Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 32
Filter
Add more filters











Publication year range
1.
Cell ; 187(20): 5587-5603.e19, 2024 Oct 03.
Article in English | MEDLINE | ID: mdl-39293445

ABSTRACT

Filoviruses, including the Ebola and Marburg viruses, cause hemorrhagic fevers with up to 90% lethality. The viral nucleocapsid is assembled by polymerization of the nucleoprotein (NP) along the viral genome, together with the viral proteins VP24 and VP35. We employed cryo-electron tomography of cells transfected with viral proteins and infected with model Ebola virus to illuminate assembly intermediates, as well as a 9 Å map of the complete intracellular assembly. This structure reveals a previously unresolved third and outer layer of NP complexed with VP35. The intrinsically disordered region, together with the C-terminal domain of this outer layer of NP, provides the constant width between intracellular nucleocapsid bundles and likely functions as a flexible tether to the viral matrix protein in the virion. A comparison of intracellular nucleocapsids with prior in-virion nucleocapsid structures reveals that the nucleocapsid further condenses vertically in the virion. The interfaces responsible for nucleocapsid assembly are highly conserved and offer targets for broadly effective antivirals.


Subject(s)
Ebolavirus , Electron Microscope Tomography , Nucleocapsid , Virus Assembly , Ebolavirus/ultrastructure , Ebolavirus/chemistry , Ebolavirus/metabolism , Ebolavirus/physiology , Nucleocapsid/metabolism , Nucleocapsid/ultrastructure , Nucleocapsid/chemistry , Humans , Cryoelectron Microscopy/methods , Nucleocapsid Proteins/chemistry , Nucleocapsid Proteins/metabolism , Nucleocapsid Proteins/ultrastructure , Nucleoproteins/chemistry , Nucleoproteins/metabolism , Nucleoproteins/ultrastructure , Animals , Viral Proteins/metabolism , Viral Proteins/chemistry , Viral Proteins/ultrastructure , Models, Molecular , Virion/ultrastructure , Virion/metabolism , Hemorrhagic Fever, Ebola/virology , Chlorocebus aethiops
2.
Sci Rep ; 14(1): 14099, 2024 06 18.
Article in English | MEDLINE | ID: mdl-38890308

ABSTRACT

We report the first cryoEM structure of the Hendra henipavirus nucleoprotein in complex with RNA, at 3.5 Å resolution, derived from single particle analysis of a double homotetradecameric RNA-bound N protein ring assembly exhibiting D14 symmetry. The structure of the HeV N protein adopts the common bi-lobed paramyxoviral N protein fold; the N-terminal and C-terminal globular domains are bisected by an RNA binding cleft containing six RNA nucleotides and are flanked by the N-terminal and C-terminal arms, respectively. In common with other paramyxoviral nucleocapsids, the lateral interface between adjacent Ni and Ni+1 protomers involves electrostatic and hydrophobic interactions mediated primarily through the N-terminal arm and globular domains with minor contribution from the C-terminal arm. However, the HeV N multimeric assembly uniquely identifies an additional protomer-protomer contact between the Ni+1 N-terminus and Ni-1 C-terminal arm linker. The model presented here broadens the understanding of RNA-bound paramyxoviral nucleocapsid architectures and provides a platform for further insight into the molecular biology of HeV, as well as the development of antiviral interventions.


Subject(s)
Cryoelectron Microscopy , Hendra Virus , Nucleocapsid , Nucleoproteins , Hendra Virus/chemistry , Nucleoproteins/chemistry , Nucleoproteins/ultrastructure , Nucleoproteins/metabolism , Nucleocapsid/chemistry , Nucleocapsid/ultrastructure , Nucleocapsid/metabolism , Models, Molecular , RNA, Viral/chemistry , RNA, Viral/metabolism , RNA, Viral/genetics , Nucleocapsid Proteins/chemistry , Nucleocapsid Proteins/ultrastructure , Nucleocapsid Proteins/metabolism
3.
J Biol Chem ; 299(12): 105362, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37863261

ABSTRACT

The nucleocapsid (N) protein of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) compacts the RNA genome into viral ribonucleoprotein (vRNP) complexes within virions. Assembly of vRNPs is inhibited by phosphorylation of the N protein serine/arginine (SR) region. Several SARS-CoV-2 variants of concern carry N protein mutations that reduce phosphorylation and enhance the efficiency of viral packaging. Variants of the dominant B.1.1 viral lineage also encode a truncated N protein, termed N∗ or Δ(1-209), that mediates genome packaging despite lacking the N-terminal RNA-binding domain and SR region. Here, we use mass photometry and negative stain electron microscopy to show that purified Δ(1-209) and viral RNA assemble into vRNPs that are remarkably similar in size and shape to those formed with full-length N protein. We show that assembly of Δ(1-209) vRNPs requires the leucine-rich helix of the central disordered region and that this helix promotes N protein oligomerization. We also find that fusion of a phosphomimetic SR region to Δ(1-209) inhibits RNA binding and vRNP assembly. Our results provide new insights into the mechanisms by which RNA binding promotes N protein self-association and vRNP assembly, and how this process is modulated by phosphorylation.


Subject(s)
Nucleocapsid Proteins , SARS-CoV-2 , Humans , COVID-19/virology , Nucleocapsid Proteins/genetics , Nucleocapsid Proteins/metabolism , Nucleocapsid Proteins/ultrastructure , RNA, Viral/metabolism , RNA, Viral/ultrastructure , SARS-CoV-2/genetics , SARS-CoV-2/metabolism , SARS-CoV-2/ultrastructure , Phosphorylation , Virus Assembly/genetics
4.
PLoS Pathog ; 17(7): e1009740, 2021 07.
Article in English | MEDLINE | ID: mdl-34270629

ABSTRACT

Nipah and its close relative Hendra are highly pathogenic zoonotic viruses, storing their ssRNA genome in a helical nucleocapsid assembly formed by the N protein, a major viral immunogen. Here, we report the first cryoEM structure for a Henipavirus RNA-bound nucleocapsid assembly, at 3.5 Å resolution. The helical assembly is stabilised by previously undefined N- and C-terminal segments, contributing to subunit-subunit interactions. RNA is wrapped around the nucleocapsid protein assembly with a periodicity of six nucleotides per protomer, in the "3-bases-in, 3-bases-out" conformation, with protein plasticity enabling non-sequence specific interactions. The structure reveals commonalities in RNA binding pockets and in the conformation of bound RNA, not only with members of the Paramyxoviridae family, but also with the evolutionarily distant Filoviridae Ebola virus. Significant structural differences with other Paramyxoviridae members are also observed, particularly in the position and length of the exposed α-helix, residues 123-139, which may serve as a valuable epitope for surveillance and diagnostics.


Subject(s)
Nipah Virus/ultrastructure , Nucleocapsid Proteins/ultrastructure , Nucleocapsid/ultrastructure , Cryoelectron Microscopy , Models, Molecular , Molecular Conformation , Nipah Virus/chemistry , Nucleocapsid/chemistry , Nucleocapsid Proteins/chemistry , RNA, Viral/chemistry , RNA, Viral/ultrastructure
5.
Brief Bioinform ; 22(6)2021 11 05.
Article in English | MEDLINE | ID: mdl-34143202

ABSTRACT

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), a causative agent of the coronavirus disease (COVID-19), is a part of the $\beta $-Coronaviridae family. The virus contains five major protein classes viz., four structural proteins [nucleocapsid (N), membrane (M), envelop (E) and spike glycoprotein (S)] and replicase polyproteins (R), synthesized as two polyproteins (ORF1a and ORF1ab). Due to the severity of the pandemic, most of the SARS-CoV-2-related research are focused on finding therapeutic solutions. However, studies on the sequences and structure space throughout the evolutionary time frame of viral proteins are limited. Besides, the structural malleability of viral proteins can be directly or indirectly associated with the dysfunctionality of the host cell proteins. This dysfunctionality may lead to comorbidities during the infection and may continue at the post-infection stage. In this regard, we conduct the evolutionary sequence-structure analysis of the viral proteins to evaluate their malleability. Subsequently, intrinsic disorder propensities of these viral proteins have been studied to confirm that the short intrinsically disordered regions play an important role in enhancing the likelihood of the host proteins interacting with the viral proteins. These interactions may result in molecular dysfunctionality, finally leading to different diseases. Based on the host cell proteins, the diseases are divided in two distinct classes: (i) proteins, directly associated with the set of diseases while showing similar activities, and (ii) cytokine storm-mediated pro-inflammation (e.g. acute respiratory distress syndrome, malignancies) and neuroinflammation (e.g. neurodegenerative and neuropsychiatric diseases). Finally, the study unveils that males and postmenopausal females can be more vulnerable to SARS-CoV-2 infection due to the androgen-mediated protein transmembrane serine protease 2.


Subject(s)
COVID-19/genetics , Genome, Viral/genetics , Protein Conformation , SARS-CoV-2/ultrastructure , COVID-19/virology , Coronavirus Envelope Proteins/genetics , Coronavirus Envelope Proteins/ultrastructure , Humans , Membrane Proteins/genetics , Membrane Proteins/ultrastructure , Nucleocapsid Proteins/genetics , Nucleocapsid Proteins/ultrastructure , SARS-CoV-2/genetics , SARS-CoV-2/pathogenicity , Spike Glycoprotein, Coronavirus/genetics , Spike Glycoprotein, Coronavirus/ultrastructure , Viral Replicase Complex Proteins/genetics , Viral Replicase Complex Proteins/ultrastructure , Viral Structural Proteins/genetics , Viral Structural Proteins/ultrastructure
6.
Viruses ; 12(9)2020 08 29.
Article in English | MEDLINE | ID: mdl-32872471

ABSTRACT

Rhabdoviruses, as single-stranded, negative-sense RNA viruses within the order Mononegavirales, are characterised by bullet-shaped or bacteroid particles that contain a helical ribonucleoprotein complex (RNP). Here, we review the components of the RNP and its higher-order structural assembly.


Subject(s)
Rhabdoviridae/chemistry , Ribonucleoproteins/chemistry , Viral Proteins/chemistry , Nucleocapsid Proteins/chemistry , Nucleocapsid Proteins/ultrastructure , Protein Conformation , Rhabdoviridae/genetics , Ribonucleoproteins/ultrastructure , Viral Matrix Proteins/chemistry , Viral Matrix Proteins/ultrastructure , Viral Proteins/ultrastructure , Viral Replicase Complex Proteins/chemistry , Viral Replicase Complex Proteins/ultrastructure , Virion/chemistry
7.
Virology ; 527: 159-168, 2019 01 15.
Article in English | MEDLINE | ID: mdl-30529564

ABSTRACT

Viral genomes are protected and organized by virally encoded packaging proteins. Heterologous production of these proteins often results in formation of particles resembling the authentic viral capsid or nucleocapsid, with cellular nucleic acids packaged in place of the viral genome. Quantifying the total protein and nucleic acid content of particle preparations is a recurrent biochemical problem. We describe a method for resolving this problem, developed when characterizing particles resembling the Menangle Virus nucleocapsid. The protein content was quantified using the biuret assay, which is largely independent of amino acid composition. Bound nucleic acids were quantified by determining the phosphorus content, using inductively coupled plasma mass spectrometry. Estimates for the amount of RNA packaged within the particles were consistent with the structurally-characterized packaging mechanism. For a bacterially-produced nucleoprotein complex, phosphorus usually provides a unique elemental marker of bound nucleic acids, hence this method of analysis should be routinely applicable.


Subject(s)
Chemistry Techniques, Analytical/methods , Nucleocapsid Proteins/analysis , Paramyxoviridae/chemistry , Biuret Reaction , Escherichia coli/genetics , Escherichia coli/metabolism , Mass Spectrometry , Nucleic Acids/analysis , Nucleic Acids/metabolism , Nucleocapsid Proteins/isolation & purification , Nucleocapsid Proteins/metabolism , Nucleocapsid Proteins/ultrastructure , Paramyxoviridae/genetics , Paramyxoviridae/metabolism , Paramyxoviridae/ultrastructure , Phosphorus/analysis , Phosphorylation , Protein Binding , Recombinant Proteins/analysis , Recombinant Proteins/isolation & purification , Recombinant Proteins/metabolism , Recombinant Proteins/ultrastructure
8.
Nature ; 551(7680): 394-397, 2017 11 16.
Article in English | MEDLINE | ID: mdl-29144446

ABSTRACT

Ebola and Marburg viruses are filoviruses: filamentous, enveloped viruses that cause haemorrhagic fever. Filoviruses are within the order Mononegavirales, which also includes rabies virus, measles virus, and respiratory syncytial virus. Mononegaviruses have non-segmented, single-stranded negative-sense RNA genomes that are encapsidated by nucleoprotein and other viral proteins to form a helical nucleocapsid. The nucleocapsid acts as a scaffold for virus assembly and as a template for genome transcription and replication. Insights into nucleoprotein-nucleoprotein interactions have been derived from structural studies of oligomerized, RNA-encapsidating nucleoprotein, and cryo-electron microscopy of nucleocapsid or nucleocapsid-like structures. There have been no high-resolution reconstructions of complete mononegavirus nucleocapsids. Here we apply cryo-electron tomography and subtomogram averaging to determine the structure of Ebola virus nucleocapsid within intact viruses and recombinant nucleocapsid-like assemblies. These structures reveal the identity and arrangement of the nucleocapsid components, and suggest that the formation of an extended α-helix from the disordered carboxy-terminal region of nucleoprotein-core links nucleoprotein oligomerization, nucleocapsid condensation, RNA encapsidation, and accessory protein recruitment.


Subject(s)
Cryoelectron Microscopy , Ebolavirus/chemistry , Ebolavirus/ultrastructure , Electron Microscope Tomography , Nucleocapsid Proteins/ultrastructure , Nucleocapsid/chemistry , Nucleocapsid/ultrastructure , Animals , Chlorocebus aethiops , HEK293 Cells , Humans , Marburgvirus/chemistry , Models, Molecular , Molecular Conformation , Nucleocapsid Proteins/chemistry , RNA, Viral/chemistry , RNA, Viral/ultrastructure , Vero Cells
9.
Acta Crystallogr D Struct Biol ; 73(Pt 8): 650-659, 2017 Aug 01.
Article in English | MEDLINE | ID: mdl-28777080

ABSTRACT

Toscana virus (TOSV) is an arthropod-borne virus belonging to the Phlebovirus genus within the Bunyaviridae family. As in other bunyaviruses, the genome of TOSV is made up of three RNA segments. They are encapsidated by the nucleoprotein (N), which also plays an essential role in virus replication. To date, crystallographic structures of phlebovirus N have systematically revealed closed-ring organizations which do not fully match the filamentous organization of the ribonucleoprotein (RNP) complex observed by electron microscopy. In order to further bridge the gap between crystallographic data on N and observations of the RNP by electron microscopy, the structural organization of recombinant TOSV N was investigated by an integrative approach combining X-ray diffraction crystallography, transmission electron microscopy, small-angle X-ray scattering, size-exclusion chromatography and multi-angle laser light scattering. It was found that in solution TOSV N forms open oligomers consistent with the encapsidation mechanism of phlebovirus RNA.


Subject(s)
Nucleocapsid Proteins/chemistry , Nucleoproteins/chemistry , Sandfly fever Naples virus/chemistry , Bunyaviridae Infections/virology , Crystallography, X-Ray , Models, Molecular , Nucleocapsid Proteins/metabolism , Nucleocapsid Proteins/ultrastructure , Nucleoproteins/metabolism , Nucleoproteins/ultrastructure , Protein Conformation , Protein Multimerization , RNA, Viral/metabolism , Sandfly fever Naples virus/metabolism , Scattering, Small Angle , Solutions , X-Ray Diffraction
10.
J Virol ; 90(2): 1048-61, 2016 01 15.
Article in English | MEDLINE | ID: mdl-26559827

ABSTRACT

UNLABELLED: Hantaviruses, which belong to the genus Hantavirus in the family Bunyaviridae, infect mammals, including humans, causing either hemorrhagic fever with renal syndrome (HFRS) or hantavirus cardiopulmonary syndrome (HCPS) in humans with high mortality. Hantavirus encodes a nucleocapsid protein (NP) to encapsidate the genome and form a ribonucleoprotein complex (RNP) together with viral polymerase. Here, we report the crystal structure of the core domains of NP (NPcore) encoded by Sin Nombre virus (SNV) and Andes virus (ANDV), which are two representative members that cause HCPS in the New World. The constructs of SNV and ANDV NPcore exclude the N- and C-terminal portions of full polypeptide to obtain stable proteins for crystallographic study. The structure features an N lobe and a C lobe to clamp RNA-binding crevice and exhibits two protruding extensions in both lobes. The positively charged residues located in the RNA-binding crevice play a key role in RNA binding and virus replication. We further demonstrated that the C-terminal helix and the linker region connecting the N-terminal coiled-coil domain and NPcore are essential for hantavirus NP oligomerization through contacts made with two adjacent protomers. Moreover, electron microscopy (EM) visualization of native RNPs extracted from the virions revealed that a monomer-sized NP-RNA complex is the building block of viral RNP. This work provides insight into the formation of hantavirus RNP and provides an understanding of the evolutionary connections that exist among bunyaviruses. IMPORTANCE: Hantaviruses are distributed across a wide and increasing range of host reservoirs throughout the world. In particular, hantaviruses can be transmitted via aerosols of rodent excreta to humans or from human to human and cause HFRS and HCPS, with mortalities of 15% and 50%, respectively. Hantavirus is therefore listed as a category C pathogen. Hantavirus encodes an NP that plays essential roles both in RNP formation and in multiple biological functions. NP is also the exclusive target for the serological diagnoses. This work reveals the structure of hantavirus NP, furthering the knowledge of hantavirus RNP formation, revealing the relationship between hantavirus NP and serological specificity and raising the potential for the development of new diagnosis and therapeutics targeting hantavirus infection.


Subject(s)
Nucleocapsid Proteins/chemistry , Orthohantavirus/chemistry , Ribonucleoproteins/chemistry , Sin Nombre virus/chemistry , Crystallography, X-Ray , Microscopy, Electron , Models, Molecular , Nucleocapsid Proteins/ultrastructure , Protein Conformation , Ribonucleoproteins/ultrastructure
11.
Protein Expr Purif ; 116: 19-29, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26196500

ABSTRACT

Hendra virus (family Paramyxoviridae) is a negative sense single-stranded RNA virus (NSRV) which has been found to cause disease in humans, horses, and experimentally in other animals, e.g. pigs and cats. Pteropid bats commonly known as flying foxes have been identified as the natural host reservoir. The Hendra virus nucleocapsid protein (HeV N) represents the most abundant viral protein produced by the host cell, and is highly immunogenic with naturally infected humans and horses producing specific antibodies towards this protein. The purpose of this study was to express and purify soluble, functionally active recombinant HeV N, suitable for use as an immunodiagnostic reagent to detect antibodies against HeV. We expressed both full-length HeV N, (HeV NFL), and a C-terminal truncated form, (HeV NCORE), using a bacterial heterologous expression system. Both HeV N constructs were engineered with an N-terminal Hisx6 tag, and purified using a combination of immobilized metal affinity chromatography (IMAC) and size exclusion chromatography (SEC). Purified recombinant HeV N proteins self-assembled into soluble higher order oligomers as determined by SEC and negative-stain transmission electron microscopy. Both HeV N proteins were highly immuno-reactive with sera from animals and humans infected with either HeV or the closely related Nipah virus (NiV), but displayed no immuno-reactivity towards sera from animals infected with a non-pathogenic paramyxovirus (CedPV), or animals receiving Equivac® (HeV G glycoprotein subunit vaccine), using a Luminex-based multiplexed microsphere assay.


Subject(s)
Hendra Virus/chemistry , Hendra Virus/immunology , Nucleocapsid Proteins/chemistry , Nucleocapsid Proteins/immunology , Amino Acid Sequence , Animals , Antibodies, Viral/immunology , Cloning, Molecular , Escherichia coli/genetics , Gene Expression , Hendra Virus/genetics , Hendra Virus/ultrastructure , Henipavirus Infections/immunology , Henipavirus Infections/virology , Horses , Humans , Molecular Sequence Data , Nucleocapsid Proteins/genetics , Nucleocapsid Proteins/ultrastructure , Plasmids/genetics , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/immunology , Recombinant Proteins/ultrastructure , Swine
12.
Biochim Biophys Acta ; 1854(3): 229-38, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25523747

ABSTRACT

Intrinsically disordered proteins contain some residual structures, which may fold further upon binding to the partner protein for function. The residual structures observed in two intrinsically disordered proteins, including the C-terminal segment of peripherin-2 (63 residues) and measles virus nucleocapsid protein Ntail (125 residues), were compared using NMR. Differences in the chemical shifts of alpha-, beta- and carbonyl carbons between the observed structure and calculated random coil revealed the existence of a helix and some possible beta-structures in both proteins. The intensity of signals in the C-terminal segment of peripherin-2 in NMR spectra was informative and locally low, particularly in the middle and N-terminal parts: this suggested the broadening of the signals caused by the formation of residual structures in those areas. Furthermore, the protection of exchange of amide protons was significantly observed at the N-terminus. Conversely, the intensities of signals for Ntail were random beyond the overall areas of protein, and indicated no characteristic pattern. Only a faint protection of amide-proton exchange in Ntail was observed in the C-terminus. It was concluded that Ntail was more intrinsically disordered than the C-terminal segment of peripherin-2. The combination of chemical shifts with the amide-proton exchanges and signal intensities was useful for the analyses of the remaining secondary structures. The beta-structure might be more detectable by the protection of amide-proton exchange than the helical structure, although the changes in chemical shifts were sensitive for the detection of elements of both secondary structures.


Subject(s)
Amino Acids/chemistry , Magnetic Resonance Spectroscopy/methods , Nucleocapsid Proteins/chemistry , Nucleocapsid Proteins/ultrastructure , Peripherins/chemistry , Peripherins/ultrastructure , Xenopus Proteins/chemistry , Xenopus Proteins/ultrastructure , Amino Acid Sequence , Crystallography , Molecular Sequence Data , Protein Structure, Secondary
13.
J Virol ; 88(24): 14148-60, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25275119

ABSTRACT

UNLABELLED: The hexameric lattice of an immature retroviral particle consists of Gag polyprotein, which is the precursor of all viral structural proteins. Lentiviral and alpharetroviral Gag proteins contain a peptide sequence called the spacer peptide (SP), which is localized between the capsid (CA) and nucleocapsid (NC) domains. SP plays a critical role in intermolecular interactions during the assembly of immature particles of several retroviruses. Published models of supramolecular structures of immature particles suggest that in lentiviruses and alpharetroviruses, SP adopts a rod-like six-helix bundle organization. In contrast, Mason-Pfizer monkey virus (M-PMV), a betaretrovirus that assembles in the cytoplasm, does not contain a distinct SP sequence, and the CA-NC connecting region is not organized into a clear rod-like structure. Nevertheless, the CA-NC junction comprises a sequence critical for assembly of immature M-PMV particles. In the present work, we characterized this region, called the SP-like domain, in detail. We provide biochemical data confirming the critical role of the M-PMV SP-like domain in immature particle assembly, release, processing, and infectivity. Circular dichroism spectroscopy revealed that, in contrast to the SP regions of other retroviruses, a short SP-like domain-derived peptide (SPLP) does not form a purely helical structure in aqueous or helix-promoting solution. Using 8-Å cryo-electron microscopy density maps of immature M-PMV particles, we prepared computational models of the SP-like domain and indicate the structural features required for M-PMV immature particle assembly. IMPORTANCE: Retroviruses such as HIV-1 are of great medical importance. Using Mason-Pfizer monkey virus (M-PMV) as a model retrovirus, we provide biochemical and structural data confirming the general relevance of a short segment of the structural polyprotein Gag for retrovirus assembly and infectivity. Although this segment is critical for assembly of immature particles of lentiviruses, alpharetroviruses, and betaretroviruses, the organization of this domain is strikingly different. A previously published electron microscopic structure of an immature M-PMV particle allowed us to model this important region into the electron density map. The data presented here help explain the different packing of the Gag segments of various retroviruses, such as HIV, Rous sarcoma virus (RSV), and M-PMV. Such knowledge contributes to understanding the importance of this region and its structural flexibility among retroviral species. The region might play a key role in Gag-Gag interactions, leading to different morphological pathways of immature particle assembly.


Subject(s)
Capsid Proteins/metabolism , Mason-Pfizer monkey virus/physiology , Nucleocapsid Proteins/metabolism , Virus Assembly , Capsid Proteins/chemistry , Capsid Proteins/genetics , Capsid Proteins/ultrastructure , Circular Dichroism , Cryoelectron Microscopy , Models, Molecular , Nucleocapsid Proteins/chemistry , Nucleocapsid Proteins/genetics , Nucleocapsid Proteins/ultrastructure , Protein Conformation , Virus Release
14.
RNA ; 19(8): 1129-36, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23798666

ABSTRACT

Schmallenberg virus (SBV) is a newly emerged orthobunyavirus (family Bunyaviridae) that has caused severe disease in the offspring of farm animals across Europe. Like all orthobunyaviruses, SBV contains a tripartite negative-sense RNA genome that is encapsidated by the viral nucleocapsid (N) protein in the form of a ribonucleoprotein complex (RNP). We recently reported the three-dimensional structure of SBV N that revealed a novel fold. Here we report the crystal structure of the SBV N protein in complex with a 42-nt-long RNA to 2.16 Å resolution. The complex comprises a tetramer of N that encapsidates the RNA as a cross-shape inside the protein ring structure, with each protomer bound to 11 ribonucleotides. Eight bases are bound in the positively charged cleft between the N- and C-terminal domains of N, and three bases are shielded by the extended N-terminal arm. SBV N appears to sequester RNA using a different mechanism compared with the nucleoproteins of other negative-sense RNA viruses. Furthermore, the structure suggests that RNA binding results in conformational changes of some residues in the RNA-binding cleft and the N- and C-terminal arms. Our results provide new insights into the novel mechanism of RNA encapsidation by orthobunyaviruses.


Subject(s)
Nucleocapsid Proteins/chemistry , Orthobunyavirus/chemistry , RNA, Viral/chemistry , Animals , Binding Sites , Crystallography, X-Ray , Macromolecular Substances/chemistry , Macromolecular Substances/ultrastructure , Microscopy, Electron , Models, Molecular , Nucleic Acid Conformation , Nucleocapsid Proteins/ultrastructure , Orthobunyavirus/pathogenicity , Orthobunyavirus/ultrastructure , Protein Structure, Quaternary , Static Electricity
15.
PLoS Pathog ; 7(5): e1002030, 2011 May.
Article in English | MEDLINE | ID: mdl-21589902

ABSTRACT

Rift Valley fever virus (RVFV), a Phlebovirus with a genome consisting of three single-stranded RNA segments, is spread by infected mosquitoes and causes large viral outbreaks in Africa. RVFV encodes a nucleoprotein (N) that encapsidates the viral RNA. The N protein is the major component of the ribonucleoprotein complex and is also required for genomic RNA replication and transcription by the viral polymerase. Here we present the 1.6 Å crystal structure of the RVFV N protein in hexameric form. The ring-shaped hexamers form a functional RNA binding site, as assessed by mutagenesis experiments. Electron microscopy (EM) demonstrates that N in complex with RNA also forms rings in solution, and a single-particle EM reconstruction of a hexameric N-RNA complex is consistent with the crystallographic N hexamers. The ring-like organization of the hexamers in the crystal is stabilized by circular interactions of the N terminus of RVFV N, which forms an extended arm that binds to a hydrophobic pocket in the core domain of an adjacent subunit. The conformation of the N-terminal arm differs from that seen in a previous crystal structure of RVFV, in which it was bound to the hydrophobic pocket in its own core domain. The switch from an intra- to an inter-molecular interaction mode of the N-terminal arm may be a general principle that underlies multimerization and RNA encapsidation by N proteins from Bunyaviridae. Furthermore, slight structural adjustments of the N-terminal arm would allow RVFV N to form smaller or larger ring-shaped oligomers and potentially even a multimer with a super-helical subunit arrangement. Thus, the interaction mode between subunits seen in the crystal structure would allow the formation of filamentous ribonucleocapsids in vivo. Both the RNA binding cleft and the multimerization site of the N protein are promising targets for the development of antiviral drugs.


Subject(s)
Nucleocapsid Proteins/chemistry , Protein Multimerization , RNA, Viral/chemistry , Ribonucleoproteins/chemistry , Rift Valley fever virus/physiology , Amino Acid Sequence , Animals , Crystallography, X-Ray/methods , DNA, Complementary/genetics , Humans , Microscopy, Electron , Models, Molecular , Mutagenesis, Site-Directed , Nucleocapsid Proteins/genetics , Nucleocapsid Proteins/isolation & purification , Nucleocapsid Proteins/ultrastructure , Protein Interaction Domains and Motifs , RNA, Viral/genetics , RNA, Viral/metabolism , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/isolation & purification , Recombinant Fusion Proteins/ultrastructure , Rift Valley fever virus/chemistry , Rift Valley fever virus/genetics , Rift Valley fever virus/ultrastructure , Sequence Alignment , Surface Plasmon Resonance/methods , Virus Assembly
16.
Proc Natl Acad Sci U S A ; 107(26): 11769-74, 2010 Jun 29.
Article in English | MEDLINE | ID: mdl-20547879

ABSTRACT

Rift Valley fever virus (RVFV) is a negative-sense RNA virus (genus Phlebovirus, family Bunyaviridae) that infects livestock and humans and is endemic to sub-Saharan Africa. Like all negative-sense viruses, the segmented RNA genome of RVFV is encapsidated by a nucleocapsid protein (N). The 1.93-A crystal structure of RVFV N and electron micrographs of ribonucleoprotein (RNP) reveal an encapsidated genome of substantially different organization than in other negative-sense RNA virus families. The RNP polymer, viewed in electron micrographs of both virus RNP and RNP reconstituted from purified N with a defined RNA, has an extended structure without helical symmetry. N-RNA species of approximately 100-kDa apparent molecular weight and heterogeneous composition were obtained by exhaustive ribonuclease treatment of virus RNP, by recombinant expression of N, and by reconstitution from purified N and an RNA oligomer. RNA-free N, obtained by denaturation and refolding, has a novel all-helical fold that is compact and well ordered at both the N and C termini. Unlike N of other negative-sense RNA viruses, RVFV N has no positively charged surface cleft for RNA binding and no protruding termini or loops to stabilize a defined N-RNA oligomer or RNP helix. A potential protein interaction site was identified in a conserved hydrophobic pocket. The nonhelical appearance of phlebovirus RNP, the heterogeneous approximately 100-kDa N-RNA multimer, and the N fold differ substantially from the RNP and N of other negative-sense RNA virus families and provide valuable insights into the structure of the encapsidated phlebovirus genome.


Subject(s)
Nucleocapsid Proteins/chemistry , RNA, Viral/chemistry , Rift Valley fever virus/chemistry , Amino Acid Sequence , Animals , Base Sequence , Conserved Sequence , Crystallography, X-Ray , DNA Primers/genetics , Humans , Macromolecular Substances/chemistry , Microscopy, Electron, Transmission , Models, Molecular , Molecular Sequence Data , Mutagenesis, Site-Directed , Nucleocapsid Proteins/genetics , Nucleocapsid Proteins/ultrastructure , Phlebovirus/genetics , Protein Interaction Domains and Motifs , Protein Multimerization , RNA, Viral/genetics , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/ultrastructure , Rift Valley fever virus/genetics , Rift Valley fever virus/physiology , Rift Valley fever virus/ultrastructure , Sequence Homology, Amino Acid , Static Electricity , Virus Assembly
17.
Arq. Inst. Biol. (Online) ; 77(1): 1-9, jan-mar, 2010. ilus, tab
Article in Portuguese | VETINDEX, LILACS | ID: biblio-1381388

ABSTRACT

O gene da proteína de nucleocapsídeo (1.230 pb) da estirpe M41 do vírus da bronquite infecciosa (VBI) foi amplificado pelas reações de transcrição reversa e em cadeia da polimerase (RT-PCR) e clonado, em seguida, em dois sistemas; pET28a - Escherichia coli e pFLD -Pichia pastoris. Os produtos recombinantes construídos para expressão (pET28a-N ou pFLD-N) foram identificados por análises de PCR e de sequenciamento de nucleotídeos. Os clones transformantes da linhagem BL21 de E. coli e da linhagem GS115 de P. pastoris foram submetidos aos protocolos apropriados de indução. A expressão da proteína N de fusão com etiqueta de poli-histidina e com massa molecular de 54 kDa foi determinada pelas técnicas de SDS-PAGE e de Western blotting, confirmando-se que ambas proteínas N recombinantes apresentaram tamanhos e antigenicidade compatíveis com a proteína N nativa do próprio VBI. O sistema E. coli expressou uma quantidade relevante da proteína N recombinante, enquanto que o sistema P. pastoris produziu uma baixa recuperação dessa proteína recombinante. A proteína N recombinante gerada pelo sistema bacteriano foi purificada em resina de níquel-sepharose. O conjunto de resultados indica que o sistema de expressão constituído por pET28a ­ E. coli é mais efetivo para produzir a proteína N recombinante do VBI destinada ao uso como antígeno para detectar anticorpos anti-virais específicos em ensaios de imunodiagnóstico para essa infecção viral.


The nucleocapsid protein (N) gene (1,230 bp) of the M41 strain of infectious bronchitis virus (IBV) was amplified by reverse transcriptase-polymerase chain reaction (RT-PCR), and cloned in two systems; pET28a Escherichia coli and pFLD Pichia pastoris. The recombinant expression constructs (pET28a-N or pFLD-N) were identified by PCR and sequencing analysis. The transformant clones of BL21 strain of E. coli or GS115 of P. pastoris were submitted to appropriate inducting protocols. Expression of histidine-tagged fusion N proteins with a molecular mass of 54 kDa was determined by SDS-PAGE and Western blotting analysis, confirming that both recombinant N proteins were comparable in size and antigenicity to native IBV N protein. The E. coli system overexpressed the recombinant N protein, while the P. pastoris system produced a low yield of this recombinant protein. The bacteria expressed N protein was purified by chromatography on nickel-sepharose resin. These results indicated that the pET28a E. coli expression system is more effective to generate N recombinant protein for using as an antigen to detect anti-IBV antibodies in immuno-assays for this viral infection.


Subject(s)
Pichia/genetics , Infectious bronchitis virus/ultrastructure , Nucleocapsid Proteins/ultrastructure , Escherichia coli/genetics , Enzyme-Linked Immunosorbent Assay , Cloning, Molecular
18.
Science ; 327(5966): 689-93, 2010 Feb 05.
Article in English | MEDLINE | ID: mdl-20133572

ABSTRACT

Vesicular stomatitis virus (VSV) is a bullet-shaped rhabdovirus and a model system of negative-strand RNA viruses. Through direct visualization by means of cryo-electron microscopy, we show that each virion contains two nested, left-handed helices: an outer helix of matrix protein M and an inner helix of nucleoprotein N and RNA. M has a hub domain with four contact sites that link to neighboring M and N subunits, providing rigidity by clamping adjacent turns of the nucleocapsid. Side-by-side interactions between neighboring N subunits are critical for the nucleocapsid to form a bullet shape, and structure-based mutagenesis results support this description. Together, our data suggest a mechanism of VSV assembly in which the nucleocapsid spirals from the tip to become the helical trunk, both subsequently framed and rigidified by the M layer.


Subject(s)
Nucleocapsid Proteins/chemistry , RNA, Viral/chemistry , Vesiculovirus/chemistry , Vesiculovirus/ultrastructure , Viral Matrix Proteins/chemistry , Cryoelectron Microscopy , Crystallography, X-Ray , Image Processing, Computer-Assisted , Lipid Bilayers , Models, Molecular , Mutagenesis , Nucleocapsid Proteins/genetics , Nucleocapsid Proteins/ultrastructure , Protein Conformation , Protein Structure, Secondary , Protein Structure, Tertiary , Protein Subunits/chemistry , RNA, Viral/ultrastructure , Vesiculovirus/physiology , Viral Matrix Proteins/ultrastructure , Virion/chemistry , Virion/ultrastructure , Virus Assembly
19.
J Virol ; 83(21): 11402-6, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19692473

ABSTRACT

The nucleocapsid protein (NP) of mumps virus (MuV), a paramyxovirus, was coexpressed with the phosphoprotein (P) in Escherichia coli. The NP and P proteins form a soluble complex containing RNA. Under a transmission electron microscope, the NP-RNA complex appears as a nucleocapsidlike ring that has a diameter of approximately 20 nm with 13 subunits. There is a piece of single-stranded RNA with a length of 78 nucleotides in the NP-RNA ring. Shorter RNA pieces are also visible. The MuV NP protein may provide weaker protection of the RNA than the NP protein of some other negative-strand RNA viruses, reflecting the degree of NP protein association.


Subject(s)
Mumps virus/metabolism , Nucleocapsid Proteins/metabolism , Nucleocapsid , Phosphoproteins/metabolism , Virion , Humans , Microscopy, Electron, Transmission , Multiprotein Complexes , Mumps virus/ultrastructure , Nucleocapsid/metabolism , Nucleocapsid/ultrastructure , Nucleocapsid Proteins/genetics , Nucleocapsid Proteins/ultrastructure , Particle Size , Phosphoproteins/genetics , Phosphoproteins/ultrastructure , RNA, Viral/metabolism , RNA, Viral/ultrastructure , Virion/metabolism , Virion/ultrastructure
20.
Virus Res ; 145(1): 92-6, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19559738

ABSTRACT

Tioman virus (TioV) was isolated from a number of pooled urine samples of Tioman Island flying foxes (Pteropus hypomelanus) during the search for the reservoir host of Nipah virus. Studies have established TioV as a new virus in the family Paramyxoviridae. This novel paramyxovirus is antigenically related to Menangle virus that was isolated in Australia in 1997 during disease outbreak in pigs. TioV causes mild disease in pigs and has a predilection for lymphoid tissues. Recent serosurvey showed that 1.8% of Tioman Islanders had neutralizing antibodies against TioV, indicating probable past infection. For the development of convenient serological tests for this virus, recombinant TioV nucleocapsid (N) protein was expressed in the yeast Saccharomyces cerevisiae. High yields of recombinant TioV N protein were obtained. Electron microscopy demonstrated that purified recombinant N protein self-assembled into nucleocapsid-like particles which were identical in density and morphology to authentic nucleocapsids from paramyxovirus-infected cells. Different size nucleocapsid-like particles were stable and readily purified by CsCl gradient ultracentrifugation. Polyclonal sera raised in rabbits after immunization with recombinant TioV N protein reacted reliably with TioV infected tissues in immunohistochemistry tests. It confirmed that the antigenic properties of yeast derived TioV N protein are identical to authentic viral protein.


Subject(s)
Nucleocapsid Proteins/biosynthesis , Pneumovirinae/genetics , Recombinant Proteins/biosynthesis , Saccharomyces cerevisiae/metabolism , Amino Acid Sequence , Animals , Antibodies, Viral/immunology , Antigens, Viral/immunology , Chiroptera , Immunohistochemistry , Mice , Microscopy, Electron, Transmission , Molecular Sequence Data , Nucleocapsid Proteins/genetics , Nucleocapsid Proteins/immunology , Nucleocapsid Proteins/ultrastructure , Paramyxoviridae Infections/immunology , Paramyxoviridae Infections/virology , Pneumovirinae/immunology , Pneumovirinae/isolation & purification , Rabbits , Recombinant Proteins/genetics , Recombinant Proteins/immunology , Recombinant Proteins/ultrastructure , Swine
SELECTION OF CITATIONS
SEARCH DETAIL