Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 48
Filter
Add more filters










Publication year range
1.
Viruses ; 12(9)2020 09 22.
Article in English | MEDLINE | ID: mdl-32971895

ABSTRACT

Coronaviruses (CoVs) are enveloped, positive sense, single strand RNA viruses that cause respiratory, intestinal and neurological diseases in mammals and birds. Following replication, CoVs assemble on intracellular membranes including the endoplasmic reticulum Golgi intermediate compartment (ERGIC) where the envelope protein (E) functions in virus assembly and release. In consequence, E potentially contains membrane-modifying peptides. To search for such peptides, the E coding sequence of Mouse Hepatitis Virus (MHV) was inspected for its amino acid conservation, proximity to the membrane and/or predicted amphipathic helices. Peptides identified in silico were synthesized and tested for membrane-modifying activity in the presence of giant unilamellar vesicles (GUVs) consisting of 1,2-dipalmitoyl-sn-glycero-3-phosphocholine (DPPC), sphingomyelin and cholesterol. To confirm the presence of membrane binding peptides identified in the context of a full-length E protein, the wild type and a number of mutants in the putative membrane binding peptide were expressed in Lenti-X-293T mammalian and insect cells, and the distribution of E antigen within the expressing cell was assessed. Our data identify a role for the post-transmembrane region of MHV E in membrane binding.


Subject(s)
Murine hepatitis virus/chemistry , Peptides/chemistry , Viral Envelope Proteins/chemistry , Amino Acid Sequence , Animals , Cell Line , Coronavirus Infections , Humans , Intracellular Membranes/metabolism , Mice , Murine hepatitis virus/genetics , Murine hepatitis virus/metabolism , Mutation , Peptides/chemical synthesis , Peptides/metabolism , Sf9 Cells , Spodoptera , Unilamellar Liposomes/metabolism , Viral Envelope Proteins/genetics , Viral Envelope Proteins/metabolism
2.
PLoS Pathog ; 16(3): e1008392, 2020 03.
Article in English | MEDLINE | ID: mdl-32150576

ABSTRACT

Coronaviruses recognize a variety of receptors using different domains of their envelope-anchored spike protein. How these diverse receptor recognition patterns affect viral entry is unknown. Mouse hepatitis coronavirus (MHV) is the only known coronavirus that uses the N-terminal domain (NTD) of its spike to recognize a protein receptor, CEACAM1a. Here we determined the cryo-EM structure of MHV spike complexed with mouse CEACAM1a. The trimeric spike contains three receptor-binding S1 heads sitting on top of a trimeric membrane-fusion S2 stalk. Three receptor molecules bind to the sides of the spike trimer, where three NTDs are located. Receptor binding induces structural changes in the spike, weakening the interactions between S1 and S2. Using protease sensitivity and negative-stain EM analyses, we further showed that after protease treatment of the spike, receptor binding facilitated the dissociation of S1 from S2, allowing S2 to transition from pre-fusion to post-fusion conformation. Together these results reveal a new role of receptor binding in MHV entry: in addition to its well-characterized role in viral attachment to host cells, receptor binding also induces the conformational change of the spike and hence the fusion of viral and host membranes. Our study provides new mechanistic insight into coronavirus entry and highlights the diverse entry mechanisms used by different viruses.


Subject(s)
Carcinoembryonic Antigen/chemistry , Murine hepatitis virus/chemistry , Murine hepatitis virus/physiology , Receptors, Virus/chemistry , Spike Glycoprotein, Coronavirus/chemistry , Virus Internalization , Animals , Carcinoembryonic Antigen/metabolism , Carcinoembryonic Antigen/ultrastructure , Cell Line, Tumor , Cryoelectron Microscopy , HEK293 Cells , Humans , Membrane Fusion , Mice , Models, Molecular , Protein Binding , Protein Conformation , Protein Conformation, alpha-Helical , Protein Domains , Protein Multimerization , Proteolysis , Receptors, Virus/metabolism , Receptors, Virus/ultrastructure , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Severe acute respiratory syndrome-related coronavirus/chemistry , Spike Glycoprotein, Coronavirus/metabolism , Spike Glycoprotein, Coronavirus/ultrastructure , Virus Attachment
3.
Biochem Biophys Res Commun ; 511(4): 794-799, 2019 04 16.
Article in English | MEDLINE | ID: mdl-30833083

ABSTRACT

Mouse hepatitis virus A59 (MHV-A59) is a representative member of the genus betacoronavirus within the subfamily Coronavirinae, which infects the liver, brain and respiratory tract. Through different inoculation routes, MHV-A59 can provide animal models for encephalitis, hepatitis and pneumonia to explore viral life machinery and virus-host interactions. In viral replication, non-structural protein 5 (Nsp5), also termed main protease (Mpro), plays a dominant role in processing coronavirus-encoded polyproteins and is thus recognized as an ideal target of anti-coronavirus agents. However, no structure of the MHV-A59 Mpro has been reported, and molecular exploration of the catalysis mechanism remains hindered. Here, we solved the crystal structure of the MHV-A59 Mpro complexed with a Michael acceptor-based inhibitor, N3. Structural analysis revealed that the Cß of the vinyl group of N3 covalently bound to C145 of the catalytic dyad of Mpro, which irreversibly inactivated cysteine protease activity. The lactam ring of the P1 side chain and the isobutyl group of the P2 side chain, which mimic the conserved residues at the same positions of the substrate, fit well into the S1 and S2 pockets. Through a comparative study with Mpro of other coronaviruses, we observed that the substrate-recognition pocket and enzyme inhibitory mechanism is highly conservative. Altogether, our study provided structural features of MHV-A59 Mpro and indicated that a Michael acceptor inhibitor is an ideal scaffold for antiviral drugs.


Subject(s)
Murine hepatitis virus/chemistry , Peptide Hydrolases/chemistry , Protease Inhibitors/pharmacology , Viral Nonstructural Proteins/antagonists & inhibitors , Viral Nonstructural Proteins/chemistry , Amino Acid Sequence , Animals , Coronavirus Infections/virology , Crystallography, X-Ray , Humans , Mice , Models, Molecular , Murine hepatitis virus/drug effects , Murine hepatitis virus/metabolism , Peptide Hydrolases/metabolism , Protein Conformation/drug effects , Sequence Alignment , Viral Nonstructural Proteins/metabolism
4.
J Biol Chem ; 294(20): 8064-8087, 2019 05 17.
Article in English | MEDLINE | ID: mdl-30824541

ABSTRACT

Fusion peptides (FPs) in spike proteins are key players mediating early events in cell-to-cell fusion, vital for intercellular viral spread. A proline residue located at the central FP region has often been suggested to have a distinctive role in this fusion event. The spike glycoprotein from strain RSA59 (PP) of mouse hepatitis virus (MHV) contains two central, consecutive prolines in the FP. Here, we report that deletion of one of these proline residues, resulting in RSA59 (P), significantly affected neural cell syncytia formation and viral titers postinfection in vitro Transcranial inoculation of C57Bl/6 mice with RSA59 (PP) or RSA59 (P) yielded similar degrees of necrotizing hepatitis and meningitis, but only RSA59 (PP) produced widespread encephalitis that extended deeply into the brain parenchyma. By day 6 postinfection, both virus variants were mostly cleared from the brain. Interestingly, inoculation with the RSA59 (P)-carrying MHV significantly reduced demyelination at the chronic stage. We also found that the presence of two consecutive prolines in FP promotes a more ordered, compact, and rigid structure in the spike protein. These effects on FP structure were due to proline's unique stereochemical properties intrinsic to its secondary amino acid structure, revealed by molecular dynamics and NMR experiments. We therefore propose that the differences in the severity of encephalitis and demyelination between RSA59 (PP) and RSA59 (P) arise from the presence or absence, respectively, of the two consecutive prolines in FP. Our studies define a structural determinant of MHV entry in the brain parenchyma important for altered neuropathogenesis.


Subject(s)
Brain , Demyelinating Diseases , INDEL Mutation , Meningitis, Viral , Murine hepatitis virus , Viral Envelope Proteins , Animals , Brain/metabolism , Brain/pathology , Brain/virology , Cell Line , Demyelinating Diseases/genetics , Demyelinating Diseases/metabolism , Demyelinating Diseases/pathology , Demyelinating Diseases/virology , Meningitis, Viral/genetics , Meningitis, Viral/metabolism , Meningitis, Viral/pathology , Meningitis, Viral/virology , Mice , Murine hepatitis virus/chemistry , Murine hepatitis virus/genetics , Murine hepatitis virus/metabolism , Nuclear Magnetic Resonance, Biomolecular , Proline , Protein Domains , Structure-Activity Relationship , Viral Envelope Proteins/chemistry , Viral Envelope Proteins/genetics , Viral Envelope Proteins/metabolism
5.
mBio ; 9(3)2018 05 01.
Article in English | MEDLINE | ID: mdl-29717007

ABSTRACT

Selective packaging is a mechanism used by multiple virus families to specifically incorporate genomic RNA (gRNA) into virions and exclude other types of RNA. Lineage A betacoronaviruses incorporate a 95-bp stem-loop structure, the packaging signal (PS), into the nsp15 locus of ORF1b that is both necessary and sufficient for the packaging of RNAs. However, unlike other viral PSs, where mutations generally resulted in viral replication defects, mutation of the coronavirus (CoV) PS results in large increases in subgenomic RNA packaging with minimal effects on gRNA packaging in vitro and on viral titers. Here, we show that selective packaging is also required for viral evasion of the innate immune response and optimal pathogenicity. We engineered two distinct PS mutants in two different strains of murine hepatitis virus (MHV) that packaged increased levels of subgenomic RNAs, negative-sense genomic RNA, and even cellular RNAs. All PS mutant viruses replicated normally in vitro but caused dramatically reduced lethality and weight loss in vivo PS mutant virus infection of bone marrow-derived macrophages resulted in increased interferon (IFN) production, indicating that the innate immune system limited the replication and/or pathogenesis of PS mutant viruses in vivo PS mutant viruses remained attenuated in MAVS-/- and Toll-like receptor 7-knockout (TLR7-/-) mice, two well-known RNA sensors for CoVs, but virulence was restored in interferon alpha/beta receptor-knockout (IFNAR-/-) mice or in MAVS-/- mice treated with IFNAR-blocking antibodies. Together, these data indicate that coronaviruses promote virulence by utilizing selective packaging to avoid innate immune detection.IMPORTANCE Coronaviruses (CoVs) produce many types of RNA molecules during their replication cycle, including both positive- and negative-sense genomic and subgenomic RNAs. Despite this, coronaviruses selectively package only positive-sense genomic RNA into their virions. Why CoVs selectively package their genomic RNA is not clear, as disruption of the packaging signal in MHV, which leads to loss of selective packaging, does not affect genomic RNA packaging or virus replication in cultured cells. This contrasts with other viruses, where disruption of selective packaging generally leads to altered replication. Here, we demonstrate that in the absence of selective packaging, the virulence of MHV was significantly reduced. Importantly, virulence was restored in the absence of interferon signaling, indicating that selective packaging is a mechanism used by CoVs to escape innate immune detection.


Subject(s)
Coronavirus Infections/veterinary , Interferon Type I/immunology , Murine hepatitis virus/physiology , Murine hepatitis virus/pathogenicity , Rodent Diseases/immunology , Virus Assembly , Animals , Coronavirus Infections/genetics , Coronavirus Infections/immunology , Coronavirus Infections/virology , Host-Pathogen Interactions , Interferon Type I/genetics , Inverted Repeat Sequences , Male , Mice , Murine hepatitis virus/chemistry , Murine hepatitis virus/genetics , Open Reading Frames , RNA, Viral/chemistry , RNA, Viral/genetics , RNA, Viral/metabolism , Rodent Diseases/genetics , Rodent Diseases/virology , Viral Nonstructural Proteins/genetics , Viral Nonstructural Proteins/metabolism , Virulence , Virus Replication
6.
J Virol ; 90(12): 5586-5600, 2016 06 15.
Article in English | MEDLINE | ID: mdl-27030273

ABSTRACT

UNLABELLED: The fusion peptides (FP) play an essential role in fusion of viral envelope with cellular membranes. The location and properties of the FPs in the spike (S) glycoproteins of different coronaviruses (CoV) have not yet been determined. Through amino acid sequence analysis of S proteins of representative CoVs, we identified a common region as a possible FP (pFP) that shares the characteristics of FPs of class I viral fusion proteins, including high Ala/Gly content, intermediate hydrophobicity, and few charged residues. To test the hypothesis that this region contains the CoV FP, we systemically mutated every residue in the pFP of Middle East respiratory syndrome betacoronavirus (MERS-CoV) and found that 11 of the 22 residues in the pFP (from G953 to L964, except for A956) were essential for S protein-mediated cell-cell fusion and virus entry. The synthetic MERS-CoV pFP core peptide (955IAGVGWTAGL964) induced extensive fusion of liposome membranes, while mutant peptide failed to induce any lipid mixing. We also selectively mutated residues in pFPs of two other ß-CoVs, severe acute respiratory syndrome coronavirus (SARS-CoV) and mouse hepatitis virus (MHV). Although the amino acid sequences of these two pFPs differed significantly from that of MERS-CoV and each other, most of the pFP mutants of SARS-CoV and MHV also failed to mediate membrane fusion, suggesting that these pFPs are also the functional FPs. Thus, the FPs of 3 different lineages of ß-CoVs are conserved in location within the S glycoproteins and in their functions, although their amino acid sequences have diverged significantly during CoV evolution. IMPORTANCE: Within the class I viral fusion proteins of many enveloped viruses, the FP is the critical mediator of fusion of the viral envelope with host cell membranes leading to virus infection. FPs from within a virus family, like influenza viruses or human immunodeficiency viruses (HIV), tend to share high amino acid sequence identity. In this study, we determined the location and amino acid sequences of the FPs of S glycoproteins of 3 ß-CoVs, MERS-CoV, SARS-CoV, and MHV, and demonstrated that they were essential for mediating cell-cell fusion and virus entry. Interestingly, in marked contrast to the FPs of influenza and HIV, the primary amino acid sequences of the FPs of ß-CoVs in 3 different lineages differed significantly. Thus, during evolution the FPs of ß-CoVs have diverged significantly in their primary sequences while maintaining the same essential biological functions. Our findings identify a potential new target for development of drugs against CoVs.


Subject(s)
Middle East Respiratory Syndrome Coronavirus/chemistry , Murine hepatitis virus/chemistry , Peptides/chemistry , Severe acute respiratory syndrome-related coronavirus/chemistry , Spike Glycoprotein, Coronavirus/chemistry , Spike Glycoprotein, Coronavirus/genetics , Amino Acid Sequence , Animals , Evolution, Molecular , HEK293 Cells , Humans , Membrane Fusion , Mice , Middle East Respiratory Syndrome Coronavirus/genetics , Murine hepatitis virus/genetics , Mutation , Peptides/chemical synthesis , Peptides/genetics , Severe acute respiratory syndrome-related coronavirus/genetics , Sequence Alignment , Spike Glycoprotein, Coronavirus/metabolism , Virus Internalization
7.
Nature ; 531(7592): 114-117, 2016 Mar 03.
Article in English | MEDLINE | ID: mdl-26855426

ABSTRACT

The tremendous pandemic potential of coronaviruses was demonstrated twice in the past few decades by two global outbreaks of deadly pneumonia. Entry of coronaviruses into cells is mediated by the transmembrane spike glycoprotein S, which forms a trimer carrying receptor-binding and membrane fusion functions. S also contains the principal antigenic determinants and is the target of neutralizing antibodies. Here we present the structure of a mouse coronavirus S trimer ectodomain determined at 4.0 Å resolution by single particle cryo-electron microscopy. It reveals the metastable pre-fusion architecture of S and highlights key interactions stabilizing it. The structure shares a common core with paramyxovirus F proteins, implicating mechanistic similarities and an evolutionary connection between these viral fusion proteins. The accessibility of the highly conserved fusion peptide at the periphery of the trimer indicates potential vaccinology strategies to elicit broadly neutralizing antibodies against coronaviruses. Finally, comparison with crystal structures of human coronavirus S domains allows rationalization of the molecular basis for species specificity based on the use of spatially contiguous but distinct domains.


Subject(s)
Cryoelectron Microscopy , Murine hepatitis virus/chemistry , Murine hepatitis virus/ultrastructure , Spike Glycoprotein, Coronavirus/chemistry , Spike Glycoprotein, Coronavirus/ultrastructure , Amino Acid Sequence , Animals , Antibodies, Neutralizing/immunology , Cell Line , Coronavirus Infections/immunology , Coronavirus Infections/virology , Drosophila melanogaster , Mice , Models, Molecular , Molecular Sequence Data , Murine hepatitis virus/immunology , Protein Multimerization , Protein Structure, Tertiary , Spike Glycoprotein, Coronavirus/immunology , Viral Vaccines/chemistry , Viral Vaccines/immunology , Virus Internalization
8.
J Gen Virol ; 97(4): 880-886, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26757803

ABSTRACT

Prior studies have demonstrated that the mouse hepatitis virus (MHV) A59 strain ns2 protein is a member of the 2H phosphoesterase family and exhibits 2',5'-phosphodiesterase (PDE) activity. During the IFN antiviral response, ns2 cleaves 2',5'-oligoadenylate (2-5A), a key mediator of RNase L activation, thereby subverting the activation of RNase L and evading host innate immunity. However, the mechanism of 2-5A cleavage by ns2 remains unclear. Here, we present the crystal structure of the MHV ns2 PDE domain and demonstrate a PDE fold similar to that of the cellular protein, a kinase anchoring protein 7 central domain (AKAP7(CD)) and rotavirus VP3 carboxy-terminal domain. The structure displays a pair of strictly conserved HxT/Sx motifs and forms a deep, positively charged catalytic groove with ß-sheets and an arginine-containing loop. These findings provide insight into the structural basis for 2-5A binding of MHV ns2.


Subject(s)
Endoribonucleases/chemistry , Murine hepatitis virus/chemistry , Phosphoric Diester Hydrolases/chemistry , Viral Nonstructural Proteins/chemistry , A Kinase Anchor Proteins/chemistry , A Kinase Anchor Proteins/genetics , A Kinase Anchor Proteins/metabolism , Amino Acid Motifs , Animals , Catalytic Domain , Cloning, Molecular , Conserved Sequence , Crystallography, X-Ray , Endoribonucleases/genetics , Endoribonucleases/metabolism , Escherichia coli/genetics , Escherichia coli/metabolism , Gene Expression , Mice , Models, Molecular , Molecular Sequence Data , Murine hepatitis virus/enzymology , Phosphoric Diester Hydrolases/genetics , Phosphoric Diester Hydrolases/metabolism , Protein Interaction Domains and Motifs , Protein Structure, Secondary , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Rotavirus/chemistry , Structural Homology, Protein , Viral Nonstructural Proteins/genetics , Viral Nonstructural Proteins/metabolism
9.
J Biol Chem ; 290(42): 25293-306, 2015 Oct 16.
Article in English | MEDLINE | ID: mdl-26296883

ABSTRACT

Murine hepatitis virus (MHV) has long served as a model system for the study of coronaviruses. Non-structural protein 3 (nsp3) is the largest nsp in the coronavirus genome, and it contains multiple functional domains that are required for coronavirus replication. Despite the numerous functional studies on MHV and its nsp3 domain, the structure of only one domain in nsp3, the small ubiquitin-like domain 1 (Ubl1), has been determined. We report here the x-ray structure of three tandemly linked domains of MHV nsp3, including the papain-like protease 2 (PLP2) catalytic domain, the ubiquitin-like domain 2 (Ubl2), and a third domain that we call the DPUP (domain preceding Ubl2 and PLP2) domain. DPUP has close structural similarity to the severe acute respiratory syndrome coronavirus unique domain C (SUD-C), suggesting that this domain may not be unique to the severe acute respiratory syndrome coronavirus. The PLP2 catalytic domain was found to have both deubiquitinating and deISGylating isopeptidase activities in addition to proteolytic activity. A computationally derived model of MHV PLP2 bound to ubiquitin was generated, and the potential interactions between ubiquitin and PLP2 were probed by site-directed mutagenesis. These studies extend substantially our structural knowledge of MHV nsp3, providing a platform for further investigation of the role of nsp3 domains in MHV viral replication.


Subject(s)
Murine hepatitis virus/chemistry , Viral Nonstructural Proteins/chemistry , Amino Acid Sequence , Crystallography, X-Ray , Molecular Sequence Data , Protein Conformation , Sequence Homology, Amino Acid , Substrate Specificity , Viral Nonstructural Proteins/physiology
10.
J Virol ; 88(8): 4451-65, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24501403

ABSTRACT

UNLABELLED: The coronavirus nucleocapsid (N) protein forms a helical ribonucleoprotein with the viral positive-strand RNA genome and binds to the principal constituent of the virion envelope, the membrane (M) protein, to facilitate assembly and budding. Besides these structural roles, N protein associates with a component of the replicase-transcriptase complex, nonstructural protein 3, at a critical early stage of infection. N protein has also been proposed to participate in the replication and selective packaging of genomic RNA and the transcription and translation of subgenomic mRNA. Coronavirus N proteins contain two structurally distinct RNA-binding domains, an unusual characteristic among RNA viruses. To probe the functions of these domains in the N protein of the model coronavirus mouse hepatitis virus (MHV), we constructed mutants in which each RNA-binding domain was replaced by its counterpart from the N protein of severe acute respiratory syndrome coronavirus (SARS-CoV). Mapping of revertants of the resulting chimeric viruses provided evidence for extensive intramolecular interactions between the two RNA-binding domains. Through analysis of viral RNA that was packaged into virions we identified the second of the two RNA-binding domains as a principal determinant of MHV packaging signal recognition. As expected, the interaction of N protein with M protein was not affected in either of the chimeric viruses. Moreover, the SARS-CoV N substitutions did not alter the fidelity of leader-body junction formation during subgenomic mRNA synthesis. These results more clearly delineate the functions of N protein and establish a basis for further exploration of the mechanism of genomic RNA packaging. IMPORTANCE: This work describes the interactions of the two RNA-binding domains of the nucleocapsid protein of a model coronavirus, mouse hepatitis virus. The main finding is that the second of the two domains plays an essential role in recognizing the RNA structure that allows the selective packaging of genomic RNA into assembled virions.


Subject(s)
Coronaviridae Infections/veterinary , Genome, Viral , Murine hepatitis virus/physiology , Nucleocapsid Proteins/chemistry , Nucleocapsid Proteins/metabolism , RNA, Viral/metabolism , Rodent Diseases/virology , Virus Assembly , Animals , Cell Line , Coronaviridae Infections/virology , Coronavirus Nucleocapsid Proteins , Mice , Murine hepatitis virus/chemistry , Murine hepatitis virus/genetics , Nucleocapsid Proteins/genetics , Protein Binding , Protein Structure, Tertiary , RNA, Viral/genetics
11.
Virology ; 442(1): 74-81, 2013 Jul 20.
Article in English | MEDLINE | ID: mdl-23628137

ABSTRACT

Coronavirus spike (S) protein assembles into virions via its carboxy-terminus, which is composed of a transmembrane domain and an endodomain. Here, the carboxy-terminal charge-rich motif in the endodomain was verified to be critical for the specificity of S assembly into mouse hepatitis virus (MHV). Recombinant MHVs exhibited a range of abilities to accommodate the homologous S endodomains from the betacoronaviruses bovine coronavirus and human SARS-associated coronavirus, the alphacoronavirus porcine transmissible gastroenteritis virus (TGEV), and the gammacoronavirus avian infectious bronchitis virus respectively. Interestingly, in TGEV endodomain chimeras the reverting mutations resulted in stronger S incorporation into virions, and a net gain of negatively charged residues in the charge-rich motif accounted for the improvement. Additionally, MHV S assembly could also be rescued by the acidic carboxy-terminal domain of the nucleocapsid protein. These results indicate an important role for negatively charged endodomain residues in the incorporation of MHV S protein into assembled virions.


Subject(s)
Membrane Glycoproteins/chemistry , Membrane Glycoproteins/metabolism , Murine hepatitis virus/metabolism , Viral Envelope Proteins/chemistry , Viral Envelope Proteins/metabolism , Virus Assembly , Amino Acid Sequence , Animals , Cattle , Cell Line , Coronavirus, Bovine/genetics , Coronavirus, Bovine/metabolism , Humans , Membrane Glycoproteins/genetics , Mice , Molecular Sequence Data , Murine hepatitis virus/chemistry , Murine hepatitis virus/genetics , Mutation , Protein Structure, Tertiary/genetics , Severe acute respiratory syndrome-related coronavirus , Spike Glycoprotein, Coronavirus , Transmissible gastroenteritis virus/genetics , Transmissible gastroenteritis virus/metabolism , Viral Envelope Proteins/genetics
12.
J Virol ; 87(6): 3502-15, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23302895

ABSTRACT

Coronaviruses (CoVs) are positive-sense, single-stranded, enveloped RNA viruses that infect a variety of vertebrate hosts. The CoV nucleocapsid (N) protein contains two structurally independent RNA binding domains, designated the N-terminal domain (NTD) and the dimeric C-terminal domain (CTD), joined by a charged linker region rich in serine and arginine residues (SR-rich linker). An important goal in unraveling N function is to molecularly characterize N-protein interactions. Recent genetic evidence suggests that N interacts with nsp3a, a component of the viral replicase. Here we present the solution nuclear magnetic resonance (NMR) structure of mouse hepatitis virus (MHV) nsp3a and show, using isothermal titration calorimetry, that MHV N219, an N construct that extends into the SR-rich linker (residues 60 to 219), binds cognate nsp3a with high affinity (equilibrium association constant [K(a)], [1.4 ± 0.3] × 10(6) M(-1)). In contrast, neither N197, an N construct containing only the folded NTD (residues 60 to 197), nor the CTD dimer (residues 260 to 380) binds nsp3a with detectable affinity. This indicates that the key nsp3a binding determinants localize to the SR-rich linker, a finding consistent with those of reverse genetics studies. NMR chemical shift perturbation analysis reveals that the N-terminal region of an MHV N SR-rich linker peptide (residues 198 to 230) binds to the acidic face of MHV nsp3a containing the acidic α2 helix with an affinity (expressed as K(a)) of 8.1 × 10(3) M(-1). These studies reveal that the SR-rich linker of MHV N is necessary but not sufficient to maintain this high-affinity binding to N.


Subject(s)
Murine hepatitis virus/chemistry , Murine hepatitis virus/physiology , Nucleocapsid Proteins/metabolism , Viral Nonstructural Proteins/chemistry , Viral Nonstructural Proteins/metabolism , Magnetic Resonance Spectroscopy , Models, Molecular , Protein Binding , Protein Conformation
13.
PLoS Pathog ; 8(1): e1002492, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22291594

ABSTRACT

The hemagglutinin-esterases (HEs), envelope glycoproteins of corona-, toro- and orthomyxoviruses, mediate reversible virion attachment to O-acetylated sialic acids (O-Ac-Sias). They do so through concerted action of distinct receptor-binding ("lectin") and receptor-destroying sialate O-acetylesterase ("esterase") domains. Most HEs target 9-O-acetylated Sias. In one lineage of murine coronaviruses, however, HE esterase substrate and lectin ligand specificity changed dramatically as these viruses evolved to use 4-O-acetylated Sias instead. Here we present the crystal structure of the lectin domain of mouse hepatitis virus (MHV) strain S HE, resolved both in its native state and in complex with a receptor analogue. The data show that the shift from 9-O- to 4-O-Ac-Sia receptor usage primarily entailed a change in ligand binding topology and, surprisingly, only modest changes in receptor-binding site architecture. Our findings illustrate the ease with which viruses can change receptor-binding specificity with potential consequences for host-, organ and/or cell tropism, and for pathogenesis.


Subject(s)
Hemagglutinins, Viral/chemistry , Murine hepatitis virus/chemistry , N-Acetylneuraminic Acid/chemistry , Receptors, Virus/chemistry , Viral Fusion Proteins/chemistry , Animals , Binding Sites , Crystallography, X-Ray , Hemagglutinins, Viral/metabolism , Humans , Mice , Murine hepatitis virus/physiology , N-Acetylneuraminic Acid/metabolism , Rats , Rats, Wistar , Receptors, Virus/metabolism , Viral Fusion Proteins/metabolism , Viral Tropism/physiology
14.
J Biol Chem ; 287(10): 7063-73, 2012 Mar 02.
Article in English | MEDLINE | ID: mdl-22241479

ABSTRACT

Coronavirus (CoV) nucleocapsid (N) protein contains two structurally independent RNA binding domains. These are denoted N-terminal domain (NTD) and C-terminal domain and are joined by a charged linker region rich in serine and arginine residues (SR linker). In mouse hepatitis virus (MHV), the NTD binds the transcriptional regulatory sequence (TRS) RNA, a conserved hexanucleotide sequence required for subgenomic RNA synthesis. The NTD is also capable of disrupting a short RNA duplex. We show here that three residues on the ß3 (Arg-125 and Tyr-127) and ß5 (Tyr-190) strands play key roles in TRS RNA binding and helix destabilization with Ala substitutions of these residues lethal to the virus. NMR studies of the MHV NTD·TRS complex revealed that this region defines a major RNA binding interface in MHV with site-directed spin labeling studies consistent with a model in which the adenosine-rich 3'-region of TRS is anchored by Arg-125, Tyr-127, and Tyr-190 in a way that is critical for efficient subgenomic RNA synthesis in MHV. Characterization of CoV N NTDs from infectious bronchitis virus and from severe acute respiratory syndrome CoV revealed that, although detailed NTD-TRS determinants are distinct from those of MHV NTD, rapid helix destabilization activity of CoV N NTDs is most strongly correlated with CoV function and virus viability.


Subject(s)
Models, Molecular , Murine hepatitis virus/chemistry , Nucleocapsid Proteins/chemistry , RNA, Viral/chemistry , RNA-Binding Proteins/chemistry , Amino Acid Substitution , Animals , Cell Line , Mice , Murine hepatitis virus/genetics , Murine hepatitis virus/metabolism , Mutation, Missense , Nucleocapsid Proteins/genetics , Nucleocapsid Proteins/metabolism , Protein Structure, Secondary , Protein Structure, Tertiary , RNA, Viral/genetics , RNA, Viral/metabolism , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism
15.
Article in English | MEDLINE | ID: mdl-20516597

ABSTRACT

Mouse hepatitis virus (MHV) belongs to the group II coronaviruses. The virus produces nine genes encoding 11 proteins that could be recognized as structural proteins and nonstructural proteins and are crucial for viral RNA synthesis. The nucleocapsid (N) protein, one of the structural proteins, interacts with the 30.4 kb virus genomic RNA to form the helical nucleocapsid and associates with the membrane glycoprotein via its C-terminus to stabilize virion assembly. Here, the expression and crystallization of the MHV nucleocapsid protein C-terminal domain are reported. The crystals diffracted to 2.20 A resolution and belonged to space group P422, with unit-cell parameters a = 66.6, c = 50.8 A. Assuming the presence of two molecules in the asymmetric unit, the solvent content is 43.0% (V(M) = 2.16 A(3) Da(-1)).


Subject(s)
Murine hepatitis virus/chemistry , Nucleocapsid Proteins/chemistry , Crystallization , Crystallography, X-Ray , Gene Expression , Nucleocapsid Proteins/genetics
16.
Protein Cell ; 1(7): 688-97, 2010 Jul.
Article in English | MEDLINE | ID: mdl-21203940

ABSTRACT

Coronaviruses are the causative agent of respiratory and enteric diseases in animals and humans. One example is SARS, which caused a worldwide health threat in 2003. In coronaviruses, the structural protein N (nucleocapsid protein) associates with the viral RNA to form the filamentous nucleocapsid and plays a crucial role in genome replication and transcription. The structure of N-terminal domain of MHV N protein also implicated its specific affinity with transcriptional regulatory sequence (TRS) RNA. Here we report the crystal structures of the two proteolytically resistant N- (NTD) and C-terminal (CTD) domains of the N protein from murine hepatitis virus (MHV). The structure of NTD in two different crystal forms was solved to 1.5 Å. The higher resolution provides more detailed structural information than previous reports, showing that the NTD structure from MHV shares a similar overall and topology structure with that of SARS-CoV and IBV, but varies in its potential surface, which indicates a possible difference in RNA-binding module. The structure of CTD was solved to 2.0-Å resolution and revealed a tightly intertwined dimer. This is consistent with analytical ultracentrifugation experiments, suggesting a dimeric assembly of the N protein. The similarity between the structures of these two domains from SARS-CoV, IBV and MHV corroborates a conserved mechanism of nucleocapsid formation for coronaviruses.


Subject(s)
Murine hepatitis virus/chemistry , Nucleocapsid Proteins/chemistry , Phosphoproteins/chemistry , RNA/metabolism , Amino Acid Sequence , Binding Sites , Crystallography, X-Ray , Molecular Sequence Data , Murine hepatitis virus/metabolism , Nucleocapsid Proteins/metabolism , Phosphoproteins/metabolism , Protein Binding , Protein Folding , Protein Multimerization , Protein Structure, Secondary , Protein Structure, Tertiary , Sequence Alignment
17.
J Virol ; 84(1): 280-90, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19846526

ABSTRACT

Positive-strand RNA viruses induce modifications of cytoplasmic membranes to form replication complexes. For coronaviruses, replicase nonstructural protein 4 (nsp4) has been proposed to function in the formation and organization of replication complexes. Murine hepatitis virus (MHV) nsp4 is glycosylated at residues Asn176 (N176) and N237 during plasmid expression of nsp4 in cells. To test if MHV nsp4 residues N176 and N237 are glycosylated during virus replication and to determine the effects of N176 and N237 on nsp4 function and MHV replication, alanine substitutions of nsp4 N176, N237, or both were engineered into the MHV-A59 genome. The N176A, N237A, and N176A/N237A mutant viruses were viable, and N176 and N237 were glycosylated during infection of wild-type (wt) and mutant viruses. The nsp4 glycosylation mutants exhibited impaired virus growth and RNA synthesis, with the N237A and N176A/N237A mutant viruses demonstrating more profound defects in virus growth and RNA synthesis. Electron microscopic analysis of ultrastructure from infected cells demonstrated that the nsp4 mutants had aberrant morphology of virus-induced double-membrane vesicles (DMVs) compared to those infected with wt virus. The degree of altered DMV morphology directly correlated with the extent of impairment in viral RNA synthesis and virus growth of the nsp4 mutant viruses. The results indicate that nsp4 plays a critical role in the organization and stability of DMVs. The results also support the conclusion that the structure of DMVs is essential for efficient RNA synthesis and optimal replication of coronaviruses.


Subject(s)
Intracellular Membranes , Murine hepatitis virus/physiology , Murine hepatitis virus/ultrastructure , Viral Nonstructural Proteins/physiology , Virus Replication , Coronaviridae , Glycosylation , Microscopy, Electron , Murine hepatitis virus/chemistry , Mutant Proteins , Mutation, Missense , RNA, Viral/biosynthesis
18.
PLoS One ; 4(7): e6217, 2009 Jul 10.
Article in English | MEDLINE | ID: mdl-19593433

ABSTRACT

BACKGROUND: The replication of coronaviruses takes place on cytoplasmic double membrane vesicles (DMVs) originating in the endoplasmic reticulum (ER). Three trans-membrane non-structural proteins, nsp3, nsp4 and nsp6, are understood to be membrane anchors of the coronavirus replication complex. Nsp4 is localized to the ER membrane when expressed alone but is recruited into the replication complex in infected cells. It is revealed to contain four trans-membrane regions and its N- and C-termini are exposed to the cytosol. METHODOLOGY/PRINCIPAL FINDINGS: We have determined the crystal structures of the C-terminal hydrophilic domain of nsp4 (nsp4C) from MHV strain A59 and a C425S site-directed mutant. The highly conserved 89 amino acid region from T408 to Q496 is shown to possess a new fold. The wild-type (WT) structure features two monomers linked by a Cys425-Cys425 disulfide bond in one asymmetric unit. The monomers are arranged with their N- and C-termini in opposite orientations to form an "open" conformation. Mutation of Cys425 to Ser did not affect the monomer structure, although the mutant dimer adopts strikingly different conformations by crystal packing, with the cross-linked C-termini and parallel N-termini of two monomers forming a "closed" conformation. The WT nsp4C exists as a dimer in solution and can dissociate easily into monomers in a reducing environment. CONCLUSIONS/SIGNIFICANCE: As nsp4C is exposed in the reducing cytosol, the monomer of nsp4C should be physiological. This structure may serve as a basis for further functional studies of nsp4.


Subject(s)
Murine hepatitis virus/chemistry , Viral Nonstructural Proteins/chemistry , Amino Acid Sequence , Animals , Dimerization , Endoplasmic Reticulum/chemistry , Mice , Models, Molecular , Molecular Sequence Data , Mutagenesis, Site-Directed , Protein Conformation , Protein Folding , Sequence Homology, Amino Acid , Viral Nonstructural Proteins/genetics
19.
J Virol ; 83(14): 7221-34, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19420077

ABSTRACT

The coronavirus nucleocapsid protein (N), together with the large, positive-strand RNA viral genome, forms a helically symmetric nucleocapsid. This ribonucleoprotein structure becomes packaged into virions through association with the carboxy-terminal endodomain of the membrane protein (M), which is the principal constituent of the virion envelope. Previous work with the prototype coronavirus mouse hepatitis virus (MHV) has shown that a major determinant of the N-M interaction maps to the carboxy-terminal domain 3 of the N protein. To explore other domain interactions of the MHV N protein, we expressed a series of segments of the MHV N protein as fusions with green fluorescent protein (GFP) during the course of viral infection. We found that two of these GFP-N-domain fusion proteins were selectively packaged into virions as the result of tight binding to the N protein in the viral nucleocapsid, in a manner that did not involve association with either M protein or RNA. The nature of each type of binding was further explored through genetic analysis. Our results defined two strongly interacting regions of the N protein. One is the same domain 3 that is critical for M protein recognition during assembly. The other is domain N1b, which corresponds to the N-terminal domain that has been structurally characterized in detail for two other coronaviruses, infectious bronchitis virus and the severe acute respiratory syndrome coronavirus.


Subject(s)
Coronavirus Infections/metabolism , Coronavirus Infections/veterinary , Murine hepatitis virus/physiology , Nucleocapsid Proteins/chemistry , Nucleocapsid Proteins/metabolism , Rodent Diseases/metabolism , Amino Acid Sequence , Animals , Cell Line , Cell Membrane/metabolism , Cell Membrane/virology , Coronavirus Infections/virology , Coronavirus Nucleocapsid Proteins , Mice , Molecular Sequence Data , Murine hepatitis virus/chemistry , Murine hepatitis virus/genetics , Mutation , Nucleocapsid Proteins/genetics , Protein Binding , Protein Structure, Tertiary , Rodent Diseases/virology , Virus Assembly
20.
Proc Natl Acad Sci U S A ; 106(2): 582-7, 2009 Jan 13.
Article in English | MEDLINE | ID: mdl-19124777

ABSTRACT

Coronaviruses are enveloped viruses containing the largest reported RNA genomes. As a result of their pleomorphic nature, our structural insight into the coronavirion is still rudimentary, and it is based mainly on 2D electron microscopy. Here we report the 3D virion structure of coronaviruses obtained by cryo-electron tomography. Our study focused primarily on the coronavirus prototype murine hepatitis virus (MHV). MHV particles have a distinctly spherical shape and a relatively homogenous size ( approximately 85 nm envelope diameter). The viral envelope exhibits an unusual thickness (7.8 +/- 0.7 nm), almost twice that of a typical biological membrane. Focal pairs revealed the existence of an extra internal layer, most likely formed by the C-terminal domains of the major envelope protein M. In the interior of the particles, coiled structures and tubular shapes are observed, consistent with a helical nucleocapsid model. Our reconstructions provide no evidence of a shelled core. Instead, the ribonucleoprotein seems to be extensively folded onto itself, assuming a compact structure that tends to closely follow the envelope at a distance of approximately 4 nm. Focal contact points and thread-like densities connecting the envelope and the ribonucleoprotein are revealed in the tomograms. Transmissible gastroenteritis coronavirion tomograms confirm all the general features and global architecture observed for MHV. We propose a general model for the structure of the coronavirion in which our own and published observations are combined.


Subject(s)
Cryoelectron Microscopy , Electron Microscope Tomography , Murine hepatitis virus/ultrastructure , Virion/ultrastructure , Animals , Coronavirus , Mice , Murine hepatitis virus/chemistry , Nucleocapsid/chemistry , Nucleocapsid/ultrastructure , Viral Envelope Proteins/chemistry , Virion/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...