1
|
Lu N, Li Z, Su D, Chen J, Zhao J, Gao Y, Liu Q, Liu G, Luo X, Luo R, Deng X, Zhu H, Luo Z. Design of novel chiral self-assembling peptides to explore the efficiency and mechanism of mRNA-FIPV vaccine delivery vehicles. Int J Pharm 2024; 660:124344. [PMID: 38885779 DOI: 10.1016/j.ijpharm.2024.124344] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Revised: 06/09/2024] [Accepted: 06/14/2024] [Indexed: 06/20/2024]
Abstract
The enhancement of conventional liposome and lipid nanoparticle (LNP) methodologies in the formulation and deployment of messenger RNA (mRNA) vaccines necessitates further refinement to augment both their effectiveness and biosafety profiles. Additionally, researching these innovative delivery carrier materials represents both a prominent focus and a significant challenge in the current scientific landscape. Here we designed new chiral self-assembling peptides as the delivery carrier for RNA vaccines to study the underlying mechanisms in the feline infectious peritonitis virus (FIPV) model system. Firstly, we successfully transcribed mature enhanced green fluorescent protein (EGFP) mRNA and feline infectious peritonitis virus nucleocapsid (FIPV N) mRNA in vitro from optimized vectors. Subsequently, we developed chiral self-assembling peptide-1 (CSP-1) and chiral self-assembling peptide-2 (CSP-2) peptides, taking into account the physical and chemical characteristics of nucleic acid molecules as well as the principles of self-assembling peptides, with the aim of improving the delivery efficiency of mRNA molecule complexes. We determined the optimal coating ratio between CSP and mRNA by electrophoretic mobility shift assay. We found that the peptides and mRNA complexes can protect the mRNA from RNase A enzyme and efficiently deliver mRNA into cells for target antigen proteins expression. Animal experiments confirmed that CSP-1/mRNA complex can effectively trigger immune response mechanisms involving IFN-γ and T cell activation. It can also stimulate CD4+ and CD8+ T cell proliferation and induce serum antibody titers up to 10,000 times higher. And no pathological changes were observed by immunohistochemistry in liver, spleen, and kidney, indicating that CSP-1 may be a safe and promising delivery system for mRNA vaccines. Methodologically, this research represents a novel endeavor in the utilization of chiral self-assembling peptides within the realm of mRNA vaccines. This approach not only introduces fresh prospects for employing such nanomaterials in various mRNA vaccines but also expands the potential for developing small molecules, proteins, and antibodies. Furthermore, it paves the way for new clinical applications of existing pharmaceuticals.
Collapse
Affiliation(s)
- Na Lu
- College of Basic Medical Sciences, Molecular Medicine and Cancer Research Center, Chongqing Medical University, Chongqing, China
| | - Zhaoxu Li
- College of Basic Medical Sciences, Molecular Medicine and Cancer Research Center, Chongqing Medical University, Chongqing, China; Department of Materials Science and Engineering, University of California, Irvine, CA, United States
| | - Di Su
- College of Basic Medical Sciences, Molecular Medicine and Cancer Research Center, Chongqing Medical University, Chongqing, China
| | - Jialei Chen
- College of Basic Medical Sciences, Molecular Medicine and Cancer Research Center, Chongqing Medical University, Chongqing, China
| | - Jiawei Zhao
- College of Basic Medical Sciences, Molecular Medicine and Cancer Research Center, Chongqing Medical University, Chongqing, China
| | - Yu Gao
- College of Basic Medical Sciences, Molecular Medicine and Cancer Research Center, Chongqing Medical University, Chongqing, China
| | - Qichen Liu
- College of Basic Medical Sciences, Molecular Medicine and Cancer Research Center, Chongqing Medical University, Chongqing, China
| | - Guicen Liu
- College of Basic Medical Sciences, Molecular Medicine and Cancer Research Center, Chongqing Medical University, Chongqing, China
| | - Xinyi Luo
- College of Basic Medical Sciences, Molecular Medicine and Cancer Research Center, Chongqing Medical University, Chongqing, China; Department of Physiology and Biophysics, University of California, Irvine, CA, United States
| | - Ruyue Luo
- College of Basic Medical Sciences, Molecular Medicine and Cancer Research Center, Chongqing Medical University, Chongqing, China; Department of Medicine, Northwestern University Feinberg School of Medicine,Chicago, IL,United States
| | - Xiaoyan Deng
- College of Basic Medical Sciences, Molecular Medicine and Cancer Research Center, Chongqing Medical University, Chongqing, China
| | - Huifang Zhu
- College of Basic Medical Sciences, Molecular Medicine and Cancer Research Center, Chongqing Medical University, Chongqing, China
| | - Zhongli Luo
- College of Basic Medical Sciences, Molecular Medicine and Cancer Research Center, Chongqing Medical University, Chongqing, China.
| |
Collapse
|
2
|
Xu L, Ren J, Li L, Wang M, Zhu G, Zheng H, Zeng Q, Shang Y, Li D. Vimentin inhibits peste des petits ruminants virus replication by interaction with nucleocapsid protein. Virology 2024; 595:110056. [PMID: 38552409 DOI: 10.1016/j.virol.2024.110056] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2023] [Revised: 02/29/2024] [Accepted: 03/07/2024] [Indexed: 05/18/2024]
Abstract
The Peste des petits ruminant virus (PPRV) is a member of the Paramyxoviridae family and is classified into the genus Measles virus. PPRV predominantly infects small ruminants, leading to mortality rates of nearly 100%, which have caused significant economic losses in developing countries. Host proteins are important in virus replication, but the PPRV nucleocapsid (N) protein-host interacting partners for regulating PPRV replication remain unclear. The present study confirmed the interaction between PPRV-N and the host protein vimentin by co-immunoprecipitation and co-localization experiments. Overexpression of vimentin suppressed PPRV replication, whereas vimentin knockdown had the opposite effect. Mechanistically, N was subjected to degradation via the ubiquitin/proteasome pathway, where vimentin recruits the E3 ubiquitin ligase NEDD4L to fulfill N-ubiquitination, resulting in the degradation of the N protein. These findings suggest that the host protein vimentin and E3 ubiquitin ligase NEDD4L have an anti-PPRV effect.
Collapse
Affiliation(s)
- Long Xu
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, 730070, China; State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, 730000, China; Gansu Province Research Center for Basic Disciplines of Pathogen Biology, Lanzhou 730046, China
| | - Jingjing Ren
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, 730000, China; Gansu Province Research Center for Basic Disciplines of Pathogen Biology, Lanzhou 730046, China
| | - Lingxia Li
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, 730000, China; Gansu Province Research Center for Basic Disciplines of Pathogen Biology, Lanzhou 730046, China
| | - Mengyi Wang
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, 730000, China; Gansu Province Research Center for Basic Disciplines of Pathogen Biology, Lanzhou 730046, China
| | - Guoqiang Zhu
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, 730000, China; Gansu Province Research Center for Basic Disciplines of Pathogen Biology, Lanzhou 730046, China
| | - Haixue Zheng
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, 730000, China; Gansu Province Research Center for Basic Disciplines of Pathogen Biology, Lanzhou 730046, China
| | - Qiaoying Zeng
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, 730070, China.
| | - Youjun Shang
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, 730000, China; Gansu Province Research Center for Basic Disciplines of Pathogen Biology, Lanzhou 730046, China.
| | - Dan Li
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, 730000, China; Gansu Province Research Center for Basic Disciplines of Pathogen Biology, Lanzhou 730046, China.
| |
Collapse
|
3
|
van den Hurk S, Regmi G, Naikare HK, Velayudhan BT. Advances in Laboratory Diagnosis of Coronavirus Infections in Cattle. Pathogens 2024; 13:524. [PMID: 39057751 PMCID: PMC11279749 DOI: 10.3390/pathogens13070524] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2024] [Revised: 06/17/2024] [Accepted: 06/18/2024] [Indexed: 07/28/2024] Open
Abstract
Coronaviruses cause infections in humans and diverse species of animals and birds with a global distribution. Bovine coronavirus (BCoV) produces predominantly two forms of disease in cattle: a respiratory form and a gastrointestinal form. All age groups of cattle are affected by the respiratory form of coronavirus, whereas the gastroenteric form causes neonatal diarrhea or calf scours in young cattle and winter dysentery in adult cattle. The tremendous impacts of bovine respiratory disease and the associated losses are well-documented and underscore the importance of this pathogen. Beyond this, studies have demonstrated significant impacts on milk production associated with outbreaks of winter dysentery, with up to a 30% decrease in milk yield. In North America, BCoV was identified for the first time in 1972, and it continues to be a significant economic concern for the cattle industry. A number of conventional and molecular diagnostic assays are available for the detection of BCoV from clinical samples. Conventional assays for BCoV detection include virus isolation, which is challenging from clinical samples, electron microscopy, fluorescent antibody assays, and various immunoassays. Molecular tests are mainly based on nucleic acid detection and predominantly include conventional and real-time polymerase chain reaction (PCR) assays. Isothermal amplification assays and genome sequencing have gained increased interest in recent years for the detection, characterization, and identification of BCoV. It is believed that isothermal amplification assays, such as loop-mediated isothermal amplification and recombinase polymerase amplification, among others, could aid the development of barn-side point-of-care tests for BCoV. The present study reviewed the literature on coronavirus infections in cattle from the last three and a half decades and presents information mainly on the current and advancing diagnostics in addition to epidemiology, clinical presentations, and the impact of the disease on the cattle industry.
Collapse
Affiliation(s)
- Shaun van den Hurk
- Athens Veterinary Diagnostic Laboratory, College of Veterinary Medicine, University of Georgia, Athens, GA 30602, USA;
| | - Girija Regmi
- Tifton Veterinary Diagnostic and Investigational Laboratory, College of Veterinary Medicine, University of Georgia, Tifton, GA 30602, USA;
| | - Hemant K. Naikare
- University of Minnesota Veterinary Diagnostic Laboratory, Saint Paul, MN 55108, USA;
| | - Binu T. Velayudhan
- Athens Veterinary Diagnostic Laboratory, College of Veterinary Medicine, University of Georgia, Athens, GA 30602, USA;
| |
Collapse
|
4
|
Kamboj A, Dumka S, Saxena MK, Singh Y, Kaur BP, da Silva SJR, Kumar S. A Comprehensive Review of Our Understanding and Challenges of Viral Vaccines against Swine Pathogens. Viruses 2024; 16:833. [PMID: 38932126 PMCID: PMC11209531 DOI: 10.3390/v16060833] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2024] [Revised: 05/18/2024] [Accepted: 05/22/2024] [Indexed: 06/28/2024] Open
Abstract
Pig farming has become a strategically significant and economically important industry across the globe. It is also a potentially vulnerable sector due to challenges posed by transboundary diseases in which viral infections are at the forefront. Among the porcine viral diseases, African swine fever, classical swine fever, foot and mouth disease, porcine reproductive and respiratory syndrome, pseudorabies, swine influenza, and transmissible gastroenteritis are some of the diseases that cause substantial economic losses in the pig industry. It is a well-established fact that vaccination is undoubtedly the most effective strategy to control viral infections in animals. From the period of Jenner and Pasteur to the recent new-generation technology era, the development of vaccines has contributed significantly to reducing the burden of viral infections on animals and humans. Inactivated and modified live viral vaccines provide partial protection against key pathogens. However, there is a need to improve these vaccines to address emerging infections more comprehensively and ensure their safety. The recent reports on new-generation vaccines against swine viruses like DNA, viral-vector-based replicon, chimeric, peptide, plant-made, virus-like particle, and nanoparticle-based vaccines are very encouraging. The current review gathers comprehensive information on the available vaccines and the future perspectives on porcine viral vaccines.
Collapse
Affiliation(s)
- Aman Kamboj
- College of Veterinary and Animal Sciences, G. B. Pant University of Agriculture and Technology, Pantnagar 263145, Uttarakhand, India; (A.K.); (M.K.S.); (Y.S.)
| | - Shaurya Dumka
- Department of Biosciences and Bioengineering, Indian Institute of Technology, Guwahati 781039, Assam, India; (S.D.); (B.P.K.)
| | - Mumtesh Kumar Saxena
- College of Veterinary and Animal Sciences, G. B. Pant University of Agriculture and Technology, Pantnagar 263145, Uttarakhand, India; (A.K.); (M.K.S.); (Y.S.)
| | - Yashpal Singh
- College of Veterinary and Animal Sciences, G. B. Pant University of Agriculture and Technology, Pantnagar 263145, Uttarakhand, India; (A.K.); (M.K.S.); (Y.S.)
| | - Bani Preet Kaur
- Department of Biosciences and Bioengineering, Indian Institute of Technology, Guwahati 781039, Assam, India; (S.D.); (B.P.K.)
| | | | - Sachin Kumar
- Department of Biosciences and Bioengineering, Indian Institute of Technology, Guwahati 781039, Assam, India; (S.D.); (B.P.K.)
| |
Collapse
|
5
|
Cao L, Kong X, Li X, Suo X, Duan Y, Yuan C, Zhang Y, Zheng H, Wang Q. A Customized Novel Blocking ELISA for Detection of Bat-Origin Swine Acute Diarrhea Syndrome Coronavirus Infection. Microbiol Spectr 2023; 11:e0393022. [PMID: 37272819 PMCID: PMC10434073 DOI: 10.1128/spectrum.03930-22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2022] [Accepted: 05/09/2023] [Indexed: 06/06/2023] Open
Abstract
Swine acute diarrhea syndrome coronavirus (SADS-CoV) is a newly discovered emerging alphacoronavirus. SADS-CoV shares over 90% genome sequence identity with bat alphacoronavirus HKU2. SADS-CoV was associated with severe diarrhea and high mortality rates in piglets. Accurate serological diagnosis of SADS-CoV infection is key in managing the emerging SADS-CoV. However, thus far there have been no effective antibody-based diagnostic tests for diagnose of SADS-CoV exposure. Here, monoclonal antibody (MAb) 6E8 against SADS-CoV N protein accurately recognized SADS-CoV infection. Then, MAb 6E8 was utilized as a blocking antibody to develop blocking ELISA (bELISA). We customized the rN coating antigen with concentration 0.25 μg/mL. According to receiver operator characteristic curve analysis, the cutoff value of the bELISA was determined as 38.19% when the max Youden index was 0.955, and specificity was 100%, and sensitivity was 95.5%. Specificity testing showed that there was no cross-reactivity with other serum positive swine enteric coronaviruses, such as porcine epidemic diarrhea virus (PEDV), transmissible gastroenteritis virus (TGEV), porcine deltacoronavirus (PDCoV), porcine rotavirus (PoRV), and porcine sapelovirus (PSV). In conclusion, we customized a novel and high-quality blocking ELISA for detection of SADS-CoV infection, and the current bELISA will be linked to a clinical and epidemiological assessment of SADS-CoV infection. IMPORTANCE SADS-CoV was reported to be of high potential for dissemination among various of host species. Accurate serological diagnosis of SADS-CoV infection is key in managing the emerging SADS-CoV. However, thus far there have been no effective antibody-based diagnostic tests for diagnose of SADS-CoV exposure. We customed a novel and high-quality bELISA assay for detection of SADS-CoV N protein antibodies, and the current bELISA will be linked to a clinical and epidemiological assessment of SADS-CoV infection.
Collapse
Affiliation(s)
- Liyan Cao
- Institute of Urban Agriculture, Chinese Academy of Agricultural Sciences, Chengdu, China
- State Key Laboratory of Veterinary Etiological Biology, Lanzhou Veterinary Research Institute, National Foot and Mouth Diseases Reference Laboratory, Chinese Academy of Agricultural Sciences, Lanzhou, China
- Chengdu National Agricultural Science and Technology Center, Chengdu, China
| | - Xiangyu Kong
- Institute of Urban Agriculture, Chinese Academy of Agricultural Sciences, Chengdu, China
- State Key Laboratory of Veterinary Etiological Biology, Lanzhou Veterinary Research Institute, National Foot and Mouth Diseases Reference Laboratory, Chinese Academy of Agricultural Sciences, Lanzhou, China
- Chengdu National Agricultural Science and Technology Center, Chengdu, China
| | - Xiangtong Li
- Institute of Urban Agriculture, Chinese Academy of Agricultural Sciences, Chengdu, China
- State Key Laboratory of Veterinary Etiological Biology, Lanzhou Veterinary Research Institute, National Foot and Mouth Diseases Reference Laboratory, Chinese Academy of Agricultural Sciences, Lanzhou, China
- Chengdu National Agricultural Science and Technology Center, Chengdu, China
| | - Xuepeng Suo
- Institute of Urban Agriculture, Chinese Academy of Agricultural Sciences, Chengdu, China
- State Key Laboratory of Veterinary Etiological Biology, Lanzhou Veterinary Research Institute, National Foot and Mouth Diseases Reference Laboratory, Chinese Academy of Agricultural Sciences, Lanzhou, China
- Chengdu National Agricultural Science and Technology Center, Chengdu, China
| | - Yueyue Duan
- Institute of Urban Agriculture, Chinese Academy of Agricultural Sciences, Chengdu, China
- State Key Laboratory of Veterinary Etiological Biology, Lanzhou Veterinary Research Institute, National Foot and Mouth Diseases Reference Laboratory, Chinese Academy of Agricultural Sciences, Lanzhou, China
- Chengdu National Agricultural Science and Technology Center, Chengdu, China
| | - Cong Yuan
- Institute of Urban Agriculture, Chinese Academy of Agricultural Sciences, Chengdu, China
- State Key Laboratory of Veterinary Etiological Biology, Lanzhou Veterinary Research Institute, National Foot and Mouth Diseases Reference Laboratory, Chinese Academy of Agricultural Sciences, Lanzhou, China
- Chengdu National Agricultural Science and Technology Center, Chengdu, China
| | - Yu Zhang
- Institute of Urban Agriculture, Chinese Academy of Agricultural Sciences, Chengdu, China
- State Key Laboratory of Veterinary Etiological Biology, Lanzhou Veterinary Research Institute, National Foot and Mouth Diseases Reference Laboratory, Chinese Academy of Agricultural Sciences, Lanzhou, China
- Chengdu National Agricultural Science and Technology Center, Chengdu, China
| | - Haixue Zheng
- State Key Laboratory of Veterinary Etiological Biology, Lanzhou Veterinary Research Institute, National Foot and Mouth Diseases Reference Laboratory, Chinese Academy of Agricultural Sciences, Lanzhou, China
| | - Qi Wang
- Institute of Urban Agriculture, Chinese Academy of Agricultural Sciences, Chengdu, China
- State Key Laboratory of Veterinary Etiological Biology, Lanzhou Veterinary Research Institute, National Foot and Mouth Diseases Reference Laboratory, Chinese Academy of Agricultural Sciences, Lanzhou, China
- Chengdu National Agricultural Science and Technology Center, Chengdu, China
| |
Collapse
|
6
|
Characterization and epitope mapping of monoclonal antibodies against PEDV N protein. Virology 2023; 579:29-37. [PMID: 36592554 DOI: 10.1016/j.virol.2022.12.011] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Revised: 12/20/2022] [Accepted: 12/20/2022] [Indexed: 12/30/2022]
Abstract
Porcine epidemic diarrhea virus (PEDV) causes acute diarrhea, vomiting, dehydration and high mortality in neonatal piglets. The nucleocapsid (N) protein of PEDV is a highly conserved protein with strong immunogenicity and palys an important role in PEDV diagnosis. However, epitopes on the PEDV N protein have not yet been well characterized. Here, 32 monoclonal antibodies (mAbs) against the PEDV N protein were produced and identified. Six new epitopes were first identified by using a high-throughput epitope mapping method named AbMap. Sequence analysis revealed that among the six epitopes five epitopes were highly conserved among different PEDV strains. We also confirmed that the mAbs derived from the six epitopes of PEDV N protein, have no cross-reactivity with transmissible gastro enteritis virus or porcine delta coronavirus. These mAbs and their defined epitopes will help to understand the N protein structure and immunological characteristics, and to develop a rapid, accurate PEDV diagnosis method.
Collapse
|
7
|
Irungbam M, Chitkara A, Singh VK, Sonkar SC, Dubey A, Bansal A, Shrivastava R, Goswami B, Manchanda V, Saxena S, Saxena R, Garg S, Husain F, Talukdar T, Kumar D, Koner BC. Evaluation of Performance of Detection of Immunoglobulin G and Immunoglobulin M Antibody Against Spike Protein of SARS-CoV-2 by a Rapid Kit in a Real-Life Hospital Setting. Front Microbiol 2022; 13:802292. [PMID: 35558113 PMCID: PMC9087894 DOI: 10.3389/fmicb.2022.802292] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Accepted: 01/26/2022] [Indexed: 12/04/2022] Open
Abstract
Background Antibody testing is often used for serosurveillance of coronavirus disease 2019 (COVID-19). Enzyme-linked immunosorbent assay and chemiluminescence-based antibody tests are quite sensitive and specific for such serological testing. Rapid antibody tests against different antigens are developed and effectively used for this purpose. However, their diagnostic efficiency, especially in real-life hospital setting, needs to be evaluated. Thus, the present study was conducted in a dedicated COVID-19 hospital in New Delhi, India, to evaluate the diagnostic efficacy of a rapid antibody kit against the receptor-binding domain (RBD) of the spike protein of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Methods Sixty COVID-19 confirmed cases by reverse transcriptase–polymerase chain reaction (RT-PCR) were recruited and categorized as early, intermediate, and late cases based on the days passed after their first RT-PCR–positive test report, with 20 subjects in each category. Twenty samples from pre-COVID era and 20 RT-PCR–negative collected during the study period were taken as controls. immunoglobulin M (IgM) and immunoglobulin G (IgG) antibodies against the RBD of the spike (S) protein of SARS-CoV-2 virus were detected by rapid antibody test and compared with the total antibody against the nucleocapsid (N) antigen of SARS-CoV-2 by electrochemiluminescence-based immunoassay (ECLIA). Results The detection of IgM against the RBD of the spike protein by rapid kit was less sensitive and less specific for the diagnosis of SARS-CoV-2 infection. However, diagnostic efficacy of IgG by rapid kit was highly sensitive and specific when compared with the total antibody against N antigen measured by ECLIA. Conclusion It can be concluded that detection of IgM against the RBD of S protein by rapid kit is less effective, but IgG detection can be used as an effective diagnostic tool for SARS-CoV-2 infection in real-life hospital setting.
Collapse
Affiliation(s)
- Monica Irungbam
- Department of Biochemistry, Maulana Azad Medical College and Associated Hospitals, New Delhi, India
| | - Anubhuti Chitkara
- Department of Biochemistry, Maulana Azad Medical College and Associated Hospitals, New Delhi, India
| | - Vijay Kumar Singh
- Multidisciplinary Research Unit (MRU), Maulana Azad Medical College and Associated Hospitals, New Delhi, India
| | - Subash Chandra Sonkar
- Multidisciplinary Research Unit (MRU), Maulana Azad Medical College and Associated Hospitals, New Delhi, India
| | - Abhisek Dubey
- Department of Biochemistry, Maulana Azad Medical College and Associated Hospitals, New Delhi, India
| | - Aastha Bansal
- Department of Biochemistry, Maulana Azad Medical College and Associated Hospitals, New Delhi, India
| | - Ritika Shrivastava
- Department of Biochemistry, Maulana Azad Medical College and Associated Hospitals, New Delhi, India
| | - Binita Goswami
- Department of Biochemistry, Maulana Azad Medical College and Associated Hospitals, New Delhi, India.,Multidisciplinary Research Unit (MRU), Maulana Azad Medical College and Associated Hospitals, New Delhi, India
| | - Vikas Manchanda
- Department of Microbiology, Maulana Azad Medical College and Associated Hospitals, New Delhi, India
| | - Sonal Saxena
- Department of Microbiology, Maulana Azad Medical College and Associated Hospitals, New Delhi, India
| | - Ritu Saxena
- Emergency Department, Lok Nayak Jai Prakash Narayan (LNJP) Hospital, New Delhi, India
| | - Sandeep Garg
- Department of Medicine, Lok Nayak Jai Prakash Narayan (LNJP) Hospital, New Delhi, India
| | - Farah Husain
- Department of Anesthesiology, Lok Nayak Jai Prakash Narayan (LNJP) Hospital, New Delhi, India
| | - Tanmay Talukdar
- Department of TB & Chest Diseases/Pulmonary Medicine, Lady Hardinge Medical College (LHMC), New Delhi, India
| | - Dinesh Kumar
- Food Safety and Standards Authority of India, Ministry of Health and Family Welfare (MoHFW), New Delhi, India
| | - Bidhan Chandra Koner
- Department of Biochemistry, Maulana Azad Medical College and Associated Hospitals, New Delhi, India.,Multidisciplinary Research Unit (MRU), Maulana Azad Medical College and Associated Hospitals, New Delhi, India
| |
Collapse
|
8
|
Tran TX, Lien NTK, Thu HT, Duy ND, Duong BTT, Quyen DV. Changes in the spike and nucleocapsid protein of porcine epidemic diarrhea virus strain in Vietnam-a molecular potential for the vaccine development? PeerJ 2021; 9:e12329. [PMID: 34721997 PMCID: PMC8530102 DOI: 10.7717/peerj.12329] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Accepted: 09/27/2021] [Indexed: 11/20/2022] Open
Abstract
Background Porcine epidemic diarrhea virus (PEDV) is a dangerous virus causing large piglet losses. PEDV spread rapidly between pig farms and caused the death of up to 90% of infected piglets. Current vaccines are only partially effective in providing immunity to suckling due to the rapid dissemination and ongoing evolution of PEDV. Methods In this study, the complete genome of a PEDV strain in Vietnam 2018 (IBT/VN/2018 strain) has been sequenced. The nucleotide sequence of each fragment was assembled to build a continuous complete sequence using the DNASTAR program. The complete nucleotide sequences and amino acid sequences of S, N, and ORF3 genes were aligned and analyzed to detect the mutations. Results The full-length genome was determined with 28,031 nucleotides in length which consisted of the 5'UTR, ORF1ab, S protein, ORF3, E protein, M protein, N protein, and 3'UTR region. The phylogenetic analysis showed that the IBT/VN/2018 strain was highly virulent belonged to the G2b subgroup along with the Northern American and Asian S-INDEL strains. Multiple sequence alignment of deduced amino acids revealed numerous mutations in the S, N, and ORF3 regions including one substitution 766P > L766 in the epitope SS6; two in the S0subdomain (135DN136 > 135SI136 and N144> D144); two in subdomain SHR1 at aa 1009L > M1009 and 1089S > L1089; one at aa 1279P > S1279 in subdomain SHR2 of the S protein; two at aa 364N > I364 and 378N > S378 in the N protein; four at aa 25L > S25, 70I > V70, 107C > F107, and 168D > N168 in the ORF3 protein. We identified two insertions (at aa 59NQGV62 and aa 145N) and one deletion (at aa 168DI169) in S protein. Remarkable, eight amino acid substitutions (294I > M294, 318A > S318, 335V > I335, 361A > T361, 497R > T497, 501SH502 > 501IY502, 506I > T506, 682V > I682, and 777P > L777) were found in SA subdomain. Besides, N- and O-glycosylation analysis of S, N, and ORF3 protein reveals three known sites (25G+, 123N+, and 62V+) and three novel sites (144D+, 1009M+, and 1279L+) in the IBT/VN/2018 strain compared with the vaccine strains. Taken together, the results showed that mutations in the S, N, and ORF3 genes can affect receptor specificity, viral pathogenicity, and the ability to evade the host immune system of the IBT/VN/2018 strain. Our results highlight the importance of molecular characterization of field strains of PEDV for the development of an effective vaccine to control PEDV infections in Vietnam.
Collapse
Affiliation(s)
- Thach Xuan Tran
- Dept of Molecular Microbiology, Institute of Biotechnology, Hanoi, Vietnam
| | - Nguyen T K Lien
- Functional of Genomics Lab, Institute of Genome Research, Vietnam Academy of Science and Technology, Hanoi, Vietnam
| | - Ha T Thu
- Dept of Molecular Microbiology, Institute of Biotechnology, Hanoi, Vietnam
| | - Nguyen Dinh Duy
- Dept of Molecular Microbiology, Institute of Biotechnology, Hanoi, Vietnam
| | - Bui T T Duong
- Dept of Molecular Microbiology, Institute of Biotechnology, Hanoi, Vietnam
| | - Dong Van Quyen
- Dept of Molecular Microbiology, Institute of Biotechnology, Hanoi, Vietnam.,University of Science and Technology of Ha Noi, Vietnam Academy of Science and Technology, Hanoi, Vietnam
| |
Collapse
|
9
|
Low ZY, Yip AJW, Sharma A, Lal SK. SARS coronavirus outbreaks past and present-a comparative analysis of SARS-CoV-2 and its predecessors. Virus Genes 2021; 57:307-317. [PMID: 34061288 PMCID: PMC8167830 DOI: 10.1007/s11262-021-01846-9] [Citation(s) in RCA: 7] [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: 03/17/2021] [Accepted: 05/10/2021] [Indexed: 02/08/2023]
Abstract
The Coronavirus Disease 2019 (COVID-19), a pneumonic disease caused by the SARS Coronavirus 2 (SARS-CoV-2), is the 7th Coronavirus to have successfully infected and caused an outbreak in humans. Genome comparisons have shown that previous isolates, the SARS-related coronavirus (SARSr-CoV), including the SARS-CoV are closely related, yet different in disease manifestation. Several explanations were suggested for the undetermined origin of SARS-CoV-2, in particular, bats, avian and Malayan pangolins as reservoir hosts, owing to the high genetic similarity. The general morphology and structure of all these viral isolates overlap with analogous disease symptoms such as fever, dry cough, fatigue, dyspnoea and headache, very similar to the current SARS-CoV-2. Chest CT scans for SARS-CoV-2, SARS-CoV and MERS-CoV reveal pulmonary lesions, bilateral ground-glass opacities, and segmental consolidation in the lungs, a common pathological trait. With greatly overlapping similarities among the previous coronavirus, the SARS-CoV, it becomes interesting to observe marked differences in disease severity of the SARS-CoV-2 thereby imparting it the ability to rapidly transmit, exhibit greater stability, bypass innate host defences, and increasingly adapt to their new host thereby resulting in the current pandemic. The most recent B.1.1.7, B.1.351 and P.1 variants of SARS-CoV-2, highlight the fact that changes in amino acids in the Spike protein can contribute to enhanced infection and transmission efficiency. This review covers a comparative analysis of previous coronavirus outbreaks and highlights the differences and similarities among different coronaviruses, including the most recent isolates that have evolved to become easily transmissible with higher replication efficiency in humans.
Collapse
Affiliation(s)
- Zheng Yao Low
- School of Science, Monash University, Sunway Campus, 47500, Bandar Sunway, Selangor DE, Malaysia
| | - Ashley Jia Wen Yip
- School of Science, Monash University, Sunway Campus, 47500, Bandar Sunway, Selangor DE, Malaysia
| | - Anshika Sharma
- School of Science, Monash University, Sunway Campus, 47500, Bandar Sunway, Selangor DE, Malaysia
| | - Sunil K Lal
- School of Science, Monash University, Sunway Campus, 47500, Bandar Sunway, Selangor DE, Malaysia.
- Tropical Medicine and Biology Platform, Monash University, Sunway Campus, 47500, Bandar Sunway, Selangor DE, Malaysia.
| |
Collapse
|
10
|
Feng Y, Jiang H, Qiu M, Liu L, Zou S, Li Y, Guo Q, Han N, Sun Y, Wang K, Lu L, Zhuang X, Zhang S, Chen S, Mo F. Multi-Epitope Vaccine Design Using an Immunoinformatic Approach for SARS-CoV-2. Pathogens 2021; 10:pathogens10060737. [PMID: 34208061 PMCID: PMC8230658 DOI: 10.3390/pathogens10060737] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2021] [Revised: 06/04/2021] [Accepted: 06/08/2021] [Indexed: 12/14/2022] Open
Abstract
Through 4 June 2021, COVID-19 has caused over 172.84 million cases of infection and 3.71 million deaths worldwide. Due to its rapid dissemination and high mutation rate, it is essential to develop a vaccine harboring multiple epitopes and efficacious against multiple variants to prevent the immune escape of SARS-CoV-2. An in silico approach based on the viral genome was applied to identify 19 high-immunogenic B-cell epitopes and 499 human leukocyte antigen (HLA)-restricted T-cell epitopes. Thirty multi-epitope peptide vaccines were designed by iNeo-Suite and manufactured by solid-phase synthesis. Docking analysis confirmed stable hydrogen bonds of epitopes with their corresponding HLA alleles. When four peptide candidates derived from the spike protein of SARS-CoV-2 were selected to immunize mice, a significantly larger amount of total IgG in serum, as well as an increase of CD19+ cells in the inguinal lymph nodes, were observed in the peptide-immunized mice compared to the control. The ratios of IFN-γ-secreting lymphocytes in CD4+ or CD8+ T-cells in the peptide-immunized mice were higher than those in the control mice. There were also a larger number of IFN-γ-secreting T-cells in the spleens of peptide-immunized mice. The peptide vaccines in this study successfully elicited antigen-specific humoral and cellular immune responses in mice. To further validate the safety and efficacy of this vaccine, animal studies using a primate model, as well as clinical trials in humans, are required.
Collapse
Affiliation(s)
- Ye Feng
- Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou 310001, China; (Y.F.); (S.Z.)
- Institute for Translational Medicine, Zhejiang University School of Medicine, Hangzhou 310002, China
| | - Haiping Jiang
- The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310007, China;
| | - Min Qiu
- Hangzhou Neoantigen Therapeutics Co., Ltd., Hangzhou 310058, China; (M.Q.); (L.L.); (Q.G.); (N.H.); (Y.S.); (K.W.); (L.L.); (S.Z.)
| | - Liang Liu
- Hangzhou Neoantigen Therapeutics Co., Ltd., Hangzhou 310058, China; (M.Q.); (L.L.); (Q.G.); (N.H.); (Y.S.); (K.W.); (L.L.); (S.Z.)
| | - Shengmei Zou
- Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou 310001, China; (Y.F.); (S.Z.)
- Institute for Translational Medicine, Zhejiang University School of Medicine, Hangzhou 310002, China
| | - Yun Li
- Zhejiang Forest Resources Monitoring Center, Hangzhou 310020, China;
| | - Qianpeng Guo
- Hangzhou Neoantigen Therapeutics Co., Ltd., Hangzhou 310058, China; (M.Q.); (L.L.); (Q.G.); (N.H.); (Y.S.); (K.W.); (L.L.); (S.Z.)
| | - Ning Han
- Hangzhou Neoantigen Therapeutics Co., Ltd., Hangzhou 310058, China; (M.Q.); (L.L.); (Q.G.); (N.H.); (Y.S.); (K.W.); (L.L.); (S.Z.)
| | - Yingqiang Sun
- Hangzhou Neoantigen Therapeutics Co., Ltd., Hangzhou 310058, China; (M.Q.); (L.L.); (Q.G.); (N.H.); (Y.S.); (K.W.); (L.L.); (S.Z.)
| | - Kui Wang
- Hangzhou Neoantigen Therapeutics Co., Ltd., Hangzhou 310058, China; (M.Q.); (L.L.); (Q.G.); (N.H.); (Y.S.); (K.W.); (L.L.); (S.Z.)
| | - Lantian Lu
- Hangzhou Neoantigen Therapeutics Co., Ltd., Hangzhou 310058, China; (M.Q.); (L.L.); (Q.G.); (N.H.); (Y.S.); (K.W.); (L.L.); (S.Z.)
| | - Xinlei Zhuang
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China;
| | - Shanshan Zhang
- Hangzhou Neoantigen Therapeutics Co., Ltd., Hangzhou 310058, China; (M.Q.); (L.L.); (Q.G.); (N.H.); (Y.S.); (K.W.); (L.L.); (S.Z.)
- Zhejiang California International Nanosystems Institute, Zhejiang University, Hangzhou 310058, China
| | - Shuqing Chen
- Hangzhou Neoantigen Therapeutics Co., Ltd., Hangzhou 310058, China; (M.Q.); (L.L.); (Q.G.); (N.H.); (Y.S.); (K.W.); (L.L.); (S.Z.)
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China;
- Zhejiang California International Nanosystems Institute, Zhejiang University, Hangzhou 310058, China
- Correspondence: (S.C.); (F.M.); Tel.: +86-571-8820-8411 (S.C.); +86-571-8608-8519 (F.M.)
| | - Fan Mo
- Hangzhou Neoantigen Therapeutics Co., Ltd., Hangzhou 310058, China; (M.Q.); (L.L.); (Q.G.); (N.H.); (Y.S.); (K.W.); (L.L.); (S.Z.)
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China;
- Vancouver Prostate Centre, University of British Columbia, Vancouver, BC V6H 3Z6, Canada
- Hangzhou AI-Force Therapeutics Co., Ltd., Hangzhou 310000, China
- Correspondence: (S.C.); (F.M.); Tel.: +86-571-8820-8411 (S.C.); +86-571-8608-8519 (F.M.)
| |
Collapse
|
11
|
Low ZY, Yip AJW, Sharma A, Lal SK. SARS coronavirus outbreaks past and present-a comparative analysis of SARS-CoV-2 and its predecessors. Virus Genes 2021. [PMID: 34061288 DOI: 10.1007/s11262‐021‐01846‐9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The Coronavirus Disease 2019 (COVID-19), a pneumonic disease caused by the SARS Coronavirus 2 (SARS-CoV-2), is the 7th Coronavirus to have successfully infected and caused an outbreak in humans. Genome comparisons have shown that previous isolates, the SARS-related coronavirus (SARSr-CoV), including the SARS-CoV are closely related, yet different in disease manifestation. Several explanations were suggested for the undetermined origin of SARS-CoV-2, in particular, bats, avian and Malayan pangolins as reservoir hosts, owing to the high genetic similarity. The general morphology and structure of all these viral isolates overlap with analogous disease symptoms such as fever, dry cough, fatigue, dyspnoea and headache, very similar to the current SARS-CoV-2. Chest CT scans for SARS-CoV-2, SARS-CoV and MERS-CoV reveal pulmonary lesions, bilateral ground-glass opacities, and segmental consolidation in the lungs, a common pathological trait. With greatly overlapping similarities among the previous coronavirus, the SARS-CoV, it becomes interesting to observe marked differences in disease severity of the SARS-CoV-2 thereby imparting it the ability to rapidly transmit, exhibit greater stability, bypass innate host defences, and increasingly adapt to their new host thereby resulting in the current pandemic. The most recent B.1.1.7, B.1.351 and P.1 variants of SARS-CoV-2, highlight the fact that changes in amino acids in the Spike protein can contribute to enhanced infection and transmission efficiency. This review covers a comparative analysis of previous coronavirus outbreaks and highlights the differences and similarities among different coronaviruses, including the most recent isolates that have evolved to become easily transmissible with higher replication efficiency in humans.
Collapse
Affiliation(s)
- Zheng Yao Low
- School of Science, Monash University, Sunway Campus, 47500, Bandar Sunway, Selangor DE, Malaysia
| | - Ashley Jia Wen Yip
- School of Science, Monash University, Sunway Campus, 47500, Bandar Sunway, Selangor DE, Malaysia
| | - Anshika Sharma
- School of Science, Monash University, Sunway Campus, 47500, Bandar Sunway, Selangor DE, Malaysia
| | - Sunil K Lal
- School of Science, Monash University, Sunway Campus, 47500, Bandar Sunway, Selangor DE, Malaysia. .,Tropical Medicine and Biology Platform, Monash University, Sunway Campus, 47500, Bandar Sunway, Selangor DE, Malaysia.
| |
Collapse
|
12
|
Almofti YA, Abd-Elrahman KA, Eltilib EEM. Vaccinomic approach for novel multi epitopes vaccine against severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2). BMC Immunol 2021; 22:22. [PMID: 33765919 PMCID: PMC7992937 DOI: 10.1186/s12865-021-00412-0] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Accepted: 03/10/2021] [Indexed: 12/26/2022] Open
Abstract
BACKGROUND The spread of a novel coronavirus termed severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) in China and other countries is of great concern worldwide with no effective vaccine. This study aimed to design a novel vaccine construct against SARS-CoV-2 from the spike S protein and orf1ab polyprotein using immunoinformatics tools. The vaccine was designed from conserved epitopes interacted against B and T lymphocytes by the combination of highly immunogenic epitopes with suitable adjuvant and linkers. RESULTS The proposed vaccine composed of 526 amino acids and was shown to be antigenic in Vaxigen server (0.6194) and nonallergenic in Allertop server. The physiochemical properties of the vaccine showed isoelectric point of 10.19. The instability index (II) was 31.25 classifying the vaccine as stable. Aliphatic index was 84.39 and the grand average of hydropathicity (GRAVY) was - 0.049 classifying the vaccine as hydrophilic. Vaccine tertiary structure was predicted, refined and validated to assess the stability of the vaccine via Ramachandran plot and ProSA-web servers. Moreover, solubility of the vaccine construct was greater than the average solubility provided by protein sol and SOLpro servers indicating the solubility of the vaccine construct. Disulfide engineering was performed to reduce the high mobile regions in the vaccine to enhance stability. Docking of the vaccine construct with TLR4 demonstrated efficient binding energy with attractive binding energy of - 338.68 kcal/mol and - 346.89 kcal/mol for TLR4 chain A and chain B respectively. Immune simulation significantly provided high levels of immunoglobulins, T-helper cells, T-cytotoxic cells and INF-γ. Upon cloning, the vaccine protein was reverse transcribed into DNA sequence and cloned into pET28a(+) vector to ensure translational potency and microbial expression. CONCLUSION A unique vaccine construct from spike S protein and orf1ab polyprotein against B and T lymphocytes was generated with potential protection against the pandemic. The present study might assist in developing a suitable therapeutics protocol to combat SARSCoV-2 infection.
Collapse
Affiliation(s)
- Yassir A Almofti
- Department of Molecular Biology and Bioinformatics, College of Veterinary Medicine, University of Bahri, Khartoum, Sudan.
| | - Khoubieb Ali Abd-Elrahman
- Department of Pharmaceutical Technology, College of Pharmacy, University of Medical Science and Technology (MUST), Khartoum, Sudan
| | - Elsideeq E M Eltilib
- Department of Molecular Biology and Bioinformatics, College of Veterinary Medicine, University of Bahri, Khartoum, Sudan
| |
Collapse
|
13
|
Turlewicz-Podbielska H, Pomorska-Mól M. Porcine Coronaviruses: Overview of the State of the Art. Virol Sin 2021; 36:833-851. [PMID: 33723809 PMCID: PMC7959302 DOI: 10.1007/s12250-021-00364-0] [Citation(s) in RCA: 81] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Accepted: 11/19/2020] [Indexed: 12/12/2022] Open
Abstract
Like RNA viruses in general, coronaviruses (CoV) exhibit high mutation rates which, in combination with their strong tendency to recombine, enable them to overcome the host species barrier and adapt to new hosts. It is currently known that six CoV are able to infect pigs. Four of them belong to the genus Alphacoronavirus [transmissible gastroenteritis coronavirus (TEGV), porcine respiratory coronavirus (PRCV), porcine epidemic diarrhea virus (PEDV), swine acute diarrhea syndrome coronavirus (SADS-CoV)], one of them to the genus Betacoronavirus [porcine hemagglutinating encephalomyelitis virus (PHEV)] and the last one to the genus Deltacoronavirus (PDCoV). PHEV was one of the first identified swine CoV and is still widespread, causing subclinical infections in pigs in several countries. PRCV, a spike deletion mutant of TGEV associated with respiratory tract infection, appeared in the 1980s. PRCV is considered non-pathogenic since its infection course is mild or subclinical. Since its appearance, pig populations have become immune to both PRCV and TGEV, leading to a significant reduction in the clinical and economic importance of TGEV. TGEV, PEDV and PDCoV are enteropathogenic CoV and cause clinically indistinguishable acute gastroenteritis in all age groups of pigs. PDCoV and SADS-CoV have emerged in 2014 (US) and in 2017 (China), respectively. Rapid diagnosis is crucial for controlling CoV infections and preventing them from spreading. Since vaccines are available only for some porcine CoV, prevention should focus mainly on a high level of biosecurity. In view of the diversity of CoV and the potential risk factors associated with zoonotic emergence, updating the knowledge concerning this area is essential.
Collapse
Affiliation(s)
- Hanna Turlewicz-Podbielska
- Department of Preclinical Sciences and Infectious Diseases, Faculty of Veterinary Medicine and Animal Sciences, Poznan University of Life Sciences, ul. Wołyńska 35, 60-637, Poznan, Poland
| | - Małgorzata Pomorska-Mól
- Department of Preclinical Sciences and Infectious Diseases, Faculty of Veterinary Medicine and Animal Sciences, Poznan University of Life Sciences, ul. Wołyńska 35, 60-637, Poznan, Poland.
| |
Collapse
|
14
|
Wong NA, Saier MH. The SARS-Coronavirus Infection Cycle: A Survey of Viral Membrane Proteins, Their Functional Interactions and Pathogenesis. Int J Mol Sci 2021; 22:1308. [PMID: 33525632 PMCID: PMC7865831 DOI: 10.3390/ijms22031308] [Citation(s) in RCA: 61] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Revised: 01/21/2021] [Accepted: 01/22/2021] [Indexed: 02/07/2023] Open
Abstract
Severe Acute Respiratory Syndrome Coronavirus-2 (SARS-CoV-2) is a novel epidemic strain of Betacoronavirus that is responsible for the current viral pandemic, coronavirus disease 2019 (COVID-19), a global health crisis. Other epidemic Betacoronaviruses include the 2003 SARS-CoV-1 and the 2009 Middle East Respiratory Syndrome Coronavirus (MERS-CoV), the genomes of which, particularly that of SARS-CoV-1, are similar to that of the 2019 SARS-CoV-2. In this extensive review, we document the most recent information on Coronavirus proteins, with emphasis on the membrane proteins in the Coronaviridae family. We include information on their structures, functions, and participation in pathogenesis. While the shared proteins among the different coronaviruses may vary in structure and function, they all seem to be multifunctional, a common theme interconnecting these viruses. Many transmembrane proteins encoded within the SARS-CoV-2 genome play important roles in the infection cycle while others have functions yet to be understood. We compare the various structural and nonstructural proteins within the Coronaviridae family to elucidate potential overlaps and parallels in function, focusing primarily on the transmembrane proteins and their influences on host membrane arrangements, secretory pathways, cellular growth inhibition, cell death and immune responses during the viral replication cycle. We also offer bioinformatic analyses of potential viroporin activities of the membrane proteins and their sequence similarities to the Envelope (E) protein. In the last major part of the review, we discuss complement, stimulation of inflammation, and immune evasion/suppression that leads to CoV-derived severe disease and mortality. The overall pathogenesis and disease progression of CoVs is put into perspective by indicating several stages in the resulting infection process in which both host and antiviral therapies could be targeted to block the viral cycle. Lastly, we discuss the development of adaptive immunity against various structural proteins, indicating specific vulnerable regions in the proteins. We discuss current CoV vaccine development approaches with purified proteins, attenuated viruses and DNA vaccines.
Collapse
Affiliation(s)
- Nicholas A. Wong
- Department of Molecular Biology, Division of Biological Sciences, University of California at San Diego, La Jolla, CA 92093-0116, USA
| | - Milton H. Saier
- Department of Molecular Biology, Division of Biological Sciences, University of California at San Diego, La Jolla, CA 92093-0116, USA
| |
Collapse
|
15
|
Deletion in the S1 Region of Porcine Epidemic Diarrhea Virus Reduces the Virulence and Influences the Virus-Neutralizing Activity of the Antibody Induced. Viruses 2020; 12:v12121378. [PMID: 33276502 PMCID: PMC7761297 DOI: 10.3390/v12121378] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Revised: 11/30/2020] [Accepted: 11/30/2020] [Indexed: 01/31/2023] Open
Abstract
Porcine epidemic diarrhea virus (PEDV) causes severe diarrhea and a high rate of mortality in suckling pigs. The epidemic of PEDV that occurred after 2013 was caused by non-insertion and deletion of S gene (S-INDEL) PEDV strains. During this epidemic, a variant of the non-S-INDEL PEDV strain with a large deletion of 205 amino acids on the spike gene (5-17-V) was also found to co-exist with a non-S-INDEL PEDV without deletion (5-17-O). Herein, we describe the differences in the complete genome, distribution, virulence, and antigenicity between strain 5-17-O and variant strain 5-17-V. The deletion of 205 amino acids was primarily located in the S1O domain and was associated with milder clinical signs and lower mortality in suckling pigs than those of the 5-17-O strain. The 5-17-V strain-induced antibody did not completely cross-neutralize the 5-17-O strain. In conclusion, the deletion in the S1 region reduces the virulence of PEDV and influences the virus-neutralizing activities of the antibody it induces.
Collapse
|
16
|
Lin P, Wang M, Wei Y, Kim T, Wei X. Coronavirus in human diseases: Mechanisms and advances in clinical treatment. MedComm (Beijing) 2020; 1:270-301. [PMID: 33173860 PMCID: PMC7646666 DOI: 10.1002/mco2.26] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Revised: 07/20/2020] [Accepted: 07/21/2020] [Indexed: 02/05/2023] Open
Abstract
Coronaviruses (CoVs), a subfamily of coronavirinae, are a panel of single-stranded RNA virus. Human coronavirus (HCoV) strains (HCoV-229E, HCoV-OC43, HCoV-HKU1, HCoV-NL63) usually cause mild upper respiratory diseases and are believed to be harmless. However, other HCoVs, associated with severe acute respiratory syndrome, Middle East respiratory syndrome, and COVID-19, have been identified as important pathogens due to their potent infectivity and lethality worldwide. Moreover, currently, no effective antiviral drugs treatments are available so far. In this review, we summarize the biological characters of HCoVs, their association with human diseases, and current therapeutic options for the three severe HCoVs. We also highlight the discussion about novel treatment strategies for HCoVs infections.
Collapse
Affiliation(s)
- Panpan Lin
- Laboratory of Aging Research and Cancer Drug Target State Key Laboratory of Biotherapy and Cancer Center National Clinical Research Center for Geriatrics West China Hospital Sichuan University Chengdu China
| | - Manni Wang
- Laboratory of Aging Research and Cancer Drug Target State Key Laboratory of Biotherapy and Cancer Center National Clinical Research Center for Geriatrics West China Hospital Sichuan University Chengdu China
| | - Yuquan Wei
- Laboratory of Aging Research and Cancer Drug Target State Key Laboratory of Biotherapy and Cancer Center National Clinical Research Center for Geriatrics West China Hospital Sichuan University Chengdu China
| | - Taewan Kim
- Wexner Medical Center The Ohio State University Columbus Ohio 43210 USA
| | - Xiawei Wei
- Laboratory of Aging Research and Cancer Drug Target State Key Laboratory of Biotherapy and Cancer Center National Clinical Research Center for Geriatrics West China Hospital Sichuan University Chengdu China
| |
Collapse
|
17
|
Saha RP, Sharma AR, Singh MK, Samanta S, Bhakta S, Mandal S, Bhattacharya M, Lee SS, Chakraborty C. Repurposing Drugs, Ongoing Vaccine, and New Therapeutic Development Initiatives Against COVID-19. Front Pharmacol 2020; 11:1258. [PMID: 32973505 PMCID: PMC7466451 DOI: 10.3389/fphar.2020.01258] [Citation(s) in RCA: 75] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2020] [Accepted: 07/30/2020] [Indexed: 12/15/2022] Open
Abstract
As the COVID-19 is still growing throughout the globe, a thorough investigation into the specific immunopathology of SARS-CoV-2, its interaction with the host immune system and pathogen evasion mechanism may provide a clear picture of how the pathogen can breach the host immune defenses in elderly patients and patients with comorbid conditions. Such studies will also reveal the underlying mechanism of how children and young patients can withstand the disease better. The study of the immune defense mechanisms and the prolonged immune memory from patients population with convalescent plasma may help in designing a suitable vaccine candidate not only for the current outbreak but also for similar outbreaks in the future. The vital drug candidates, which are being tested as potential vaccines or therapeutics against COVID-19, include live attenuated vaccine, inactivated or killed vaccine, subunit vaccine, antibodies, interferon treatment, repurposing existing drugs, and nucleic acid-based vaccines. Several organizations around the world have fast-tracked the development of a COVID-19 vaccine, and some drugs already went to phase III of clinical trials. Hence, here, we have tried to take a quick glimpse of the development stages of vaccines or therapeutic approaches to treat this deadly disease.
Collapse
Affiliation(s)
- Rudra P. Saha
- Department of Biotechnology, School of Life Science & Biotechnology, Adamas University, Kolkata, India
| | - Ashish Ranjan Sharma
- Institute for Skeletal Aging & Orthopedic Surgery, Hallym University-Chuncheon Sacred Heart Hospital, Chuncheon-si, South Korea
| | - Manoj K. Singh
- Department of Biotechnology, School of Life Science & Biotechnology, Adamas University, Kolkata, India
| | - Saikat Samanta
- Department of Biotechnology, School of Life Science & Biotechnology, Adamas University, Kolkata, India
| | - Swarnav Bhakta
- Department of Biotechnology, School of Life Science & Biotechnology, Adamas University, Kolkata, India
| | - Snehasish Mandal
- Department of Biotechnology, School of Life Science & Biotechnology, Adamas University, Kolkata, India
| | - Manojit Bhattacharya
- Institute for Skeletal Aging & Orthopedic Surgery, Hallym University-Chuncheon Sacred Heart Hospital, Chuncheon-si, South Korea
| | - Sang-Soo Lee
- Institute for Skeletal Aging & Orthopedic Surgery, Hallym University-Chuncheon Sacred Heart Hospital, Chuncheon-si, South Korea
| | - Chiranjib Chakraborty
- Department of Biotechnology, School of Life Science & Biotechnology, Adamas University, Kolkata, India
- Institute for Skeletal Aging & Orthopedic Surgery, Hallym University-Chuncheon Sacred Heart Hospital, Chuncheon-si, South Korea
| |
Collapse
|
18
|
Molecular Characterization of Porcine Epidemic Diarrhea Virus and Its New Genetic Classification Based on the Nucleocapsid Gene. Viruses 2020; 12:v12080790. [PMID: 32717934 PMCID: PMC7472284 DOI: 10.3390/v12080790] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2020] [Revised: 07/19/2020] [Accepted: 07/21/2020] [Indexed: 11/17/2022] Open
Abstract
Porcine epidemic diarrhea virus (PEDV) causes continuous, significant damage to the swine industry worldwide. By RT-PCR-based methods, this study demonstrated the ongoing presence of PEDV in pigs of all ages in Korea at the average detection rate of 9.92%. By the application of Bayesian phylogenetic analysis, it was found that the nucleocapsid (N) gene of PEDV could evolve at similar rates to the spike (S) gene at the order of 10-4 substitutions/site/year. Based on branching patterns of PEDV strains, three main N gene-base genogroups (N1, N2, and N3) and two sub-genogroups (N3a, N3b) were proposed in this study. By analyzing the antigenic index, possible antigenic differences also emerged in both the spike and nucleocapsid proteins between the three genogroups. The antigenic indexes of genogroup N3 strains were significantly lower compared with those of genogroups N1 and N2 strains in the B-cell epitope of the nucleocapsid protein. Similarly, significantly lower antigenic indexes in some parts of the B-cell epitope sequences of the spike protein (COE, S1D, and 2C10) were also identified. PEDV mutants derived from genetic mutations of the S and N genes may cause severe damage to swine farms by evading established host immunities.
Collapse
|
19
|
Identification and characterization of linear B cell epitopes on the nucleocapsid protein of porcine epidemic diarrhea virus using monoclonal antibodies. Virus Res 2020; 281:197912. [PMID: 32142743 PMCID: PMC7114562 DOI: 10.1016/j.virusres.2020.197912] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2019] [Revised: 03/02/2020] [Accepted: 03/02/2020] [Indexed: 01/19/2023]
Abstract
The nucleocapsid (N) protein of porcine epidemic diarrhea virus (PEDV), the most important pathogen causing severe diarrhea in piglets, is a highly conserved structural protein. In this study, 5 monoclonal antibodies (McAbs) against the PEDV N-protein were prepared and identified. Three new epitopes, 56QIRWRMRRGERI67, 318GYAQIASLAPNVAALLFGGNVA VRE342 and 398HEEAIYDDV406, were firstly identified in the viral N-protein, by using McAbs 3F10, 6A11, and 1C9. The epitope 398HEEAIYDDV406 was deleted in SH strain (isolated by our lab) and different between CV777 and YZ strain (isolated by our lab). To study the characters of this epitope, four peptides were synthesized according to the sequence of SH and CV777 and used in the study. The result showed that the 398th amino acid maybe an important amino acid of the epitope. Biological information analysis showed that the three B cell linear epitopes are highly conserved among different PEDV isolates. In addition, McAb 1C9, which attached to the epitope 398HEEAIYDDV406, showed variant reactivity with PEDV CV777, SH, YZ and MS strains. McAb 1C9 reacted with PEDV strains CV777 and YZ, but not with SH which had a deletion from 399 to 410 amino acids in N-protein (No. MK841494). Among the three McAbs, 6A11, 3F10 and 1C9, only 6A11 reacted with porcine transmissible gastroenteritis virus (TGEV) in immunofluorescence assay, therefore the other two could be used to distinguish TGEV and PEDV. These mAbs and their defined epitopes may provide useful tool for the study of the PEDV N-protein structure and function, and facilitate the development of diagnostic methods for PEDV.
Collapse
|
20
|
Chang CY, Peng JY, Cheng YH, Chang YC, Wu YT, Tsai PS, Chiou HY, Jeng CR, Chang HW. Development and comparison of enzyme-linked immunosorbent assays based on recombinant trimeric full-length and truncated spike proteins for detecting antibodies against porcine epidemic diarrhea virus. BMC Vet Res 2019; 15:421. [PMID: 31775769 PMCID: PMC6880432 DOI: 10.1186/s12917-019-2171-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2019] [Accepted: 11/08/2019] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Since 2010, outbreaks of genotype 2 (G2) porcine epidemic diarrhea virus (PEDV) have caused high mortality in neonatal piglets and have had devastating impacts on the swine industry in many countries. A reliable serological assay for evaluating the PEDV-specific humoral and mucosal immune response is important for disease survey, monitoring the efficacy of immunization, and designing strategies for the prevention and control of PED. Two PEDV spike (S) glycoprotein-based indirect enzyme-linked immunosorbent assays (ELISAs) were developed using G2b PEDV-Pintung 52 (PEDV-PT) trimeric full-length S and truncated S1-501 proteins derived from the human embryonic kidney (HEK)-293 cell expression system. The truncated S1-501 protein was selected from a superior expressed stable cell line. The sensitivity and specificity of these two ELISAs were compared to immunostaining of G2b PEDV-PT infected cells and to a commercial nucleocapsid (N)-based indirect ELISA kit using a panel of PEDV negative and hyperimmune sera. RESULTS The commercial N-based ELISA exhibited a sensitivity of 37%, a specificity of 100%, and a fair agreement (kappa = 0.37) with the immunostaining result. In comparison, the full-length S-based ELISA showed a sensitivity of 97.8%, a specificity of 94%, and an almost perfect agreement (kappa = 0.90) with the immunostaining result. Interestingly, the S1-501-based ELISA had even higher sensitivity of 98.9% and specificity of 99.1%, and an almost perfect agreement (kappa = 0.97) with the immunostaining result. A fair agreement (kappa< 0.4) was seen between the commercial N-based ELISA and either of our S-based ELISAs. However, the results of the full-length S-based ELISA shared an almost perfect agreement (kappa = 0.92) with that of S1-501-based ELISA. CONCLUSIONS Both full-length S-based and S1-501-based ELISAs exhibit high sensitivity and high specificity for detecting antibodies against PEDVs. Considering the high protein yield and cost-effectiveness, the S1-501-based ELISA could be used as a reliable, sensitive, specific, and economic serological test for PEDV.
Collapse
Affiliation(s)
- Chia-Yu Chang
- Graduate Institute of Molecular and Comparative Pathobiology, School of Veterinary Medicine, National Taiwan University, No. 1, Section 4, Roosevelt Rd., Taipei, 10617, Taiwan.,School of Veterinary Medicine, National Taiwan University, Taipei, 10617, Taiwan
| | - Ju-Yi Peng
- Graduate Institute of Molecular and Comparative Pathobiology, School of Veterinary Medicine, National Taiwan University, No. 1, Section 4, Roosevelt Rd., Taipei, 10617, Taiwan
| | - Yun-Han Cheng
- School of Veterinary Medicine, National Taiwan University, Taipei, 10617, Taiwan
| | - Yen-Chen Chang
- Graduate Institute of Molecular and Comparative Pathobiology, School of Veterinary Medicine, National Taiwan University, No. 1, Section 4, Roosevelt Rd., Taipei, 10617, Taiwan.,School of Veterinary Medicine, National Taiwan University, Taipei, 10617, Taiwan
| | - Yen-Tse Wu
- School of Veterinary Medicine, National Taiwan University, Taipei, 10617, Taiwan
| | - Pei-Shiue Tsai
- School of Veterinary Medicine, National Taiwan University, Taipei, 10617, Taiwan
| | - Hue-Ying Chiou
- Graduate Institute of Veterinary Pathobiology, College of Veterinary Medicine, National Chung Hsing University, Taichung, 402, Taiwan
| | - Chian-Ren Jeng
- Graduate Institute of Molecular and Comparative Pathobiology, School of Veterinary Medicine, National Taiwan University, No. 1, Section 4, Roosevelt Rd., Taipei, 10617, Taiwan.,School of Veterinary Medicine, National Taiwan University, Taipei, 10617, Taiwan
| | - Hui-Wen Chang
- Graduate Institute of Molecular and Comparative Pathobiology, School of Veterinary Medicine, National Taiwan University, No. 1, Section 4, Roosevelt Rd., Taipei, 10617, Taiwan. .,School of Veterinary Medicine, National Taiwan University, Taipei, 10617, Taiwan.
| |
Collapse
|
21
|
Generation, identification, and functional analysis of monoclonal antibodies against porcine epidemic diarrhea virus nucleocapsid. Appl Microbiol Biotechnol 2019; 103:3705-3714. [PMID: 30877355 PMCID: PMC7079923 DOI: 10.1007/s00253-019-09702-5] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2018] [Revised: 02/15/2019] [Accepted: 02/15/2019] [Indexed: 12/18/2022]
Abstract
The variant strains of porcine epidemic diarrhea virus (PEDV) severely threaten the pig industry worldwide and cause up to 100% mortality in suckling piglets. It is critically important and urgent to develop tools for detection of PEDV infection. In this study, we developed six monoclonal antibodies (mAbs) targeting N protein of PEDV and analyzed their applications on enzyme-linked immunosorbent assay (ELISA), indirect immunofluorescence assay (IFA), western blot assay, and flow cytometry assay. The results demonstrated that all these six mAbs were IgG1 isotype and κ chain. Among these six mAbs, 3F12 recognizes a linear epitope (VAAVKDALKSLGI) while the other five mAbs recognize different conformational epitopes formed by a specific peptide fragment or the full length of N protein. The functional analysis showed that all these six mAbs were applicable to ELISA, western blot, IFA, and flow cytometry assay. In conclusion, we developed six mAbs against PEDV-N protein to facilitate the early detection of PEDV infection using ELISA, western blot, IFA, and flow cytometry.
Collapse
|
22
|
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: 2.0] [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
|
23
|
Shan Y, Liu ZQ, Li GW, Chen C, Luo H, Liu YJ, Zhuo XH, Shi XF, Fang WH, Li XL. Nucleocapsid protein from porcine epidemic diarrhea virus isolates can antagonize interferon-λ production by blocking the nuclear factor-κB nuclear translocation. J Zhejiang Univ Sci B 2018; 19:570-580. [PMID: 29971995 PMCID: PMC6052364 DOI: 10.1631/jzus.b1700283] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Porcine epidemic diarrhea virus (PEDV) is a highly infectious pathogen that can cause severe diseases in pigs and result in enormous economic losses in the worldwide swine industry. Previous studies revealed that PEDV exhibits an obvious capacity for modulating interferon (IFN) signaling or expression. The newly discovered type III IFN, which plays a crucial role in antiviral immunity, has strong antiviral activity against PEDV proliferation in IPEC-J2 cells. In this study, we aimed to investigate the effect of PEDV nucleocapsid (N) protein on type III IFN-λ. We found that the N proteins of ten PEDV strains isolated between 2013 and 2017 from different local farms shared high nucleotide identities, while the N protein of the CV777 vaccine strain formed a monophyletic branch in the phylogenetic tree. The N protein of the epidemic strain could antagonize type III IFN, but not type I or type II IFN expression induced by polyinosinic-polycytidylic acid (poly(I:C)) in IPEC-J2 cells. Subsequently, we demonstrated that the inhibition of poly(I:C)-induced IFN-λ3 production by PEDV N protein was dependent on the blocking of nuclear factor-κB (NF-κB) nuclear translocation. These findings might help increase understanding of the pathogenesis of PEDV and its mechanisms for evading the host immune response.
Collapse
Affiliation(s)
- Ying Shan
- Zhejiang Province Key Lab of Preventive Veterinary Medicine, Institute of Preventive Veterainary Medicine, College of Animal Sciences, Zhejiang University, Hangzhou 310058, China
| | - Zi-Qi Liu
- Zhejiang Province Key Lab of Preventive Veterinary Medicine, Institute of Preventive Veterainary Medicine, College of Animal Sciences, Zhejiang University, Hangzhou 310058, China
| | - Guo-Wei Li
- Zhejiang Province Key Lab of Preventive Veterinary Medicine, Institute of Preventive Veterainary Medicine, College of Animal Sciences, Zhejiang University, Hangzhou 310058, China
| | - Cong Chen
- Zhejiang Province Key Lab of Preventive Veterinary Medicine, Institute of Preventive Veterainary Medicine, College of Animal Sciences, Zhejiang University, Hangzhou 310058, China
| | - Hao Luo
- Zhejiang Province Key Lab of Preventive Veterinary Medicine, Institute of Preventive Veterainary Medicine, College of Animal Sciences, Zhejiang University, Hangzhou 310058, China
| | - Ya-Jie Liu
- Zhejiang Province Key Lab of Preventive Veterinary Medicine, Institute of Preventive Veterainary Medicine, College of Animal Sciences, Zhejiang University, Hangzhou 310058, China
| | - Xun-Hui Zhuo
- Institute of Parasitic Disease, Zhejiang Academy of Medical Sciences, Hangzhou 310013, China
| | - Xing-Fen Shi
- Animal Products Quality Testing Center of Zhejiang Province, Hangzhou 310020, China
| | - Wei-Huan Fang
- Zhejiang Province Key Lab of Preventive Veterinary Medicine, Institute of Preventive Veterainary Medicine, College of Animal Sciences, Zhejiang University, Hangzhou 310058, China
| | - Xiao-Liang Li
- Zhejiang Province Key Lab of Preventive Veterinary Medicine, Institute of Preventive Veterainary Medicine, College of Animal Sciences, Zhejiang University, Hangzhou 310058, China
| |
Collapse
|
24
|
Han X, Qi J, Song H, Wang Q, Zhang Y, Wu Y, Lu G, Yuen KY, Shi Y, Gao GF. Structure of the S1 subunit C-terminal domain from bat-derived coronavirus HKU5 spike protein. Virology 2017; 507:101-109. [PMID: 28432925 PMCID: PMC7111649 DOI: 10.1016/j.virol.2017.04.016] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2017] [Revised: 04/10/2017] [Accepted: 04/15/2017] [Indexed: 02/05/2023]
Abstract
Accumulating evidence indicates that MERS-CoV originated from bat coronaviruses (BatCoVs). Previously, we demonstrated that both MERS-CoV and BatCoV HKU4 use CD26 as a receptor, but how the BatCoVs evolved to bind CD26 is an intriguing question. Here, we solved the crystal structure of the S1 subunit C-terminal domain of HKU5 (HKU5-CTD), another BatCoV that is phylogenetically related to MERS-CoV but cannot bind to CD26. We observed that the conserved core subdomain and those of other betacoronaviruses (betaCoVs) have a similar topology of the external subdomain, indicating the same ancestor of lineage C betaCoVs. However, two deletions in two respective loops located in HKU5-CTD result in conformational variations in CD26-binding interface and are responsible for the non-binding of HKU5-CTD to CD26. Combined with sequence variation in the HKU5-CTD receptor binding interface, we propose the necessity for surveilling the mutation in BatCoV HKU5 spike protein in case of bat-to-human interspecies transmission.
Collapse
Affiliation(s)
- Xue Han
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Jianxun Qi
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, China
| | - Hao Song
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, China; Research Network of Immunity and Health (RNIH), Beijing Institutes of Life Science, Chinese Academy of Sciences, Beijing 100101, China
| | - Qihui Wang
- CAS Key Laboratory of Microbial Physiological and Metabolic Engineering, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, China; Shenzhen Key Laboratory of Pathogen and Immunity, Shenzhen Third People's Hospital, Shenzhen 518112, China
| | - Yanfang Zhang
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Ying Wu
- Shenzhen Key Laboratory of Pathogen and Immunity, Shenzhen Third People's Hospital, Shenzhen 518112, China; School of Basic Medical Sciences, Wuhan University, Wuhan 430071, China
| | - Guangwen Lu
- West China Hospital Emergency Department (WCHED), State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, and Collaborative Innovation Center of Biotherapy, Chengdu, Sichuan 610041, China
| | - Kwok-Yung Yuen
- State Key Laboratory for Emerging Infectious Diseases, The University of Hong Kong, Pokfulam 999077, Hong Kong Special Administration Region; Department of Microbiology, The University of Hong Kong, Pokfulam 999077, Hong Kong Special Administration Region; Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Zhejiang University, Hangzhou 310003, China
| | - Yi Shi
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China; Shenzhen Key Laboratory of Pathogen and Immunity, Shenzhen Third People's Hospital, Shenzhen 518112, China; Center for Influenza Research and Early-warning (CASCIRE), Chinese Academy of Sciences, Beijing 100101, China.
| | - George F Gao
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China; Research Network of Immunity and Health (RNIH), Beijing Institutes of Life Science, Chinese Academy of Sciences, Beijing 100101, China; Shenzhen Key Laboratory of Pathogen and Immunity, Shenzhen Third People's Hospital, Shenzhen 518112, China; Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Zhejiang University, Hangzhou 310003, China; Center for Influenza Research and Early-warning (CASCIRE), Chinese Academy of Sciences, Beijing 100101, China; National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention (China CDC), Beijing 102206, China.
| |
Collapse
|
25
|
Cell Attachment Domains of the Porcine Epidemic Diarrhea Virus Spike Protein Are Key Targets of Neutralizing Antibodies. J Virol 2017; 91:JVI.00273-17. [PMID: 28381581 DOI: 10.1128/jvi.00273-17] [Citation(s) in RCA: 115] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2017] [Accepted: 03/24/2017] [Indexed: 01/12/2023] Open
Abstract
Porcine epidemic diarrhea virus (PEDV) causes enteric disease in pigs, resulting in significant economic losses to the swine industry worldwide. Current vaccination approaches against this emerging coronavirus are only partially effective, though natural infection protects pigs against reinfection and provides lactogenic immunity to suckling piglets. The viral spike (S) glycoprotein, responsible for receptor binding and cell entry, is the major target for neutralizing antibodies. However, knowledge of antibody epitopes, their nature and location in the spike structure, and the mechanisms by which the antibodies interfere with infection is scarce. Here we describe the generation and characterization of 10 neutralizing and nonneutralizing mouse monoclonal antibodies raised against the S1 receptor binding subunit of the S protein. By expression of different S1 protein fragments, six antibody epitope classes distributed over the five structural domains of the S1 subunit were identified. Characterization of antibodies for cross-reactivity and cross-neutralization revealed antigenic differences among PEDV strains. The epitopes of potent neutralizing antibodies segregated into two epitope classes and mapped within the N-terminal sialic acid binding domain and in the more C-terminal receptor binding domain. Antibody neutralization escape mutants displayed single amino acid substitutions that impaired antibody binding and neutralization and defined the locations of the epitopes. Our observations picture the antibody epitope landscape of the PEDV S1 subunit and reveal that its cell attachment domains are key targets of neutralizing antibodies.IMPORTANCE Porcine epidemic diarrhea virus (PEDV), an emerging porcine coronavirus, causes an economically important enteric disease in pigs. Effective PEDV vaccines for disease control are currently lacking. The spike (S) glycoprotein on the virion surface is the key player in virus cell entry and, therefore, the main target of neutralizing antibodies. To understand the antigenic landscape of the PEDV spike protein, we developed monoclonal antibodies against the spike protein's S1 receptor binding region and characterized their epitopes, neutralizing activity, and cross-reactivity toward multiple PEDV strains. Epitopes of antibodies segregated into six epitope classes dispersed over the multidomain S1 structure. Monoclonal antibodies revealed antigenic variability in B-cell epitopes between PEDV strains. The epitopes of neutralizing antibodies mapped to two distinct domains in S1 that are involved in binding to carbohydrate and proteinaceous cell surface molecules, respectively, indicating the importance of these cell attachment sites on the PEDV spike protein in eliciting a protective humoral immune response.
Collapse
|
26
|
Quinteros JA, Lee SW, Markham PF, Noormohammadi AH, Hartley CA, Legione AR, Coppo MJC, Vaz PK, Browning GF. Full genome analysis of Australian infectious bronchitis viruses suggests frequent recombination events between vaccine strains and multiple phylogenetically distant avian coronaviruses of unknown origin. Vet Microbiol 2016; 197:27-38. [PMID: 27938680 PMCID: PMC7117259 DOI: 10.1016/j.vetmic.2016.11.003] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2016] [Revised: 11/03/2016] [Accepted: 11/04/2016] [Indexed: 01/30/2023]
Abstract
Australian strains of infectious bronchitis virus (IBV) have been evolving independently for many years, with control achieved by vaccination with local attenuated strains. Previous studies have documented the emergence of recombinants over the last 20 years, with the most recent one, Ck/Aus/N1/08, detected in 2008. These recombinants did not appear to be controlled by the vaccines currently in use. In this study we sequenced the complete genomes of three emergent Australian strains of IBV (IBV/Ck/Aus/N1/88, IBV/Ck/Aus/N1/03 and IBV/Ck/Aus/N1/08) and a previously incompletely characterised vaccine strain, IBV/Ck/Aus/Armidale, and compared them to the genome of the vaccine strain VicS. We detected multiple recombination events throughout the genome between wild type viruses and the vaccine strains in all three emergent isolates. Moreover, we found that strain N1/88 was not entirely exogenous, as was previously hypothesised. Rather, it originated from a recombination event involving the VicS vaccine strain. The S glycoprotein genes of N1/88 and N1/03 were known to be genetically distinct from previously characterised circulating strains and from each other, and the original donors of these genes remains unknown. The S1 glycoprotein gene of N1/88, a subgroup 2 strain, shares a high nucleotide identity with the sequence of the S1 gene of the recent isolate N1/08. As the subgroup 2 strains have not been isolated for at least 20 years, it appears likely that an unknown avian coronavirus that was the donor of the S1 glycoprotein sequence of N1/88 in the 1980s is still recombining with IBV strains in the field.
Collapse
Affiliation(s)
- José A Quinteros
- Asia-Pacific Centre for Animal Health, Faculty of Veterinary and Agricultural Sciences, The University of Melbourne, Parkville, 3010, Victoria, Australia.
| | - Sang-Won Lee
- Asia-Pacific Centre for Animal Health, Faculty of Veterinary and Agricultural Sciences, The University of Melbourne, Parkville, 3010, Victoria, Australia; College of Veterinary Medicine, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul 143-701, Republic of Korea, Republic of Korea
| | - Philip F Markham
- Asia-Pacific Centre for Animal Health, Faculty of Veterinary and Agricultural Sciences, The University of Melbourne, Parkville, 3010, Victoria, Australia
| | - Amir H Noormohammadi
- Asia-Pacific Centre for Animal Health, Faculty of Veterinary and Agricultural Sciences, The University of Melbourne, Werribee, 3030, Victoria, Australia
| | - Carol A Hartley
- Asia-Pacific Centre for Animal Health, Faculty of Veterinary and Agricultural Sciences, The University of Melbourne, Parkville, 3010, Victoria, Australia
| | - Alistair R Legione
- Asia-Pacific Centre for Animal Health, Faculty of Veterinary and Agricultural Sciences, The University of Melbourne, Parkville, 3010, Victoria, Australia
| | - Mauricio J C Coppo
- Asia-Pacific Centre for Animal Health, Faculty of Veterinary and Agricultural Sciences, The University of Melbourne, Parkville, 3010, Victoria, Australia
| | - Paola K Vaz
- Asia-Pacific Centre for Animal Health, Faculty of Veterinary and Agricultural Sciences, The University of Melbourne, Parkville, 3010, Victoria, Australia
| | - Glenn F Browning
- Asia-Pacific Centre for Animal Health, Faculty of Veterinary and Agricultural Sciences, The University of Melbourne, Parkville, 3010, Victoria, Australia
| |
Collapse
|
27
|
Wang Q, Wong G, Lu G, Yan J, Gao GF. MERS-CoV spike protein: Targets for vaccines and therapeutics. Antiviral Res 2016; 133:165-77. [PMID: 27468951 PMCID: PMC7113765 DOI: 10.1016/j.antiviral.2016.07.015] [Citation(s) in RCA: 80] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2016] [Revised: 07/07/2016] [Accepted: 07/22/2016] [Indexed: 02/05/2023]
Abstract
The disease outbreak caused by Middle East respiratory syndrome coronavirus (MERS-CoV) is still ongoing in the Middle East. Over 1700 people have been infected since it was first reported in September 2012. Despite great efforts, licensed vaccines or therapeutics against MERS-CoV remain unavailable. The MERS-CoV spike (S) protein is an important viral antigen known to mediate host-receptor binding and virus entry, as well as induce robust humoral and cell-mediated responses in humans during infection. In this review, we highlight the importance of the S protein in the MERS-CoV life cycle, summarize recent advances in the development of vaccines and therapeutics based on the S protein, and discuss strategies that can be explored to develop new medical countermeasures against MERS-CoV. A licensed vaccine or therapeutic against MERS-CoV remains unavailable to date. The S protein plays a pivotal role for virus entry and thus is an ideal target for vaccine and antiviral development. DNA vaccines expressing the S protein merit further development for potential human application. nAbs and peptides targeting the S protein needs to be evaluated in NHPs before clinical trials.
Collapse
MESH Headings
- Animals
- Antibodies, Neutralizing/immunology
- Antibodies, Viral/immunology
- Antiviral Agents/pharmacology
- Antiviral Agents/therapeutic use
- Coronavirus Infections/prevention & control
- Coronavirus Infections/therapy
- Drug Discovery
- Humans
- Middle East Respiratory Syndrome Coronavirus/immunology
- Middle East Respiratory Syndrome Coronavirus/physiology
- Receptors, Virus/chemistry
- Receptors, Virus/metabolism
- Spike Glycoprotein, Coronavirus/chemistry
- Spike Glycoprotein, Coronavirus/genetics
- Spike Glycoprotein, Coronavirus/immunology
- Spike Glycoprotein, Coronavirus/metabolism
- Structure-Activity Relationship
- Vaccines, DNA/immunology
- Vaccines, Subunit/immunology
- Vaccines, Virus-Like Particle/immunology
- Viral Vaccines/immunology
- Virus Internalization
Collapse
Affiliation(s)
- Qihui Wang
- CAS Key Laboratory of Microbial Physiological and Metabolic Engineering, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, China; Shenzhen Key Laboratory of Pathogen and Immunity, Shenzhen Third People's Hospital, Shenzhen 518112, China.
| | - Gary Wong
- Shenzhen Key Laboratory of Pathogen and Immunity, Shenzhen Third People's Hospital, Shenzhen 518112, China; CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, China; CAS Center for Influenza Research and Early-warning (CASCIRE), Chinese Academy of Sciences, Beijing 100101, China
| | - Guangwen Lu
- West China Hospital Emergency Department (WCHED), State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, and Collaborative Innovation Center of Biotherapy, Chengdu, Sichuan 610041, China
| | - Jinghua Yan
- CAS Key Laboratory of Microbial Physiological and Metabolic Engineering, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, China; Shenzhen Key Laboratory of Pathogen and Immunity, Shenzhen Third People's Hospital, Shenzhen 518112, China; CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - George F Gao
- Shenzhen Key Laboratory of Pathogen and Immunity, Shenzhen Third People's Hospital, Shenzhen 518112, China; CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, China; CAS Center for Influenza Research and Early-warning (CASCIRE), Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China; National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention (China CDC), Beijing 102206, China; Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Zhejiang University, Hangzhou 310003, China; Savaid Medical School, University of Chinese Academy of Sciences, Beijing 100049, China.
| |
Collapse
|
28
|
Lin CM, Saif LJ, Marthaler D, Wang Q. Evolution, antigenicity and pathogenicity of global porcine epidemic diarrhea virus strains. Virus Res 2016; 226:20-39. [PMID: 27288724 PMCID: PMC7111424 DOI: 10.1016/j.virusres.2016.05.023] [Citation(s) in RCA: 190] [Impact Index Per Article: 23.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2016] [Revised: 05/21/2016] [Accepted: 05/21/2016] [Indexed: 12/16/2022]
Abstract
Evolution of global PEDV strains. Cross-reactivity between PEDV and other coronaviruses and antigenic variations among different PEDV strains. Pathologic features of different PEDV strains. Considerations for vaccine strain selection: PEDV virulence attenuation and in vivo cross-protection among PEDV variants.
Emerging and re-emerging coronaviruses cause morbidity and mortality in human and animal populations, resulting in serious public and animal health threats and economic losses. The ongoing outbreak of a highly contagious and deadly porcine epidemic diarrhea virus (PEDV) in Asia, the Americas and Europe is one example. Genomic sequence analyses of PEDV variants have revealed important insights into the evolution of PEDV. However, the antigenic variations among different PEDV strains are less explored, although they may contribute to the failure of PEDV vaccines in Asian countries. In addition, the evolution of PEDV results in variants with distinct genetic features and virulence differences; thus PEDV can serve as a model to explore the molecular mechanisms of coronavirus evolution and pathogenesis. In this article, we review the evolution, antigenic relationships and pathologic features of PEDV strains. This information and review of researches will aid in the development of strategies for control and prevention of PED.
Collapse
Affiliation(s)
- Chun-Ming Lin
- Food Animal Health Research Program, Ohio Agricultural Research and Development Center, College of Food, Agricultural and Environmental Sciences, Department of Veterinary Preventive Medicine, The Ohio State University, Wooster, OH, USA.
| | - Linda J Saif
- Food Animal Health Research Program, Ohio Agricultural Research and Development Center, College of Food, Agricultural and Environmental Sciences, Department of Veterinary Preventive Medicine, The Ohio State University, Wooster, OH, USA.
| | - Douglas Marthaler
- Department of Veterinary Population Medicine and Veterinary Diagnostic Laboratory, University of Minnesota, 1333 Gortner Avenue, St. Paul, MN 55108, United States.
| | - Qiuhong Wang
- Food Animal Health Research Program, Ohio Agricultural Research and Development Center, College of Food, Agricultural and Environmental Sciences, Department of Veterinary Preventive Medicine, The Ohio State University, Wooster, OH, USA.
| |
Collapse
|
29
|
Song Q, Stone S, Drebes D, Greiner LL, Dvorak CMT, Murtaugh MP. Characterization of anti-porcine epidemic diarrhea virus neutralizing activity in mammary secretions. Virus Res 2016; 226:85-92. [PMID: 27287711 PMCID: PMC7126973 DOI: 10.1016/j.virusres.2016.06.002] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2016] [Revised: 06/01/2016] [Accepted: 06/02/2016] [Indexed: 11/15/2022]
Abstract
Colostrum uptake is important for early establishment of lactogenic immunity. Neutralizing activity in milk and colostrum is associated with anti-spike IgA. Sow milk is a continuous supply of IgA with neutralizing activity. Temporal patterns of neutralizing antibody production in milk are variable.
Porcine epidemic diarrhea virus (PEDV) causes a severe clinical enteric disease in suckling neonates with up to 100% mortality, resulting in devastating economic losses to the pork industry in recent years. Maternal immunity via colostrum and milk is a vital source to neonates of passive protection against diarrhea, dehydration and death caused by PEDV. Comprehensive information on neutralizing activity (NA) against PEDV in mammary secretions is critically important for assessing the protective capacity of sows. Therefore, the objectives of this study were to characterize anti-PEDV neutralizing activity in mammary secretions. Anti-PEDV NA was present in colostrum, milk and serum from PEDV-infected sows as determined both by immunofluorescence and ELISA-based neutralizing assays, with neutralization levels higher in colostrum and milk than in serum. The highest NA was observed in colostrum on day 1, and decreased rapidly in milk at day 3, then gradually declined from day 3 to day 19 post-farrowing. Notably, the NA in mammary secretions showed various patterns of decline over time of lactation that may contribute to variation in sow protective capacities. The kinetics of NA decline were associated with total IgA and IgG antibody levels. Neutralizing activity significantly correlated with specific IgA primarily to spike domain 1 (S1) and domain 2 (S2) proteins of PEDV rather than to specific IgG in colostrum. Subsequently, the NA in milk was mainly related to specific IgA to S1 and S2 during lactation.
Collapse
Affiliation(s)
- Qinye Song
- Department of Veterinary and Biomedical Sciences, College of Veterinary Medicine, University of Minnesota, Saint Paul, MN, United States.
| | - Suzanne Stone
- Department of Veterinary and Biomedical Sciences, College of Veterinary Medicine, University of Minnesota, Saint Paul, MN, United States
| | - Donna Drebes
- Department of Veterinary and Biomedical Sciences, College of Veterinary Medicine, University of Minnesota, Saint Paul, MN, United States
| | - Laura L Greiner
- Carthage Innovative Swine Solutions, LLC, Carthage, IL, United States
| | - Cheryl M T Dvorak
- Department of Veterinary and Biomedical Sciences, College of Veterinary Medicine, University of Minnesota, Saint Paul, MN, United States
| | - Michael P Murtaugh
- Department of Veterinary and Biomedical Sciences, College of Veterinary Medicine, University of Minnesota, Saint Paul, MN, United States.
| |
Collapse
|
30
|
Loa CC, Wu CC, Lin TL. A Multiplex Polymerase Chain Reaction for Differential Detection of Turkey Coronavirus from Chicken Infectious Bronchitis Virus and Bovine Coronavirus. SPRINGER PROTOCOLS HANDBOOKS 2016. [PMCID: PMC7122580 DOI: 10.1007/978-1-4939-3414-0_12] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/01/2022]
Abstract
A multiplex polymerase chain reaction (PCR) method for differential detection of turkey coronavirus (TCoV), infectious bronchitis virus (IBV), and bovine coronavirus (BCoV) is presented in this chapter. Primers are designed from the conserved or variable regions of nucleocapsid (N) or spike (S) protein genes of TCoV, IBV, and BCoV and used in the same PCR reaction. Reverse transcription followed by PCR reaction is used to amplify a portion of N or S gene of the corresponding coronaviruses. Two PCR products, a 356-bp band corresponding to N gene and a 727-bp band corresponding to S gene, are obtained for TCoV. In contrast, one PCR product of 356 bp corresponding to a fragment of N gene is obtained for IBV strains and one PCR product of 568 bp corresponding to a fragment of S gene is obtained for BCoV.
Collapse
|
31
|
Wang L. Recombinant Turkey Coronavirus Nucleocapsid Protein Expressed in Escherichia coli. SPRINGER PROTOCOLS HANDBOOKS 2016. [PMCID: PMC7122433 DOI: 10.1007/978-1-4939-3414-0_4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Expression and purification of turkey coronavirus (TCoV) nucleocapsid (N) protein from a prokaryotic expression system as histidine-tagged fusion protein are presented in this chapter. Expression of histidine-tagged fusion N protein with a molecular mass of 57 kDa is induced with isopropyl β-d-1-thiogalactopyranoside (IPTG). The expressed N protein inclusion body is extracted and purified by chromatography on nickel-agarose column to near homogeneity. The protein recovery can be 10 mg from 100 ml of bacterial culture. The purified N protein is a superior source of TCoV antigen for antibody-capture ELISA for detection of antibodies to TCoV.
Collapse
Affiliation(s)
- Leyi Wang
- Animal Disease Diagnostic Lab, Ohio Department of Agriculture, Reynoldsburg, Ohio USA
| |
Collapse
|
32
|
Ouyang K, Shyu DL, Dhakal S, Hiremath J, Binjawadagi B, Lakshmanappa YS, Guo R, Ransburgh R, Bondra KM, Gauger P, Zhang J, Specht T, Gilbertie A, Minton W, Fang Y, Renukaradhya GJ. Evaluation of humoral immune status in porcine epidemic diarrhea virus (PEDV) infected sows under field conditions. Vet Res 2015; 46:140. [PMID: 26667229 PMCID: PMC4699368 DOI: 10.1186/s13567-015-0285-x] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2015] [Accepted: 11/17/2015] [Indexed: 11/21/2022] Open
Abstract
Porcine epidemic diarrhea virus (PEDV) is an economically devastating enteric disease in the swine industry. The virus infects pigs of all ages, but it cause severe clinical disease in neonatal suckling pigs with up to 100% mortality. Currently, available vaccines are not completely effective and feedback methods utilizing PEDV infected material has variable success in preventing reinfection. Comprehensive information on the levels and duration of effector/memory IgA and IgG antibody secreting B cell response in the intestines and lymphoid organs of PEDV-infected sows, and their association with specific antibody levels in clinical samples such as plasma, oral fluid, and feces is important. Therefore, our goal in this study was to quantify PEDV specific IgA and IgG B cell responses in sows at approximately 1 and 6 months post-infection in commercial swine herds, including parity one and higher sows. Our data indicated that evaluation of both PEDV specific IgA and IgG antibody levels in the plasma and oral fluid (but not feces) samples is beneficial in disease diagnosis. PEDV specific B cell response in the intestine and spleen of infected sows decline by 6 months, and this associates with specific antibody levels in the plasma and oral fluid samples; but the virus neutralization titers in plasma remains high beyond 6 months post-infection. In conclusion, in sows infected with PEDV the presence of effector/memory B cell response and strong virus neutralization titers in plasma up to 6 months post-infection, suggests their potential to protect sows from reinfection and provide maternal immunity to neonates, but challenge studies are required to confirm such responses.
Collapse
Affiliation(s)
- Kang Ouyang
- Food Animal Health Research Program (FAHRP), OARDC, Department of Veterinary Preventive Medicine, The Ohio State University, Wooster, OH, 44691, USA. .,College of Animal Science and Technology, Guangxi University, Nanning, China.
| | - Duan-Liang Shyu
- Food Animal Health Research Program (FAHRP), OARDC, Department of Veterinary Preventive Medicine, The Ohio State University, Wooster, OH, 44691, USA.
| | - Santosh Dhakal
- Food Animal Health Research Program (FAHRP), OARDC, Department of Veterinary Preventive Medicine, The Ohio State University, Wooster, OH, 44691, USA.
| | - Jagadish Hiremath
- Food Animal Health Research Program (FAHRP), OARDC, Department of Veterinary Preventive Medicine, The Ohio State University, Wooster, OH, 44691, USA.
| | - Basavaraj Binjawadagi
- Food Animal Health Research Program (FAHRP), OARDC, Department of Veterinary Preventive Medicine, The Ohio State University, Wooster, OH, 44691, USA.
| | - Yashavanth S Lakshmanappa
- Food Animal Health Research Program (FAHRP), OARDC, Department of Veterinary Preventive Medicine, The Ohio State University, Wooster, OH, 44691, USA.
| | - Rui Guo
- Department of Diagnostic Medicine and Pathobiology, College of Veterinary Medicine, Kansas State University, Manhattan, KS, 66506, USA.
| | - Russell Ransburgh
- Department of Diagnostic Medicine and Pathobiology, College of Veterinary Medicine, Kansas State University, Manhattan, KS, 66506, USA.
| | - Kathryn M Bondra
- Food Animal Health Research Program (FAHRP), OARDC, Department of Veterinary Preventive Medicine, The Ohio State University, Wooster, OH, 44691, USA.
| | - Phillip Gauger
- Veterinary Diagnostic and Production Animal Medicine, Iowa State University, Ames, IA, USA.
| | - Jianqiang Zhang
- Veterinary Diagnostic and Production Animal Medicine, Iowa State University, Ames, IA, USA.
| | - Terry Specht
- Four Star Veterinary Services, Chickasaw, OH, 45826, USA.
| | | | - William Minton
- Four Star Veterinary Services, Chickasaw, OH, 45826, USA.
| | - Ying Fang
- Department of Diagnostic Medicine and Pathobiology, College of Veterinary Medicine, Kansas State University, Manhattan, KS, 66506, USA.
| | - Gourapura J Renukaradhya
- Food Animal Health Research Program (FAHRP), OARDC, Department of Veterinary Preventive Medicine, The Ohio State University, Wooster, OH, 44691, USA.
| |
Collapse
|
33
|
Shen H, Zhang C, Guo P, Liu Z, Zhang J. Effective inhibition of porcine epidemic diarrhea virus by RNA interference in vitro. Virus Genes 2015; 51:252-9. [PMID: 26329934 PMCID: PMC7088742 DOI: 10.1007/s11262-015-1242-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2015] [Accepted: 08/20/2015] [Indexed: 11/25/2022]
Abstract
Porcine epidemic diarrhea virus (PEDV) is a member of the coronaviridae family, which can cause acute and highly contagious enteric disease of swine characterized by severe entero-pathogenic diarrhea in piglets. Currently, the vaccines of PEDV are only partially effective and there is no specific drug available for treatment of PEDV infection. To exploit the possibility of using RNA interference (RNAi) as a strategy against PEDV infection, five shRNA-expressing plasmids targeting the N, M, and S genes of PEDV were constructed and transfected into Vero cells. The cytopathic effect and MTS assays demonstrated that two shRNAs (pSilencer4.1-M1 and pSilencer4.1-N) were capable of protecting cells against PEDV invasion with very high specificity and efficiency. The two shRNA expression plasmids were also able to inhibit the PEDV replication significantly, as shown by detection of virus titers (TCID50/mL). A real-time quantitative RT-PCR further confirmed that the amounts of viral RNAs in cell cultures pre-transfected with these two plasmids were reduced by 95.0 %. Our results suggest that RNAi might be a promising new strategy against PEDV infection.
Collapse
Affiliation(s)
- Haiyan Shen
- Institute of Animal Health, Guangdong Academy of Agricultural Sciences, Guangdong Open Laboratory of Veterinary Public Health, Guangdong Provincial Key Laboratory of Livestock Disease Prevention, Guangzhou, 510640, Guangdong, China
| | - Chunhong Zhang
- Institute of Animal Health, Guangdong Academy of Agricultural Sciences, Guangdong Open Laboratory of Veterinary Public Health, Guangdong Provincial Key Laboratory of Livestock Disease Prevention, Guangzhou, 510640, Guangdong, China
| | - Pengju Guo
- Guangdong Laboratory Animals Monitoring Institute, Guangzhou, 510640, Guangdong, China
| | - Zhicheng Liu
- Institute of Animal Health, Guangdong Academy of Agricultural Sciences, Guangdong Open Laboratory of Veterinary Public Health, Guangdong Provincial Key Laboratory of Livestock Disease Prevention, Guangzhou, 510640, Guangdong, China
| | - Jianfeng Zhang
- Institute of Animal Health, Guangdong Academy of Agricultural Sciences, Guangdong Open Laboratory of Veterinary Public Health, Guangdong Provincial Key Laboratory of Livestock Disease Prevention, Guangzhou, 510640, Guangdong, China.
| |
Collapse
|
34
|
Development of an indirect ELISA, blocking ELISA, fluorescent microsphere immunoassay and fluorescent focus neutralization assay for serologic evaluation of exposure to North American strains of Porcine Epidemic Diarrhea Virus. BMC Vet Res 2015; 11:180. [PMID: 26232106 PMCID: PMC4522128 DOI: 10.1186/s12917-015-0500-z] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2015] [Accepted: 07/20/2015] [Indexed: 11/10/2022] Open
Abstract
Background Recent, severe outbreaks of porcine epidemic diarrhea virus (PEDV) in Asia and North America highlight the need for well-validated diagnostic tests for the identification of PEDV infected animals and evaluation of their immune status to this virus. PEDV was first detected in the U.S. in May 2013 and spread rapidly across the country. Some serological assays for PEDV have been previously described, but few were readily available in the U.S. Several U.S. laboratories quickly developed indirect fluorescent antibody (IFA) assays for the detection of antibodies to PEDV in swine serum, indicating prior exposure. However, the IFA has several disadvantages, including low throughput and relatively subjective interpretation. Different serologic test formats have advantages and disadvantages, depending on the questions being asked, so a full repertoire of tests is useful. Therefore, the objective of this study was to develop and validate multiple improved serological assays for PEDV, including an indirect ELISA (iELISA); a highly specific monoclonal antibody-based blocking ELISA (bELISA); fluorescent microsphere immunoassays (FMIA) that can be multiplexed to monitor exposure to multiple antigens and pathogens simultaneously; and a fluorescent focus neutralization assay (FFN) to measure functional virus neutralizing antibodies. Results A recombinant North American nucleoprotein (NP) based iELISA was developed and validated along with a bELISA using newly developed PEDV-NP specific biotinylated monoclonal antibodies (mAbs) and an FMIA using magnetic beads coupled with expressed NA PEDV-NP. Receiver operating characteristic (ROC) analysis was performed using swine serum samples (iELISA n = 1486, bELISA n = 1186, FMIA n = 1420). The ROC analysis for the FMIA showed estimated sensitivity and specificity of 98.2 and 99.2 %, respectively. The iELISA and bELISA showed a sensitivity and specificity of 97.9 and 97.6 %; and 98.2 and 98.9 %, respectively. Inter-rater (kappa) agreement was calculated to be 0.941 between iELISA and IFA, 0.945 between bELISA and IFA and 0.932 between FMIA and IFA. Similar comparative kappa values were observed between the iELISA, bELISA and FMIA, which demonstrated a significant level of testing agreement among the three assays. No cross-reactivity with the closely related coronaviruses, transmissible gastroenteritis virus (TGEV) or porcine respiratory coronavirus (PRCV) was noted with these assays. All three assays detected seroconversion of naïve animals within 6–9 days post exposure. The FFN assay allows relative quantitation of functional neutralizing antibodies in serum, milk or colostrum samples. Conclusion Well-validated iELISA, bELISA and FMIA assays for the detection of PEDV antibodies were developed and showed good correlation with IFA and each other. Each assay format has advantages that dictate how they will be used in the field. Newly developed mAbs to the PEDV-NP were used in the bELISA and for expediting FFN testing in the detection and quantitation of neutralizing antibodies. In addition, these PEDV mAbs are useful for immunohistochemistry, fluorescent antibody staining and other antigen-based tests. Measurement of neutralizing antibody responses using the FFN assay may provide a valuable tool for assessment of vaccine candidates or protective immunity.
Collapse
|
35
|
Yu X, Shi L, Lv X, Yao W, Cao M, Yu H, Wang X, Zheng S. Development of a real-time reverse transcription loop-mediated isothermal amplification method for the rapid detection of porcine epidemic diarrhea virus. Virol J 2015; 12:76. [PMID: 25972083 PMCID: PMC4459462 DOI: 10.1186/s12985-015-0297-1] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2014] [Accepted: 04/09/2015] [Indexed: 01/21/2023] Open
Abstract
Background Porcine epidemic diarrhea (PED) is an acute and highly contagious enteric disease characterized by severe enteritis, vomiting and watery diarrhea in swine. Recently, the outbreak of the epidemic disease has been a serious problem in swine industry. The objective of this study is to develop a rapid, sensitive, and real-time reverse transcription loop-mediated isothermal amplification (RT-LAMP) method for the detection of porcine epidemic diarrhea virus (PEDV) in less equipped laboratories. Results The optimal reaction condition of the current real-time RT-LAMP for PEDV was 62 °C for 45 min. It was capable of detecting PEDV from clinical samples and differentiating PEDV from several related porcine viruses, while it did not require additional expensive equipment. The minimum detection limit of the real-time RT-LAMP assay was 0.07PFU per reaction for PEDV RNA, making this assay approximately 100-fold more sensitive than that of one-step RT-PCR. By screening a panel of clinical specimens, the results showed that this method presented a similar sensitivity with real-time RT-PCR and was somewhat sensitive than one-step RT-PCR in detection of clinical samples. Conclusions In this study, we have developed a new real-time RT-LAMP method, which is rapid, sensitive and efficient to detect PEDV.This method holds great promises not only in laboratory detection and discrimination of PEDV but also in large scale field and clinical studies.
Collapse
Affiliation(s)
- Xuewu Yu
- College of Veterinary Medicine, Northeast Agricultural University, No.59, Mucai street, Xiangfan District, Harbin, 150030, China. .,Animal Epidemic Diseases Control and Prevention Center of Liaoning Province, Shenyang, China.
| | - Lin Shi
- College of Veterinary Medicine, Northeast Agricultural University, No.59, Mucai street, Xiangfan District, Harbin, 150030, China. .,Animal Epidemic Diseases Control and Prevention Center of Liaoning Province, Shenyang, China.
| | - Xiaoping Lv
- College of Veterinary Medicine, Northeast Agricultural University, No.59, Mucai street, Xiangfan District, Harbin, 150030, China.
| | - Wei Yao
- Animal Epidemic Diseases Control and Prevention Center of Liaoning Province, Shenyang, China.
| | - Minghui Cao
- China Institute of Veterinary Drugs Control, Beijing, China.
| | - Hanxun Yu
- Animal Epidemic Diseases Control and Prevention Center of Dalian city, Dalian, China.
| | - Xiurong Wang
- Animal Influenza Laboratory of the Ministry of Agriculture and State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, China.
| | - Shimin Zheng
- College of Veterinary Medicine, Northeast Agricultural University, No.59, Mucai street, Xiangfan District, Harbin, 150030, China.
| |
Collapse
|
36
|
Antigenic relationships among porcine epidemic diarrhea virus and transmissible gastroenteritis virus strains. J Virol 2015; 89:3332-42. [PMID: 25589635 DOI: 10.1128/jvi.03196-14] [Citation(s) in RCA: 83] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
UNLABELLED Porcine epidemic diarrhea virus (PEDV) and transmissible gastroenteritis virus (TGEV) are economically important swine enteropathogenic coronaviruses. These two viruses belong to two distinct species of the Alphacoronavirus genus within Coronaviridae and induce similar clinical signs and pathological lesions in newborn piglets, but they are presumed to be antigenically distinct. In the present study, two-way antigenic cross-reactivity examinations between the prototype PEDV CV777 strain, three distinct U.S. PEDV strains (the original highly virulent PC22A, S indel Iowa106, and S 197del PC177), and two representative U.S. TGEV strains (Miller and Purdue) were conducted by cell culture immunofluorescent (CCIF) and viral neutralization (VN) assays. None of the pig TGEV antisera neutralized PEDV and vice versa. One-way cross-reactions were observed by CCIF between TGEV Miller hyperimmune pig antisera and all PEDV strains. Enzyme-linked immunosorbent assays, immunoblotting using monoclonal antibodies and Escherichia coli-expressed recombinant PEDV and TGEV nucleocapsid (N) proteins, and sequence analysis suggested at least one epitope on the N-terminal region of PEDV/TGEV N protein that contributed to this cross-reactivity. Biologically, PEDV strain CV777 induced greater cell fusion in Vero cells than did U.S. PEDV strains. Consistent with the reported genetic differences, the results of CCIF and VN assays also revealed higher antigenic variation between PEDV CV777 and U.S. strains. IMPORTANCE Evidence of antigenic cross-reactivity between porcine enteric coronaviruses, PEDV and TGEV, in CCIF assays supports the idea that these two species are evolutionarily related, but they are distinct species defined by VN assays. Identification of PEDV- or TGEV-specific antigenic regions allows the development of more specific immunoassays for each virus. Antigenic and biologic variations between the prototype and current PEDV strains could explain, at least partially, the recurrence of PEDV epidemics. Information on the conserved antigenicity among PEDV strains is important for the development of PEDV vaccines to protect swine from current highly virulent PEDV infections.
Collapse
|
37
|
Whole-genome analysis of porcine epidemic diarrhea virus (PEDV) from eastern China. Arch Virol 2014; 159:2777-85. [PMID: 24818713 PMCID: PMC7086842 DOI: 10.1007/s00705-014-2102-7] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2014] [Accepted: 04/23/2014] [Indexed: 11/23/2022]
Abstract
The complete genome sequence of a porcine epidemic diarrhea virus variant, strain SHQP/YM/2013, from China was determined and compared with those of other porcine epidemic diarrhea viruses. The full-length genome was 28,038 nucleotides (nt) in length without the poly (A) tail, and it was similar to that of other reported PEDV strains, with the characteristic gene order 5′-replicase (1a/1b) -S-ORF3-E-M-N-3′. Nucleotide sequence analysis based on individual virus genes indicated a close relationship between the S gene of SHQP/YM/2013 and those of the four Korean field strains from 2008–2009. Its ORF3 gene, however, fell into three groups. Recent prevalent Chinese PEDV field isolates were divided between group 1 and group 3, which suggests that the recent prevalent Chinese PEDV field isolates represent a new genotype that differs from the genotype that includes the vaccine strains. Based on phylogenetic analysis of the M gene, ORF3 gene and S gene, our study demonstrated that prevalent PEDV isolates in China may have originated from Korean strains. This report describes the complete genome sequence of SHQP/YM/2013, and the data will promote a better understanding of the molecular epidemiology and genetic diversity of PEDV field isolates in eastern China.
Collapse
|
38
|
Sequence and phylogenetic analysis of nucleocapsid genes of porcine epidemic diarrhea virus (PEDV) strains in China. Arch Virol 2013; 158:1267-73. [PMID: 23389550 PMCID: PMC3668129 DOI: 10.1007/s00705-012-1592-4] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2012] [Accepted: 11/29/2012] [Indexed: 11/23/2022]
Abstract
Porcine epidemic diarrhea virus (PEDV) causes acute diarrhea and dehydration with high mortality rates in swine. It has become increasingly problematic in China. Since the nucleocapsid (N) protein is highly conserved, it is a candidate protein for early diagnosis and vaccine development. In this study, the N genes of 15 PEDV strains were amplified by RT-PCR and cloned into the pMT-19T vector, sequenced, and compared to each other as well as to PEDV reference strains. The nucleotide sequences of the N gene of the Chinese PEDV strains consist of 1326 nucleotides and encode a 441-aa-long peptide. The nucleotide sequences of the fifteen PEDV strains in our study were 96.1-100 % identical to each other, and the deduced amino acid sequences were 94.8-100 % identical. Sequence comparison with other PEDV strains selected from GenBank revealed that their nucleotide sequences were 94.2-99.7 % identical to those of the Chinese PEDV strains, and their deduced amino acid sequences were 94.1-99.5 % identical. In addition, the fifteen strains showed a high degree of nucleotide sequence identity to the early domestic strains (98.4-99.7 %) except the LZC strain, but less sequence identity to the vaccine strain (CV777) used in China (94.7-97.7 %). Phylogenetic analysis showed that the Chinese PEDV strains are composed of a separate cluster including three early domestic strains (JS-2004-02, LJB/03 and DX) but differ genetically from the vaccine strain (CV777) and the early Korean strains (Chinju99 and SM98).
Collapse
|
39
|
Xu X, Zhang H, Zhang Q, Huang Y, Dong J, Liang Y, Liu HJ, Tong D. Porcine epidemic diarrhea virus N protein prolongs S-phase cell cycle, induces endoplasmic reticulum stress, and up-regulates interleukin-8 expression. Vet Microbiol 2013; 164:212-21. [PMID: 23562137 PMCID: PMC7117426 DOI: 10.1016/j.vetmic.2013.01.034] [Citation(s) in RCA: 99] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2012] [Revised: 01/14/2013] [Accepted: 01/21/2013] [Indexed: 01/07/2023]
Abstract
Porcine epidemic diarrhea (PED) is an acute and highly contagious enteric disease of swine caused by porcine epidemic diarrhea virus (PEDV). The porcine intestinal epithelial cell is the PEDV target cell. In this study, we established a porcine intestinal epithelial cell (IEC) line which can stably express PEDV N protein. We also investigate the subcellular localization and function of PEDV N protein by examining its effects on cell growth, cycle progression, interleukin-8 (IL-8) expression, and survival. The results show that the PEDV N protein localizes in the endoplasmic reticulum (ER), inhibits the IEC growth and prolongs S-phase cell cycle. The S-phase is prolonged which is associated with a decrease of cyclin A transcription level and an increase of cyclin A degradation. The IEC expressing PEDV N protein can express higher levels of IL-8 than control cells. Further studies show that PEDV N protein induces ER stress and activates NF-κB, which is responsible for the up-regulation of IL-8 and Bcl-2 expression. This is the first report to demonstrate that PEDV N protein can induce cell cycle prolongation at the S-phase, ER stress and up-regulation interleukin-8 expression. These findings provide novel information on the function of the PEDV N protein and are likely to be very useful in understanding the molecular mechanisms responsible for PEDV pathogenesis.
Collapse
Affiliation(s)
- Xingang Xu
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi 712100, China
| | | | | | | | | | | | | | | |
Collapse
|
40
|
Gao Y, Kou Q, Ge X, Zhou L, Guo X, Yang H. Phylogenetic analysis of porcine epidemic diarrhea virus field strains prevailing recently in China. Arch Virol 2012; 158:711-5. [PMID: 23151819 DOI: 10.1007/s00705-012-1541-2] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2012] [Accepted: 10/05/2012] [Indexed: 11/26/2022]
Abstract
Porcine epidemic diarrhea virus (PEDV) is the causative agent of porcine epidemic diarrhea (PED), which is characterized by severe diarrhea, dehydration and high mortality in the affected pigs. Recently, clinical outbreaks of diarrhea in suckling piglets emerged in pig-producing areas of China. In this study, molecular detection of PEDV was conducted using RT-PCR (targeting the M gene) on samples collected from piglets with watery diarrhea from 15 pig farms, and phylogenetic analysis of PEDV field strains was carried out based on their M and S genes. In addition, the complete genome sequence of a PEDV field strain was determined. PEDV was detected in 92.7 % of the samples (267/288). The 15 M genes that were amplified shared 99.6-100 % nucleotide identity and 99.1-100 % amino acid similarity with each other. The 15 S genes exhibited 98.6-99.9 % homology, both at the nucleotide level and at the deduced amino acid level. Phylogenetic analysis showed that all of the amplified M genes grouped in cluster 3, together with some Chinese, Korean and Thai strains, while all of the amplified S genes were in cluster 3 and were closely related to Korean strains. Compared with previous PEDV strains, all of the S genes have common characteristics, namely, a 4-aa (GENQ) insertion between positions 55 and 56, a 1-aa (N) insertion between positions 135 and 136, and a 2-aa (DG) deletion between positions 155 and 156, similar or identical to Korean KNU-serial strains reported in recent years. The genome of the sequenced PEDV field strain is 28,038 nucleotides in length, excluding the poly (A) tail. Our findings suggest that a novel PEDV with a characteristic variant S gene is responsible for recent outbreaks of clinical diarrhea in piglets in China.
Collapse
Affiliation(s)
- Yueyi Gao
- Key Laboratory of Animal Epidemiology and Zoonosis of Ministry of Agriculture, College of Veterinary Medicine and State Key Laboratory of Agrobiotechnology, China Agricultural University, Beijing, People's Republic of China
| | | | | | | | | | | |
Collapse
|
41
|
Pan Y, Tian X, Li W, Zhou Q, Wang D, Bi Y, Chen F, Song Y. Isolation and characterization of a variant porcine epidemic diarrhea virus in China. Virol J 2012; 9:195. [PMID: 22967434 PMCID: PMC3487931 DOI: 10.1186/1743-422x-9-195] [Citation(s) in RCA: 76] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2011] [Accepted: 09/05/2012] [Indexed: 11/29/2022] Open
Abstract
An outbreak of diarrhea in pigs started in Guangdong, South China in January 2011. Cases were characterized by watery diarrhea, dehydration and vomiting, with 80–100% morbidity and 50–90% mortality in suckling piglets. The causative agent of the diarrhea was ultimately identified as porcine epidemic diarrhea virus (PEDV). In this study, we isolated a PEDV strain designated CHGD-01 from piglet intestines using Vero cell cultures, and its specific cytopathic effects were confirmed in susceptible cells by direct immunofluorescence testing and electron microscopy. The complete genome of CHGD-01 was shown to be 28,035 nucleotides in length, with a similar structure to that of PEDV reference strains. Phylogenetic analyses based on the whole genome revealed that CHGD-01 shared nucleotide sequence identities of 98.2–98.4% with two other Chinese isolates reported in the same year, thus constituting a new cluster. Amino acid sequence analysis based on individual virus genes indicated a close relationship between the spike protein gene of CHGD-01 and the field strain KNU0802 in Korea. Its ORF3 and nucleoprotein genes, however, were divergent from all other sequenced PEDV isolate clusters and therefore formed a new group, suggesting a new variant PEDV isolate in China. Further studies will be required to determine the immunogenicity and pathogenicity of this new variant.
Collapse
Affiliation(s)
- Yongfei Pan
- Guangdong Wen's Group Academy, Guangdong Wen's Foodstuffs Group Co, Ltd, Xinxing, Guangdong, 527400, China
| | | | | | | | | | | | | | | |
Collapse
|
42
|
Izzo MM, Kirkland PD, Gu X, Lele Y, Gunn AA, House JK. Comparison of three diagnostic techniques for detection of rotavirus and coronavirus in calf faeces in Australia. Aust Vet J 2012; 90:122-9. [PMID: 22443326 PMCID: PMC7159673 DOI: 10.1111/j.1751-0813.2011.00891.x] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Objective Compare real‐time reverse transcription polymerase chain reaction (qRT‐PCR), a commercially available enzyme‐linked immunosorbent assay (ELISA) and lateral flow immunochromatography assay (LAT) for the detection of rotavirus and coronavirus in faecal samples collected from diarrhoeic calves. Design Prospective survey. Method Samples were tested at two separate facilities using a commercial ELISA and an in‐house qRT‐PCR. Simple logistic regression was performed to examine the relationship between the two tests. A subset of samples was screened using qRT‐PCR, ELISA and a commercial LAT dipstick (132 faecal samples were tested for coronavirus and 122 samples for rotavirus). Results Of the 586 samples tested, 131 (22.39%) and 468 (79.86%) were positive for coronavirus and group A rotavirus, respectively, using qRT‐PCR. The number of samples positive on ELISA for coronavirus and rotavirus was 73 (12.46%) and 225 (38.40%), respectively. Using LAT, 30 (22.73%) and 43 (35.35%) samples were positive for coronavirus and rotavirus, respectively. Simple linear regression revealed a statistically significant (P < 0.05) but weak (r2=−0.07 and −0.40) correlation between the rotavirus/coronavirus qRT‐PCR and ELISA, respectively. There was also poor agreement between the LAT and qRT‐PCR assays. Conclusion The sensitivity and specificity of the commercial ELISA and LAT assays evaluated in this study were low compared with qRT‐PCR. The low positive and negative predictive values of the assays suggests that they were of limited diagnostic benefit in the population sampled.
Collapse
Affiliation(s)
- M M Izzo
- Livestock Veterinary Teaching and Research Unit, University of Sydney, 410 Werombi Rd Camden, New South Wales 2570, Australia.
| | | | | | | | | | | |
Collapse
|
43
|
Song D, Park B. Porcine epidemic diarrhoea virus: a comprehensive review of molecular epidemiology, diagnosis, and vaccines. Virus Genes 2012; 44:167-75. [PMID: 22270324 PMCID: PMC7089188 DOI: 10.1007/s11262-012-0713-1] [Citation(s) in RCA: 497] [Impact Index Per Article: 41.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2011] [Accepted: 01/04/2012] [Indexed: 01/20/2023]
Abstract
The porcine epidemic diarrhoea virus (PEDV), a member of the Coronaviridae family, causes acute diarrhoea and dehydration in pigs. Although it was first identified in Europe, it has become increasingly problematic in many Asian countries, including Korea, China, Japan, the Philippines, and Thailand. The economic impacts of the PEDV are substantial, given that it results in significant morbidity and mortality in neonatal piglets and is associated with increased costs related to vaccination and disinfection. Recently, progress has been made in understanding the molecular epidemiology of PEDV, thereby leading to the development of new vaccines. In the current review, we first describe the molecular and genetic characteristics of the PEDV. Then we discuss its molecular epidemiology and diagnosis, what vaccines are available, and how PEDV can be treated.
Collapse
Affiliation(s)
- Daesub Song
- Viral Infectious Disease Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, 305-806 South Korea
- University of Science and Technology, Daejeon, 305-350 South Korea
| | - Bongkyun Park
- Department of Veterinary Virology, College of Veterinary Medicine and BK21 Program for Veterinary Science, Seoul National University, Seoul, 151-742 Korea
| |
Collapse
|
44
|
Park SJ, Kim HK, Song DS, Moon HJ, Park BK. Molecular characterization and phylogenetic analysis of porcine epidemic diarrhea virus (PEDV) field isolates in Korea. Arch Virol 2011; 156:577-85. [PMID: 21210162 PMCID: PMC7086862 DOI: 10.1007/s00705-010-0892-9] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2010] [Accepted: 12/10/2010] [Indexed: 11/25/2022]
Abstract
Porcine epidemic diarrhea virus (PEDV) has caused enteric disease with devastating impact since the first identification of PEDV in 1992 in Korea. In this study, we investigated molecular epidemiology, showed genetic diversity, and analyzed phylogenetic relationships of Korean PEDV field isolates with other PEDV reference strains. Genetic analysis of the complete M and ORF3 genes showed that each PEDV group had several unique characteristics, and this indicated that specific groups of PEDVs may be differentiated from the other PEDVs by specific nucleotide differences. Especially, ORF3 gene analysis can be used for discrimination between vaccine and wild-type PEDVs. Sequence and phylogenetic analysis showed that recent, prevalent Korean PEDV field isolates have close relationships to Chinese field strains and differ genetically from European strains and vaccine strains used in Korea. These results raise questions as to whether a new type of PEDV vaccine may be necessary for preventing PEDV infection more effectively in Korea.
Collapse
Affiliation(s)
- Seong-Jun Park
- Department of Veterinary Medicine Virology Lab, College of Veterinary Medicine and BK21 Program for Veterinary Science, Seoul National University, Gwanak-gu, Seoul, 151-742 Korea
| | - Hye-Kwon Kim
- Department of Veterinary Medicine Virology Lab, College of Veterinary Medicine and BK21 Program for Veterinary Science, Seoul National University, Gwanak-gu, Seoul, 151-742 Korea
| | - Dae-Sub Song
- Viral Infectious Disease Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, 305-806 Korea
| | - Hyoung-Joon Moon
- Research Unit, Green Cross Veterinary Products, Yongin, 449-903 Korea
| | - Bong-Kyun Park
- Department of Veterinary Medicine Virology Lab, College of Veterinary Medicine and BK21 Program for Veterinary Science, Seoul National University, Gwanak-gu, Seoul, 151-742 Korea
| |
Collapse
|
45
|
Yang Z, Wu N, Fu Y, Yang G, Wang W, Zu Y, Efferth T. Anti-infectious bronchitis virus (IBV) activity of 1,8-cineole: effect on nucleocapsid (N) protein. J Biomol Struct Dyn 2011; 28:323-30. [PMID: 20919748 DOI: 10.1080/07391102.2010.10507362] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
In the present study, anti-IBV (infectious bronchitis virus) activity of 1,8-cineole was studied by MTT assay, as well as docking and molecular dynamic (MD) simulations. The CC50 of 1,8-cineole was above 10 mM. And the maximum noncytotoxic concentration (TD0) of 1,8-cineole was determined to be 3.90 ± 0.22 mM, which was much higher than that of ribavirin (0.78 ± 0.15 mM). 1,8-cineole could inhibit IBV with an IC(50) of 0.61 mM. MTT assay showed that the inhibition of IBV by 1, 8-cineole appears to occur moderately before entering the cell but much strongly after penetration of the virus into the cell. In silico simulations indicated that the binding site of 1,8-cineole was located at the N terminus of phosphorylated nucleocapsid (N) protein, with interaction energy equaling -40.33 kcal mol(-1). The residues TyrA92, ProA134, PheA137, AspA138 and TyrA140 had important roles during the binding process and are fully or partially conserved in various IBV strains. Based on spatial and energetic criteria, 1,8-cineole interfered with the binding between RNA and IBV N-protein. Results presented here may suggest that 1,8-cineole possesses anti-IBV properties, and therefore is a potential source of anti-IBV ingredients for the pharmaceutical industry.
Collapse
Affiliation(s)
- Zhiwei Yang
- Key Laboratory of Forest Plant Ecology, Ministry of Education, Northeast Forestry University, Harbin 150040, PR China
| | | | | | | | | | | | | |
Collapse
|
46
|
Comparative anti-infectious bronchitis virus (IBV) activity of (-)-pinene: effect on nucleocapsid (N) protein. Molecules 2011; 16:1044-54. [PMID: 21350392 PMCID: PMC6259611 DOI: 10.3390/molecules16021044] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2010] [Revised: 12/23/2010] [Accepted: 01/17/2011] [Indexed: 01/25/2023] Open
Abstract
In the present study, anti-IBV (infectious bronchitis virus) activities of (-)-pinenes were studied by MTT assay, as well as docking and molecular dynamic (MD) simulations. The CC₅₀ values of (-)-α-pinene and (-)-β-pinene were above 10 mM. And the maximum noncytotoxic concentrations (TD₀) of (-)-α-pinene and (-)-β-pinene were determined as 7.88 ± 0.06 and 6.09 ± 0.31 mM, respectively. The two compounds were found to inhibit IBV with an IC₅₀ of 0.98 ± 0.25 and 1.32 ± 0.11 mM. The MTT assay showed that the inhibitions of (-)-pinenes against IBV appear to occur moderately before entering the cell but are much stronger occur after penetration of the virus into the cell. Molecular simulations indicated that (-)-α-pinene and (-)-β-pinene specifically interact with the active site which is located at the N terminus of phosphorylated nucleocapsid (N) protein, with the former being more potent than the latter. The binding energies of them are -36.83 and -35.59 kcal mol-1, respectively. Results presented here may suggest that (-)-α-pinene and (-)-β-pinene possess anti-IBV properties, and therefore are a potential source of anti-IBV ingredients for the pharmaceutical industry.
Collapse
|
47
|
Farsang A, Ros C, Renström LHM, Baule C, Soós T, Belák S. Molecular epizootiology of infectious bronchitis virus in Sweden indicating the involvement of a vaccine strain. Avian Pathol 2010; 31:229-36. [PMID: 12396345 PMCID: PMC7154300 DOI: 10.1080/03079450220136530] [Citation(s) in RCA: 78] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
To improve the detection and molecular identification of infectious bronchitis virus (avian coronavirus), two reverse transcriptase-polymerase chain reaction (PCR) assays were developed. As 'diagnostic#10; PCR', a set of consensus nested primers was selected from highly conserved stretches of the nucleocapsid (N) gene. As 'phylogeny' PCR, a fragment of the spike protein gene (S1) was amplified and the PCR products were directly sequenced. To study the phylogenetic relationships of the viruses from various outbreaks, studies of molecular epizootiology were performed in Sweden, a Nordic region, where the occurrence of natural cases of the disease is relatively low and the occasional use of live vaccine(s) is well recorded and monitored. The disease appeared in the region in 1994, associated with production problems among layers of various ages. During outbreaks in 1995 and 1997, both layers and broilers were affected. To reduce losses, a live attenuated vaccine has been applied since 1997. By examining 12 cases between 1994 and 1998, molecular epizootiology revealed that, before 1997, the viruses had gene sequences very similar to strains of the Massachusetts serotype. However, comparative sequence analysis of the S1 gene revealed that the identity was not 100% to any of the strains of this serotype that we analysed. A virus related to the Dutch-type strain, D274, was also identified on one farm. Surprisingly, from 1997, the year that vaccination commenced with a live Massachusetts serotype vaccine, the majority of viruses detected had S1 sequences identical to the live Massachusetts vaccine strain. This genetic relation to the vaccine virus was also confirmed by N gene sequence analysis. The studies of molecular epizootiology reveal a strong probability that the vaccination had lead to the spread of the vaccine virus, causing various disease manifestations and a confusing epizootiological situation in the poultry population.
Collapse
Affiliation(s)
- A. Farsang
- Institute for Veterinary Medicinal Products, H-1107, Budapest, Szállás utca 8, Hungary
| | - C. Ros
- Department of Chemistry and Biochemistry, University of Bern, Freistsrasse 3, 3012 Bern and ZLB Bioplasma AG, Bern, Switzerland
| | - Lena H. M. Renström
- Department of Virology, The National Veterinary Institute, Biomedical Center, S-751 23 Uppsala, Sweden
| | - Claudia Baule
- Department of Virology, The National Veterinary Institute, Biomedical Center, S-751 23 Uppsala, Sweden
| | - T. Soós
- Institute for Veterinary Medicinal Products, H-1107, Budapest, Szállás utca 8, Hungary
| | - S. Belák
- Department of Virology, The National Veterinary Institute, Biomedical Center, S-751 23 Uppsala, Sweden
- To whom correspondence should be sent. E-mail:
| |
Collapse
|
48
|
Gibertoni A, Gonçalves M, Montassier M, Fernandes C, Montassier H. CLONAGEM, EXPRESSÃO E CARACTERIZAÇÃO DA NUCLEOPROTEÍNA RECOMBINANTE DO VÍRUS DA BRONQUITE INFECCIOSA EM ESCHERICHIA COLI E EM PICHIA PASTORIS. ARQUIVOS DO INSTITUTO BIOLÓGICO 2010. [DOI: 10.1590/1808-1657v77p0012010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
RESUMO O gene da proteína de nucleocapsídeo (1.230 pb) da estirpe M41 do vírus da bronquite infecciosa (VBI) foi amplificado pelas reações de transcrição reversa e em cadeia da polimerase (RT-PCR) e clonado, em seguida, em dois sistemas; pET28a - Escherichia coli e pFLD -Pichia pastoris. Os produtos recombinantes construídos para expressão (pET28a-N ou pFLD-N) foram identificados por análises de PCR e de sequenciamento de nucleotídeos. Os clones transformantes da linhagem BL21 de E. coli e da linhagem GS115 de P. pastoris foram submetidos aos protocolos apropriados de indução. A expressão da proteína N de fusão com etiqueta de poli-histidina e com massa molecular de 54 kDa foi determinada pelas técnicas de SDS-PAGE e de Western blotting, confirmando-se que ambas proteínas N recombinantes apresentaram tamanhos e antigenicidade compatíveis com a proteína N nativa do próprio VBI. O sistema E. coli expressou uma quantidade relevante da proteína N recombinante, enquanto que o sistema P. pastoris produziu uma baixa recuperação dessa proteína recombinante. A proteína N recombinante gerada pelo sistema bacteriano foi purificada em resina de níquel-sepharose. O conjunto de resultados indica que o sistema de expressão constituído por pET28a – E. coli é mais efetivo para produzir a proteína N recombinante do VBI destinada ao uso como antígeno para detectar anticorpos anti-virais específicos em ensaios de imunodiagnóstico para essa infecção viral.
Collapse
|
49
|
Pak JE, Sharon C, Satkunarajah M, Auperin TC, Cameron CM, Kelvin DJ, Seetharaman J, Cochrane A, Plummer FA, Berry JD, Rini JM. Structural insights into immune recognition of the severe acute respiratory syndrome coronavirus S protein receptor binding domain. J Mol Biol 2009; 388:815-23. [PMID: 19324051 PMCID: PMC7094495 DOI: 10.1016/j.jmb.2009.03.042] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2008] [Revised: 03/16/2009] [Accepted: 03/16/2009] [Indexed: 11/02/2022]
Abstract
The spike (S) protein of the severe acute respiratory syndrome coronavirus (SARS-CoV) is responsible for host cell attachment and fusion of the viral and host cell membranes. Within S the receptor binding domain (RBD) mediates the interaction with angiotensin-converting enzyme 2 (ACE2), the SARS-CoV host cell receptor. Both S and the RBD are highly immunogenic and both have been found to elicit neutralizing antibodies. Reported here is the X-ray crystal structure of the RBD in complex with the Fab of a neutralizing mouse monoclonal antibody, F26G19, elicited by immunization with chemically inactivated SARS-CoV. The RBD-F26G19 Fab complex represents the first example of the structural characterization of an antibody elicited by an immune response to SARS-CoV or any fragment of it. The structure reveals that the RBD surface recognized by F26G19 overlaps significantly with the surface recognized by ACE2 and, as such, suggests that F26G19 likely neutralizes SARS-CoV by blocking the virus-host cell interaction.
Collapse
Affiliation(s)
- John E Pak
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Pimentel TAPF, Yan Z, Jeffers SA, Holmes KV, Hodges RS, Burkhard P. Peptide nanoparticles as novel immunogens: design and analysis of a prototypic severe acute respiratory syndrome vaccine. Chem Biol Drug Des 2009; 73:53-61. [PMID: 19152635 PMCID: PMC2756483 DOI: 10.1111/j.1747-0285.2008.00746.x] [Citation(s) in RCA: 129] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Severe acute respiratory syndrome (SARS) is an infectious disease caused by a novel coronavirus that cost nearly 800 lives. While there have been no recent outbreaks of the disease, the threat remains as SARS coronavirus (SARS‐CoV) like strains still exist in animal reservoirs. Therefore, the development of a vaccine against SARS is in grave need. Here, we have designed and produced a prototypic SARS vaccine: a self‐assembling polypeptide nanoparticle that repetitively displays a SARS B‐cell epitope from the C‐terminal heptad repeat of the virus’ spike protein. Biophysical analyses with circular dichroism, transmission electron microscopy and dynamic light scattering confirmed the computational design showing α‐helcial nanoparticles with sizes of about 25 nm. Immunization experiments with no adjuvants were performed with BALB/c mice. An investigation of the binding properties of the elicited antibodies showed that they were highly conformation specific for the coiled‐coil epitope because they specifically recognized the native trimeric conformation of C‐terminal heptad repeat region. Consequently, the antisera exhibited neutralization activity in an in vitro infection inhibition assay. We conclude that these peptide nanoparticles represent a promising platform for vaccine design, in particular for diseases that are characterized by neutralizing epitopes with coiled‐coil conformation such as SARS‐CoV or other enveloped viruses.
Collapse
Affiliation(s)
- Tais A P F Pimentel
- Institute of Materials Science, University of Connecticut, Storrs, CT 06269-3136, USA
| | | | | | | | | | | |
Collapse
|