101
|
Reed ML, Howell G, Harrison SM, Spencer KA, Hiscox JA. Characterization of the nuclear export signal in the coronavirus infectious bronchitis virus nucleocapsid protein. J Virol 2007; 81:4298-304. [PMID: 17202223 PMCID: PMC1866115 DOI: 10.1128/jvi.02239-06] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2006] [Accepted: 12/27/2006] [Indexed: 01/13/2023] Open
Abstract
The nucleocapsid (N) protein of infectious bronchitis virus (IBV) localizes to the cytoplasm and nucleolus and contains an eight-amino-acid nucleolar retention motif. In this study, a leucine-rich nuclear export signal (NES) (291-LQLDGLHL-298) present in the C-terminal region of the IBV N protein was analyzed by using alanine substitution and deletion mutagenesis to investigate the relative contributions that leucine residues make to nuclear export and where these residues are located on the structure of the IBV N protein. The analysis indicated that Leu296 and Leu298 are required for efficient nuclear export of the protein. Structural information indicated that both of these amino acids are available for interaction with protein complexes involved in this process. However, export of N protein from the nucleus/nucleolus was not inhibited by leptomycin B treatment, indicating that N protein nuclear export is independent of the CRM1-mediated export pathway.
Collapse
Affiliation(s)
- Mark L Reed
- Institute of Molecular and Cellular Biology, Faculty of Biology Sciences, Garstang Building, University of Leeds, Leeds LS2 6JT, United Kingdom
| | | | | | | | | |
Collapse
|
102
|
Abstract
The nucleolus is a dynamic subnuclear structure that is crucial to the successful functioning of a cell. Its functions include ribosomal RNA synthesis, cell growth and cell-cycle control as well as responding to cellular stress. Recent studies show that the nucleolus is not a steady-state structure but instead is made up of numerous protein–protein and protein–nucleic-acid interactions that are constantly changing in response to the metabolic conditions of the cell. Many different viruses target the nucleolus to disrupt host-cell function and to recruit cellular proteins to aid in virus replication. The study of viral-protein trafficking to the nucleolus and the interaction of viral proteins with nucleolar proteins is providing many insights into the cell biology of the nucleolus. Because the nucleolus is fundamental to the life cycle of many viruses, disrupting the interaction between the nucleolus and the virus could lead to the design of novel therapeutic strategies.
RNA viruses, particularly positive-strand RNA viruses, interact with the nucleolus to usurp host-cell functions and recruit nucleolar proteins to facilitate virus replication. Here, Julian Hiscox reviews the latest data on RNA-virus interactions with this dynamic subnuclear structure. The nucleolus is a dynamic subnuclear structure with roles in ribosome subunit biogenesis, mediation of cell-stress responses and regulation of cell growth. The proteome and structure of the nucleolus are constantly changing in response to metabolic conditions. RNA viruses interact with the nucleolus to usurp host-cell functions and recruit nucleolar proteins to facilitate virus replication. Investigating the interactions between RNA viruses and the nucleolus will facilitate the design of novel anti-viral therapies, such as recombinant vaccines and therapeutic molecular interventions, and also contribute to a more detailed understanding of the cell biology of the nucleolus.
Collapse
Affiliation(s)
- Julian A Hiscox
- Institute of Molecular and Cellular Biology, Faculty of Biological Sciences, and Astbury Centre for Structural Molecular Biology, Garstang Building, University of Leeds, Leeds, LS2 9JT, UK.
| |
Collapse
|
103
|
Abreu JT, Resende JS, Flatschart RB, Folgueras-Flatschart AV, Mendes ACR, Martins NRS, Silva CBA, Ferreira BMC, Resende M. Molecular analysis of Brazilian infectious bronchitis field isolates by reverse transcription-polymerase chain reaction, restriction fragment length polymorphism, and partial sequencing of the N gene. Avian Dis 2007; 50:494-501. [PMID: 17274284 DOI: 10.1637/7525-030706r.1] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Molecular analysis of 15 Brazilian infectious bronchitis virus (IBV) isolates, obtained from clinical outbreaks of the disease in chickens (broilers or layers) in the state of Minas Gerais (Brazil) between 1972 and 1989, is reported. Using the N protein gene as target, IBVs were analyzed by reverse transcription-polymerase chain reaction/restriction fragment length polymorphism (RT-PCR/RFLP) with the restriction enzymes AvaII, HphI, Sau96I, and Tsp509I and cDNA sequencing. Results obtained from those isolates were compared to 19 sequences available in GenBank. N gene RFLP profiles, cDNA sequences, and predicted amino acid composition were used for the construction of dendrograms. Brazilian isolates were grouped into one distinct group. Identity of predicted N protein amino acid composition varied from 45% (between isolates G and 208) up to 99% (PM 1 and PM2), and, when compared to the other IBVs, the amino acid identity was from 42% (Q3/88 and G) up to 97% (D41 and PM1). The great genetic diversity was shown to occur before the official use of vaccination in Brazil and has remained thereafter.
Collapse
Affiliation(s)
- Josiane T Abreu
- Laboratório de Virologia Comparada, Departamento de Microbiologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, P.O. Box 486, Belo Horizonte, Minas Gerais, Brasil 31270-901
| | | | | | | | | | | | | | | | | |
Collapse
|
104
|
Harrison SM, Dove BK, Rothwell L, Kaiser P, Tarpey I, Brooks G, Hiscox JA. Characterisation of cyclin D1 down-regulation in coronavirus infected cells. FEBS Lett 2007; 581:1275-86. [PMID: 17359980 PMCID: PMC7094712 DOI: 10.1016/j.febslet.2007.02.039] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2006] [Revised: 01/30/2007] [Accepted: 02/13/2007] [Indexed: 01/02/2023]
Abstract
The positive strand RNA coronavirus, infectious bronchitis virus (IBV), induces a G2/M phase arrest and reduction in the G1 and G1/S phase transition regulator cyclin D1. Quantitative real-time RT-PCR and Western blot analysis demonstrated that cyclin D1 was reduced post-transcriptionally within infected cells independently of the cell-cycle stage at the time of infection. Confocal microscopy revealed that cyclin D1 decreased in IBV-infected cells as infection progressed and inhibition studies indicated that a population of cyclin D1 could be targeted for degradation by a virus mediated pathway. In contrast to the SARS-coronavirus, IBV nucleocapsid protein did not interact with cyclin D1.
Collapse
Affiliation(s)
- Sally M. Harrison
- Institute of Molecular and Cellular Biology, Faculty of Biological Sciences, University of Leeds, Leeds, LS2 9JT, UK
| | - Brian K. Dove
- Institute of Molecular and Cellular Biology, Faculty of Biological Sciences, University of Leeds, Leeds, LS2 9JT, UK
| | - Lisa Rothwell
- Institute for Animal Health (Compton Laboratory), UK
| | - Pete Kaiser
- Institute for Animal Health (Compton Laboratory), UK
| | | | - Gavin Brooks
- School of Pharmacy, University of Reading, Reading, UK
| | - Julian A. Hiscox
- Institute of Molecular and Cellular Biology, Faculty of Biological Sciences, University of Leeds, Leeds, LS2 9JT, UK
- Astbury Centre for Structural Molecular Biology, University of Leeds, Leeds, UK
| |
Collapse
|
105
|
Hodgson T, Britton P, Cavanagh D. Neither the RNA nor the proteins of open reading frames 3a and 3b of the coronavirus infectious bronchitis virus are essential for replication. J Virol 2007; 80:296-305. [PMID: 16352554 PMCID: PMC1317528 DOI: 10.1128/jvi.80.1.296-305.2006] [Citation(s) in RCA: 72] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Gene 3 of infectious bronchitis virus is tricistronic; open reading frames (ORFs) 3a and 3b encode two small nonstructural (ns) proteins, 3a and 3b, of unknown function, and a third, structural protein E, is encoded by ORF 3c. To determine if either the 3a or the 3b protein is required for replication, we first modified their translation initiation codons to prevent translation of the 3a and 3b proteins from recombinant infectious bronchitis viruses (rIBVs). Replication in primary chick kidney (CK) cells and in chicken embryos was not affected. In chicken tracheal organ cultures (TOCs), the recombinant rIBVs reached titers similar to those of the wild-type virus, but in the case of viruses lacking the 3a protein, the titer declined reproducibly earlier. Translation of the IBV E protein is believed to be initiated by internal entry of ribosomes at a structure formed by the sequences corresponding to ORFs 3a and 3b. To assess the necessity of this mechanism, we deleted most of the sequence representing 3a and 3b to produce a gene in which ORF 3c (E) was adjacent to the gene 3 transcription-associated sequence. Western blot analysis revealed that the recombinant IBV produced fivefold less E protein. Nevertheless, titers produced in CK cells, embryos, and TOCs were similar to those of the wild-type virus, although they declined earlier in TOCs, probably due to the absence of the 3a protein. Thus, neither the tricistronic arrangement of gene 3, the internal initiation of translation of E protein, nor the 3a and 3b proteins are essential for replication per se, suggesting that these proteins are accessory proteins that may have roles in vivo.
Collapse
Affiliation(s)
- Teri Hodgson
- Institute for Animal Health, Compton Laboratory, Compton, Newbury, Berkshire RG20 7NN, United Kingdom
| | | | | |
Collapse
|
106
|
Enjuanes L, Almazán F, Sola I, Zuñiga S. Biochemical aspects of coronavirus replication and virus-host interaction. Annu Rev Microbiol 2006; 60:211-30. [PMID: 16712436 DOI: 10.1146/annurev.micro.60.080805.142157] [Citation(s) in RCA: 157] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Infection by different coronaviruses (CoVs) causes alterations in the transcriptional and translational patterns, cell cycle, cytoskeleton, and apoptosis pathways of the host cells. In addition, CoV infection may cause inflammation, alter immune and stress responses, and modify the coagulation pathways. The balance between the up- and downregulated genes could explain the pathogenesis caused by these viruses. We review specific aspects of CoV-host interactions. CoV genome replication takes place in the cytoplasm in a membrane-protected microenvironment and may control the cell machinery by locating some of their proteins in the host cell nucleus. CoVs initiate translation by cap-dependent and cap-independent mechanisms. CoV transcription involves a discontinuous RNA synthesis (template switching) during the extension of a negative copy of the subgenomic mRNAs. The requirement for base-pairing during transcription has been formally demonstrated in arteriviruses and CoVs. CoV N proteins have RNA chaperone activity that may help initiate template switching. Both viral and cellular proteins are required for replication and transcription, and the role of selected proteins is addressed.
Collapse
Affiliation(s)
- Luis Enjuanes
- Department of Molecular and Cell Biology, CNB, CSIC, 28049 Madrid, Spain.
| | | | | | | |
Collapse
|
107
|
Fan Z, Zhuo Y, Tan X, Zhou Z, Yuan J, Qiang B, Yan J, Peng X, Gao GF. SARS-CoV nucleocapsid protein binds to hUbc9, a ubiquitin conjugating enzyme of the sumoylation system. J Med Virol 2006; 78:1365-73. [PMID: 16998888 PMCID: PMC7167196 DOI: 10.1002/jmv.20707] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
SARS‐CoV is a newly identified coronavirus (CoV) that causes severe acute respiratory syndrome (SARS). The SARS‐CoV nucleocapsid (N) protein is an important structural and functional protein. To identify cellular proteins that interact with the SARS‐CoV N protein and to elucidate the possible involvement of N protein in SARS‐CoV pathogenesis, a human lymphocyte cDNA library was screened using a yeast two‐hybrid system assay. hUbc9, a ubiquitin conjugating enzyme of sumoylation system, was found to interact specifically with the N protein, implying the post‐translational sumoylation of the N protein. Mapping studies localized the critical N sequences for this interaction to amino acids 170–210, which includes the SR‐rich motif. However, the consensus motif of sumoylation GK62EE in the N protein is not responsible for binding to hUbc9. Mutations of hUbc9 at the enzyme active site C93A or C93S severely impair the interaction with the N protein. The two proteins were also shown to colocalize in the cytoplasm of the transfected 293T cells. This is the first report demonstrating the interaction of hUbc9 with a structural protein of plus‐strand RNA viruses, indicating a new drug target for SARS‐CoV. J. Med. Virol. 78:1365–1373, 2006. © 2006 Wiley‐Liss, Inc.
Collapse
Affiliation(s)
- Zheng Fan
- Center for Molecular Virology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, China
| | - Yue Zhuo
- Center for Molecular Virology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, China
- Department of Biochemistry, Anhui Agricultural University, Hefei, China
| | - Xinyu Tan
- National Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Zhi Zhou
- Center for Molecular Virology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, China
- College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Jiangang Yuan
- National Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Boqin Qiang
- National Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jinghua Yan
- Center for Molecular Virology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, China
| | - Xiaozhong Peng
- National Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - George F. Gao
- Center for Molecular Virology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
108
|
Abstract
Coronaviruses are large, enveloped RNA viruses of both medical and veterinary importance. Interest in this viral family has intensified in the past few years as a result of the identification of a newly emerged coronavirus as the causative agent of severe acute respiratory syndrome (SARS). At the molecular level, coronaviruses employ a variety of unusual strategies to accomplish a complex program of gene expression. Coronavirus replication entails ribosome frameshifting during genome translation, the synthesis of both genomic and multiple subgenomic RNA species, and the assembly of progeny virions by a pathway that is unique among enveloped RNA viruses. Progress in the investigation of these processes has been enhanced by the development of reverse genetic systems, an advance that was heretofore obstructed by the enormous size of the coronavirus genome. This review summarizes both classical and contemporary discoveries in the study of the molecular biology of these infectious agents, with particular emphasis on the nature and recognition of viral receptors, viral RNA synthesis, and the molecular interactions governing virion assembly.
Collapse
Affiliation(s)
- Paul S Masters
- Wadsworth Center, New York State Department of Health, Albany, 12201, USA
| |
Collapse
|
109
|
Bussmann BM, Reiche S, Jacob LH, Braun JM, Jassoy C. Antigenic and cellular localisation analysis of the severe acute respiratory syndrome coronavirus nucleocapsid protein using monoclonal antibodies. Virus Res 2006; 122:119-26. [PMID: 16920216 PMCID: PMC7114340 DOI: 10.1016/j.virusres.2006.07.005] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2006] [Revised: 07/07/2006] [Accepted: 07/12/2006] [Indexed: 01/19/2023]
Abstract
A member of the family of coronaviruses has previously been identified as the cause of the severe acute respiratory syndrome (SARS). In this study, several monoclonal antibodies against the nucleocapsid protein have been generated to examine distribution of the nucleocapsid in virus-infected cells and to study antigenic regions of the protein. Confocal microscopic analysis identified nucleocapsids packaged in vesicles in the perinuclear area indicating viral synthesis at the endoplasmic reticulum and Golgi apparatus. The monoclonal antibodies bound to the central and carboxyterminal half of the nucleocapsid protein indicating prominent exposure and immunogenicity of this part of the protein. Antibodies recognised both linear and conformational epitopes. Predictions of antigenicity using mathematical modelling based on hydrophobicity analysis of SARS nucleoprotein could not be confirmed fully. Antibody binding to discontinuous peptides provides evidence that amino acids 274–283 and 373–382 assemble to a structural unit particularly rich in basic amino acids. In addition, amino acids 286–295, 316–325 and 361–367 that represent the epitope recognised by monoclonal antibody 6D11C1 converge indicating a well-structured C-terminal region of the SARS virus nucleocapsid protein and functional relationship of the peptide regions involved. Alternatively, dimerisation of the nucleocapsid protein may result in juxtaposition of the amino acid sequences 316–325 and 361–367 on one nucleoprotein molecule to amino acid 286–295 on the second peptide. The monoclonal antibodies will be available to assess antigenicity and immunological variabilities between different SARS CoV strains.
Collapse
Affiliation(s)
- Bianca M. Bussmann
- Institute of Virology, University of Leipzig, Johannisallee 30, 04103 Leipzig, Germany
| | - Sven Reiche
- Institute of Virology, University of Leipzig, Johannisallee 30, 04103 Leipzig, Germany
| | - Lotta H. Jacob
- Institute of Virology, University of Leipzig, Johannisallee 30, 04103 Leipzig, Germany
| | - Jan Matthias Braun
- Institute of Clinical Immunology and Transfusion Medicine (IKIT) and Interdisciplinary Centre of Clinical Research (IZKF), Faculty of Medicine, University of Leipzig, Johannisallee 30, 04103 Leipzig, Germany
| | - Christian Jassoy
- Institute of Virology, University of Leipzig, Johannisallee 30, 04103 Leipzig, Germany
- Corresponding author. Tel.: +49 341 9714314; fax: +49 341 9714309.
| |
Collapse
|
110
|
Dove BK, You JH, Reed ML, Emmett SR, Brooks G, Hiscox JA. Changes in nucleolar morphology and proteins during infection with the coronavirus infectious bronchitis virus. Cell Microbiol 2006; 8:1147-57. [PMID: 16819967 PMCID: PMC7162191 DOI: 10.1111/j.1462-5822.2006.00698.x] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
The nucleolus is a dynamic subnuclear structure involved in ribosome subunit biogenesis, cell cycle control and mediating responses to cell stress, among other functions. While many different viruses target proteins to the nucleolus and recruit nucleolar proteins to facilitate virus replication, the effect of infection on the nucleolus in terms of morphology and protein content is unknown. Previously we have shown that the coronavirus nucleocapsid protein will localize to the nucleolus. In this study, using the avian infectious bronchitis coronavirus, we have shown that virus infection results in a number of changes to the nucleolus both in terms of gross morphology and protein content. Using confocal microscopy coupled with fluorescent labelled nucleolar marker proteins we observed changes in the morphology of the nucleolus including an enlarged fibrillar centre. We found that the tumour suppressor protein, p53, which localizes normally to the nucleus and nucleolus, was redistributed predominately to the cytoplasm.
Collapse
Affiliation(s)
- Brian K Dove
- Institute of Molecular and Cellular Biology, Faculty of Biological Sciences, University of Leeds, Leeds, UK
| | | | | | | | | | | |
Collapse
|
111
|
Peng H, Yang LT, Wang LY, Li J, Huang J, Lu ZQ, Koup RA, Bailer RT, Wu CY. Long-lived memory T lymphocyte responses against SARS coronavirus nucleocapsid protein in SARS-recovered patients. Virology 2006; 351:466-75. [PMID: 16690096 PMCID: PMC7111820 DOI: 10.1016/j.virol.2006.03.036] [Citation(s) in RCA: 112] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2006] [Revised: 03/09/2006] [Accepted: 03/22/2006] [Indexed: 01/28/2023]
Abstract
The nucleocapsid (N) protein is a structural component of severe acute respiratory syndrome (SARS) coronavirus (SARS-CoV) and can induce antibody responses in SARS patients during infection. However, it is not known whether SARS-CoV N protein can induce a long persistence of memory T-cell response in human. In this study, we found that peripheral blood mononuclear cells (PBMCs) from fully recovered SARS individuals rapidly produced IFN-gamma and IL-2 following stimulation with a pool of overlapping peptides that cover the entire N protein sequence. The N-specific IFN-gamma(+)CD4(+) T cells were mainly composed of CD45RA(-)CCR7(+)CD62L(-) cells, whereas IFN-gamma(+)CD8(+) memory T cells were mostly contained within CD45RA(+)CCR7(-)CD62L(-) cell population. Epitope mapping study indicated that a cluster of overlapping peptides located in the C-terminal region (amino acids [aa] 331 to 362) of N protein contained at least two different T-cell epitopes. The results indicated that human memory T-cell responses specific for SARS-CoV N protein could persist for 2 years in the absence of antigen, which would be a valuable for the design of effective vaccines against SARS-CoV and for basic studies of human T-cell memory.
Collapse
Affiliation(s)
- Hui Peng
- Department of Immunology, Zhongshan Medical School, Sun Yat-sen University, No. 74 Zhongshan Road II, Guangzhou 510089, China
| | | | | | | | | | | | | | | | | |
Collapse
|
112
|
Oh W, Yang MR, Lee EW, Park KM, Pyo S, Yang JS, Lee HW, Song J. Jab1 mediates cytoplasmic localization and degradation of West Nile virus capsid protein. J Biol Chem 2006; 281:30166-74. [PMID: 16882664 DOI: 10.1074/jbc.m602651200] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
The clinical manifestations of West Nile virus (WNV), a member of the Flavivirus family, include febrile illness, sporadic encephalitis, and paralysis. The capsid (Cp) of WNV is thought to participate in these processes by inducing apoptosis through mitochondrial dysfunction and activation of caspase-9 and caspase-3. To further identify the molecular mechanism of the WNV capsid protein (WNVCp), yeast two-hybrid assays were employed using WNV-Cp as bait. Jab1, the fifth subunit of the COP9 signalosome, was subsequently identified as a molecule that interacts with WNVCp. Immunoprecipitation and glutathione S-transferase pulldown assays confirmed that direct interaction could occur between WNVCp and Jab1. Immunofluorescence microscopy demonstrated that the overexpressed WNVCp, which localized to the nucleolus, was translocated to the cytoplasm upon its co-expression with Jab1. When treated with leptomycin B, Jab1-facilitated nuclear exclusion of WNVCp was prevented, which indicated that the CRM1 complex is required for Jab1-facilitated nuclear export of WNVCp. Moreover, Jab1 promoted the degradation of WNVCp in a proteasome-dependent way. Consistent with this, WNVCp-mediated cell cycle arrest at the G(2) phase in H1299 was prevented by exogenous Jab1. Finally, an analysis of WNVCp deletion mutants indicated that the first 15 amino acids were required for interaction with Jab1. Furthermore, the double-point mutant of the WNVCp, P5A/P8A, was incapable of binding to Jab1. These results indicate that Jab1 has a potential protective effect against pathogenic WNVCp and might provide a novel target site for the treatment of disease caused by WNV.
Collapse
Affiliation(s)
- Wonkyung Oh
- Department of Food Science and Biotechnology, Sungkyunkwan University, Suwon 440-746, Republic of Korea
| | | | | | | | | | | | | | | |
Collapse
|
113
|
Oh WK, Song J. Hsp70 functions as a negative regulator of West Nile virus capsid protein through direct interaction. Biochem Biophys Res Commun 2006; 347:994-1000. [PMID: 16854374 PMCID: PMC7117540 DOI: 10.1016/j.bbrc.2006.06.190] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2006] [Accepted: 06/29/2006] [Indexed: 12/15/2022]
Abstract
West Nile virus (WNV) is a member of the Flavivirus family and induces febrile illness, sporadic encephalitis, and paralysis. The capsid (Cp) of WNV is thought to play a role in inducing these symptoms through caspase-3- and caspase-9-dependent apoptosis. Using WNVCp as bait for a yeast two-hybrid assay, we identified that Hsp70 interacted with WNVCp. The interaction between Hsp70 and WNVCp was further substantiated using purified proteins. Deletion analysis of Hsp70 indicated that WNVCp could bind to the substrate binding domain of Hsp70. The presence of WNVCp in the Hsp70-dependent folding system inhibited the refolding of β-galactosidase (β-gal), which showed that WNVCp might function as a negative regulator of Hsp70. Finally, the cytotoxic effect of WNVCp in 293T cells was prevented by ectopic Hsp70, suggesting a negative regulatory role of Hsp70 on WNVCp. Our findings suggest a possible negative regulatory role of Hps70 in the pathway of WNV infection.
Collapse
Affiliation(s)
- Won-kyung Oh
- Department of Food Science and Biotechnology, Faculty of Life Science and Technology Sungkyunkwan University, Suwon, Kyunggi-do 440-746, Republic of Korea
| | | |
Collapse
|
114
|
Reed ML, Dove BK, Jackson RM, Collins R, Brooks G, Hiscox JA. Delineation and modelling of a nucleolar retention signal in the coronavirus nucleocapsid protein. Traffic 2006; 7:833-48. [PMID: 16734668 PMCID: PMC7488588 DOI: 10.1111/j.1600-0854.2006.00424.x] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Unlike nuclear localization signals, there is no obvious consensus sequence for the targeting of proteins to the nucleolus. The nucleolus is a dynamic subnuclear structure which is crucial to the normal operation of the eukaryotic cell. Studying nucleolar trafficking signals is problematic as many nucleolar retention signals (NoRSs) are part of classical nuclear localization signals (NLSs). In addition, there is no known consensus signal with which to inform a study. The avian infectious bronchitis virus (IBV), coronavirus nucleocapsid (N) protein, localizes to the cytoplasm and the nucleolus. Mutagenesis was used to delineate a novel eight amino acid motif that was necessary and sufficient for nucleolar retention of N protein and colocalize with nucleolin and fibrillarin. Additionally, a classical nuclear export signal (NES) functioned to direct N protein to the cytoplasm. Comparison of the coronavirus NoRSs with known cellular and other viral NoRSs revealed that these motifs have conserved arginine residues. Molecular modelling, using the solution structure of severe acute respiratory (SARS) coronavirus N‐protein, revealed that this motif is available for interaction with cellular factors which may mediate nucleolar localization. We hypothesise that the N‐protein uses these signals to traffic to and from the nucleolus and the cytoplasm.
Collapse
Affiliation(s)
- Mark L. Reed
- Institute of Molecular and Cellular Biology, University of Leeds, Leeds, UK
| | - Brian K. Dove
- Institute of Molecular and Cellular Biology, University of Leeds, Leeds, UK
| | - Richard M. Jackson
- Institute of Molecular and Cellular Biology, University of Leeds, Leeds, UK
- Astbury Centre for Structural Molecular Biology, University of Leeds, Leeds, UK
| | - Rebecca Collins
- Institute of Molecular and Cellular Biology, University of Leeds, Leeds, UK
| | - Gavin Brooks
- School of Pharmacy, University of Reading, Reading, UK
| | - Julian A. Hiscox
- Institute of Molecular and Cellular Biology, University of Leeds, Leeds, UK
- Astbury Centre for Structural Molecular Biology, University of Leeds, Leeds, UK
| |
Collapse
|
115
|
Kim H, Lee SH, Han JH, Lee JA, Cheang YH, Chang DJ, Lee YS, Kaang BK. A nucleolar protein ApLLP induces ApC/EBP expression required for long-term synaptic facilitation in aplysia neurons. Neuron 2006; 49:707-18. [PMID: 16504946 DOI: 10.1016/j.neuron.2006.01.035] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2005] [Revised: 06/07/2005] [Accepted: 01/26/2006] [Indexed: 10/25/2022]
Abstract
In Aplysia, long-term synaptic plasticity is induced by serotonin (5-HT) or neural activity and requires gene expression. Here, we demonstrate that ApLLP, a novel nucleolus protein, is critically involved in both long-term facilitation (LTF) and behavioral sensitization. Membrane depolarization induced ApLLP expression, which activated ApC/EBP expression through a direct binding to CRE. LTF was produced by a single pulse of 5-HT 30 min after the membrane depolarization. This LTF was blocked when either ApLLP or ApC/EBP were blocked by specific antibodies. In contrast, ApLLP overexpression induced LTF in response to a single 5-HT treatment. Simultaneously, a siphon noxious stimulus (SNS) to intact Aplysia induced ApLLP and ApC/EBP expression, and single tail shock 30 min after SNS transformed short-term sensitization to long-term sensitization of siphon withdrawal reflex. These results suggest that ApLLP is an activity-dependent transcriptional activator that switches short-term facilitation to long-term facilitation.
Collapse
Affiliation(s)
- Hyoung Kim
- National Research Laboratory, Institute of Molecular Biology and Genetics, School of Biological Sciences, College of Natural Sciences, Seoul National University, San 56-1 Silim-dong Kwanak-gu, Seoul 151-742, Korea
| | | | | | | | | | | | | | | |
Collapse
|
116
|
Perlman S, Holmes KV. Infectious bronchitis coronavirus induces cell-cycle perturbations. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2006; 581:357-62. [PMID: 17037559 PMCID: PMC7123543 DOI: 10.1007/978-0-387-33012-9_63] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/02/2022]
Affiliation(s)
- Stanley Perlman
- Department of Pediatrics, University of Iowa, 52242 Iowa City, IA USA
| | - Kathryn V. Holmes
- Department of Microbiology, University of Colorado Health Sciences Center at Fitzsimons, 80045-8333 Aurora, CO USA
| |
Collapse
|
117
|
You J, Dove BK, Enjuanes L, DeDiego ML, Alvarez E, Howell G, Heinen P, Zambon M, Hiscox JA. Subcellular localization of the severe acute respiratory syndrome coronavirus nucleocapsid protein. J Gen Virol 2005; 86:3303-3310. [PMID: 16298975 DOI: 10.1099/vir.0.81076-0] [Citation(s) in RCA: 69] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
The coronavirus nucleocapsid (N) protein is a viral RNA-binding protein with multiple functions in terms of virus replication and modulating cell signalling pathways. N protein is composed of three distinct regions containing RNA-binding motif(s), and appropriate signals for modulating cell signalling. The subcellular localization of severe acute respiratory syndrome coronavirus (SARS-CoV) N protein was studied. In infected cells, SARS-CoV N protein localized exclusively to the cytoplasm. In contrast to the avian coronavirus N protein, overexpressed SARS-CoV N protein remained principally localized to the cytoplasm, with very few cells exhibiting nucleolar localization. Bioinformatic analysis and deletion mutagenesis coupled to confocal microscopy and live-cell imaging, revealed that SARS-CoV N protein regions I and III contained nuclear localization signals and region II contained a nucleolar retention signal. However, cytoplasmic localization was directed by region III and was the dominant localization signal in the protein.
Collapse
Affiliation(s)
- Jaehwan You
- Institute of Molecular and Cellular Biology, Faculty of Biological Sciences, Garstang Building, University of Leeds, Leeds LS2 9JT, UK
| | - Brian K Dove
- Institute of Molecular and Cellular Biology, Faculty of Biological Sciences, Garstang Building, University of Leeds, Leeds LS2 9JT, UK
| | - Luis Enjuanes
- Department of Molecular and Cell Biology, Centro Nacional de Biotecnología (CNB, CSIC), Campus Univ. Autonoma, 3 Darwin Street, Cantoblanco, 28049 Madrid, Spain
| | - Marta L DeDiego
- Department of Molecular and Cell Biology, Centro Nacional de Biotecnología (CNB, CSIC), Campus Univ. Autonoma, 3 Darwin Street, Cantoblanco, 28049 Madrid, Spain
| | - Enrique Alvarez
- Department of Molecular and Cell Biology, Centro Nacional de Biotecnología (CNB, CSIC), Campus Univ. Autonoma, 3 Darwin Street, Cantoblanco, 28049 Madrid, Spain
| | - Gareth Howell
- Astbury Centre for Structural Molecular Biology, University of Leeds, Leeds LS2 9JT, UK
| | - Paul Heinen
- Health Protection Agency, London NW9 5HT, UK
| | | | - Julian A Hiscox
- Astbury Centre for Structural Molecular Biology, University of Leeds, Leeds LS2 9JT, UK
- Institute of Molecular and Cellular Biology, Faculty of Biological Sciences, Garstang Building, University of Leeds, Leeds LS2 9JT, UK
| |
Collapse
|
118
|
Lee C, Hodgins D, Calvert JG, Welch SKW, Jolie R, Yoo D. Mutations within the nuclear localization signal of the porcine reproductive and respiratory syndrome virus nucleocapsid protein attenuate virus replication. Virology 2005; 346:238-50. [PMID: 16330065 PMCID: PMC7172752 DOI: 10.1016/j.virol.2005.11.005] [Citation(s) in RCA: 82] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2005] [Revised: 09/27/2005] [Accepted: 11/04/2005] [Indexed: 11/17/2022]
Abstract
Porcine reproductive and respiratory syndrome virus (PRRSV) is an RNA virus replicating in the cytoplasm, but the nucleocapsid (N) protein is specifically localized to the nucleus and nucleolus in virus-infected cells. A 'pat7' motif of 41-PGKK(N/S)KK has previously been identified in the N protein as the functional nuclear localization signal (NLS); however, the biological consequences of N protein nuclear localization are unknown. In the present study, the role of N protein nuclear localization during infection was investigated in pigs using an NLS-null mutant virus. When two lysines at 43 and 44 at the NLS locus were substituted to glycines, the modified NLS with 41-PGGGNKK restricted the N protein to the cytoplasm. This NLS-null mutation was introduced into a full-length infectious cDNA clone of PRRSV. Upon transfection of cells, the NLS-null full-length clone induced cytopathic effects and produced infectious progeny. The NLS-null virus grew to a titer 100-fold lower than that of wild-type virus. To examine the response to NLS-null PRRSV in the natural host, three groups of pigs, consisting of seven animals per group, were intranasally inoculated with wild-type, placebo, or NLS-null virus, and the animals were maintained for 4 weeks. The NLS-null-infected pigs had a significantly shorter mean duration of viremia than wild-type-infected pigs but developed significantly higher titers of neutralizing antibodies. Mutations occurred at the NLS locus in one pig during viremia, and four types of mutations were identified: 41-PGRGNKK, 41-PGGRNKK, and 41-PGRRNKK, and 41-PGKKSKK. Both wild-type and NLS-null viruses persisted in the tonsils for at least 4 weeks, and the NLS-null virus persisting in the tonsils was found to be mutated to either 41-PGRGNKK or 41-PGGRNKK in all pigs. No other mutation was found in the N gene. All types of reversions which occurred during viremia and persistence were able to translocate the mutated N proteins to the nucleus, indicating a strong selection pressure for reversion at the NLS locus of the N protein in vivo. Reversions from NLS-null to functional NLS in the tonsils suggest a possible correlation of viral persistence with N protein nuclear localization. These results show that N protein nuclear localization is non-essential for PRRSV multiplication but may play an important role in viral attenuation and in pathogenesis in vivo.
Collapse
Affiliation(s)
- Changhee Lee
- Department of Pathobiology, Ontario Veterinary College, University of Guelph, Guelph, Ontario, Canada N1G 2W1
| | - Douglas Hodgins
- Department of Pathobiology, Ontario Veterinary College, University of Guelph, Guelph, Ontario, Canada N1G 2W1
| | | | | | - Rika Jolie
- Pfizer Animal Health, Kalamazoo, MI 49001, USA
| | - Dongwan Yoo
- Department of Pathobiology, Ontario Veterinary College, University of Guelph, Guelph, Ontario, Canada N1G 2W1
- Corresponding author. Fax: +1 519 824 5930.
| |
Collapse
|
119
|
Yuan X, Yao Z, Shan Y, Chen B, Yang Z, Wu J, Zhao Z, Chen J, Cong Y. Nucleolar localization of non-structural protein 3b, a protein specifically encoded by the severe acute respiratory syndrome coronavirus. Virus Res 2005; 114:70-9. [PMID: 16046244 PMCID: PMC7114190 DOI: 10.1016/j.virusres.2005.06.001] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2005] [Revised: 06/01/2005] [Accepted: 06/06/2005] [Indexed: 12/20/2022]
Abstract
The open reading frame 3 (ORF3) of the severe acute respiratory syndrome coronavirus (SARS-CoV) genome encodes a predicted 154-amino acid protein, which lacks similarities to any known protein, and is named 3b. In this study, it was shown that 3b protein was predominately localized to nucleus with EGFP tag at its N- or C-terminus. The localization patterns were similar in different transfected cells. Immuno-fluorescence assay revealed that 3b protein was co-localized well with C23 in nucleolus. C23, B23 and fibrillarin all are important nucleolar proteins, which localize in the region of the nucleolus. Co-transfection of p3b-EGFP with pC23-DsRed, pB23-DsRed and pfibrillarin-DsRed further confirmed 3b's nucleolus localization. With construction of serial truncated mutants of 3b, a region (residues 134-154 aa) responsible for nucleolar localization was determinated in 3b protein. These results provide a new insight for further functional studies of SARS-CoV 3b protein.
Collapse
Key Words
- dmem, dulbecco's modified eagel's medium
- ecl, enhanced chemiluminescence
- egfp, enhanced green fluorescent protein
- n, nucleocapsid
- nls, nuclear localization signal
- nols, nucleolus localization signal
- orf, open reading frame
- pbs, phosphate-buffered saline
- sars, severe acute respiratory syndrome
- sars-cov, sars coronavirus
Collapse
Affiliation(s)
- Xiaoling Yuan
- Department of Pathophysiology, Beijing Institute of Radiation Medicine, No. 27 Taiping Road, Beijing 100850, China
| | | | | | | | | | | | | | | | | |
Collapse
|
120
|
Fang X, Ye L, Timani KA, Li S, Zen Y, Zhao M, Zheng H, Wu Z. Peptide domain involved in the interaction between membrane protein and nucleocapsid protein of SARS-associated coronavirus. BMB Rep 2005; 38:381-5. [PMID: 16053703 DOI: 10.5483/bmbrep.2005.38.4.381] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Severe acute respiratory syndrome (SARS) is an emerging infectious disease associated with a novel coronavirus (CoV) that was identified and molecularly characterized in 2003. Previous studies on various coronaviruses indicate that protein-protein interactions amongst various coronavirus proteins are critical for viral assembly and morphogenesis. It is necessary to elucidate the molecular mechanism of SARS-CoV replication and rationalize the anti-SARS therapeutic intervention. In this study, we employed an in vitro GST pull-down assay to investigate the interaction between the membrane (M) and the nucleocapsid (N) proteins. Our results show that the interaction between the M and N proteins does take place in vitro. Moreover, we provide an evidence that 12 amino acids domain (194-205) in the M protein is responsible for binding to N protein. Our work will help shed light on the molecular mechanism of the virus assembly and provide valuable information pertaining to rationalization of future anti-viral strategies.
Collapse
Affiliation(s)
- Xiaonan Fang
- State Key Laboratory of Virology, College of Life Science, Wuhan University, Wuhan, Hubei 430072, China
| | | | | | | | | | | | | | | |
Collapse
|
121
|
Rowland RRR, Chauhan V, Fang Y, Pekosz A, Kerrigan M, Burton MD. Intracellular localization of the severe acute respiratory syndrome coronavirus nucleocapsid protein: absence of nucleolar accumulation during infection and after expression as a recombinant protein in vero cells. J Virol 2005; 79:11507-12. [PMID: 16103202 PMCID: PMC1193611 DOI: 10.1128/jvi.79.17.11507-11512.2005] [Citation(s) in RCA: 65] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The nucleocapsid (N) protein of several members within the order Nidovirales localizes to the nucleolus during infection and after transfection of cells with N genes. However, confocal microscopy of N protein localization in Vero cells infected with the severe acute respiratory syndrome coronavirus (SARS-CoV) or transfected with the SARS-CoV N gene failed to show the presence of N in the nucleoplasm or nucleolus. Amino acids 369 to 389, which contain putative nuclear localization signal (NLS) and nucleolar localization signal motifs, failed to restore nuclear localization to an NLS-minus mutant Rev protein. These data indicate that nuclear localization is not a conserved property among all nidoviruses.
Collapse
Affiliation(s)
- Raymond R R Rowland
- Diagnostic Medicine and Pathobiology, Kansas State University, Manhattan, 66506, USA.
| | | | | | | | | | | |
Collapse
|
122
|
Takiuchi E, Stipp DT, Alfieri AF, Alfieri AA. Improved detection of bovine coronavirus N gene in faeces of calves infected naturally by a semi-nested PCR assay and an internal control. J Virol Methods 2005; 131:148-54. [PMID: 16182383 PMCID: PMC7112777 DOI: 10.1016/j.jviromet.2005.08.005] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2005] [Revised: 08/12/2005] [Accepted: 08/18/2005] [Indexed: 11/29/2022]
Abstract
Bovine coronavirus (BCoV), a positive sense single-stranded RNA virus, is an important causative agent of neonatal diarrhoea in calves from beef and dairy cattle worldwide. The routine detection and diagnosis of BCoV have been mainly dependent on assays with low sensitivity. The aim of the present study was to develop and evaluate a semi-nested PCR (SN-PCR) to amplify a 251 bp fragment of BCoV N gene from fresh (n = 25) and frozen (n = 25) diarrhoeic faecal samples of naturally infected calves. To improve detection of BCoV in faecal samples by the SN-PCR an internal control was developed, and the results were compared with a conventional RT-PCR assay. The rates of positive samples by SN-PCR and RT-PCR were 24% (12/50) and 8% (4/50), respectively (K = 0.43). Only fresh samples were positive in RT-PCR while the SN-PCR detected BCoV in both fresh and frozen faecal samples. The sensitivity of SN-PCR was determined by 10-fold serial dilutions of the BCoV Kakegawa strain (HA titre: 256) that was detected until 10−7 dilution. The specificity of the amplicons was assessed by restriction fragment length polymorphism and sequence analysis. The inclusion of an internal control provides a way to detect assay inhibition in faecal samples and failure of nucleic acid extraction that allow reduction of the number of false-negative results.
Collapse
Affiliation(s)
| | | | | | - Amauri A. Alfieri
- Corresponding author. Tel.: +55 43 3371 4485; fax: +55 43 3371 4714.
| |
Collapse
|
123
|
Timani KA, Liao Q, Ye L, Zeng Y, Liu J, Zheng Y, Ye L, Yang X, Lingbao K, Gao J, Zhu Y. Nuclear/nucleolar localization properties of C-terminal nucleocapsid protein of SARS coronavirus. Virus Res 2005; 114:23-34. [PMID: 15992957 PMCID: PMC7114095 DOI: 10.1016/j.virusres.2005.05.007] [Citation(s) in RCA: 82] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2005] [Revised: 05/10/2005] [Accepted: 05/17/2005] [Indexed: 01/17/2023]
Abstract
A novel coronavirus (CoV) has recently been identified as the aetiological agent of severe acute respiratory syndrome (SARS). Nucleocapsid (N) proteins of the Coronaviridae family have no discernable homology, but they share a common nucleolar-cytoplasmic distribution pattern. There are three putative nuclear localization signal (NLS) motifs present in the N. To determine the role of these putative NLSs in the intracellular localization of the SARS–CoV N, we performed a confocal microscopy analysis using rabbit anti-N antisera. In this report, we show that the wild type N was distributed mainly in the cytoplasm. The N-terminal of the N, which contains the NLS1 (aa38–44), was localized to the nucleus. The C-terminus of the N, which contains both NLS2 (aa257–265) and NLS3 (aa369–390) was localized to the cytoplasm and the nucleolus. Results derived from analysis of various deletion mutations show that the region containing amino acids 226–289 is able to mediate nucleolar localization. The deletion of two hydrophobic regions that flanked the NLS3 recovered its activity and localized to the nucleus. Furthermore, deletion of leucine rich region (220-LALLLLDRLNRL) resulted in the accumulation of N to the cytoplasm and nucleolus, and when fusing this peptide to EGFP localization was cytoplasmic, suggesting that the N may act as a shuttle protein. Differences in nuclear/nucleolar localization properties of N from other members of coronavirus family suggest a unique function for N, which may play an important role in the pathogenesis of SARS.
Collapse
Affiliation(s)
- Khalid Amine Timani
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan, Hubei Province 430072, PR China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
124
|
Jonassen CM, Kofstad T, Larsen IL, Løvland A, Handeland K, Follestad A, Lillehaug A. Molecular identification and characterization of novel coronaviruses infecting graylag geese (Anser anser), feral pigeons (Columbia livia) and mallards (Anas platyrhynchos). J Gen Virol 2005; 86:1597-1607. [PMID: 15914837 DOI: 10.1099/vir.0.80927-0] [Citation(s) in RCA: 111] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
In light of the finding of a previously unknown coronavirus as the aetiology of the severe acute respiratory syndrome (SARS), it is probable that other coronaviruses, than those recognized to date, are circulating in animal populations. Here, the results of a screening for coronavirus are presented, using a universal coronavirus RT-PCR, of the bird species graylag goose (Anser anser), feral pigeon (Columbia livia) and mallard (Anas platyrhynchos). Coronaviruses were found in cloacal swab samples from all the three bird species. In the graylag goose, 40 of 163 sampled birds were coronavirus positive, whereas two of 100 sampled pigeons and one of five sampled mallards tested positive. The infected graylag geese showed lower body weights compared with virus-negative birds, suggesting clinical significance of the infection. Phylogenetic analyses performed on the replicase gene and nucleocapsid protein sequences, indicated that the novel coronaviruses described in the present study all branch off from group III coronaviruses. All the novel avian coronaviruses harboured the conserved s2m RNA structure in their 3′ untranslated region, like other previously described group III coronaviruses, and like the SARS coronavirus. Sequencing of the complete nucleocapsid gene and downstream regions of goose and pigeon coronaviruses, evidenced the presence of two additional open reading frames for the goose coronavirus with no sequence similarity to known proteins, but with predicted transmembrane domains for one of the encoded proteins, and one additional open reading frame for the pigeon coronavirus, with a predicted transmembrane domain, downstream of the nucleocapsid gene.
Collapse
Affiliation(s)
| | - Tone Kofstad
- Section for Virology and Serology, National Veterinary Institute, PO Box 8156 Dep., N-0033 Oslo, Norway
| | - Inger-Lise Larsen
- Section for Virology and Serology, National Veterinary Institute, PO Box 8156 Dep., N-0033 Oslo, Norway
| | - Atle Løvland
- Section for Pathology, National Veterinary Institute, PO Box 8156 Dep., N-0033 Oslo, Norway
| | - Kjell Handeland
- Section for Wildlife Diseases, National Veterinary Institute, PO Box 8156 Dep., N-0033 Oslo, Norway
| | - Arne Follestad
- Norwegian Institute for Nature Research, Tungasletta 2, N-7485 Trondheim, Norway
| | - Atle Lillehaug
- Section for Wildlife Diseases, National Veterinary Institute, PO Box 8156 Dep., N-0033 Oslo, Norway
| |
Collapse
|
125
|
Li FQ, Xiao H, Tam JP, Liu D. Sumoylation of the nucleocapsid protein of severe acute respiratory syndrome coronavirus. FEBS Lett 2005; 579:2387-96. [PMID: 15848177 PMCID: PMC7094623 DOI: 10.1016/j.febslet.2005.03.039] [Citation(s) in RCA: 75] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2004] [Revised: 03/08/2005] [Accepted: 03/09/2005] [Indexed: 11/30/2022]
Abstract
Severe acute respiratory syndrome coronavirus (SARS‐CoV) encodes a highly basic nucleocapsid (N) protein of 422 amino acids. Similar to other coronavirus N proteins, SARS‐CoV N protein is predicted to be phosphorylated and may contain nuclear localization signals, serine/arginine‐rich motif, RNA binding domain and regions responsible for self‐association and homo‐oligomerization. In this study, we demonstrate that the protein is posttranslationally modified by covalent attachment to the small ubiquitin‐like modifier. The major sumoylation site was mapped to the 62lysine residue of the N protein. Further expression and characterization of wild type N protein and K62A mutant reveal that sumoylation of the N protein drastically promotes its homo‐oligomerization, and plays certain roles in the N protein‐mediated interference of host cell division. This is the first report showing that a coronavirus N protein undergoes posttranslational modification by sumoylation, and the functional implication of this modification in the formation of coronavirus ribouncleoprotein complex, virion assembly and virus–host interactions.
Collapse
Affiliation(s)
- Frank Qisheng Li
- School of Biological Sciences, Nanyang Technological University, 60 Nanyang Drive, Singapore 637551, Singapore
| | - Han Xiao
- Institute of Molecular and Cell Biology, 61 Biopolis Drive, Proteos, Singapore 138673, Singapore
| | - James P. Tam
- School of Biological Sciences, Nanyang Technological University, 60 Nanyang Drive, Singapore 637551, Singapore
| | - D.X. Liu
- School of Biological Sciences, Nanyang Technological University, 60 Nanyang Drive, Singapore 637551, Singapore
- Institute of Molecular and Cell Biology, 61 Biopolis Drive, Proteos, Singapore 138673, Singapore
| |
Collapse
|
126
|
Tan YJ, Lim SG, Hong W. Characterization of viral proteins encoded by the SARS-coronavirus genome. Antiviral Res 2005; 65:69-78. [PMID: 15708633 PMCID: PMC7114173 DOI: 10.1016/j.antiviral.2004.10.001] [Citation(s) in RCA: 79] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2004] [Accepted: 10/20/2004] [Indexed: 12/12/2022]
Abstract
A new disease, termed severe acute respiratory syndrome (SARS), emerged at the end of 2002 and caused profound disturbances in over 30 countries worldwide in 2003. A novel coronavirus was identified as the aetiological agent of SARS and the 30 kb viral genome was deciphered with unprecedented speed in a coordinated manner by the global community. Since then, much progress has been made in the virological and molecular characterization of the proteins encoded by SARS-coronavirus (SARS-CoV) genome, which contains 14 potential open reading frames (ORFs). These investigations can be broadly classified into three groups: (a) studies on the replicase 1a/1b gene products which are important for viral replication, (b) studies on the structural proteins, spike, nucleocapsid, membrane and envelope, which have homologues in all coronaviruses, and are important for viral assembly and (c) expression and functional studies of the “accessory” proteins that are specifically encoded by SARS-CoV. A comparison of the properties of these three groups of SARS-CoV proteins with the knowledge that coronavirologists have generated over more than 30 years of research can help us in the prevention and treatment of SARS in the event of the re-emergence of this new infectious disease.
Collapse
Affiliation(s)
- Yee-Joo Tan
- Institute of Molecular and Cell Biology, 61 Biopolis Drive, Proteos 138673, Singapore.
| | | | | |
Collapse
|
127
|
Chou THW, Wang S, Sakhatskyy PV, Mboudjeka I, Mboudoudjeck I, Lawrence JM, Huang S, Coley S, Yang B, Li J, Zhu Q, Lu S. Epitope mapping and biological function analysis of antibodies produced by immunization of mice with an inactivated Chinese isolate of severe acute respiratory syndrome-associated coronavirus (SARS-CoV). Virology 2005; 334:134-43. [PMID: 15749129 PMCID: PMC7111783 DOI: 10.1016/j.virol.2005.01.035] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2004] [Revised: 01/10/2005] [Accepted: 01/25/2005] [Indexed: 11/26/2022]
Abstract
Inactivated severe acute respiratory syndrome-associated coronavirus (SARS-CoV) has been tested as a candidate vaccine against the re-emergence of SARS. In order to understand the efficacy and safety of this approach, it is important to know the antibody specificities generated with inactivated SARS-CoV. In the current study, a panel of twelve monoclonal antibodies (mAbs) was established by immunizing Balb/c mice with the inactivated BJ01 strain of SARS-CoV isolated from the lung tissue of a SARS-infected Chinese patient. These mAbs could recognize SARS-CoV-infected cells by immunofluorescence analysis (IFA). Seven of them were mapped to the specific segments of recombinant spike (S) protein: six on S1 subunit (aa 12-798) and one on S2 subunit (aa 797-1192). High neutralizing titers against SARS-CoV were detected with two mAbs (1A5 and 2C5) targeting at a subdomain of S protein (aa 310-535), consistent with the previous report that this segment of S protein contains the major neutralizing domain. Some of these S-specific mAbs were able to recognize cleaved products of S protein in SARS-CoV-infected Vero E6 cells. None of the remaining five mAbs could recognize either of the recombinant S, N, M, or E antigens by ELISA. This study demonstrated that the inactivated SARS-CoV was able to preserve the immunogenicity of S protein including its major neutralizing domain. The relative ease with which these mAbs were generated against SARS-CoV virions further supports that subunit vaccination with S constructs may also be able to protect animals and perhaps humans. It is somewhat unexpected that no N-specific mAbs were identified albeit anti-N IgG was easily identified in SARS-CoV-infected patients. The availability of this panel of mAbs also provided potentially useful agents with applications in therapy, diagnosis, and basic research of SARS-CoV.
Collapse
Affiliation(s)
- Te-hui W Chou
- Laboratory of Nucleic Acid Vaccines, Department of Medicine, University of Massachusetts Medical School, 364 Plantation Street, Lazare Research Building, Worcester, MA 01605-2397, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
128
|
Mori Y, Okabayashi T, Yamashita T, Zhao Z, Wakita T, Yasui K, Hasebe F, Tadano M, Konishi E, Moriishi K, Matsuura Y. Nuclear localization of Japanese encephalitis virus core protein enhances viral replication. J Virol 2005; 79:3448-58. [PMID: 15731239 PMCID: PMC1075736 DOI: 10.1128/jvi.79.6.3448-3458.2005] [Citation(s) in RCA: 117] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Japanese encephalitis virus (JEV) core protein was detected in both the nucleoli and cytoplasm of mammalian and insect cell lines infected with JEV or transfected with the expression plasmid of the core protein. Mutation analysis revealed that Gly(42) and Pro(43) in the core protein are essential for the nuclear and nucleolar localization. A mutant M4243 virus in which both Gly(42) and Pro(43) were replaced by Ala was recovered by plasmid-based reverse genetics. In C6/36 mosquito cells, the M4243 virus exhibited RNA replication and protein synthesis comparable to wild-type JEV, whereas propagation in Vero cells was impaired. The mutant core protein was detected in the cytoplasm but not in the nucleus of either C6/36 or Vero cell lines infected with the M4243 virus. The impaired propagation of M4243 in mammalian cells was recovered by the expression of wild-type core protein in trans but not by that of the mutant core protein. Although M4243 mutant virus exhibited a high level of neurovirulence comparable to wild-type JEV in spite of the approximately 100-fold-lower viral propagation after intracerebral inoculation to 3-week-old mice of strain Jcl:ICR, no virus was recovered from the brain after intraperitoneal inoculation of the mutant. These results indicate that nuclear localization of JEV core protein plays crucial roles not only in the replication in mammalian cells in vitro but also in the pathogenesis of encephalitis induced by JEV in vivo.
Collapse
Affiliation(s)
- Yoshio Mori
- Research Center for Emerging Infectious Diseases, Research Institute for Microbial Diseases, Osaka University, 3-1 Yamada-oka, Suita, Osaka 565-0871, Japan
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
129
|
WU XD, SHANG B, YANG RF, YU H, MA ZH, SHEN X, JI YY, LIN Y, WU YD, LIN GM, TIAN L, GAN XQ, YANG S, JIANG WH, DAI EH, WANG XY, JIANG HL, XIE YH, ZHU XL, PEI G, LI L, WU JR, SUN B. The spike protein of severe acute respiratory syndrome (SARS) is cleaved in virus infected Vero-E6 cells. Cell Res 2005; 14:400-6. [PMID: 15450134 PMCID: PMC7091875 DOI: 10.1038/sj.cr.7290240] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2004] [Accepted: 08/24/2004] [Indexed: 01/05/2023] Open
Abstract
Spike protein is one of the major structural proteins of severe acute respiratory syndrome-coronavirus. It is essential for the interaction of the virons with host cell receptors and subsequent fusion of the viral envelop with host cell membrane to allow infection. Some spike proteins of coronavirus, such as MHV, HCoV-OC43, AIBV and BcoV, are proteolytically cleaved into two subunits, S1 and S2. In contrast, TGV, FIPV and HCoV-229E are not. Many studies have shown that the cleavage of spike protein seriously affects its function. In order to investigate the maturation and proteolytic processing of the S protein of SARS CoV, we generated S1 and S2 subunit specific antibodies (Abs) as well as N, E and 3CL protein-specific Abs. Our results showed that the antibodies could efficiently and specifically bind to their corresponding proteins from E.coli expressed or lysate of SARS-CoV infected Vero-E6 cells by Western blot analysis. Furthermore, the anti-S1 and S2 Abs were proved to be capable of binding to SARS CoV under electron microscope observation. When S2 Ab was used to perform immune precipitation with lysate of SARS-CoV infected cells, a cleaved S2 fragment was detected with S2-specific mAb by Western blot analysis. The data demonstrated that the cleavage of S protein was observed in the lysate, indicating that proteolytic processing of S protein is present in host cells.
Collapse
Affiliation(s)
- Xiao Dong WU
- Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, 320 Yueyang Road, Shanghai, 200031 China
| | - Bo SHANG
- Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, 320 Yueyang Road, Shanghai, 200031 China
| | - Rui Fu YANG
- Institute of Microbiology and Epidemiology, Institute of Basic Medical Sciences, Academy of Military Medical Sciences, Beijing, 100071 China
| | - Hao YU
- Institute of Plant Physiology and Ecology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, 300 Fenglin Road, Shanghai, 200031 China
| | - Zhi Hai MA
- Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, 320 Yueyang Road, Shanghai, 200031 China
| | - Xu SHEN
- Institute of Plant Physiology and Ecology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, 300 Fenglin Road, Shanghai, 200031 China
| | - Yong Yong JI
- Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, 320 Yueyang Road, Shanghai, 200031 China
| | - Ying LIN
- Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, 320 Yueyang Road, Shanghai, 200031 China
| | - Ya Di WU
- Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, 320 Yueyang Road, Shanghai, 200031 China
| | - Guo Mei LIN
- Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, 320 Yueyang Road, Shanghai, 200031 China
| | - Lin TIAN
- Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, 320 Yueyang Road, Shanghai, 200031 China
| | - Xiao Qing GAN
- Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, 320 Yueyang Road, Shanghai, 200031 China
| | - Sheng YANG
- Institute of Microbiology and Epidemiology, Institute of Basic Medical Sciences, Academy of Military Medical Sciences, Beijing, 100071 China
| | - Wei Hong JIANG
- Institute of Microbiology and Epidemiology, Institute of Basic Medical Sciences, Academy of Military Medical Sciences, Beijing, 100071 China
| | - Er Hei DAI
- Institute of Materia Medica, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, 320 Taiyuan Road, Shanghai, 200031 China
| | - Xiao Yi WANG
- Institute of Materia Medica, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, 320 Taiyuan Road, Shanghai, 200031 China
| | - Hua Liang JIANG
- Institute of Plant Physiology and Ecology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, 300 Fenglin Road, Shanghai, 200031 China
| | - You Hua XIE
- Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, 320 Yueyang Road, Shanghai, 200031 China
| | - Xue Liang ZHU
- Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, 320 Yueyang Road, Shanghai, 200031 China
| | - Gang PEI
- Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, 320 Yueyang Road, Shanghai, 200031 China
| | - Lin LI
- Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, 320 Yueyang Road, Shanghai, 200031 China
| | - Jia Rui WU
- Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, 320 Yueyang Road, Shanghai, 200031 China
| | - Bing SUN
- Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, 320 Yueyang Road, Shanghai, 200031 China
| |
Collapse
|
130
|
He R, Leeson A, Ballantine M, Andonov A, Baker L, Dobie F, Li Y, Bastien N, Feldmann H, Strocher U, Theriault S, Cutts T, Cao J, Booth TF, Plummer FA, Tyler S, Li X. Characterization of protein-protein interactions between the nucleocapsid protein and membrane protein of the SARS coronavirus. Virus Res 2005; 105:121-5. [PMID: 15351485 PMCID: PMC7127797 DOI: 10.1016/j.virusres.2004.05.002] [Citation(s) in RCA: 77] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2004] [Revised: 05/07/2004] [Accepted: 05/10/2004] [Indexed: 02/04/2023]
Abstract
The human coronavirus, associated with severe acute respiratory syndrome (SARS-CoV), was identified and molecularly characterized in 2003. Sequence analysis of the virus indicates that there is only 20% amino acid (aa) identity with known coronaviruses. Previous studies indicate that protein-protein interactions amongst various coronavirus proteins are critical for viral assembly. Yet, little sequence homology between the newly identified SARS-CoV and those previously studied coronaviruses suggests that determination of protein-protein interaction and identification of amino acid sequences, responsible for such interaction in SARS-CoV, are necessary for the elucidation of the molecular mechanism of SARS-CoV replication and rationalization of anti-SARS therapeutic intervention. In this study, we employed mammalian two-hybrid system to investigate possible interactions between SARS-CoV nucleocapsid (N) and the membrane (M) proteins. We found that interaction of the N and M proteins takes place in vivo and identified that a stretch of amino acids (168-208) in the N protein may be critical for such protein-protein interactions. Importantly, the same region has been found to be required for multimerization of the N protein (He et al., 2004) suggesting this region may be crucial in maintaining correct conformation of the N protein for self-interaction and interaction with the M protein.
Collapse
Affiliation(s)
- Runtao He
- National Microbiology Laboratory, Health Canada, 1015 Arlington St., Winnipeg, Man., Canada R3E 3R2.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
131
|
Britton P, Evans S, Dove B, Davies M, Casais R, Cavanagh D. Generation of a recombinant avian coronavirus infectious bronchitis virus using transient dominant selection. J Virol Methods 2005; 123:203-11. [PMID: 15620403 PMCID: PMC7112893 DOI: 10.1016/j.jviromet.2004.09.017] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2004] [Revised: 09/22/2004] [Accepted: 09/29/2004] [Indexed: 02/08/2023]
Abstract
A reverse genetics system for the avian coronavirus infectious bronchitis virus (IBV) has been described in which a full-length cDNA, corresponding to the IBV (Beaudette-CK) genome, was inserted into the vaccinia virus genome following in vitro assembly of three contiguous cDNAs [Casais, R., Thiel, V., Siddell, S.G., Cavanagh, D., Britton, P., 2001. Reverse genetics system for the avian coronavirus infectious bronchitis virus. J. Virol. 75, 12359-12369]. The method has subsequently been used to generate a recombinant IBV expressing a chimaeric S gene [Casais, R., Dove, B., Cavanagh, D., Britton, P., 2003. Recombinant avian infectious bronchitis virus expressing a heterologous spike gene demonstrates that the spike protein is a determinant of cell tropism. J. Virol. 77, 9084-9089]. Use of vaccinia virus as a vector for the full-length cDNA of the IBV genome has the advantage that modifications can be made to the IBV cDNA using homologous recombination, a method frequently used to insert and delete sequences from the vaccinia virus genome. We describe the use of homologous recombination as a method for modifying the Beaudette full-length cDNA, within the vaccinia virus genome, without the requirement for in vitro assembly of the IBV cDNA. To demonstrate the feasibility of the method we exchanged the ectodomain of the Beaudette spike gene for the corresponding region from IBV M41 and generated two recombinant infectious bronchitis viruses (rIBVs) expressing the chimaeric S protein, validating the method as an alternative way for generating rIBVs.
Collapse
Affiliation(s)
- Paul Britton
- Division of Molecular Biology, Institute for Animal Health, Compton Laboratory, Compton, Newbury, Berkshire RG20 7NN, UK.
| | | | | | | | | | | |
Collapse
|
132
|
Abstract
This chapter describes the interactions between the different structural components of the viruses and discusses their relevance for the process of virion formation. Two key factors determine the efficiency of the assembly process: intracellular transport and molecular interactions. Many viruses have evolved elaborate strategies to ensure the swift and accurate delivery of the virion components to the cellular compartment(s) where they must meet and form (sub) structures. Assembly of viruses starts in the nucleus by the encapsidation of viral DNA, using cytoplasmically synthesized capsid proteins; nucleocapsids then migrate to the cytosol, by budding at the inner nuclear membrane followed by deenvelopment, to pick up the tegument proteins.
Collapse
Affiliation(s)
- Cornelis A M de Haan
- Virology Division, Department of Infectious Diseases and Immunology, Faculty of Veterinary Medicine, Utrecht University, 3584 CL Utrecht, The Netherlands
| | | |
Collapse
|
133
|
Lu DD, Chen SH, Zhang SM, Zhang ML, Zhang W, Bo XC, Wang SQ. Screening of specific antigens for SARS clinical diagnosis using a protein microarray. Analyst 2005; 130:474-82. [PMID: 15776156 DOI: 10.1039/b415888a] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
In this study several SARS-CoV structural proteins and fragments were expressed in E. coli as GST or TRX fusion proteins. They were fabricated on a microarray and tested with sera from SARS patients. Antigenic screening indicated that recombinant GST-N2 fusion protein, the carboxy-terminus 213aa-423aa of N protein, was strongest positive and weakest non-specific compared with others. An indirect antibody ELISA method was developed and clinical positive and negative sera for their antibodies against GST-N2 fusion protein were assayed. 311 out of the 442 sera from clinical SARS inpatients, as well as 229 out of 302 sera from convalescent patients gave positive reactivities; positive rates were 70.4% and 75.8% respectively. Sera from a total of 2726 non-SARS patients and healthy individuals were tested and the false positive rate was only 0.07%. When the sensitivity control sample was diluted 1 : 64, it yielded OD values above the cutoff value. Reported data showed that this was a relatively high degree of sensitivity and specificity for SARS-CoV antibody testing. The data indicate that GST-N2 fusion protein, which was screened by protein microarray, may be a valuable diagnostic antigen for the development of serological assays for SARS. In addition, protein microarray assay presents a higher positive rate and sensitivity (86.1% and 1 : 200) compared with the traditional ELISA screening method, and could provide a rapid, parallel and high-throughput antigen screening platform.
Collapse
Affiliation(s)
- Dan-Dan Lu
- Beijing Institute of Radiation Medicine, Beijing, 100850, People's Republic of China
| | | | | | | | | | | | | |
Collapse
|
134
|
Chen H, Gill A, Dove BK, Emmett SR, Kemp CF, Ritchie MA, Dee M, Hiscox JA. Mass spectroscopic characterization of the coronavirus infectious bronchitis virus nucleoprotein and elucidation of the role of phosphorylation in RNA binding by using surface plasmon resonance. J Virol 2005; 79:1164-79. [PMID: 15613344 PMCID: PMC538594 DOI: 10.1128/jvi.79.2.1164-1179.2005] [Citation(s) in RCA: 95] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2003] [Accepted: 07/05/2004] [Indexed: 12/15/2022] Open
Abstract
Phosphorylation of the coronavirus nucleoprotein (N protein) has been predicted to play a role in RNA binding. To investigate this hypothesis, we examined the kinetics of RNA binding between nonphosphorylated and phosphorylated infectious bronchitis virus N protein with nonviral and viral RNA by surface plasmon resonance (Biacore). Mass spectroscopic analysis of N protein identified phosphorylation sites that were proximal to RNA binding domains. Kinetic analysis, by surface plasmon resonance, indicated that nonphosphorylated N protein bound with the same affinity to viral RNA as phosphorylated N protein. However, phosphorylated N protein bound to viral RNA with a higher binding affinity than nonviral RNA, suggesting that phosphorylation of N protein determined the recognition of virus RNA. The data also indicated that a known N protein binding site (involved in transcriptional regulation) consisting of a conserved core sequence present near the 5' end of the genome (in the leader sequence) functioned by promoting high association rates of N protein binding. Further analysis of the leader sequence indicated that the core element was not the only binding site for N protein and that other regions functioned to promote high-affinity binding.
Collapse
Affiliation(s)
- Hongying Chen
- School of Animal and Microbial Sciences, University of Reading, Reading, United Kingdom
| | | | | | | | | | | | | | | |
Collapse
|
135
|
Ning Q, Yan WM, Wang ZM, Xi D, Liu MF, Levy G, Luo XP. Domain I of nucleocapsid protein of murine hepatitis virus strain 3 upregulates transcription of mfgl2 prothrimbinase/fibroleukin gene. Shijie Huaren Xiaohua Zazhi 2004; 12:594-599. [DOI: 10.11569/wcjd.v12.i3.594] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
AIM: To investigate the responsible domain(s) of N protein and the I gene within the N gene of MHV-3 or MHV-A59 in the activation of mfgl2.
METHODS: To investigate the responsible domain(s) of N protein of MHV-3 or MHV-A59 in the activation of fgl2 gene, four ways comparison of the N protein was carried out and the site directed mutated N gene expression constructs within domain I and domain III were cotransfected respectively with mfgl2 promoter/luciferase reporter gene in CHO cells. Macrophages from Balb/cJ mice were infected with I gene mutated MHV virus Alb110 and its isogenic Alb111 for 8-10 hours, procoagulant activity (PCA) were measured. MHV-A59 I gene expression construct was cotransfected with mfgl2 promoter-reporter gene in Chinese hamster ovary (CHO) cells, and luciferase activity was detected for the assessment of promoter function.
RESULTS: Mutations of residues Gly-12, Pro-38, Asn-40, Gln-41 and Asn42 within domain I of the N protein of MHV-A59 to their corresponding residues were found in MHV-2 abrogated mfgl2 transcription, whereas mutation of other N protein domain III did not affect mfgl2 gene transcription. Alb 110 and Alb 111 infected macrophages showed a remarkable increasing in PCA activity compared with no virus or MHV-2 or MHV-JHM infected macrophages. There was no significant difference in PCA activity between Alb 110, Alb 111 infected group and MHV-A59 group. Cotransfection I gene expression construct with a reporter construct containing mfgl2 promoter in CHO cells displayed no significant difference in luciferase activity compared with nontransfected CHO cells.
CONCLUSION: Domain I of nucleocapsid protein of murine hepatitis virus strain 3 upregulates the transcription of mfgl2 prothrimbinase/fibroleukin gene. The MHV-A59 I gene is not essential for activation of mfgl2 gene. Our study may shed lights on the investigation of current worldwide-distributed disease, severe acute respiratory syndrome (SARS).
Collapse
|
136
|
Chang MS, Lu YT, Ho ST, Wu CC, Wei TY, Chen CJ, Hsu YT, Chu PC, Chen CH, Chu JM, Jan YL, Hung CC, Fan CC, Yang YC. Antibody detection of SARS-CoV spike and nucleocapsid protein. Biochem Biophys Res Commun 2004; 314:931-6. [PMID: 14751221 PMCID: PMC7111193 DOI: 10.1016/j.bbrc.2003.12.195] [Citation(s) in RCA: 51] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2003] [Indexed: 12/11/2022]
Abstract
Early detection and identification of SARS-CoV-infected patients and actions to prevent transmission are absolutely critical to prevent another SARS outbreak. Antibodies that specifically recognize the SARS-CoV spike and nucleocapsid proteins may provide a rapid screening method to allow accurate identification and isolation of patients with the virus early in their infection. For this reason, we raised peptide-induced polyclonal antibodies against SARS-CoV spike protein and polyclonal antibodies against SARS-CoV nucleocapsid protein using 6x His nucleocapsid recombinant protein. Western blot analysis and immunofluorescent staining showed that these antibodies specifically recognized SARS-CoV.
Collapse
Affiliation(s)
- Mau-Sun Chang
- Department of Medical Research, Mackay Memorial Hospital, Taipei, Taiwan, ROC
- National Taipei University of Technology, Taipei, Taiwan, ROC
| | - Yen-Ta Lu
- Department of Medical Research, Mackay Memorial Hospital, Taipei, Taiwan, ROC
- Taipei Medical University, Taipei, Taiwan, ROC
| | - Shin-Tsung Ho
- Department of Laboratory Medicine, Mackay Memorial Hospital, Taipei, Taiwan, ROC
| | - Chao-Chih Wu
- Department of Medical Research, Mackay Memorial Hospital, Taipei, Taiwan, ROC
| | - Tsai-Yin Wei
- Department of Medical Research, Mackay Memorial Hospital, Taipei, Taiwan, ROC
| | - Chia-Ju Chen
- Department of Medical Research, Mackay Memorial Hospital, Taipei, Taiwan, ROC
| | - Yun-Ting Hsu
- Department of Medical Research, Mackay Memorial Hospital, Taipei, Taiwan, ROC
| | - Po-Chen Chu
- Department of Medical Research, Mackay Memorial Hospital, Taipei, Taiwan, ROC
| | - Ching-Hsin Chen
- Department of Medical Research, Mackay Memorial Hospital, Taipei, Taiwan, ROC
| | - Jien-Ming Chu
- Department of Medical Research, Mackay Memorial Hospital, Taipei, Taiwan, ROC
| | - Ya-Lin Jan
- Department of Medical Research, Mackay Memorial Hospital, Taipei, Taiwan, ROC
| | - Chia-Chien Hung
- Department of Laboratory Medicine, Mackay Memorial Hospital, Taipei, Taiwan, ROC
| | - Chi-Chen Fan
- Department of Pathology, Mackay Memorial Hospital, Taipei, Taiwan, ROC
| | - Yuh-Cheng Yang
- Department of Medical Research, Mackay Memorial Hospital, Taipei, Taiwan, ROC
- Taipei Medical University, Taipei, Taiwan, ROC
- Department of Obstetrics and Gynecology, Mackay Memorial Hospital, Taipei, Taiwan, ROC
| |
Collapse
|
137
|
He R, Leeson A, Andonov A, Li Y, Bastien N, Cao J, Osiowy C, Dobie F, Cutts T, Ballantine M, Li X. Activation of AP-1 signal transduction pathway by SARS coronavirus nucleocapsid protein. Biochem Biophys Res Commun 2004; 311:870-6. [PMID: 14623261 PMCID: PMC7111052 DOI: 10.1016/j.bbrc.2003.10.075] [Citation(s) in RCA: 99] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
In March 2003, a novel coronavirus was isolated from patients exhibiting atypical pneumonia and subsequently proven to be the causative agent of the disease now referred to as severe acute respiratory syndrome (SARS). The complete genome of the SARS coronavirus (SARS-CoV) has since been sequenced. The SARS-CoV nucleocapsid (SARS-CoV N) shares little homology with other members of the coronavirus family. To determine if the N protein is involved in the regulation of cellular signal transduction, an ELISA-based assay on transcription factors was used. We found that the amount of transcription factors binding to promoter sequences of c-Fos, ATF2, CREB-1, and FosB was increased by the expression of SARS-CoV N. Since these factors are related to AP-1 signal transduction pathway, we investigated whether the AP-1 pathway was activated by SARS-CoV N protein using the PathDetect system. The results demonstrated that the expression of N protein, not the membrane protein (M), activated AP-1 pathway. We also found that SARS-CoV N protein does not activate NF-κB pathway, demonstrating that activation of important cellular pathways by SAS-CoV N protein is selective. Thus our data for the first time indicate that SARS-CoV has encoded a strategy to regulate cellular signaling process.
Collapse
Affiliation(s)
- Runtao He
- National Microbiology Laboratory, Health Canada, 1015 Arlington Street, R3E 3R2, Winnipeg, MB, Canada.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
138
|
Abstract
Liver impairment is commonly reported in up to 60% of patients who suffer from severe acute respiratory syndrome (SARS). Here we report the clinical course and liver pathology in three SARS patients with liver impairment. Three patients who fulfilled the World Health Organization case definition of probable SARS and developed marked elevation of alanine aminotransferase were included. Percutaneous liver biopsies were performed. Liver specimens were examined by light and electron microscopy, and immunohistochemistry. Reverse-transcriptase polymerase chain reaction (RT-PCR) using enhanced real-time PCR was applied to look for evidence of SARS-associated coronavirus infection. Marked accumulation of cells in mitosis was observed in two patients and apoptosis was observed in all three patients. Other common pathologic features included ballooning of hepatocytes and mild to moderate lobular lymphocytic infiltration. No eosinophilic infiltration, granuloma, cholestasis, fibrosis, or fibrin deposition was noted. Immunohistochemical studies revealed 0.5% to 11.4% of nuclei were positive for proliferative antigen Ki-67. RT-PCR showed evidence of SARS-associated coronavirus in the liver tissues, but not in the sera of all 3 patients. However, electron microscopy could not identify viral particles. No giant mitochondria, micro- or macro-vesicular steatosis was observed. In conclusion, hepatic impairment in patients with SARS is due to SARS-associated coronavirus infection of the liver. The prominence of mitotic activity of hepatocytes is unique and may be due to a hyperproliferative state with or without disruption of cell cycle by the coronavirus. With better knowledge of pathogenesis, specific therapy may be targeted to reduce viral replication and modify the disease course.
Collapse
|
139
|
Chau TN, Lee KC, Yao H, Tsang TY, Chow TC, Yeung YC, Choi KW, Tso YK, Lau T, Lai ST, Lai CL. SARS-associated viral hepatitis caused by a novel coronavirus: report of three cases. Hepatology 2004; 39:302-10. [PMID: 14767982 PMCID: PMC7165792 DOI: 10.1002/hep.20111] [Citation(s) in RCA: 304] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/20/2003] [Accepted: 11/19/2003] [Indexed: 02/06/2023]
Abstract
Liver impairment is commonly reported in up to 60% of patients who suffer from severe acute respiratory syndrome (SARS). Here we report the clinical course and liver pathology in three SARS patients with liver impairment. Three patients who fulfilled the World Health Organization case definition of probable SARS and developed marked elevation of alanine aminotransferase were included. Percutaneous liver biopsies were performed. Liver specimens were examined by light and electron microscopy, and immunohistochemistry. Reverse-transcriptase polymerase chain reaction (RT-PCR) using enhanced real-time PCR was applied to look for evidence of SARS-associated coronavirus infection. Marked accumulation of cells in mitosis was observed in two patients and apoptosis was observed in all three patients. Other common pathologic features included ballooning of hepatocytes and mild to moderate lobular lymphocytic infiltration. No eosinophilic infiltration, granuloma, cholestasis, fibrosis, or fibrin deposition was noted. Immunohistochemical studies revealed 0.5% to 11.4% of nuclei were positive for proliferative antigen Ki-67. RT-PCR showed evidence of SARS-associated coronavirus in the liver tissues, but not in the sera of all 3 patients. However, electron microscopy could not identify viral particles. No giant mitochondria, micro- or macro-vesicular steatosis was observed. In conclusion, hepatic impairment in patients with SARS is due to SARS-associated coronavirus infection of the liver. The prominence of mitotic activity of hepatocytes is unique and may be due to a hyperproliferative state with or without disruption of cell cycle by the coronavirus. With better knowledge of pathogenesis, specific therapy may be targeted to reduce viral replication and modify the disease course.
Collapse
Affiliation(s)
- Tai-Nin Chau
- Department of Medicine and Geriatrics, Princess Margaret Hospital, Hong Kong, SAR China.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
140
|
Rowland RRR, Schneider P, Fang Y, Wootton S, Yoo D, Benfield DA. Peptide domains involved in the localization of the porcine reproductive and respiratory syndrome virus nucleocapsid protein to the nucleolus. Virology 2003; 316:135-45. [PMID: 14599798 PMCID: PMC7125632 DOI: 10.1016/s0042-6822(03)00482-3] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
The nucleocapsid (N) protein of porcine reproductive and respiratory syndrome virus (PRRSV) is the principal component of the viral nucleocapsid and localizes to the nucleolus. Peptide sequence analysis of the N protein of several North American isolates identified two potential nuclear localization signal (NLS) sequences located at amino acids 10–13 and 41–42, which were labeled NLS-1 and NLS-2, respectively. Peptides containing NLS-1 or NLS-2 were sufficient to accumulate enhanced green fluorescent protein (EGFP) in the nucleus. The inactivation of NLS-1 by site-directed mutagenesis or the deletion of the first 14 amino acids did not affect N protein localization to the nucleolus. The substitution of key lysine residues with uncharged amino acids in NLS-2 blocked nuclear/nucleolar localization. Site-directed mutagenesis within NLS-2 identified the sequence, KKNKK, as forming the core localization domain within NLS-2. Using an in vitro pull-down assay, the N protein was able to bind importin-α, importin-β nuclear transport proteins. The localization pattern of N-EGFP fusion peptides represented by a series of deletions from the C- and N-terminal ends of the N protein identified a region covering amino acids 41–72, which contained a nucleolar localization signal (NoLS) sequence. The 41–72 N peptide when fused to EGFP mimicked the nucleolar–cytoplasmic distribution of native N. These results identify a single NLS involved in the transport of N from the cytoplasm and into nucleus. An additional peptide sequence, overlapping NLS-2, is involved in the further targeting of N to the nucleolus.
Collapse
Affiliation(s)
- Raymond R R Rowland
- Department of Diagnostic Medicine and Pathobiology, 1800 Denison Avenue, Kansas State University, Manhattan, KS 66506, USA.
| | | | | | | | | | | |
Collapse
|
141
|
Yoo D, Wootton SK, Li G, Song C, Rowland RR. Colocalization and interaction of the porcine arterivirus nucleocapsid protein with the small nucleolar RNA-associated protein fibrillarin. J Virol 2003; 77:12173-83. [PMID: 14581554 PMCID: PMC254285 DOI: 10.1128/jvi.77.22.12173-12183.2003] [Citation(s) in RCA: 86] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Porcine reproductive and respiratory syndrome virus (PRRSV) replicates in the cytoplasm of infected cells, but its nucleocapsid (N) protein localizes specifically to the nucleus and nucleolus. The mechanism of nuclear translocation and whether N associates with particular nucleolar components are unknown. In the present study, we show by confocal microscopy that the PRRSV N protein colocalizes with the small nucleolar RNA (snoRNA)-associated protein fibrillarin. Direct and specific interaction of N with fibrillarin was demonstrated in vivo by the mammalian two-hybrid assay in cells cotransfected with the N and fibrillarin genes and in vitro by the glutathione S-transferase pull-down assay using the expressed fibrillarin protein. Using a series of deletion mutants, the interactive domain of N with fibrillarin was mapped to a region of amino acids 30 to 37. For fibrillarin, the first 80 amino acids, which contain the glycine-arginine-rich region (the GAR domain), was determined to be the domain interactive with N. The N protein was able to bind to the full-length genomic RNA of PRRSV, and the RNA binding domain was identified as the region overlapping with the nuclear localization signal situated at positions 41 to 47. These results suggest that the N protein nuclear transport may be controlled by the binding of RNA to N. The PRRSV N protein was also able to bind to both 28S and 18S ribosomal RNAs. The protein-protein interaction between N and fibrillarin was RNA dependent but independent of N protein phosphorylation. Taken together, our studies demonstrate a specific interaction of the PRRSV nucleocapsid protein with the host cell protein fibrillarin in the nucleolus, and they imply a potential linkage of viral strategies for the modulation of host cell functions, possibly through rRNA precursor processing and ribosome biogenesis.
Collapse
Affiliation(s)
- Dongwan Yoo
- Department of Pathobiology, Ontario Veterinary College, University of Guelph, Guelph, Ontario N1G 2W1, Canada.
| | | | | | | | | |
Collapse
|
142
|
Abstract
A number of positive and negative strand RNA viruses whose primary site of replication is the cytoplasm use the nucleus and/or nuclear components in order to facilitate their replicative processes and alter host cell function. The nucleus itself is divided into a number of different sub-domains including structures such as the nucleolus. Many of the nuclear proteins that localise to these domains are involved in RNA processing, and because of their limited coding capacity, it may be necessary for RNA viruses to sequester such cellular factors in order to facilitate the replication, transcription and translation of their genomes. Amongst the best-studied examples of this are the picornaviruses, whose infection results in the redistribution of nuclear proteins to the cytoplasm and their interaction with the internal ribosome entry site (IRES) to facilitate translation of the picornavirus polyprotein. Examples can be found of other positive and also negative strand RNA virus proteins that localise to the nucleus and sub-domains (especially the nucleolus) during virus infection, and several localisation motifs have been defined. Apart from sequestering nuclear proteins for a role in replication, such viruses may also target the nucleus to disrupt nuclear functions and to inhibit antiviral responses.
Collapse
Affiliation(s)
- Julian A Hiscox
- School of Biochemistry and Molecular Biology, University of Leeds, LS2 9JT Leeds, UK.
| |
Collapse
|
143
|
Rowland RRR, Yoo D. Nucleolar-cytoplasmic shuttling of PRRSV nucleocapsid protein: a simple case of molecular mimicry or the complex regulation by nuclear import, nucleolar localization and nuclear export signal sequences. Virus Res 2003; 95:23-33. [PMID: 12921993 PMCID: PMC7127199 DOI: 10.1016/s0168-1702(03)00161-8] [Citation(s) in RCA: 87] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
The order Nidovirales, which includes the arteriviruses and coronaviruses, incorporate a cytoplasmic replication scheme; however, the nucleocapsid (N) protein of several members of this group localizes to the nucleolus suggesting that viral proteins influence nuclear processes during replication. The relatively small, 123 amino acid, N protein of porcine reproductive and respiratory syndrome virus (PRRSV), an arterivirus, presents an ideal model system for investigating the properties and mechanism of N protein nucleolar localization. The PRRSV N protein is found in both cytoplasmic and nucleolar compartments during infection and after transfection of gene constructs that express N-enhanced green fluorescent protein (EGFP) fusion proteins. Experiments using oligopeptides, truncated polypeptides and amino acid-substituted proteins have identified several domains within PRRSV N protein that participate in nucleo-cytoplasmic shuttling, including a cryptic nuclear localization signal (NLS) called NLS-1, a functional NLS (NLS-2), a nucleolar localization sequence (NoLS), as well as a possible nuclear export signal (NES). The purpose of this paper is to review our current understanding of PRRSV N protein shuttling and propose a shuttling scheme regulated by RNA binding and post-translational modification.
Collapse
Affiliation(s)
- Raymond R R Rowland
- Department of Diagnostic Medicine and Pathobiology, Kansas State University, Manhattan, KS 66506, USA.
| | | |
Collapse
|
144
|
Casais R, Dove B, Cavanagh D, Britton P. Recombinant avian infectious bronchitis virus expressing a heterologous spike gene demonstrates that the spike protein is a determinant of cell tropism. J Virol 2003; 77:9084-9. [PMID: 12885925 PMCID: PMC167237 DOI: 10.1128/jvi.77.16.9084-9089.2003] [Citation(s) in RCA: 216] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2003] [Accepted: 05/19/2003] [Indexed: 12/11/2022] Open
Abstract
A recombinant infectious bronchitis virus (IBV), BeauR-M41(S), was generated using our reverse genetics system (R. Casais, V. Thiel, S. G. Siddell, D. Cavanagh, and P. Britton, J. Virol. 75:12359-12369, 2001), in which the ectodomain region of the spike gene from IBV M41-CK replaced the corresponding region of the IBV Beaudette genome. BeauR-M41(S) acquired the same cell tropism phenotype as IBV M41-CK in four different cell types, demonstrating that the IBV spike glycoprotein is a determinant of cell tropism.
Collapse
Affiliation(s)
- Rosa Casais
- Division of Molecular Biology, Institute for Animal Health, Compton Laboratory, Compton, Newbury, Berkshire RG20 7NN, United Kingdom
| | | | | | | |
Collapse
|
145
|
Kim H, Chang DJ, Lee JA, Lee YS, Kaang BK. Identification of nuclear/nucleolar localization signal in Aplysia learning associated protein of slug with a molecular mass of 18 kDa homologous protein. Neurosci Lett 2003; 343:134-8. [PMID: 12759182 DOI: 10.1016/s0304-3940(03)00269-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
We isolated a learning associated protein of slug with a molecular mass of 18 kDa (LAPS18) homologue from the expressed sequence tag database of Aplysia kurodai and named it Aplysia LAPS18-like protein (ApLLP). ApLLP encodes 120 amino acids and has 57% identity with LAPS18. To examine the subcellular expression pattern of ApLLP we constructed an EGFP-tagged ApLLP fusion protein and overexpressed it in both Aplysia neurons and COS-7 cells. In contrast to the previous findings, which showed that LAPS18 is secreted by COS-7 cells, ApLLP-EGFP was localized to the nucleus, and most of it to nucleoli. Analysis of deletion mutants of ApLLP-EGFP showed that the N-terminal and the C-terminal nucleolar and nucleus localization signal sequences are important for localization to the nucleus and the nucleoli.
Collapse
Affiliation(s)
- Hyoung Kim
- National Research Laboratory, Institute of Molecular Biology and Genetics, School of Biological Sciences, College of Natural Sciences, Seoul National University, San 56-1 Silim-dong Kwanak-gu, Seoul 151-742, South Korea
| | | | | | | | | |
Collapse
|
146
|
LIN Y, SHEN X, YANG RF, LI YX, JI YY, HE YY, SHI MD, LU W, SHI TL, WANG J, WANG HX, JIANG HL, SHEN JH, XIE YH, WANG Y, PEI G, SHEN BF, WU JR, SUN B. Identification of an epitope of SARS-coronavirus nucleocapsid protein. Cell Res 2003; 13:141-5. [PMID: 12862314 PMCID: PMC7091728 DOI: 10.1038/sj.cr.7290158] [Citation(s) in RCA: 74] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2003] [Revised: 06/01/2003] [Accepted: 06/01/2003] [Indexed: 12/17/2022] Open
Abstract
The nucleocapsid (N) protein of severe acute respiratory syndrome-coronavirus (SARS-CoV) is a major virion structural protein. In this study, two epitopes (N1 and N2) of the N protein of SARS-CoV were predicted by bioinformatics analysis. After immunization with two peptides, the peptides-specific antibodies were isolated from the immunized rabbits. The further experiments demonstrated that N1 peptide-induced polyclonal antibodies had a high affinity to bind to E. coli expressed N protein of SARS-CoV. Furthermore, it was confirmed that N1 peptide-specific IgG antibodies were detectable in the sera of severe acute respiratory syndrome (SARS) patients. The results indicated that an epitope of the N protein has been identified and N protein specific Abs were produced by peptide immunization, which will be usefull for the study of SARS-CoV.
Collapse
Affiliation(s)
- Ying LIN
- Institute of Biochemistry and Cell Biology, Institute of Materia Medica, Bioinformation Center, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, 200031 China
| | - Xu SHEN
- Institute of Biochemistry and Cell Biology, Institute of Materia Medica, Bioinformation Center, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, 200031 China
| | - Rui Fu YANG
- Institute of Microbiology and Epidemiology, Institute of Basic Medical Sciences, Academy of Military Medical Sciences, Beijing, 100071 China
| | - Yi Xue LI
- Institute of Biochemistry and Cell Biology, Institute of Materia Medica, Bioinformation Center, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, 200031 China
| | - Yong Yong JI
- Institute of Biochemistry and Cell Biology, Institute of Materia Medica, Bioinformation Center, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, 200031 China
| | - You Yu HE
- Institute of Biochemistry and Cell Biology, Institute of Materia Medica, Bioinformation Center, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, 200031 China
| | - Mu De SHI
- Institute of Biochemistry and Cell Biology, Institute of Materia Medica, Bioinformation Center, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, 200031 China
| | - Wei LU
- Institute of Biochemistry and Cell Biology, Institute of Materia Medica, Bioinformation Center, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, 200031 China
| | - Tie Liu SHI
- Institute of Biochemistry and Cell Biology, Institute of Materia Medica, Bioinformation Center, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, 200031 China
| | - Jin WANG
- Institute of Microbiology and Epidemiology, Institute of Basic Medical Sciences, Academy of Military Medical Sciences, Beijing, 100071 China
| | - Hong Xia WANG
- Institute of Microbiology and Epidemiology, Institute of Basic Medical Sciences, Academy of Military Medical Sciences, Beijing, 100071 China
| | - Hua Liang JIANG
- Institute of Biochemistry and Cell Biology, Institute of Materia Medica, Bioinformation Center, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, 200031 China
| | - Jian Hua SHEN
- Institute of Biochemistry and Cell Biology, Institute of Materia Medica, Bioinformation Center, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, 200031 China
| | - You Hua XIE
- Institute of Biochemistry and Cell Biology, Institute of Materia Medica, Bioinformation Center, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, 200031 China
| | - Yuan WANG
- Institute of Biochemistry and Cell Biology, Institute of Materia Medica, Bioinformation Center, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, 200031 China
| | - Gang PEI
- Institute of Biochemistry and Cell Biology, Institute of Materia Medica, Bioinformation Center, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, 200031 China
| | - Bei Fen SHEN
- Institute of Microbiology and Epidemiology, Institute of Basic Medical Sciences, Academy of Military Medical Sciences, Beijing, 100071 China
| | - Jia Rui WU
- Institute of Biochemistry and Cell Biology, Institute of Materia Medica, Bioinformation Center, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, 200031 China
| | - Bing SUN
- Institute of Biochemistry and Cell Biology, Institute of Materia Medica, Bioinformation Center, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, 200031 China
| |
Collapse
|
147
|
Wang J, Ji J, Ye J, Zhao X, Wen J, Li W, Hu J, Li D, Sun M, Zeng H, Hu Y, Tian X, Tan X, Xu N, Zeng C, Wang J, Bi S, Yang H. The structure analysis and antigenicity study of the N protein of SARS-CoV. GENOMICS, PROTEOMICS & BIOINFORMATICS 2003; 1:145-54. [PMID: 15626344 PMCID: PMC5172421 DOI: 10.1016/s1672-0229(03)01018-0] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
The Coronaviridae family is characterized by a nucleocapsid that is composed of the genome RNA molecule in combination with the nucleoprotein (N protein) within a virion. The most striking physiochemical feature of the N protein of SARS-CoV is that it is a typical basic protein with a high predicted pI and high hydrophilicity, which is consistent with its function of binding to the ribophosphate backbone of the RNA molecule. The predicted high extent of phosphorylation of the N protein on multiple candidate phosphorylation sites demonstrates that it would be related to important functions, such as RNA-binding and localization to the nucleolus of host cells. Subsequent study shows that there is an SR-rich region in the N protein and this region might be involved in the protein-protein interaction. The abundant antigenic sites predicted in the N protein, as well as experimental evidence with synthesized polypeptides, indicate that the N protein is one of the major antigens of the SARS-CoV. Compared with other viral structural proteins, the low variation rate of the N protein with regards to its size suggests its importance to the survival of the virus.
Collapse
Affiliation(s)
- Jingqiang Wang
- Beijing Genomics Institute, Chinese Academy of Sciences, Beijing 101300, China
- James D. Watson Institute of Genome Sciences, Zhijiang Campus, Zhejiang University and Hangzhou Genomics Institute, Hangzhou 310008, China
| | - Jia Ji
- Beijing Genomics Institute, Chinese Academy of Sciences, Beijing 101300, China
| | - Jia Ye
- Beijing Genomics Institute, Chinese Academy of Sciences, Beijing 101300, China
- James D. Watson Institute of Genome Sciences, Zhijiang Campus, Zhejiang University and Hangzhou Genomics Institute, Hangzhou 310008, China
| | - Xiaoqian Zhao
- Beijing Genomics Institute, Chinese Academy of Sciences, Beijing 101300, China
| | - Jie Wen
- Beijing Genomics Institute, Chinese Academy of Sciences, Beijing 101300, China
| | - Wei Li
- Beijing Genomics Institute, Chinese Academy of Sciences, Beijing 101300, China
| | - Jianfei Hu
- Beijing Genomics Institute, Chinese Academy of Sciences, Beijing 101300, China
- College of Life Sciences, Peking University, Beijing 100871, China
| | - Dawei Li
- Beijing Genomics Institute, Chinese Academy of Sciences, Beijing 101300, China
| | - Min Sun
- Beijing Genomics Institute, Chinese Academy of Sciences, Beijing 101300, China
| | - Haipan Zeng
- Beijing Genomics Institute, Chinese Academy of Sciences, Beijing 101300, China
| | - Yongwu Hu
- Beijing Genomics Institute, Chinese Academy of Sciences, Beijing 101300, China
| | - Xiangjun Tian
- Beijing Genomics Institute, Chinese Academy of Sciences, Beijing 101300, China
- James D. Watson Institute of Genome Sciences, Zhijiang Campus, Zhejiang University and Hangzhou Genomics Institute, Hangzhou 310008, China
| | - Xuehai Tan
- Beijing Genomics Institute, Chinese Academy of Sciences, Beijing 101300, China
| | - Ningzhi Xu
- Beijing Genomics Institute, Chinese Academy of Sciences, Beijing 101300, China
| | - Changqing Zeng
- Beijing Genomics Institute, Chinese Academy of Sciences, Beijing 101300, China
| | - Jian Wang
- Beijing Genomics Institute, Chinese Academy of Sciences, Beijing 101300, China
- James D. Watson Institute of Genome Sciences, Zhijiang Campus, Zhejiang University and Hangzhou Genomics Institute, Hangzhou 310008, China
| | - Shengli Bi
- Center of Disease Control and Prevention, Beijing 100050, China
| | - Huanming Yang
- Beijing Genomics Institute, Chinese Academy of Sciences, Beijing 101300, China
- James D. Watson Institute of Genome Sciences, Zhijiang Campus, Zhejiang University and Hangzhou Genomics Institute, Hangzhou 310008, China
| |
Collapse
|
148
|
Abstract
Betanodavirus greasy grouper (Epinephelus tauvina) nervous necrosis viruses (GGNNV) protein alpha, a virus capsid protein, was detected in both nucleolus and cytoplasm of infected cells of Asian sea bass (SB) and transfected cells of SB and Cos-7 with pcDNA3.1/RNA2. To study its subcellular localization, ORF of protein alpha with 338 aa was fused with enhanced green fluorescent protein (EGFP) gene and was detected in transfected cells in the absence of other viral proteins. In both SB and Cos-7 cells, protein alpha was found to localize EGFP to the nucleolus and cytoplasm. Deletion mutants of protein alpha indicated that N-terminal 43 amino acid residues were required to import EGFP-alpha protein into the nucleolus. Further deletions within the 43 amino acid backbone, EGFP/33aa(1-33) and EGFP/30aa(14-43), localized to the nucleolus, suggesting that the 20 amino acids from 14 to 33 of protein alpha were the domain of nucleolus localization. To further determine the nucleolus targeting sequence, deletion mutations within the 20 amino acids of protein alpha were constructed. It was found that the deletion of (23)RRR(25), (29)RRR(31), or (23)RRRANNRRR(31) prevented the accumulation of EGFP fusion proteins into the nucleolus, demonstrating that (23)RRRANNRRR(31) contain the signal required for nucleolar localization. A similar distribution pattern of localization of protein alpha and its deletion mutants in SB and Cos-7 cells suggested that N-terminal residues of protein alpha (23)RRRANNRRR(31) constitute a nucleolus localization signal that functions in both fish and mammalian cells.
Collapse
Affiliation(s)
- Yan Xiang Guo
- Temasek Life Sciences Laboratory, The National University of Singapore, 117604, Singapore
| | | | | |
Collapse
|
149
|
Chen H, Wurm T, Britton P, Brooks G, Hiscox JA. Interaction of the coronavirus nucleoprotein with nucleolar antigens and the host cell. J Virol 2002; 76:5233-50. [PMID: 11967337 PMCID: PMC136173 DOI: 10.1128/jvi.76.10.5233-5250.2002] [Citation(s) in RCA: 110] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2001] [Accepted: 02/06/2002] [Indexed: 11/20/2022] Open
Abstract
Coronavirus nucleoproteins (N proteins) localize to the cytoplasm and the nucleolus, a subnuclear structure, in both virus-infected primary cells and in cells transfected with plasmids that express N protein. The nucleolus is the site of ribosome biogenesis and sequesters cell cycle regulatory complexes. Two of the major components of the nucleolus are fibrillarin and nucleolin. These proteins are involved in nucleolar assembly and ribosome biogenesis and act as chaperones for the import of proteins into the nucleolus. We have found that fibrillarin is reorganized in primary cells infected with the avian coronavirus infectious bronchitis virus (IBV) and in continuous cell lines that express either IBV or mouse hepatitis virus N protein. Both N protein and a fibrillarin-green fluorescent protein fusion protein colocalized to the perinuclear region and the nucleolus. Pull-down assays demonstrated that IBV N protein interacted with nucleolin and therefore provided a possible explanation as to how coronavirus N proteins localize to the nucleolus. Nucleoli, and proteins that localize to the nucleolus, have been implicated in cell growth-cell cycle regulation. Comparison of cells expressing IBV N protein with controls indicated that cells expressing N protein had delayed cellular growth. This result could not to be attributed to apoptosis. Morphological analysis of these cells indicated that cytokinesis was disrupted, an observation subsequently found in primary cells infected with IBV. Coronaviruses might therefore delay the cell cycle in interphase, where maximum translation of viral mRNAs can occur.
Collapse
Affiliation(s)
- Hongying Chen
- Virology Group, School of Animal and Microbial Sciences, The University of Reading, United Kingdom
| | | | | | | | | |
Collapse
|
150
|
Tijms MA, van der Meer Y, Snijder EJ. Nuclear localization of non-structural protein 1 and nucleocapsid protein of equine arteritis virus. J Gen Virol 2002; 83:795-800. [PMID: 11907328 DOI: 10.1099/0022-1317-83-4-795] [Citation(s) in RCA: 67] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
RNA synthesis (genome replication and subgenomic mRNA transcription) directed by equine arteritis virus (EAV; family Arteriviridae, order Nidovirales) occurs on modified cytoplasmic membranes to which most viral replicase subunits localize. Remarkably, a fraction of non-structural protein 1 (nsp1), a protein essential for transcription but dispensable for genome replication, is present in the host cell nucleus, in particular during the earlier stages of infection. Expression of GFP-tagged fusion proteins revealed that nsp1 is actively imported into the nucleus. Although the signals responsible for nsp1 transport could not be identified, our studies revealed that another EAV protein with a partially nuclear localization, the nucleocapsid (N) protein, utilizes the CRM1-mediated nuclear export pathway. Inactivation of this pathway with the drug leptomycin B resulted in the unexpected and immediate nuclear retention of all N protein molecules, thus revealing that the protein shuttles between cytoplasm and nucleus before playing its role in cytoplasmic virus assembly.
Collapse
Affiliation(s)
- Marieke A Tijms
- Molecular Virology Laboratory, Department of Medical Microbiology, Leiden University Medical Center, LUMC E4-P, PO Box 9600, 2300 RC Leiden, The Netherlands1
| | - Yvonne van der Meer
- Molecular Virology Laboratory, Department of Medical Microbiology, Leiden University Medical Center, LUMC E4-P, PO Box 9600, 2300 RC Leiden, The Netherlands1
| | - Eric J Snijder
- Molecular Virology Laboratory, Department of Medical Microbiology, Leiden University Medical Center, LUMC E4-P, PO Box 9600, 2300 RC Leiden, The Netherlands1
| |
Collapse
|