1
|
Keep S, Carr BV, Lean FZX, Fones A, Newman J, Dowgier G, Freimanis G, Vatzia E, Polo N, Everest H, Webb I, Mcnee A, Paudyal B, Thakur N, Nunez A, MacLoughlin R, Maier H, Hammond J, Bailey D, Waters R, Charleston B, Tuthill T, Britton P, Bickerton E, Tchilian E. Porcine Respiratory Coronavirus as a Model for Acute Respiratory Coronavirus Disease. Front Immunol 2022; 13:867707. [PMID: 35418984 PMCID: PMC8995773 DOI: 10.3389/fimmu.2022.867707] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Accepted: 03/02/2022] [Indexed: 12/11/2022] Open
Abstract
In the light of the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) pandemic, we have developed a porcine respiratory coronavirus (PRCV) model for in depth mechanistic evaluation of the pathogenesis, virology and immune responses of this important family of viruses. Pigs are a large animal with similar physiology and immunology to humans and are a natural host for PRCV. Four PRCV strains were investigated and shown to induce different degrees of lung pathology. Importantly, although all four strains replicated equally well in porcine cell lines in vitro and in the upper respiratory tract in vivo, PRCV strains causing more severe lung pathology were also able to replicate in ex vivo tracheal organ cultures as well as in vivo in the trachea and lung. The time course of infection of PRCV 135, which caused the most severe pulmonary pathology, was investigated. Virus was shed from the upper respiratory tract until day 10 post infection, with infection of the respiratory mucosa, as well as olfactory and sustentacular cells, providing an excellent model to study upper respiratory tract disease in addition to the commonly known lower respiratory tract disease from PRCV. Infected animals made antibody and T cell responses that cross reacted with the four PRCV strains and Transmissible Gastroenteritis Virus. The antibody response was reproduced in vitro in organ cultures. Comparison of mechanisms of infection and immune control in pigs infected with PRCVs of differing pathogenicity with human data from SARS-CoV-2 infection and from our in vitro organ cultures, will enable key events in coronavirus infection and disease pathogenesis to be identified.
Collapse
Affiliation(s)
- Sarah Keep
- The Pirbright Institute, Pirbright, United Kingdom
| | | | - Fabian Z X Lean
- Department of Pathology, Animal and Plant Health Agency, Addlestone, United Kingdom
| | - Albert Fones
- The Pirbright Institute, Pirbright, United Kingdom
| | | | | | | | - Eleni Vatzia
- The Pirbright Institute, Pirbright, United Kingdom
| | - Noemi Polo
- The Pirbright Institute, Pirbright, United Kingdom
| | | | - Isobel Webb
- The Pirbright Institute, Pirbright, United Kingdom
| | - Adam Mcnee
- The Pirbright Institute, Pirbright, United Kingdom
| | - Basu Paudyal
- The Pirbright Institute, Pirbright, United Kingdom
| | - Nazia Thakur
- The Pirbright Institute, Pirbright, United Kingdom
| | - Alejandro Nunez
- Department of Pathology, Animal and Plant Health Agency, Addlestone, United Kingdom
| | - Ronan MacLoughlin
- Research and Development, Science and Emerging Technologies, Aerogen, Galway, Ireland
| | - Helena Maier
- The Pirbright Institute, Pirbright, United Kingdom
| | - John Hammond
- The Pirbright Institute, Pirbright, United Kingdom
| | - Dalan Bailey
- The Pirbright Institute, Pirbright, United Kingdom
| | - Ryan Waters
- The Pirbright Institute, Pirbright, United Kingdom
| | | | - Toby Tuthill
- The Pirbright Institute, Pirbright, United Kingdom
| | - Paul Britton
- The Pirbright Institute, Pirbright, United Kingdom
| | | | | |
Collapse
|
2
|
Xie W, Ao C, Yang Y, Liu Y, Liang R, Zeng Z, Ye G, Xiao S, Fu ZF, Dong W, Peng G. Two critical N-terminal epitopes of the nucleocapsid protein contribute to the cross-reactivity between porcine epidemic diarrhea virus and porcine transmissible gastroenteritis virus. J Gen Virol 2019; 100:206-216. [PMID: 30652967 DOI: 10.1099/jgv.0.001216] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Both porcine epidemic diarrhoea virus (PEDV) and porcine transmissible gastroenteritis virus (TGEV), which cause high mortality in piglets and produce similar clinical symptoms and histopathological morphology, belong to the genus Alphacoronavirus. Serological diagnosis plays an important role in distinguishing pathogen species. Together with the spike (S) protein, the nucleocapsid (N) protein is one of the immunodominant regions among coronaviruses. In this study, two-way antigenic cross-reactivity between the N proteins of PEDV and TGEV was observed by indirect immunofluorescence assay (IFA) and Western blot analysis. Furthermore, the PEDV N protein harbouring truncations of amino acids (aa) 1 to 170 or aa 125 to 301 was demonstrated to cross-react with the anti-TGEV N polyclonal antibody (PAb), whereas the truncation-expressing aa 302 to 401 resulted in a specific reaction with the anti-PEDV N PAb but not with the anti-TGEV N PAb. Mutants of the PEDV N protein were generated based on sequence alignment and structural analysis; we then confirmed that the N-terminal residues 58-RWRMRRGERIE-68 and 78-LGTGPHAD-85 contributed to the cross-reactivity. All the results provide vital clues for the development of precise diagnostic assays for porcine coronaviruses.
Collapse
Affiliation(s)
- Wenting Xie
- 1State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, Hubei, PR China
- 2College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, PR China
- 3The Cooperative Innovation Center for Sustainable Pig Production, Huazhong Agricultural University, Wuhan, Hubei, PR China
| | - Chaojie Ao
- 1State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, Hubei, PR China
- 2College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, PR China
| | - Yilin Yang
- 1State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, Hubei, PR China
- 2College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, PR China
- 3The Cooperative Innovation Center for Sustainable Pig Production, Huazhong Agricultural University, Wuhan, Hubei, PR China
| | - Yinan Liu
- 1State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, Hubei, PR China
- 2College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, PR China
- 3The Cooperative Innovation Center for Sustainable Pig Production, Huazhong Agricultural University, Wuhan, Hubei, PR China
| | - Rui Liang
- 1State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, Hubei, PR China
- 2College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, PR China
- 3The Cooperative Innovation Center for Sustainable Pig Production, Huazhong Agricultural University, Wuhan, Hubei, PR China
| | - Zhe Zeng
- 1State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, Hubei, PR China
- 2College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, PR China
- 3The Cooperative Innovation Center for Sustainable Pig Production, Huazhong Agricultural University, Wuhan, Hubei, PR China
| | - Gang Ye
- 1State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, Hubei, PR China
- 2College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, PR China
- 3The Cooperative Innovation Center for Sustainable Pig Production, Huazhong Agricultural University, Wuhan, Hubei, PR China
| | - Shaobo Xiao
- 1State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, Hubei, PR China
- 2College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, PR China
| | - Zhen F Fu
- 1State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, Hubei, PR China
- 2College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, PR China
- 4Department of Pathology, College of Veterinary Medicine, University of Georgia, Athens, GA 30602, USA
| | - Wanyu Dong
- 5National Key Laboratory of Crop Genetic Improvement and National Centre of Plant Gene Research, College of Life Science and Technology, Huazhong Agricultural University, Wuhan 430070, PR China
- 3The Cooperative Innovation Center for Sustainable Pig Production, Huazhong Agricultural University, Wuhan, Hubei, PR China
- 2College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, PR China
- 1State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, Hubei, PR China
| | - Guiqing Peng
- 3The Cooperative Innovation Center for Sustainable Pig Production, Huazhong Agricultural University, Wuhan, Hubei, PR China
- 2College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, PR China
- 1State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, Hubei, PR China
| |
Collapse
|
3
|
Mutagenesis of Coronavirus nsp14 Reveals Its Potential Role in Modulation of the Innate Immune Response. J Virol 2016; 90:5399-5414. [PMID: 27009949 DOI: 10.1128/jvi.03259-15] [Citation(s) in RCA: 92] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2015] [Accepted: 03/15/2016] [Indexed: 11/20/2022] Open
Abstract
UNLABELLED Coronavirus (CoV) nonstructural protein 14 (nsp14) is a 60-kDa protein encoded by the replicase gene that is part of the replication-transcription complex. It is a bifunctional enzyme bearing 3'-to-5' exoribonuclease (ExoN) and guanine-N7-methyltransferase (N7-MTase) activities. ExoN hydrolyzes single-stranded RNAs and double-stranded RNAs (dsRNAs) and is part of a proofreading system responsible for the high fidelity of CoV replication. nsp14 N7-MTase activity is required for viral mRNA cap synthesis and prevents the recognition of viral mRNAs as "non-self" by the host cell. In this work, a set of point mutants affecting different motifs within the ExoN domain of nsp14 was generated, using transmissible gastroenteritis virus as a model of Alphacoronavirus Mutants lacking ExoN activity were nonviable despite being competent in both viral RNA and protein synthesis. A specific mutation within zinc finger 1 (ZF-C) led to production of a viable virus with growth and viral RNA synthesis kinetics similar to that of the parental virus. Mutant recombinant transmissible gastroenteritis virus (TGEV) ZF-C (rTGEV-ZF-C) caused decreased cytopathic effect and apoptosis compared with the wild-type virus and reduced levels of dsRNA accumulation at late times postinfection. Consequently, the mutant triggered a reduced antiviral response, which was confirmed by evaluating different stages of the dsRNA-induced antiviral pathway. The expression of beta interferon (IFN-β), tumor necrosis factor (TNF), and interferon-stimulated genes in cells infected with mutant rTGEV-ZF-C was reduced compared to the levels seen with the parental virus. Overall, our data revealed a potential role for CoV nsp14 in modulation of the innate immune response. IMPORTANCE The innate immune response is the first line of antiviral defense that culminates in the synthesis of interferon and proinflammatory cytokines to control viral replication. CoVs have evolved several mechanisms to counteract the innate immune response at different levels, but the role of CoV-encoded ribonucleases in preventing activation of the dsRNA-induced antiviral response has not been described to date. The introduction of a mutation in zinc finger 1 of the ExoN domain of nsp14 led to production of a virus that induced a weak antiviral response, most likely due to the accumulation of lower levels of dsRNA in the late phases of infection. These observations allowed us to propose a novel role for CoV nsp14 ExoN activity in counteracting the antiviral response, which could serve as a novel target for the design of antiviral strategies.
Collapse
|
4
|
Zhang D, Huang X, Zhang X, Cao S, Wen X, Wen Y, Wu R, Liang E. Construction of an oral vaccine for transmissible gastroenteritis virus based on the TGEV N gene expressed in an attenuated Salmonella typhimurium vector. J Virol Methods 2015; 227:6-13. [PMID: 26297958 DOI: 10.1016/j.jviromet.2015.08.011] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2014] [Revised: 08/17/2015] [Accepted: 08/17/2015] [Indexed: 01/16/2023]
Abstract
This research aimed to develop an oral vaccine for transmissible gastroenteritis virus (TGEV) based on the TGEV N gene expressed in an attenuated Salmonella typhimurium vector and aimed to evaluate the vaccine's immune response in piglets. Recombinant plasmid pVAX-N was transformed into competent cells of attenuated S. typhimurium SL7207 via electroporation. After it was identified via RT-PCR and double digestion, the screened recombinant bacteria presenting pVAX-N were named SL7207 (pVAX-N). To evaluate the safety and stability of the developed vaccine, different dosages (5 × 10(8), 1 × 10(9), and 2 × 10(9) CFU/mice) of SL7207 (pVAX-N) were inoculated to 6-week-old mice. Piglets below 20 days of age were dosed with 1 × 10(12) CFU. Humoral (neutralization titer and specific IgG), cellular (interleukin-4, γ-interferon, and peripheral lymphocyte proliferation), and mucosal (sIgA) immune responses were detected and evaluated. The three immunizing dosages were determined to be safe for mice and were completely eliminated 8 weeks after the first inoculation. Results of antibody and cytokine detection indicated that SL7207 (pVAX-N) could significantly induce antibody-IgG, antibody-IgA, interleukin-4, and γ-interferon, whose value was maximized on the 6th week. Results confirmed that the recombinant vaccine increased the proliferation of peripheral T lymphocyte. In conclusion, the oral vaccine was developed successfully, and the vaccine could significantly induce humoral, cellular, and mucosal immune responses in piglets.
Collapse
Affiliation(s)
- Dan Zhang
- Laboratory of Animal Infectious Disease and Microarray, Laboratory of Zoonosis and Pig Disease Research Center, College of Veterinary Medicine of Sichuan Agricultural University, Ya'an 625014, China
| | - Xiaobo Huang
- Laboratory of Animal Infectious Disease and Microarray, Laboratory of Zoonosis and Pig Disease Research Center, College of Veterinary Medicine of Sichuan Agricultural University, Ya'an 625014, China; Sichuan Science-observation Experiment Station of Veterinary Drugs and Veterinary Biological Technology, Ministry of Agriculture, Ya'an 625014, China.
| | - Xiaohui Zhang
- Laboratory of Animal Infectious Disease and Microarray, Laboratory of Zoonosis and Pig Disease Research Center, College of Veterinary Medicine of Sichuan Agricultural University, Ya'an 625014, China
| | - Sanjie Cao
- Laboratory of Animal Infectious Disease and Microarray, Laboratory of Zoonosis and Pig Disease Research Center, College of Veterinary Medicine of Sichuan Agricultural University, Ya'an 625014, China; Sichuan Science-observation Experiment Station of Veterinary Drugs and Veterinary Biological Technology, Ministry of Agriculture, Ya'an 625014, China
| | - Xintian Wen
- Laboratory of Animal Infectious Disease and Microarray, Laboratory of Zoonosis and Pig Disease Research Center, College of Veterinary Medicine of Sichuan Agricultural University, Ya'an 625014, China; Sichuan Science-observation Experiment Station of Veterinary Drugs and Veterinary Biological Technology, Ministry of Agriculture, Ya'an 625014, China
| | - Yiping Wen
- Laboratory of Animal Infectious Disease and Microarray, Laboratory of Zoonosis and Pig Disease Research Center, College of Veterinary Medicine of Sichuan Agricultural University, Ya'an 625014, China; Sichuan Science-observation Experiment Station of Veterinary Drugs and Veterinary Biological Technology, Ministry of Agriculture, Ya'an 625014, China
| | - Rui Wu
- Laboratory of Animal Infectious Disease and Microarray, Laboratory of Zoonosis and Pig Disease Research Center, College of Veterinary Medicine of Sichuan Agricultural University, Ya'an 625014, China; Sichuan Science-observation Experiment Station of Veterinary Drugs and Veterinary Biological Technology, Ministry of Agriculture, Ya'an 625014, China
| | - Entao Liang
- Laboratory of Animal Infectious Disease and Microarray, Laboratory of Zoonosis and Pig Disease Research Center, College of Veterinary Medicine of Sichuan Agricultural University, Ya'an 625014, China
| |
Collapse
|
5
|
Epitope insertion at the N-terminal molecular switch of the rabbit hemorrhagic disease virus T = 3 capsid protein leads to larger T = 4 capsids. J Virol 2012; 86:6470-80. [PMID: 22491457 DOI: 10.1128/jvi.07050-11] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Viruses need only one or a few structural capsid proteins to build an infectious particle. This is possible through the extensive use of symmetry and the conformational polymorphism of the structural proteins. Using virus-like particles (VLP) from rabbit hemorrhagic disease virus (RHDV) as a model, we addressed the basis of calicivirus capsid assembly and their application in vaccine design. The RHDV capsid is based on a T=3 lattice containing 180 identical subunits (VP1). We determined the structure of RHDV VLP to 8.0-Å resolution by three-dimensional cryoelectron microscopy; in addition, we used San Miguel sea lion virus (SMSV) and feline calicivirus (FCV) capsid subunit structures to establish the backbone structure of VP1 by homology modeling and flexible docking analysis. Based on the three-domain VP1 model, several insertion mutants were designed to validate the VP1 pseudoatomic model, and foreign epitopes were placed at the N- or C-terminal end, as well as in an exposed loop on the capsid surface. We selected a set of T and B cell epitopes of various lengths derived from viral and eukaryotic origins. Structural analysis of these chimeric capsids further validates the VP1 model to design new chimeras. Whereas most insertions are well tolerated, VP1 with an FCV capsid protein-neutralizing epitope at the N terminus assembled into mixtures of T=3 and larger T=4 capsids. The calicivirus capsid protein, and perhaps that of many other viruses, thus can encode polymorphism modulators that are not anticipated from the plane sequence, with important implications for understanding virus assembly and evolution.
Collapse
|
6
|
Transmissible gastroenteritis coronavirus RNA-dependent RNA polymerase and nonstructural proteins 2, 3, and 8 are incorporated into viral particles. J Virol 2011; 86:1261-6. [PMID: 22090122 DOI: 10.1128/jvi.06428-11] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Coronavirus replication and transcription are processes mediated by a protein complex, with the RNA-dependent RNA polymerase (RdRp) as a main component. Proteomic analysis of highly purified transmissible gastroenteritis virus showed the RdRp to be a component of the viral particles. This finding was confirmed by Western blotting, immunofluorescence, and immunoelectron microscopy analyses. Interestingly, the replicase nonstructural proteins 2, 3, and 8 colocalized with the RdRp in the viral factories and were also incorporated into the virions.
Collapse
|
7
|
Nogales A, Galán C, Márquez-Jurado S, García-Gallo M, Kremer L, Enjuanes L, Almazán F. Immunogenic characterization and epitope mapping of transmissible gastroenteritis virus RNA dependent RNA polymerase. J Virol Methods 2011; 175:7-13. [PMID: 21513742 PMCID: PMC7112926 DOI: 10.1016/j.jviromet.2011.04.007] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2010] [Revised: 03/28/2011] [Accepted: 04/05/2011] [Indexed: 11/16/2022]
Abstract
Coronavirus RNA synthesis is a sophisticated process performed by a viral multienzymatic replicase complex, together with cellular factors. A key enzyme of this replication complex is the RNA dependent RNA polymerase (RdRp). To study the replication of coronavirus genome, six monoclonal antibodies (mAbs) specific for transmissible gastroenteritis virus (TGEV) RdRp were generated and characterized. His-tagged RdRp was expressed in baculovirus, purified and used as immunogen to produce mAbs. The TGEV RdRp was recognized by these mAbs in the context of virus infection by immunofluorescence analysis and Western blot. Epitope mapping by Pepscan indicated that RdRp mAbs recognized four non-overlapping linear epitopes located in a 62-amino acid region of the N-terminal domain, suggesting that this region may constitute an immunodominant domain. The availability of TGEV RdRp mAbs will be instrumental to study coronavirus replication and to analyze the function of RdRp in pathogenesis.
Collapse
Affiliation(s)
- Aitor Nogales
- Department of Molecular and Cell Biology, Centro Nacional de Biotecnología (CNB-CSIC), C/ Darwin 3, Cantoblanco, 28049 Madrid, Spain
| | - Carmen Galán
- Department of Molecular and Cell Biology, Centro Nacional de Biotecnología (CNB-CSIC), C/ Darwin 3, Cantoblanco, 28049 Madrid, Spain
| | - Silvia Márquez-Jurado
- Department of Molecular and Cell Biology, Centro Nacional de Biotecnología (CNB-CSIC), C/ Darwin 3, Cantoblanco, 28049 Madrid, Spain
| | - Mónica García-Gallo
- Protein Tools Unit, Centro Nacional de Biotecnología (CNB-CSIC), Darwin 3, Cantoblanco, 28049 Madrid, Spain
| | - Leonor Kremer
- Protein Tools Unit, Centro Nacional de Biotecnología (CNB-CSIC), Darwin 3, Cantoblanco, 28049 Madrid, Spain
| | - Luis Enjuanes
- Department of Molecular and Cell Biology, Centro Nacional de Biotecnología (CNB-CSIC), C/ Darwin 3, Cantoblanco, 28049 Madrid, Spain
| | - Fernando Almazán
- Department of Molecular and Cell Biology, Centro Nacional de Biotecnología (CNB-CSIC), C/ Darwin 3, Cantoblanco, 28049 Madrid, Spain
| |
Collapse
|
8
|
The polypyrimidine tract-binding protein affects coronavirus RNA accumulation levels and relocalizes viral RNAs to novel cytoplasmic domains different from replication-transcription sites. J Virol 2011; 85:5136-49. [PMID: 21411518 DOI: 10.1128/jvi.00195-11] [Citation(s) in RCA: 62] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
The coronavirus (CoV) discontinuous transcription mechanism is driven by long-distance RNA-RNA interactions between transcription-regulating sequences (TRSs) located at the 5' terminal leader (TRS-L) and also preceding each mRNA-coding sequence (TRS-B). The contribution of host cell proteins to CoV transcription needs additional information. Polypyrimidine tract-binding protein (PTB) was reproducibly identified in association with positive-sense RNAs of transmissible gastroenteritis coronavirus (TGEV) TRS-L and TRS-B by affinity chromatography and mass spectrometry. A temporal regulation of PTB cytoplasmic levels was observed during infection, with a significant increase from 7 to 16 h postinfection being inversely associated with a decrease in viral replication and transcription. Silencing the expression of PTB with small interfering RNA in two cell lines (Huh7 and HEK 293T) led to a significant increase of up to 4-fold in mRNA levels and virus titer, indicating a negative effect of PTB on CoV RNA accumulation. During CoV infection, PTB relocalized from the nucleus to novel cytoplasmic structures different from replication-transcription sites in which stress granule markers T-cell intracellular antigen-1 (TIA-1) and TIA-1-related protein (TIAR) colocalized. PTB was detected in these modified stress granules in TGEV-infected swine testis cells but not in stress granules induced by oxidative stress. Furthermore, viral genomic and subgenomic RNAs were detected in association with PTB and TIAR. These cytoplasmic ribonucleoprotein complexes might be involved in posttranscriptional regulation of virus gene expression.
Collapse
|
9
|
Galán C, Sola I, Nogales A, Thomas B, Akoulitchev A, Enjuanes L, Almazán F. Host cell proteins interacting with the 3' end of TGEV coronavirus genome influence virus replication. Virology 2009; 391:304-14. [PMID: 19580983 PMCID: PMC7118768 DOI: 10.1016/j.virol.2009.06.006] [Citation(s) in RCA: 60] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2009] [Revised: 05/25/2009] [Accepted: 06/03/2009] [Indexed: 11/29/2022]
Abstract
Coronavirus RNA synthesis is performed by a multienzymatic replicase complex together with cellular factors. This process requires the specific recognition of RNA cis-acting signals located at the ends of the viral genome. To identify cellular proteins involved in coronavirus RNA synthesis, transmissible gastroenteritis coronavirus (TGEV) genome ends, harboring essential cis-acting signals for replication, were used as baits for RNA affinity protein purification. Ten proteins were preferentially pulled down with either the 5' or 3' ends of the genome and identified by proteomic analysis. Nine of them, including members of the heterogeneous ribonucleoprotein family of proteins (hnRNPs), the poly(A)-binding protein (PABP), the p100 transcriptional co-activator protein and two aminoacyl-tRNA synthetases, showed a preferential binding to the 3' end of the genome, whereas only the polypyrimidine tract-binding protein (PTB) was preferentially pulled down with the 5' end of the genome. The potential function of the 3' end-interacting proteins in virus replication was studied by analyzing the effect of their silencing using a TGEV-derived replicon and the infectious virus. Gene silencing of PABP, hnRNP Q, and glutamyl-prolyl-tRNA synthetase (EPRS) caused a significant 2 to 3-fold reduction of viral RNA synthesis. Interestingly, the silencing of glyceraldehyde 3-phosphate dehydrogenase (GAPDH), initially used as a control gene, caused a 2 to 3-fold increase in viral RNA synthesis in both systems. These data suggest that PABP, hnRNP Q, and EPRS play a positive role in virus infection that could be mediated through their interaction with the viral 3' end, and that GAPDH has a negative effect on viral infection.
Collapse
Affiliation(s)
- Carmen Galán
- Department of Molecular and Cell Biology, Centro Nacional de Biotecnología, CSIC, C/Darwin 3, Cantoblanco, 28049 Madrid, Spain
| | | | | | | | | | | | | |
Collapse
|
10
|
Abstract
abstract: Severe acute respiratory syndrome (SARS) is the first emerging infectious disease of the 21st century that has been highly transmissible and fatal and was caused by a previously unknown coronavirus (SARS‐CoV). The SARS epidemic in 2003 resulted in more than 8400 SARS cases and approximately 800 deaths. Existing in non‐identified animal reservoirs, SARS‐CoV continues to represent a threat to humans although more than four years have passed since a large outbreak of SARS, and no new cases have been reported. However, we cannot exclude the possibility of reemergence of SARS. It is hence necessary to understand the biology of the SARS‐CoV to deal adequately with the next outbreak, whenever it happens. The SARS‐CoV is a novel coronavirus with a large (∼30 thousand nucleotides) positive‐sense, single‐stranded RNA containing 14 functional open reading frames (ORFs) of which 2 large ORFs constitute the replicase gene which encodes proteins required for viral RNA syntheses. The remaining 12 ORFs encode the 4 structural proteins: spike, membrane, nucleocapsid and envelope; and eight accessory proteins. The viral genome and its expression within the host cell undergoes extensive translational and enzymatic processing to form the 4 structural, 8 accessory and 16 nonstructural proteins. In an effort to understand the molecular mechanisms or capsid assembly and viral pathogenesis, laboratories around the world have adopted a variety of approaches to answering these trivial questions. It has been our effort to consolidate all information known to date about the molecular mechanisms of the SARS‐CoV into this chapter to update our readership on the current status of research.
Collapse
Affiliation(s)
- Namita Satija
- Virology Group, International Centre for Genetic Engineering & Biotechnology, New Delhi, India
| | | |
Collapse
|
11
|
Zúñiga S, Sola I, Moreno JL, Sabella P, Plana-Durán J, Enjuanes L. Coronavirus nucleocapsid protein is an RNA chaperone. Virology 2007; 357:215-27. [PMID: 16979208 PMCID: PMC7111943 DOI: 10.1016/j.virol.2006.07.046] [Citation(s) in RCA: 120] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2006] [Revised: 07/11/2006] [Accepted: 07/29/2006] [Indexed: 01/19/2023]
Abstract
RNA chaperones are nonspecific nucleic acid binding proteins with long disordered regions that help RNA molecules to adopt its functional conformation. Coronavirus nucleoproteins (N) are nonspecific RNA-binding proteins with long disordered regions. Therefore, we investigated whether transmissible gastroenteritis coronavirus (TGEV) N protein was an RNA chaperone. Purified N protein enhanced hammerhead ribozyme self-cleavage and nucleic acids annealing, which are properties that define RNA chaperones. In contrast, another RNA-binding protein, PTB, did not show these activities. N protein chaperone activity was blocked by specific monoclonal antibodies. Therefore, it was concluded that TGEV N protein is an RNA chaperone. In addition, we have shown that purified severe acute respiratory syndrome (SARS)-CoV N protein also has RNA chaperone activity. In silico predictions of disordered domains showed a similar pattern for all coronavirus N proteins evaluated. Altogether, these data led us to suggest that all coronavirus N proteins might be RNA chaperones.
Collapse
Affiliation(s)
- Sonia Zúñiga
- Centro Nacional de Biotecnología, CSIC, Department of Molecular and Cell Biology, Campus Universidad Autónoma, Darwin 3, Cantoblanco, 28049 Madrid, Spain
| | | | | | | | | | | |
Collapse
|
12
|
Angulo E, Bárcena J. Towards a unique and transmissible vaccine against myxomatosis and rabbit haemorrhagic disease for rabbit populations. WILDLIFE RESEARCH 2007. [DOI: 10.1071/wr06160] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Currently available vaccines against myxomatosis and rabbit hemorrhagic disease virus (RHDV) are not suited to immunise wild rabbit populations, as vaccines need to be delivered individually by conventional veterinary practices. As an alternative approach, research in Spain has focused on the development of a transmissible vaccine. A recombinant virus has been constructed based on a naturally attenuated myxoma virus (MV) field strain, expressing the RHDV capsid protein (VP60). Following inoculation of rabbits, the recombinant virus (MV-VP60) induced specific antibody responses against MV and RHDV, conferring protection against lethal challenges with both viruses. Furthermore, the recombinant MV-VP60 virus showed a limited horizontal transmission capacity, either by direct contact or in a flea-mediated process, promoting immunisation of contact uninoculated animals. Efficacy and safety of the vaccine have been extensively evaluated under laboratory conditions and in a limited field trial. The development of the transmissible vaccine strategy and the steps being taken to obtain the marketing authorisation for the vaccine in the European Union are presented in this review.
Collapse
|
13
|
Cloning and expression of nucleocapsid protein gene of TGEV HB06 strain. FRONTIERS OF AGRICULTURE IN CHINA 2007; 1:357-360. [PMID: 32214985 PMCID: PMC7088836 DOI: 10.1007/s11703-007-0060-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The nucleocapsid protein gene of transmissible gastroenteritis virus, 1 149 bp in length, was amplified by RT-PCR from isolated strain HB06 and cloned into pMD18-T. Sequence comparison with other transmissible gastroenteritis virus (TGEV) strains selected from the Gene Bank revealed that the homology of N gene complete sequence shares more than 97% in nucleotide. N gene was cloned into BamHI and EcoRI multiple cloning sites of the prokaryotic expression vector pET 20 b, and named pETN. After being induced by isopropyl-β-D-thiogalactopyranoside (IPTG), the recombinant nucleocapsid protein was expressed. The result of SDS-PAGE and Western-blot showed that the recombinant nucleocapsid protein was 47 kDa and had strong positive reactions with TGEV-specific antibody.
Collapse
|
14
|
Perlman S, Holmes KV. Regulation of coronavirus transcription: viral and cellular proteins interacting with transcription-regulating sequences. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2006; 581:31-5. [PMID: 17037500 PMCID: PMC7123242 DOI: 10.1007/978-0-387-33012-9_4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 05/12/2023]
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
|
15
|
Zhang X, Hasoksuz M, Spiro D, Halpin R, Wang S, Stollar S, Janies D, Hadya N, Tang Y, Ghedin E, Saif L. Complete genomic sequences, a key residue in the spike protein and deletions in nonstructural protein 3b of US strains of the virulent and attenuated coronaviruses, transmissible gastroenteritis virus and porcine respiratory coronavirus. Virology 2006; 358:424-35. [PMID: 17023013 PMCID: PMC1850758 DOI: 10.1016/j.virol.2006.08.051] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2006] [Revised: 08/17/2006] [Accepted: 08/31/2006] [Indexed: 11/28/2022]
Abstract
Transmissible gastroenteritis virus (TGEV) isolates that have been adapted to passage in cell culture maintain their infectivity in vitro but may lose their pathogenicity in vivo. To better understand the genomic mechanisms for viral attenuation, we sequenced the complete genomes of two virulent TGEV strains and their attenuated counterparts: virulent TGEV Miller M6 and attenuated TGEV Miller M60 and virulent TGEV Purdue and attenuated TGEV Purdue P115, together with the ISU-1 strain of porcine respiratory coronavirus (PRCV-ISU-1), a naturally occurring TGEV deletion mutant with an altered respiratory tropism and reduced virulence. Pairwise comparison at both the nucleotide (nt) and amino acid (aa) levels between virulent and attenuated TGEV strains identified a common change in nt 1753 of the spike gene, resulting in a serine to alanine mutation at aa position 585 of the spike proteins of the attenuated TGEV strains. Alanine was also present in this protein in PRCV-ISU-1. Particularly noteworthy, the serine to alanine mutation resides in the region of the major antigenic site A/B (aa 506-706) that elicits neutralizing antibodies and within the domain mediating the cell surface receptor aminopeptidase N binding (aa 522-744). Comparison of the predicted polypeptide products of ORF3b showed significant deletions in the naturally attenuated PRCV-ISU-1 and TGEV Miller M60; these deletions occurred at a common break point, suggesting a related mechanism of recombination that may affect viral virulence or tropism. Sequence comparisons at both genomic and protein levels indicated that PRCV-ISU-1 had a closer relationship with TGEV Miller strains than Purdue strains. Phylogenetic analyses showed that virulence is an evolutionarily labile trait in TGEV and that TGEV strains as a group share a common ancestor with PRCV.
Collapse
Affiliation(s)
- Xinsheng Zhang
- Food Animal Health Research Program, Ohio Agricultural Research and Development Center, The Ohio State University, 1680 Madison Ave., Wooster, OH 44691, USA
| | - Mustafa Hasoksuz
- Istanbul University, Faculty of Veterinary Medicine, Department of Virology, Avcilar, 34320, Istanbul, Turkey
| | - David Spiro
- The Institute for Genomic Research (TIGR), Rockville, MD 20850, USA
| | - Rebecca Halpin
- The Institute for Genomic Research (TIGR), Rockville, MD 20850, USA
| | - Shiliang Wang
- The Institute for Genomic Research (TIGR), Rockville, MD 20850, USA
| | - Sarah Stollar
- The Institute for Genomic Research (TIGR), Rockville, MD 20850, USA
| | - Daniel Janies
- Department of Biomedical Informatics, The Ohio State University Medical Center, Columbus, OH 44691, USA
| | - Nagesh Hadya
- Food Animal Health Research Program, Ohio Agricultural Research and Development Center, The Ohio State University, 1680 Madison Ave., Wooster, OH 44691, USA
| | - Yuxin Tang
- Food Animal Health Research Program, Ohio Agricultural Research and Development Center, The Ohio State University, 1680 Madison Ave., Wooster, OH 44691, USA
| | - Elodie Ghedin
- The Institute for Genomic Research (TIGR), Rockville, MD 20850, USA
| | - Linda Saif
- Food Animal Health Research Program, Ohio Agricultural Research and Development Center, The Ohio State University, 1680 Madison Ave., Wooster, OH 44691, USA
- Corresponding author. Fax: +1 330 263 3677.
| |
Collapse
|
16
|
Calvo E, Escors D, López JA, González JM, Álvarez A, Arza E, Enjuanes L. Phosphorylation and subcellular localization of transmissible gastroenteritis virus nucleocapsid protein in infected cells. J Gen Virol 2005; 86:2255-2267. [PMID: 16033973 DOI: 10.1099/vir.0.80975-0] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
The nucleocapsid (N) protein is the only phosphorylated structural protein of the coronavirus Transmissible gastroenteritis virus (TGEV). The phosphorylation state and intracellular distribution of TGEV N protein in infected cells were characterized by a combination of techniques including: (i) subcellular fractionation and analysis of tryptic peptides by two-dimensional nano-liquid chromatography, coupled to ion-trap mass spectrometry; (ii) tandem mass-spectrometry analysis of N protein resolved by SDS-PAGE; (iii) Western blotting using two specific antisera for phosphoserine-containing motifs; and (iv) confocal microscopy. A total of four N protein-derived phosphopeptides were detected in mitochondria–Golgi–endoplasmic reticulum–Golgi intermediate compartment (ERGIC)-enriched fractions, including N-protein phosphoserines 9, 156, 254 and 256. Confocal microscopy showed that the N protein found in mitochondria–Golgi–ERGIC fractions localized within the Golgi–ERGIC compartments and not with mitochondria. Phosphorylated N protein was also present in purified virions, containing at least phosphoserines 156 and 256. Coronavirus N proteins showed a conserved pattern of secondary structural elements, including six β-strands and four α-helices. Whilst serine 9 was present in a non-conserved domain, serines 156, 254 and 256 were localized close to highly conserved secondary structural elements within the central domain of coronavirus N proteins. Serine 156 was highly conserved, whereas no clear homologous sites were found for serines 254 and 256 for other coronavirus N proteins.
Collapse
Affiliation(s)
- E Calvo
- Unidad de Proteómica, Fundación Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Sinesio Delgado 4, 28029 Madrid, Spain
| | - D Escors
- Department of Molecular and Cell Biology, Centro Nacional de Biotecnología (CNB, CSIC), Campus Univ. Autonoma, 3 Darwin St, Cantoblanco, 28049 Madrid, Spain
| | - J A López
- Unidad de Proteómica, Fundación Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Sinesio Delgado 4, 28029 Madrid, Spain
| | - J M González
- Department of Molecular and Cell Biology, Centro Nacional de Biotecnología (CNB, CSIC), Campus Univ. Autonoma, 3 Darwin St, Cantoblanco, 28049 Madrid, Spain
| | - A Álvarez
- Unidad de Citometría, Fundación Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Sinesio Delgado 4, 28029 Madrid, Spain
| | - E Arza
- Unidad de Citometría, Fundación Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Sinesio Delgado 4, 28029 Madrid, Spain
| | - L Enjuanes
- Department of Molecular and Cell Biology, Centro Nacional de Biotecnología (CNB, CSIC), Campus Univ. Autonoma, 3 Darwin St, Cantoblanco, 28049 Madrid, Spain
| |
Collapse
|
17
|
He Y, Zhou Y, Wu H, Kou Z, Liu S, Jiang S. Mapping of antigenic sites on the nucleocapsid protein of the severe acute respiratory syndrome coronavirus. J Clin Microbiol 2004; 42:5309-14. [PMID: 15528730 PMCID: PMC525273 DOI: 10.1128/jcm.42.11.5309-5314.2004] [Citation(s) in RCA: 63] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2004] [Revised: 07/18/2004] [Accepted: 07/27/2004] [Indexed: 11/20/2022] Open
Abstract
Antigenic sites on the nucleocapsid (N) protein of severe acute respiratory syndrome (SARS) coronavirus (SARS-CoV) were mapped by Pepscan analysis with overlapping peptides that span the N protein sequence. Two major immunodominant epitopes located in the C-terminal region (amino acids [aa] 362 to 412) and middle region (aa 153 to 178) reacted with more than 75% of sera from SARS patients. Several minor immunodominant epitopes were reactive with about 50% of the SARS sera. Antisera from mice immunized with inactivated SARS-CoV recognized the two major immunodominant epitopes and one antigenic site located adjacent to the N-terminal region (aa 76 to 101), which did not react with the sera from SARS patients. Several monoclonal antibodies against SARS-CoV bound to the N- or C-terminal antigenic sites. These results suggest that the above antigenic sites on the N protein are important in eliciting humoral immune response against SARS-CoV in humans and animals and can be used as antigens for developing diagnostic tests.
Collapse
Affiliation(s)
- Yuxian He
- Lindsley F. Kimball Research Institute, New York Blood Center, New York, NY 10021, USA
| | | | | | | | | | | |
Collapse
|
18
|
Escors D, Capiscol C, Enjuanes L. Immunopurification applied to the study of virus protein composition and encapsidation. J Virol Methods 2004; 119:57-64. [PMID: 15158585 PMCID: PMC7119662 DOI: 10.1016/j.jviromet.2004.03.004] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2004] [Revised: 03/04/2004] [Accepted: 03/08/2004] [Indexed: 11/24/2022]
Abstract
A protocol for obtaining small amounts of highly pure virus preparations starting from reduced volumes (<5 ml) of infected tissue culture supernatants is described. This procedure was adapted to the study of transmissible gastroenteritis coronavirus (TGEV) protein composition and RNA encapsidation. This protocol relies on virion capture by monoclonal antibodies specific for a virion membrane protein. These antibodies were bound to protein A-coated ELISA wells armed with rabbit anti-mouse IgG antibodies. As an example, the purification of 35S-labelled TGEV virions was performed. The quality of virus preparations was assessed by quantifying common contaminating RNAs by real-time RT-PCR. The most critical factors influencing the purity degree are analysed and discussed.
Collapse
Affiliation(s)
- David Escors
- Department of Molecular and Cell Biology, Centro Nacional de Biotecnología, CSIC, Campus Universidad Autónoma, Cantoblanco, 28049 Madrid, Spain
| | | | | |
Collapse
|
19
|
Bárcena J, Verdaguer N, Roca R, Morales M, Angulo I, Risco C, Carrascosa JL, Torres JM, Castón JR. The coat protein of Rabbit hemorrhagic disease virus contains a molecular switch at the N-terminal region facing the inner surface of the capsid. Virology 2004; 322:118-34. [PMID: 15063122 DOI: 10.1016/j.virol.2004.01.021] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2003] [Revised: 01/16/2004] [Accepted: 01/19/2004] [Indexed: 11/22/2022]
Abstract
To function adequately, many if not all proteins involved in macromolecular assemblies show conformational polymorphism as an intrinsic feature. This general strategy has been described for many essential cellular processes. Here we describe this structural polymorphism in a viral protein, the coat protein of Rabbit hemorrhagic disease virus (RHDV), which is required during virus capsid assembly. By combining genetic, structure modeling, and cryo-electron microscopy and image processing analysis, we have established the mechanism that allows RHDV coat protein to switch among quasi-equivalent conformational states to achieve the appropriate curvature for the formation of a closed shell. The RHDV capsid structure is based on a T = 3 lattice, containing 180 copies of identical subunits, similar to those of other caliciviruses. The quasi-equivalent interactions between the coat proteins are achieved by the N-terminal region of a subset of subunits, which faces the inner surface of the capsid shell. Mutant coat protein lacking this N-terminal sequence assembles into T = 1 capsids. Our results suggest that the polymorphism of the RHDV T = 3 capsid might bear resemblance to that of plant virus T = 3 capsids.
Collapse
Affiliation(s)
- Juan Bárcena
- Centro de Investigación en Sanidad Animal (CISA-INIA), Valdeolmos, 28130, Madrid, Spain
| | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Li S, Lin L, Wang H, Yin J, Ren Y, Zhao Z, Wen J, Zhou C, Zhang X, Li X, Wang J, Zhou Z, Liu J, Shao J, Lei T, Fang J, Xu N, Liu S. The epitope study on the SARS-CoV nucleocapsid protein. GENOMICS, PROTEOMICS & BIOINFORMATICS 2003; 1:198-206. [PMID: 15629032 PMCID: PMC5172353 DOI: 10.1016/s1672-0229(03)01025-8] [Citation(s) in RCA: 20] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
The nucleocapsid protein (N protein) has been found to be an antigenic protein in a number of coronaviruses. Whether the N protein in severe acute respiratory syndrome-associated coronavirus (SARS-CoV) is antigenic remains to be elucidated. Using Western blot and Enzyme-linked Immunosorbent Assay (ELISA), the recombinant N proteins and the synthesized peptides derived from the N protein were screened in sera from SARS patients. All patient sera in this study displayed strong positive immunoreactivities against the recombinant N proteins, whereas normal sera gave negative immunoresponses to these proteins, indicating that the N protein of SARS-CoV is an antigenic protein. Furthermore, the epitope sites in the N protein were determined by competition experiments, in which the recombinant proteins or the synthesized peptides competed against the SARS-CoV proteins to bind to the antibodies raised in SARS sera. One epitope site located at the C-terminus was confirmed as the most antigenic region in this protein. A detailed screening of peptide with ELISA demonstrated that the amino sequence from Codons 371 to 407 was the epitope site at the C-terminus of the N protein. Understanding of the epitope sites could be very significant for developing an effective diagnostic approach to SARS.
Collapse
|
21
|
Wurm T, Chen H, Hodgson T, Britton P, Brooks G, Hiscox JA. Localization to the nucleolus is a common feature of coronavirus nucleoproteins, and the protein may disrupt host cell division. J Virol 2001; 75:9345-56. [PMID: 11533198 PMCID: PMC114503 DOI: 10.1128/jvi.75.19.9345-9356.2001] [Citation(s) in RCA: 169] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2001] [Accepted: 06/22/2001] [Indexed: 12/17/2022] Open
Abstract
The subcellular localization of transmissible gastroenteritis virus (TGEV) and mouse hepatitis virus (MHV) (group I and group II coronaviruses, respectively) nucleoproteins (N proteins) were examined by confocal microscopy. The proteins were shown to localize either to the cytoplasm alone or to the cytoplasm and a structure in the nucleus. This feature was confirmed to be the nucleolus by using specific antibodies to nucleolin, a major component of the nucleolus, and by confocal microscopy to image sections through a cell expressing N protein. These findings are consistent with our previous report for infectious bronchitis virus (group III coronavirus) (J. A. Hiscox et al., J. Virol. 75:506-512, 2001), indicating that nucleolar localization of the N protein is a common feature of the coronavirus family and is possibly of functional significance. Nucleolar localization signals were identified in the domain III region of the N protein from all three coronavirus groups, and this suggested that transport of N protein to the nucleus might be an active process. In addition, our results suggest that the N protein might function to disrupt cell division. Thus, we observed that approximately 30% of cells transfected with the N protein appeared to be undergoing cell division. The most likely explanation for this is that the N protein induced a cell cycle delay or arrest, most likely in the G(2)/M phase. In a fraction of transfected cells expressing coronavirus N proteins, we observed multinucleate cells and dividing cells with nucleoli (which are only present during interphase). These findings are consistent with the possible inhibition of cytokinesis in these cells.
Collapse
Affiliation(s)
- T Wurm
- Virology Group, School of Animal and Microbial Sciences, University of Reading, Reading, Berkshire RG6 6AJ, United Kingdom
| | | | | | | | | | | |
Collapse
|
22
|
Bárcena J, Morales M, Vázquez B, Boga JA, Parra F, Lucientes J, Pagès-Manté A, Sánchez-Vizcaíno JM, Blasco R, Torres JM. Horizontal transmissible protection against myxomatosis and rabbit hemorrhagic disease by using a recombinant myxoma virus. J Virol 2000; 74:1114-23. [PMID: 10627521 PMCID: PMC111445 DOI: 10.1128/jvi.74.3.1114-1123.2000] [Citation(s) in RCA: 68] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/1999] [Accepted: 11/01/1999] [Indexed: 11/20/2022] Open
Abstract
We have developed a new strategy for immunization of wild rabbit populations against myxomatosis and rabbit hemorrhagic disease (RHD) that uses recombinant viruses based on a naturally attenuated field strain of myxoma virus (MV). The recombinant viruses expressed the RHDV major capsid protein (VP60) including a linear epitope tag from the transmissible gastroenteritis virus (TGEV) nucleoprotein. Following inoculation, the recombinant viruses induced specific antibody responses against MV, RHDV, and the TGEV tag. Immunization of wild rabbits by the subcutaneous and oral routes conferred protection against virulent RHDV and MV challenges. The recombinant viruses showed a limited horizontal transmission capacity, either by direct contact or in a flea-mediated process, promoting immunization of contact uninoculated animals.
Collapse
Affiliation(s)
- J Bárcena
- Centro de Investigación en Sanidad Animal, Valdeolmos, 28130 Madrid, Spain
| | | | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Martín Alonso JM, Casais R, Boga JA, Parra F. Processing of rabbit hemorrhagic disease virus polyprotein. J Virol 1996; 70:1261-5. [PMID: 8551592 PMCID: PMC189940 DOI: 10.1128/jvi.70.2.1261-1265.1996] [Citation(s) in RCA: 37] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
Expression of rabbit hemorrhagic disease virus (RHDV) cDNAs in vitro with rabbit reticulocyte lysates and in Escherichia coli have been used to study the proteolytic processing of RHDV polyprotein encoded by ORF1. An epitope tag was used for monitoring the gene products by a specific antibody. We have identified four gene products with molecular masses of 80, 43, 73, and 60 kDa, from the amino to the carboxy terminus of the polyprotein. The amino-terminal sequences of the 43- and 73-kDa products were determined and indicated that RHDV 3C proteinase cleaved Glu-Gly peptide bonds.
Collapse
Affiliation(s)
- J M Martín Alonso
- Departamento de Bioquímica y Biología Molecular, Universidad de Oviedo, Spain
| | | | | | | |
Collapse
|