Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
Nat Commun ; 14(1): 6538, 2023 10 20.
Article in English | MEDLINE | ID: mdl-37863901

ABSTRACT

Histamine is a biogenic amine that participates in allergic and inflammatory processes by stimulating histamine receptors. The histamine H4 receptor (H4R) is a potential therapeutic target for chronic inflammatory diseases such as asthma and atopic dermatitis. Here, we show the cryo-electron microscopy structures of the H4R-Gq complex bound with an endogenous agonist histamine or the selective agonist imetit bound in the orthosteric binding pocket. The structures demonstrate binding mode of histamine agonists and that the subtype-selective agonist binding causes conformational changes in Phe3447.39, which, in turn, form the "aromatic slot". The results provide insights into the molecular underpinnings of the agonism of H4R and subtype selectivity of histamine receptors, and show that the H4R structures may be valuable in rational drug design of drugs targeting the H4R.


Subject(s)
Histamine , Receptors, G-Protein-Coupled , Humans , Histamine/metabolism , Receptors, Histamine H4 , Cryoelectron Microscopy , Receptors, G-Protein-Coupled/metabolism , Receptors, Histamine/metabolism , Histamine Agonists/pharmacology
2.
PNAS Nexus ; 2(4): pgad120, 2023 Apr.
Article in English | MEDLINE | ID: mdl-37124400

ABSTRACT

Lloviu virus (LLOV) is a novel filovirus detected in Schreiber's bats in Europe. The isolation of the infectious LLOV from bats has raised public health concerns. However, the virological and molecular characteristics of LLOV remain largely unknown. The nucleoprotein (NP) of LLOV encapsidates the viral genomic RNA to form a helical NP-RNA complex, which acts as a scaffold for nucleocapsid formation and de novo viral RNA synthesis. In this study, using single-particle cryoelectron microscopy, we determined two structures of the LLOV NP-RNA helical complex, comprising a full-length and a C-terminally truncated NP. The two helical structures were identical, demonstrating that the N-terminal region determines the helical arrangement of the NP. The LLOV NP-RNA protomers displayed a structure similar to that in the Ebola and Marburg virus, but the spatial arrangements in the helix differed. Structure-based mutational analysis identified amino acids involved in the helical assembly and viral RNA synthesis. These structures advance our understanding of the filovirus nucleocapsid formation and provide a structural basis for the development of antifiloviral therapeutics.

3.
Cell Rep ; 40(11): 111323, 2022 09 13.
Article in English | MEDLINE | ID: mdl-36103815

ABSTRACT

Prostaglandin receptors have been implicated in a wide range of functions, including inflammation, immune response, reproduction, and cancer. Our group has previously determined the crystal structure of the active-like EP3 bound to its endogenous agonist, prostaglandin E2. Here, we present the single-particle cryoelectron microscopy (cryo-EM) structure of the human EP3-Gi signaling complex at a resolution of 3.4 Å. The structure reveals the binding mode of Gi to EP3 and the structural changes induced in EP3 by Gi binding. In addition, we compare the structure of the EP3-Gi complex with other subtypes of prostaglandin receptors (EP2 and EP4) bound to Gs that have been previously reported and examine the differences in amino acid composition at the receptor-G protein interface. Mutational analysis reveals that the selectivity of the G protein depends on specific amino acid residues in the second intracellular loop and TM5.


Subject(s)
Dinoprostone , Receptors, Prostaglandin E , Amino Acids , Cryoelectron Microscopy , Dinoprostone/pharmacology , Humans , Receptors, Prostaglandin E/agonists , Receptors, Prostaglandin E/metabolism , Receptors, Prostaglandin E, EP3 Subtype/metabolism
4.
Nat Commun ; 13(1): 1191, 2022 03 04.
Article in English | MEDLINE | ID: mdl-35246537

ABSTRACT

The nucleoprotein (NP) of Marburg virus (MARV), a close relative of Ebola virus (EBOV), encapsidates the single-stranded, negative-sense viral genomic RNA (vRNA) to form the helical NP-RNA complex. The NP-RNA complex constitutes the core structure for the assembly of the nucleocapsid that is responsible for viral RNA synthesis. Although appropriate interactions among NPs and RNA are required for the formation of nucleocapsid, the structural basis of the helical assembly remains largely elusive. Here, we show the structure of the MARV NP-RNA complex determined using cryo-electron microscopy at a resolution of 3.1 Å. The structures of the asymmetric unit, a complex of an NP and six RNA nucleotides, was very similar to that of EBOV, suggesting that both viruses share common mechanisms for the nucleocapsid formation. Structure-based mutational analysis of both MARV and EBOV NPs identified key residues for helical assembly and subsequent viral RNA synthesis. Importantly, most of the residues identified were conserved in both viruses. These findings provide a structural basis for understanding the nucleocapsid formation and contribute to the development of novel antivirals against MARV and EBOV.


Subject(s)
Ebolavirus , Marburgvirus , Cryoelectron Microscopy , Ebolavirus/genetics , Marburgvirus/genetics , Nucleoproteins/chemistry , RNA, Viral/chemistry , RNA, Viral/genetics
6.
Molecules ; 26(18)2021 Sep 08.
Article in English | MEDLINE | ID: mdl-34576938

ABSTRACT

Glycosylphosphatidylinositol (GPI) anchor modification is a posttranslational modification of proteins that has been conserved in eukaryotes. The biosynthesis and transfer of GPI to proteins are carried out in the endoplasmic reticulum. Attachment of GPI to proteins is mediated by the GPI-transamidase (GPI-TA) complex, which recognizes and cleaves the C-terminal GPI attachment signal of precursor proteins. Then, GPI is transferred to the newly exposed C-terminus of the proteins. GPI-TA consists of five subunits: PIGK, GPAA1, PIGT, PIGS, and PIGU, and the absence of any subunit leads to the loss of activity. Here, we analyzed functionally important residues of the five subunits of GPI-TA by comparing conserved sequences among homologous proteins. In addition, we optimized the purification method for analyzing the structure of GPI-TA. Using purified GPI-TA, preliminary single particle images were obtained. Our results provide guidance for the structural and functional analysis of GPI-TA.


Subject(s)
Acyltransferases/chemistry , Acyltransferases/genetics , Acyltransferases/metabolism , Amino Acids/genetics , Acyltransferases/isolation & purification , Cryoelectron Microscopy , Detergents/chemistry , HEK293 Cells , Humans , Mutation , Protein Conformation , Protein Subunits , Recombinant Proteins/genetics , Recombinant Proteins/metabolism
7.
Commun Biol ; 4(1): 858, 2021 07 09.
Article in English | MEDLINE | ID: mdl-34244608

ABSTRACT

The single-stranded, negative-sense, viral genomic RNA (vRNA) of influenza A virus is encapsidated by viral nucleoproteins (NPs) and an RNA polymerase to form a ribonucleoprotein complex (vRNP) with a helical, rod-shaped structure. The vRNP is responsible for transcription and replication of the vRNA. However, the vRNP conformation during RNA synthesis is not well understood. Here, using high-speed atomic force microscopy and cryo-electron microscopy, we investigated the native structure of influenza A vRNPs during RNA synthesis in vitro. Two distinct types of vRNPs were observed in association with newly synthesized RNAs: an intact, helical rod-shaped vRNP connected with a folded RNA and a deformed vRNP associated with a looped RNA. Interestingly, the looped RNA was a double-stranded RNA, which likely comprises a nascent RNA and the template RNA detached from NPs of the vRNP. These results suggest that while some vRNPs keep their helical structures during RNA synthesis, for the repeated cycle of RNA synthesis, others accidentally become structurally deformed, which likely results in failure to commence or continue RNA synthesis. Thus, our findings provide the ultrastructural feature of vRNPs during RNA synthesis.


Subject(s)
Influenza A virus/metabolism , RNA, Viral/metabolism , Ribonucleoproteins/metabolism , Viral Proteins/metabolism , Cryoelectron Microscopy/methods , Humans , Influenza A virus/genetics , Influenza A virus/physiology , Influenza, Human/virology , Microscopy, Atomic Force/methods , Models, Molecular , Nucleic Acid Conformation , RNA, Viral/chemistry , RNA, Viral/genetics , Ribonucleoproteins/genetics , Ribonucleoproteins/ultrastructure , Viral Proteins/genetics , Viral Proteins/ultrastructure , Virus Replication/genetics
8.
Uirusu ; 70(1): 91-100, 2020.
Article in Japanese | MEDLINE | ID: mdl-33967118

ABSTRACT

Negative-strand RNA viruses do not possess a rigid viral shell, and their structures are flexible and fragile. We have applied various electron microscopies to analyze the morphologies of influenza and Ebola virus. Our studies have revealed the native interior and exterior ultrastructures of influenza virus as well as the assembly of Ebola virus core in atomic detail.


Subject(s)
Ebolavirus , Influenza, Human , Orthomyxoviridae , RNA Viruses , Ebolavirus/genetics , Humans , Nucleocapsid , RNA Viruses/genetics , RNA, Viral/genetics
9.
Nature ; 563(7729): 137-140, 2018 11.
Article in English | MEDLINE | ID: mdl-30333622

ABSTRACT

Ebola virus causes haemorrhagic fever with a high fatality rate in humans and non-human primates. It belongs to the family Filoviridae in the order Mononegavirales, which are viruses that contain linear, non-segmented, negative-sense, single-stranded genomic RNA1,2. The enveloped, filamentous virion contains the nucleocapsid, consisting of the helical nucleoprotein-RNA complex, VP24, VP30, VP35 and viral polymerase1,3. The nucleoprotein-RNA complex acts as a scaffold for nucleocapsid formation and as a template for RNA replication and transcription by condensing RNA into the virion4,5. RNA binding and nucleoprotein oligomerization are synergistic and do not readily occur independently6. Although recent cryo-electron tomography studies have revealed the overall architecture of the nucleocapsid core4,5, there has been no high-resolution reconstruction of the nucleocapsid. Here we report the structure of a recombinant Ebola virus nucleoprotein-RNA complex expressed in mammalian cells without chemical fixation, at near-atomic resolution using single-particle cryo-electron microscopy. Our structure reveals how the Ebola virus nucleocapsid core encapsidates its viral genome, its sequence-independent coordination with RNA by nucleoprotein, and the dynamic transition between the RNA-free and RNA-bound states. It provides direct structural evidence for the role of the N terminus of nucleoprotein in subunit oligomerization, and for the hydrophobic and electrostatic interactions that lead to the formation of the helical assembly. The structure is validated as representative of the native biological assembly of the nucleocapsid core by consistent dimensions and symmetry with the full virion5. The atomic model provides a detailed mechanistic basis for understanding nucleocapsid assembly and highlights key structural features that may serve as targets for anti-viral drug development.


Subject(s)
Cryoelectron Microscopy , Ebolavirus/chemistry , Ebolavirus/ultrastructure , Nucleocapsid/chemistry , RNA, Viral/chemistry , HEK293 Cells , Humans , Models, Molecular , Nucleocapsid/ultrastructure , RNA, Viral/ultrastructure
10.
J Infect Dis ; 217(9): 1372-1382, 2018 04 11.
Article in English | MEDLINE | ID: mdl-29373693

ABSTRACT

Exosomes regulate cell-cell communication by transferring functional proteins and RNAs between cells. Here, to clarify the function of exosomes during influenza virus infection, we characterized lung-derived exosomal microRNAs (miRNAs). Among the detected miRNAs, miR-483-3p was present at high levels in bronchoalveolar lavage fluid (BALF) exosomes during infection of mice with various strains of influenza virus, and miR-483-3p transfection potentiated gene expression of type I interferon and proinflammatory cytokine upon viral infection of MLE-12 cells. RNF5, a regulator of the RIG-I signaling pathway, was identified as a target gene of miR-483-3p. Moreover, we found that CD81, another miR-483-3p target, functions as a negative regulator of RIG-I signaling in MLE-12 cells. Taken together, this study indicates that BALF exosomal miRNAs may mediate the antiviral and inflammatory response to influenza virus infection.


Subject(s)
Immunity, Innate/physiology , MicroRNAs/metabolism , Orthomyxoviridae Infections/immunology , Orthomyxoviridae/immunology , Animals , Bronchoalveolar Lavage Fluid , Cell Line , Female , Gene Expression Regulation/immunology , Lung/metabolism , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , MicroRNAs/genetics , NF-kappa B , Orthomyxoviridae Infections/virology , Tetraspanin 28/genetics , Tetraspanin 28/metabolism , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism
11.
Mol Cell ; 69(3): 385-397.e8, 2018 02 01.
Article in English | MEDLINE | ID: mdl-29336876

ABSTRACT

Heterochromatin plays important roles in transcriptional silencing and genome maintenance by the formation of condensed chromatin structures, which determine the epigenetic status of eukaryotic cells. The trimethylation of histone H3 lysine 9 (H3K9me3), a target of heterochromatin protein 1 (HP1), is a hallmark of heterochromatin formation. However, the mechanism by which HP1 folds chromatin-containing H3K9me3 into a higher-order structure has not been elucidated. Here we report the three-dimensional structure of the H3K9me3-containing dinucleosomes complexed with human HP1α, HP1ß, and HP1γ, determined by cryogenic electron microscopy with a Volta phase plate. In the structures, two H3K9me3 nucleosomes are bridged by a symmetric HP1 dimer. Surprisingly, the linker DNA between the nucleosomes does not directly interact with HP1, thus allowing nucleosome remodeling by the ATP-utilizing chromatin assembly and remodeling factor (ACF). The structure depicts the fundamental architecture of heterochromatin.


Subject(s)
Chromosomal Proteins, Non-Histone/metabolism , Heterochromatin/metabolism , Chromatin/metabolism , Chromatin Assembly and Disassembly/genetics , Chromobox Protein Homolog 5 , Chromosomal Proteins, Non-Histone/genetics , Cryoelectron Microscopy/methods , DNA/metabolism , Histones/metabolism , Humans , Jumonji Domain-Containing Histone Demethylases/metabolism , Methylation , Nucleosomes/metabolism , Protein Binding , Structure-Activity Relationship , Transcription Factors/metabolism
12.
J Vet Med Sci ; 78(1): 61-70, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26400674

ABSTRACT

In our previous study, we genetically analyzed bovine viral diarrhea viruses (BVDVs) isolated from 2000 to 2006 in Japan and reported that subgenotype 1b viruses were predominant. In the present study, 766 BVDVs isolated from 2006 to 2014 in Hokkaido, Japan, were genetically analyzed to understand recent epidemics. Phylogenetic analysis based on nucleotide sequences of the 5'-untranslated region of viral genome revealed that 766 isolates were classified as genotype 1 (BVDV-1; 544 isolates) and genotype 2 (BVDV-2; 222). BVDV-1 isolates were further divided into BVDV-1a (93), 1b (371) and 1c (80) subgenotypes, and all BVDV-2 isolates were grouped into BVDV-2a subgenotype (222). Further comparative analysis was performed with BVDV-1a, 1b and 2a viruses isolated from 2001 to 2014. Phylogenetic analysis based on nucleotide sequences of the viral glycoprotein E2 gene, a major target of neutralizing antibodies, revealed that BVDV-1a, 1b and 2a isolates were further classified into several clusters. Cross-neutralization tests showed that BVDV-1b isolates were antigenically different from BVDV-1a isolates, and almost BVDV-1a, 1b and 2a isolates were antigenically similar among each subgenotype and each E2 cluster. Taken together, BVDV-1b viruses are still predominant, and BVDV-2a viruses have increased recently in Hokkaido, Japan. Field isolates of BVDV-1a, 1b and 2a show genetic diversity on the E2 gene with antigenic conservation among each subgenotype during the last 14 years.


Subject(s)
Bovine Virus Diarrhea-Mucosal Disease/virology , Diarrhea Viruses, Bovine Viral/genetics , Animals , Bovine Virus Diarrhea-Mucosal Disease/immunology , Cattle/virology , Diarrhea Viruses, Bovine Viral/immunology , Genome, Viral/genetics , Genotype , Japan/epidemiology , Neutralization Tests/veterinary , Phylogeny
13.
J Virol ; 89(20): 10524-31, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26269174

ABSTRACT

UNLABELLED: In measles virus (MV)-infected cells, the ribonucleoprotein (RNP) complex, comprised of the viral genome and the nucleocapsid (N) protein, phosphoprotein (P protein), and large protein, assembles at the perinuclear region and synthesizes viral RNAs. The cellular proteins involved in the formation of the RNP complex are largely unknown. In this report, we show that cofilin, an actin-modulating host protein, interacts with the MV N protein and aids in the formation of the RNP complex. Knockdown of cofilin using the short hairpin RNA reduces the formation of the RNP complex after MV infection and that of the RNP complex-like structure after plasmid-mediated expression of MV N and P proteins. A lower level of formation of the RNP complex results in the reduction of viral RNA synthesis. Cofilin phosphorylation on the serine residue at position 3, an enzymatically inactive form, is increased after MV infection and the phosphorylated form of cofilin is preferentially included in the complex. These results indicate that cofilin plays an important role in MV replication by increasing formation of the RNP complex and viral RNA synthesis. IMPORTANCE: Many RNA viruses induce within infected cells the structure called the ribonucleoprotein (RNP) complex in which viral RNA synthesis occurs. It is comprised of the viral genome and proteins that include the viral RNA polymerase. The cellular proteins involved in the formation of the RNP complex are largely unknown. In this report, we show that cofilin, an actin-modulating host protein, binds to the measles virus (MV) nucleocapsid protein and plays an important role in the formation of the MV RNP complex and MV RNA synthesis. The level of the phosphorylated form of cofilin, enzymatically inactive, is increased after MV infection, and the phosphorylated form is preferentially associated with the RNP complex. Our findings determined with cofilin will help us better understand the mechanism by which the RNP complex is formed in virus-infected cells and develop new antiviral drugs targeting the RNP complex.


Subject(s)
Cofilin 1/genetics , Cytoskeletal Proteins/genetics , Genome, Viral , Measles virus/genetics , Nucleoproteins/genetics , RNA, Viral/biosynthesis , Viral Proteins/genetics , Actins/genetics , Actins/metabolism , Animals , Cell Nucleus/chemistry , Cell Nucleus/metabolism , Cell Nucleus/virology , Chlorocebus aethiops , Cofilin 1/antagonists & inhibitors , Cofilin 1/metabolism , Cytoskeletal Proteins/antagonists & inhibitors , Cytoskeletal Proteins/metabolism , Gene Expression Regulation , HEK293 Cells , HeLa Cells , Host-Pathogen Interactions , Humans , Measles virus/metabolism , Nucleocapsid Proteins/genetics , Nucleocapsid Proteins/metabolism , Nucleoproteins/chemistry , Nucleoproteins/metabolism , Phosphoproteins/genetics , Phosphoproteins/metabolism , Phosphorylation , Protein Binding , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , RNA, Viral/genetics , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Signal Transduction , Vero Cells , Viral Proteins/chemistry , Viral Proteins/metabolism , Virus Replication/genetics
14.
Cell Host Microbe ; 16(6): 795-805, 2014 Dec 10.
Article in English | MEDLINE | ID: mdl-25464832

ABSTRACT

Host factors required for viral replication are ideal drug targets because they are less likely than viral proteins to mutate under drug-mediated selective pressure. Although genome-wide screens have identified host proteins involved in influenza virus replication, limited mechanistic understanding of how these factors affect influenza has hindered potential drug development. We conducted a systematic analysis to identify and validate host factors that associate with influenza virus proteins and affect viral replication. After identifying over 1,000 host factors that coimmunoprecipitate with specific viral proteins, we generated a network of virus-host protein interactions based on the stage of the viral life cycle affected upon host factor downregulation. Using compounds that inhibit these host factors, we validated several proteins, notably Golgi-specific brefeldin A-resistant guanine nucleotide exchange factor 1 (GBF1) and JAK1, as potential antiviral drug targets. Thus, virus-host interactome screens are powerful strategies to identify targetable host factors and guide antiviral drug development.


Subject(s)
Antiviral Agents/pharmacology , Influenza, Human/metabolism , Orthomyxoviridae/drug effects , Orthomyxoviridae/metabolism , Protein Interaction Mapping/methods , Protein Interaction Maps/drug effects , Viral Proteins/metabolism , Drug Evaluation, Preclinical , Guanine Nucleotide Exchange Factors/antagonists & inhibitors , Guanine Nucleotide Exchange Factors/genetics , Guanine Nucleotide Exchange Factors/metabolism , Host-Pathogen Interactions/drug effects , Humans , Influenza, Human/drug therapy , Influenza, Human/genetics , Influenza, Human/virology , Janus Kinase 1/antagonists & inhibitors , Janus Kinase 1/genetics , Janus Kinase 1/metabolism , Orthomyxoviridae/genetics , Protein Binding/drug effects , Viral Proteins/genetics
15.
J Virol ; 87(23): 12879-84, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24067952

ABSTRACT

The influenza A virus possesses an eight-segmented, negative-sense, single-stranded RNA genome (vRNA). Each vRNA segment binds to multiple copies of viral nucleoproteins and a small number of heterotrimeric polymerase complexes to form a rod-like ribonucleoprotein complex (RNP), which is essential for the transcription and replication of the vRNAs. However, how the RNPs are organized within the progeny virion is not fully understood. Here, by focusing on polymerase complexes, we analyzed the fine structure of purified RNPs and their configuration within virions by using various electron microscopies (EM). We confirmed that the individual RNPs possess a single polymerase complex at one end of the rod-like structure and that, as determined using immune EM, some RNPs are incorporated into budding virions with their polymerase-binding ends at the budding tip, whereas others align with their polymerase-binding ends at the bottom of the virion. These data further our understanding of influenza virus virion morphogenesis.


Subject(s)
DNA-Directed RNA Polymerases/metabolism , Influenza A Virus, H1N1 Subtype/metabolism , Ribonucleoproteins/metabolism , Viral Proteins/metabolism , Virion/metabolism , Animals , Chick Embryo , DNA-Directed RNA Polymerases/genetics , Humans , Influenza A Virus, H1N1 Subtype/chemistry , Influenza A Virus, H1N1 Subtype/genetics , Influenza A Virus, H1N1 Subtype/ultrastructure , Influenza, Human/virology , Microscopy, Electron , Ribonucleoproteins/chemistry , Ribonucleoproteins/genetics , Viral Proteins/chemistry , Viral Proteins/genetics , Virion/chemistry , Virion/genetics , Virion/ultrastructure
16.
Nature ; 501(7468): 551-5, 2013 Sep 26.
Article in English | MEDLINE | ID: mdl-23842494

ABSTRACT

Avian influenza A viruses rarely infect humans; however, when human infection and subsequent human-to-human transmission occurs, worldwide outbreaks (pandemics) can result. The recent sporadic infections of humans in China with a previously unrecognized avian influenza A virus of the H7N9 subtype (A(H7N9)) have caused concern owing to the appreciable case fatality rate associated with these infections (more than 25%), potential instances of human-to-human transmission, and the lack of pre-existing immunity among humans to viruses of this subtype. Here we characterize two early human A(H7N9) isolates, A/Anhui/1/2013 (H7N9) and A/Shanghai/1/2013 (H7N9); hereafter referred to as Anhui/1 and Shanghai/1, respectively. In mice, Anhui/1 and Shanghai/1 were more pathogenic than a control avian H7N9 virus (A/duck/Gunma/466/2011 (H7N9); Dk/GM466) and a representative pandemic 2009 H1N1 virus (A/California/4/2009 (H1N1pdm09); CA04). Anhui/1, Shanghai/1 and Dk/GM466 replicated well in the nasal turbinates of ferrets. In nonhuman primates, Anhui/1 and Dk/GM466 replicated efficiently in the upper and lower respiratory tracts, whereas the replicative ability of conventional human influenza viruses is typically restricted to the upper respiratory tract of infected primates. By contrast, Anhui/1 did not replicate well in miniature pigs after intranasal inoculation. Critically, Anhui/1 transmitted through respiratory droplets in one of three pairs of ferrets. Glycan arrays showed that Anhui/1, Shanghai/1 and A/Hangzhou/1/2013 (H7N9) (a third human A(H7N9) virus tested in this assay) bind to human virus-type receptors, a property that may be critical for virus transmissibility in ferrets. Anhui/1 was found to be less sensitive in mice to neuraminidase inhibitors than a pandemic H1N1 2009 virus, although both viruses were equally susceptible to an experimental antiviral polymerase inhibitor. The robust replicative ability in mice, ferrets and nonhuman primates and the limited transmissibility in ferrets of Anhui/1 suggest that A(H7N9) viruses have pandemic potential.


Subject(s)
Influenza A virus , Influenza, Human/virology , Orthomyxoviridae Infections/virology , Virus Replication , Animals , Antiviral Agents/pharmacology , Cells, Cultured , Chickens/virology , DNA-Directed RNA Polymerases/antagonists & inhibitors , Dogs , Enzyme Inhibitors/pharmacology , Female , Ferrets/virology , Humans , Influenza A Virus, H1N1 Subtype/drug effects , Influenza A Virus, H1N1 Subtype/enzymology , Influenza A virus/chemistry , Influenza A virus/drug effects , Influenza A virus/isolation & purification , Influenza A virus/pathogenicity , Influenza, Human/drug therapy , Macaca fascicularis/virology , Madin Darby Canine Kidney Cells , Male , Mice , Mice, Inbred BALB C , Models, Molecular , Monkey Diseases/pathology , Monkey Diseases/virology , Neuraminidase/antagonists & inhibitors , Orthomyxoviridae Infections/pathology , Orthomyxoviridae Infections/transmission , Quail/virology , Swine/virology , Swine, Miniature/virology , Virus Replication/drug effects
17.
Nat Commun ; 3: 639, 2012 Jan 24.
Article in English | MEDLINE | ID: mdl-22273677

ABSTRACT

The influenza A virus genome consists of eight single-stranded negative-sense RNA (vRNA) segments. Although genome segmentation provides advantages such as genetic reassortment, which contributes to the emergence of novel strains with pandemic potential, it complicates the genome packaging of progeny virions. Here we elucidate, using electron tomography, the three-dimensional structure of ribonucleoprotein complexes (RNPs) within progeny virions. Each virion is packed with eight well-organized RNPs that possess rod-like structures of different lengths. Multiple interactions are found among the RNPs. The position of the eight RNPs is not consistent among virions, but a pattern suggests the existence of a specific mechanism for assembly of these RNPs. Analyses of budding progeny virions suggest two independent roles for the viral spike proteins: RNP association on the plasma membrane and the subsequent formation of the virion shell. Our data provide further insights into the mechanisms responsible for segmented-genome packaging into virions.


Subject(s)
Influenza A virus/metabolism , Ribonucleoproteins/chemistry , Animals , Cell Line , Dogs , Genome , Image Processing, Computer-Assisted , Imaging, Three-Dimensional , Influenza A virus/genetics , Models, Molecular , Molecular Conformation , Probability , RNA, Viral/genetics , RNA, Viral/metabolism , Ribonucleoproteins/metabolism , Viral Proteins/metabolism , Virion/genetics , Virion/metabolism , Virus Assembly
18.
J Gen Virol ; 92(Pt 11): 2485-2493, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21795472

ABSTRACT

Negatively stained influenza virions sometimes show irregular morphology and are often referred to as pleomorphic. However, this irregular morphology has not been visualized when ultrathin-section transmission and scanning electron microscopies are used. This study focused on the effects of ultracentrifugation on influenza A virion morphology, as negative staining often involves ultracentrifugation to concentrate or purify virions. The morphologies of unfixed, glutaraldehyde-fixed and osmium tetroxide-fixed virions were quantitatively compared before and after ultracentrifugation, and it was found that, without chemical fixation, approximately 30% of virions were altered from oval to irregular shapes following ultracentrifugation. By contrast, most glutaraldehyde-fixed virions remained uniformly elliptical, even after ultracentrifugation. When a virus with an 11 aa deletion at the C terminus of its M2 cytoplasmic tail was ultracentrifuged, its morphology was appreciably deformed compared with that of the wild-type virus. These results demonstrate that the native morphology of influenza A virions is regular but is disrupted by ultracentrifugation, and that the cytoplasmic tail of M2 is important for virion integrity.


Subject(s)
Influenza A virus/ultrastructure , Ultracentrifugation , Virion/ultrastructure , Animals , Chick Embryo , Fixatives/metabolism , Glutaral/metabolism , Influenza A virus/isolation & purification , Microscopy, Electron , Osmium Tetroxide/metabolism , Virion/isolation & purification
SELECTION OF CITATIONS
SEARCH DETAIL
...