chemistry , RNA-Dependent RNA Polymerase/ultrastructure , Reoviridae/chemistry , Reoviridae/ultrastructure , Reoviridae Infections/virology , Viral Proteins/chemistry , Viral Proteins/ultrastructure
14.
Cell ; 179(1): 193-204.e14, 2019 Sep 19.
Article in English | MEDLINE | ID: mdl-31495574

ABSTRACT

Numerous interventions are in clinical development for respiratory syncytial virus (RSV) infection, including small molecules that target viral transcription and replication. These processes are catalyzed by a complex comprising the RNA-dependent RNA polymerase (L) and the tetrameric phosphoprotein (P). RSV P recruits multiple proteins to the polymerase complex and, with the exception of its oligomerization domain, is thought to be intrinsically disordered. Despite their critical roles in RSV transcription and replication, structures of L and P have remained elusive. Here, we describe the 3.2-Å cryo-EM structure of RSV L bound to tetrameric P. The structure reveals a striking tentacular arrangement of P, with each of the four monomers adopting a distinct conformation. The structure also rationalizes inhibitor escape mutants and mutations observed in live-attenuated vaccine candidates. These results provide a framework for determining the molecular underpinnings of RSV replication and transcription and should facilitate the design of effective RSV inhibitors.


Subject(s)
Phosphoproteins/ultrastructure , RNA-Dependent RNA Polymerase/ultrastructure , Respiratory Syncytial Virus Infections/virology , Respiratory Syncytial Virus, Human/enzymology , Viral Proteins/ultrastructure , Acetates/chemistry , Animals , Antiviral Agents/chemistry , Antiviral Agents/therapeutic use , Catalytic Domain , Cryoelectron Microscopy , Deoxycytidine/analogs & derivatives , Deoxycytidine/chemistry , Deoxycytidine/pharmacology , Deoxycytidine/therapeutic use , Hydrogen Bonding , Hydrophobic and Hydrophilic Interactions , Phosphoproteins/chemistry , Phosphoproteins/metabolism , Protein Conformation, alpha-Helical , Protein Interaction Domains and Motifs , Quinolines/chemistry , RNA-Dependent RNA Polymerase/antagonists & inhibitors , RNA-Dependent RNA Polymerase/chemistry , RNA-Dependent RNA Polymerase/metabolism , Respiratory Syncytial Virus Infections/drug therapy , Respiratory Syncytial Virus Vaccines/chemistry , Sf9 Cells , Spodoptera , Viral Proteins/chemistry , Viral Proteins/metabolism , Virus Replication/drug effects
15.
J Biol Chem ; 294(37): 13606-13618, 2019 09 13.
Article in English | MEDLINE | ID: mdl-31350335

ABSTRACT

Coronaviruses are enveloped, single-stranded RNA viruses that are distributed worldwide. They include transmissible gastroenteritis virus (TGEV), porcine epidemic diarrhea virus (PEDV), and the human coronaviruses severe acute respiratory syndrome coronavirus (SARS-CoV) and Middle East respiratory syndrome coronavirus (MERS-CoV), many of which seriously endanger human health and well-being. Only alphacoronaviruses and betacoronaviruses harbor nonstructural protein 1 (nsp1), which performs multiple functions in inhibiting antiviral host responses. The role of the C terminus of betacoronavirus nsp1 in virulence has been characterized, but the location of the alphacoronavirus nsp1 region that is important for virulence remains unclear. Here, using TGEV nsp1 as a model to explore the function of this protein in alphacoronaviruses, we demonstrate that alphacoronavirus nsp1 inhibits host gene expression. Solving the crystal structure of full-length TGEV at 1.85-Å resolution and conducting several biochemical analyses, we observed that a specific motif (amino acids 91-95) of alphacoronavirus nsp1 is a conserved region that inhibits host protein synthesis. Using a reverse-genetics system based on CRISPR/Cas9 technology to construct a recombinant TGEV in which this specific nsp1 motif was altered, we found that this mutation does not affect virus replication in cell culture but significantly reduces TGEV pathogenicity in pigs. Taken together, our findings suggest that alphacoronavirus nsp1 is an essential virulence determinant, providing a potential paradigm for the development of a new attenuated vaccine based on modified nsp1.


Subject(s)
Alphacoronavirus/genetics , Viral Nonstructural Proteins/metabolism , Virus Replication/genetics , Alphacoronavirus/pathogenicity , Animals , Betacoronavirus , Host Microbial Interactions/genetics , Host Microbial Interactions/physiology , Humans , Middle East Respiratory Syndrome Coronavirus/genetics , Porcine epidemic diarrhea virus/genetics , Protein Biosynthesis , RNA-Dependent RNA Polymerase/genetics , RNA-Dependent RNA Polymerase/metabolism , RNA-Dependent RNA Polymerase/ultrastructure , Severe acute respiratory syndrome-related coronavirus/genetics , Swine , Synapsins/metabolism , Transmissible gastroenteritis virus/genetics , Viral Nonstructural Proteins/genetics , Viral Nonstructural Proteins/ultrastructure , Virulence , Virus Replication/physiology
16.
Nat Commun ; 10(1): 2216, 2019 05 17.
Article in English | MEDLINE | ID: mdl-31101900

ABSTRACT

Transcribing and replicating a double-stranded genome require protein modules to unwind, transcribe/replicate nucleic acid substrates, and release products. Here we present in situ cryo-electron microscopy structures of rotavirus dsRNA-dependent RNA polymerase (RdRp) in two states pertaining to transcription. In addition to the previously discovered universal "hand-shaped" polymerase core domain shared by DNA polymerases and telomerases, our results show the function of N- and C-terminal domains of RdRp: the former opens the genome duplex to isolate the template strand; the latter splits the emerging template-transcript hybrid, guides genome reannealing to form a transcription bubble, and opens a capsid shell protein (CSP) to release the transcript. These two "helicase" domains also extensively interact with CSP, which has a switchable N-terminal helix that, like cellular transcriptional factors, either inhibits or promotes RdRp activity. The in situ structures of RdRp, CSP, and RNA in action inform mechanisms of not only transcription, but also replication.


Subject(s)
DNA Replication/physiology , RNA, Messenger/ultrastructure , RNA-Dependent RNA Polymerase/ultrastructure , Rotavirus/physiology , Transcription, Genetic/physiology , Animals , Capsid Proteins/metabolism , Capsid Proteins/ultrastructure , Cell Line , Chlorocebus aethiops , Cryoelectron Microscopy , Models, Molecular , Protein Domains/genetics , RNA, Double-Stranded/metabolism , RNA, Messenger/metabolism , RNA, Viral/metabolism , RNA-Dependent RNA Polymerase/genetics , RNA-Dependent RNA Polymerase/metabolism , Rotavirus/ultrastructure , Virus Replication/physiology
17.
Elife ; 62017 05 03.
Article in English | MEDLINE | ID: mdl-28467302

ABSTRACT

We present an approach to study macromolecular assemblies by detecting component proteins' characteristic high-resolution projection patterns, calculated from their known 3D structures, in single electron cryo-micrographs. Our method detects single apoferritin molecules in vitreous ice with high specificity and determines their orientation and location precisely. Simulations show that high spatial-frequency information and-in the presence of protein background-a whitening filter are essential for optimal detection, in particular for images taken far from focus. Experimentally, we could detect small viral RNA polymerase molecules, distributed randomly among binding locations, inside rotavirus particles. Based on the currently attainable image quality, we estimate a threshold for detection that is 150 kDa in ice and 300 kDa in 100 nm thick samples of dense biological material.


Subject(s)
Cryoelectron Microscopy/methods , Image Processing, Computer-Assisted/methods , Single Molecule Imaging/methods , RNA-Dependent RNA Polymerase/ultrastructure , Rotavirus/ultrastructure
18.
J Mol Biol ; 429(1): 79-87, 2017 01 06.
Article in English | MEDLINE | ID: mdl-27914893

ABSTRACT

Single-particle cryo-electron microscopy (cryo-EM) allows the high-resolution structural determination of biological assemblies in a near-native environment. However, all high-resolution (better than 3.5Å) cryo-EM structures reported to date were obtained by using 300kV transmission electron microscopes (TEMs). We report here the structures of a cypovirus capsid of 750-Å diameter at 3.3-Å resolution and of RNA-dependent RNA polymerase (RdRp) complexes within the capsid at 3.9-Å resolution using a 200-kV TEM. The newly resolved structure revealed conformational changes of two subdomains in the RdRp. These conformational changes, which were involved in RdRp's switch from non-transcribing to transcribing mode, suggest that the RdRp may facilitate the unwinding of genomic double-stranded RNA. The possibility of 3-Å resolution structural determinations for biological assemblies of relatively small sizes using cryo-EM at 200kV was discussed.


Subject(s)
Capsid/ultrastructure , Cryoelectron Microscopy , Macromolecular Substances/ultrastructure , RNA-Dependent RNA Polymerase/chemistry , RNA-Dependent RNA Polymerase/ultrastructure , Reoviridae/ultrastructure , Models, Molecular , Protein Conformation , RNA, Viral/metabolism , RNA-Dependent RNA Polymerase/metabolism , Reoviridae/enzymology , Reoviridae/metabolism
19.
Nat Commun ; 6: 8843, 2015 Nov 04.
Article in English | MEDLINE | ID: mdl-26534841

ABSTRACT

Electron cryomicroscopy can yield near-atomic resolution structures of highly ordered macromolecular complexes. Often however some subunits bind in a flexible manner, have different symmetry from the rest of the complex, or are present in sub-stoichiometric amounts, limiting the attainable resolution. Here we report a general method for the localized three-dimensional reconstruction of such subunits. After determining the particle orientations, local areas corresponding to the subunits can be extracted and treated as single particles. We demonstrate the method using three examples including a flexible assembly and complexes harbouring subunits with either partial occupancy or mismatched symmetry. Most notably, the method allows accurate fitting of the monomeric RNA-dependent RNA polymerase bound at the threefold axis of symmetry inside a viral capsid, revealing for the first time its exact orientation and interactions with the capsid proteins. Localized reconstruction is expected to provide novel biological insights in a range of challenging biological systems.


Subject(s)
Capsid Proteins/ultrastructure , Cryoelectron Microscopy/methods , Macromolecular Substances/ultrastructure , Protein Subunits , RNA-Dependent RNA Polymerase/ultrastructure , Capsid Proteins/chemistry , Capsid Proteins/metabolism , Macromolecular Substances/chemistry , Macromolecular Substances/metabolism , RNA-Dependent RNA Polymerase/chemistry , RNA-Dependent RNA Polymerase/metabolism , Rotavirus
20.
Nature ; 527(7579): 531-534, 2015 Nov 26.
Article in English | MEDLINE | ID: mdl-26503045

ABSTRACT

Viruses in the Reoviridae, like the triple-shelled human rotavirus and the single-shelled insect cytoplasmic polyhedrosis virus (CPV), all package a genome of segmented double-stranded RNAs (dsRNAs) inside the viral capsid and carry out endogenous messenger RNA synthesis through a transcriptional enzyme complex (TEC). By direct electron-counting cryoelectron microscopy and asymmetric reconstruction, we have determined the organization of the dsRNA genome inside quiescent CPV (q-CPV) and the in situ atomic structures of TEC within CPV in both quiescent and transcribing (t-CPV) states. We show that the ten segmented dsRNAs in CPV are organized with ten TECs in a specific, non-symmetric manner, with each dsRNA segment attached directly to a TEC. The TEC consists of two extensively interacting subunits: an RNA-dependent RNA polymerase (RdRP) and an NTPase VP4. We find that the bracelet domain of RdRP undergoes marked conformational change when q-CPV is converted to t-CPV, leading to formation of the RNA template entry channel and access to the polymerase active site. An amino-terminal helix from each of two subunits of the capsid shell protein (CSP) interacts with VP4 and RdRP. These findings establish the link between sensing of environmental cues by the external proteins and activation of endogenous RNA transcription by the TEC inside the virus.


Subject(s)
Genome, Viral , Multienzyme Complexes/ultrastructure , RNA, Double-Stranded/ultrastructure , RNA, Viral/ultrastructure , RNA-Dependent RNA Polymerase/ultrastructure , Reoviridae/ultrastructure , Capsid Proteins/chemistry , Capsid Proteins/metabolism , Capsid Proteins/ultrastructure , Catalytic Domain , Cryoelectron Microscopy , Genome, Viral/genetics , Models, Molecular , Multienzyme Complexes/chemistry , Multienzyme Complexes/metabolism , Nucleoside-Triphosphatase/metabolism , Nucleoside-Triphosphatase/ultrastructure , Protein Subunits/chemistry , Protein Subunits/metabolism , RNA, Double-Stranded/genetics , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , RNA, Messenger/ultrastructure , RNA, Viral/biosynthesis , RNA, Viral/genetics , RNA-Dependent RNA Polymerase/chemistry , RNA-Dependent RNA Polymerase/metabolism , Reoviridae/enzymology , Reoviridae/genetics , Templates, Genetic , Transcription, Genetic
SELECTION OF CITATIONS
SEARCH DETAIL
...