1
|
Lu C, Chen W, Chen H, Xing G, Ma J, Zhou H, Qin L, Da L, Sun S, Peng P, Li H, Jin Y, Yan Y, Pan S, Dong W, Gu J, Zhou J. Immunological characteristics of the recombinant pseudorabies virus with chimeric PCV Cap protein in pigs. Vet Microbiol 2025; 305:110529. [PMID: 40288026 DOI: 10.1016/j.vetmic.2025.110529] [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: 02/24/2025] [Revised: 04/18/2025] [Accepted: 04/20/2025] [Indexed: 04/29/2025]
Abstract
Porcine circovirus type 2 (PCV2) is one of the main pathogens causing porcine circovirus-associated diseases (PCVAD). We recently reported the immunogenicity of the recombinant PRV with an envelope-embedded Cap protein of PCV2 (PRV-Cap) in mice. Here, we further evaluated the immunoprotective efficacy of PRV-Cap virus in pigs. Following vaccination, the PRV-Cap stimulated the production of neutralizing antibodies against PRV and PCV2, along with protected piglets from the challenge of the lethal PRV, the virulent PCV2b and PCV2d. Peripheral blood mononuclear cells analysis revealed that PRV-Cap virus effectively induced proliferation and activation of CD4 and CD8 T cells, as well as an increase in T follicular helper cells, although γδ T and B cells did not show significant differences. Compared to DMEM control piglets, the expanded CD4 and CD8 T cells exhibited an effector memory T cell phenotype, and in vitro stimulation led to PRV- and PCV2-specific IFN-γ and TNF-α secretion, peaking at 21 days post-immunization. In summary, PRV-Cap virus effectively prevents PRV, PCV2b and PCV2d challenges in piglets by simultaneously inducing both PRV- and PCV2-specific humoral and cellular immunity, indicating that PRV-Cap virus is a promising and safe candidate vaccine for combined PRV and PCV2 immunization.
Collapse
Affiliation(s)
- Chenhe Lu
- MOA Key Laboratory of Animal Virology, Zhejiang University Center for Veterinary Sciences, Hangzhou, China
| | - Wenjing Chen
- MOA Key Laboratory of Animal Virology, Zhejiang University Center for Veterinary Sciences, Hangzhou, China
| | - Heng Chen
- MOA Key Laboratory of Animal Virology, Zhejiang University Center for Veterinary Sciences, Hangzhou, China
| | - Gang Xing
- MOA Key Laboratory of Animal Virology, Zhejiang University Center for Veterinary Sciences, Hangzhou, China
| | - Jiayu Ma
- MOA Key Laboratory of Animal Virology, Zhejiang University Center for Veterinary Sciences, Hangzhou, China
| | - Hui Zhou
- MOA Key Laboratory of Animal Virology, Zhejiang University Center for Veterinary Sciences, Hangzhou, China
| | - Linglong Qin
- MOA Key Laboratory of Animal Virology, Zhejiang University Center for Veterinary Sciences, Hangzhou, China
| | - Liu Da
- MOA Key Laboratory of Animal Virology, Zhejiang University Center for Veterinary Sciences, Hangzhou, China
| | - Shiping Sun
- MOA Key Laboratory of Animal Virology, Zhejiang University Center for Veterinary Sciences, Hangzhou, China
| | - Peng Peng
- MOA Key Laboratory of Animal Virology, Zhejiang University Center for Veterinary Sciences, Hangzhou, China
| | - Haimin Li
- MOA Key Laboratory of Animal Virology, Zhejiang University Center for Veterinary Sciences, Hangzhou, China
| | - Yulan Jin
- MOA Key Laboratory of Animal Virology, Zhejiang University Center for Veterinary Sciences, Hangzhou, China
| | - Yan Yan
- MOA Key Laboratory of Animal Virology, Zhejiang University Center for Veterinary Sciences, Hangzhou, China
| | - Shiyue Pan
- MOA Key Laboratory of Animal Virology, Zhejiang University Center for Veterinary Sciences, Hangzhou, China
| | - Weiren Dong
- MOA Key Laboratory of Animal Virology, Zhejiang University Center for Veterinary Sciences, Hangzhou, China
| | - Jinyan Gu
- MOA Key Laboratory of Animal Virology, Zhejiang University Center for Veterinary Sciences, Hangzhou, China
| | - Jiyong Zhou
- MOA Key Laboratory of Animal Virology, Zhejiang University Center for Veterinary Sciences, Hangzhou, China; State Key Laboratory for Diagnosis and Treatment of Severe Infectious Diseases, First Affiliated Hospital, Zhejiang University, Hangzhou, China.
| |
Collapse
|
2
|
Li H, Zhang J, Guo R, Li J, Zhang X, Han L, Xie H, Wang X. Immunogenicity evaluation of a recombinant pseudorabies virus co-expressing PCV2 and PCV3 capsid proteins in mice and piglets. Vaccine 2025; 60:127307. [PMID: 40424702 DOI: 10.1016/j.vaccine.2025.127307] [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: 02/01/2025] [Revised: 04/15/2025] [Accepted: 05/21/2025] [Indexed: 05/29/2025]
Abstract
Porcine circovirus type 2 (PCV2), porcine circovirus type 3 (PCV3), and pseudorabies virus (PRV) are major pathogens posing significant threats to the swine industry. Viral evolution and mutations have limited the efficacy of current commercial vaccines, necessitating the development of more effective prophylactic strategies. In this study, a novel recombinant virus strain, designated as rPRV-ΔTK-PCV3(Cap)/ΔgIgE-PCV2(Cap), was generated using PRV SX-10 variant as the backbone. CRISPR/Cas9-mediated deletion of TK and gE/gI genes was performed, followed by insertion of PCV3 and PCV2 capsid protein genes into the respective loci. The engineered recombinant strain demonstrated stable proliferation in BHK-21 cells, efficiently expressed heterologous PCV3 and PCV2 capsid proteins, while maintaining biological properties comparable to its parental strain. The rPRV-ΔTK-PCV3(Cap)/ΔgIgE-PCV2(Cap) demonstrated favorable safety and immunogenicity profiles in mice and piglets, eliciting robust immune responses characterized by high titers of specific antibodies against PRV, PCV3, and PCV2, along with significantly elevated levels of cytokines (IFN-γ, IL-2, and IL-4). Histopathological analysis and viral load quantification demonstrated that rPRV-ΔTK-PCV3(Cap)/ΔgIgE-PCV2(Cap) immunization significantly attenuated tissue lesions and decreased viral copies of PRV and PCV in mice and piglets. Collectively, these findings suggest that rPRV-ΔTK-PCV3(Cap)/ΔgIgE-PCV2(Cap) serves as a promising candidate vaccine against PRV and PCV infections.
Collapse
Affiliation(s)
- Hui Li
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shanxi, China
| | - Jingnan Zhang
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shanxi, China
| | - Ruhai Guo
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shanxi, China
| | - JunDa Li
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shanxi, China
| | - Xiao Zhang
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shanxi, China
| | - Likang Han
- College of Veterinary Medicine, Gansu Agricultural University, Anning, Gansu, China
| | - Honglin Xie
- School of Life Science and Engineering, Foshan University, Guangdong 528225, PR China.
| | - Xinglong Wang
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shanxi, China.
| |
Collapse
|
3
|
Zhuang L, Gong J, Shen J, Zhao Y, Yang J, Liu Q, Zhang Y, Shen Q. Advances in molecular epidemiology and detection methods of pseudorabies virus. DISCOVER NANO 2025; 20:45. [PMID: 39992589 PMCID: PMC11850701 DOI: 10.1186/s11671-025-04217-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Accepted: 02/10/2025] [Indexed: 02/26/2025]
Abstract
Pseudorabies (PR), a highly contagious disease caused by the pseudorabies virus (PRV), represents a significant threat to the global swine industry. Despite the success of developed countries in controlling the PRV epidemic through swine pseudorabies eradication programs, wild boars, as a potential source of infection, still require sustained attention and effective control measures. Concurrently, there has been considerable global attention directed towards cases of PRV infection in humans. In consideration of the aforementioned factors, this paper presents a comprehensive review of recent developments in the PRV genome, epidemiology, vaccine research, and molecular detection methods. The epidemiology section presents an analysis of the transmission routes, susceptible animal groups, and geographic distribution of PRV, as well as an examination of the trend of the epidemic in recent years. In the field of vaccine research, the current development of genetically engineered vaccines is emphasized, and the immunogenicity and safety of vaccines are discussed. Moreover, the molecular detection techniques utilized to identify PRV, including immunological methods, nucleic acid detection methods, biosensors, and so forth, are presented in a systematic manner. Finally, this paper presents a comprehensive discussion of the current status of PRV-related research and offers insights into future directions, with the aim of providing a foundation for the scientific prevention and control of PRV.
Collapse
Affiliation(s)
- Linlin Zhuang
- School of Animal Husbandry and Veterinary Medicine, Jiangsu Vocational College of Agriculture and Forestry, Jurong, 212400, People's Republic of China
- State Key Laboratory of Digital Medical Engineering, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Science and Medical Engineering & Basic Medicine Research and Innovation Center of Ministry of Education, Zhongda Hospital, Southeast University, Nanjing, 211102, People's Republic of China
| | - Jiansen Gong
- Poultry Institute, Chinese Academy of Agricultural Sciences, Yangzhou, 225125, People's Republic of China
| | - Jingyi Shen
- School of Animal Science and Food Engineering, Jinling Institute of Technology, Nanjing, 210038, People's Republic of China
| | - Ying Zhao
- State Key Laboratory of Digital Medical Engineering, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Science and Medical Engineering & Basic Medicine Research and Innovation Center of Ministry of Education, Zhongda Hospital, Southeast University, Nanjing, 211102, People's Republic of China
| | - Jianbo Yang
- School of Animal Husbandry and Veterinary Medicine, Jiangsu Vocational College of Agriculture and Forestry, Jurong, 212400, People's Republic of China
| | - Qingxin Liu
- School of Animal Husbandry and Veterinary Medicine, Jiangsu Vocational College of Agriculture and Forestry, Jurong, 212400, People's Republic of China
| | - Yu Zhang
- State Key Laboratory of Digital Medical Engineering, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Science and Medical Engineering & Basic Medicine Research and Innovation Center of Ministry of Education, Zhongda Hospital, Southeast University, Nanjing, 211102, People's Republic of China.
| | - Qiuping Shen
- School of Animal Husbandry and Veterinary Medicine, Jiangsu Vocational College of Agriculture and Forestry, Jurong, 212400, People's Republic of China.
| |
Collapse
|
4
|
Wang H, Feng W. Current Status of Porcine Reproductive and Respiratory Syndrome Vaccines. Vaccines (Basel) 2024; 12:1387. [PMID: 39772049 PMCID: PMC11679953 DOI: 10.3390/vaccines12121387] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Revised: 12/01/2024] [Accepted: 12/06/2024] [Indexed: 01/05/2025] Open
Abstract
Porcine reproductive and respiratory syndrome (PRRS), characterized by reproductive failures in breeding pigs and respiratory diseases in growing pigs, is a widespread and challenging disease. The agent, PRRSV, is a single-strand RNA virus that is undergoing continuous mutation and evolution, resulting in the global spread of multiple strains with different genetic characteristics and variable antigens. There are currently no effective measures to eradicate PRRS, and vaccination is crucial for controlling the disease. At present, various types of vaccine are available or being studied, including inactivated vaccines, modified live virus (MLV) vaccines, vector vaccines, subunit vaccines, DNA vaccines, RNA vaccines, etc. MLV vaccines have been widely used to control PRRSV infection for more than 30 years since they were first introduced in North America in 1994, and have shown a certain efficacy. However, there are safety and efficacy issues such as virulence reversion, recombination with field strains, and a lack of protection against heterologous strains, while other types of vaccine have their own advantages and disadvantages, making the eradication of PRRS a challenge. This article reviews the latest progress of these vaccines in the prevention and control of PRRS and provides scientific inspiration for developing new strategies for the next generation of PRRS vaccines.
Collapse
Affiliation(s)
- Honglei Wang
- Department of Clinical Laboratory, Affiliated Hospital of Hebei University, Baoding 071000, China
| | - Wenhai Feng
- State Key Laboratory of Agrobiotechnology, China Agricultural University, Beijing 100193, China
- Ministry of Agriculture Key Laboratory of Soil Microbiology, China Agricultural University, Beijing 100193, China
- Department of Microbiology and Immunology, College of Biological Sciences, China Agricultural University, Beijing 100193, China
| |
Collapse
|
5
|
Jiao XQ, Liu Y, Chen XM, Wang CY, Cui JT, Zheng LL, Ma SJ, Chen HY. Construction and Immunogenicity of a Recombinant Porcine Pseudorabies Virus (PRV) Expressing the Major Neutralizing Epitope Regions of S1 Protein of Variant PEDV. Viruses 2024; 16:1580. [PMID: 39459914 PMCID: PMC11512226 DOI: 10.3390/v16101580] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Revised: 08/30/2024] [Accepted: 10/07/2024] [Indexed: 10/28/2024] Open
Abstract
Porcine epidemic diarrhea virus (PEDV) infection causes severe diarrhea and high mortality in neonatal piglets. Pseudorabies causes acute and often fatal infections in young piglets, respiratory disorders in growing pigs, and reproductive failure in sows. In late 2011, pseudorabies virus (PRV) variants occurred in Bartha-K61-vaccine-immunized swine herds, resulting in economic losses to the global pig industry. Therefore, it is essential to develop a safe and effective vaccine against both PEDV and PRV infections. In this study, we constructed a recombinant virus rPRV-PEDV S1 expressing the major neutralizing epitope region (COE, SS2, and SS6) of the PEDV S1 protein by homologous recombination technology and CRISPR/Cas9 gene editing technology, and then evaluated its biological characteristics in vitro and immunogenicity in pigs. The recombinant virus rPRV-PEDV S1 had similar growth kinetics in vitro to the parental rPRV NY-gE-/gI-/TK- strain, and was proven genetically stable in swine testicle (ST) cells and safe for piglets. PEDV S1-specific antibodies were detected in piglets immunized with rPRV-PEDV S1 on the 7th day post-immunization (dpi), and the antibody level increased rapidly at 14-21 dpi. Moreover, the immunized piglets receiving the recombinant virus exhibited alleviated clinical signs and reduced viral load compared to the unvaccinated group following a virulent PEDV HN2021 strain challenge. Also, piglets immunized with rPRV-PEDV S1 developed a PRV-specific humoral immune response and elicited complete protection against a lethal PRV NY challenge. These data indicate that the recombinant rPRV-PEDV S1 is a promising vaccine candidate strain for the prevention and control of PEDV and PRV infections.
Collapse
MESH Headings
- Animals
- Porcine epidemic diarrhea virus/immunology
- Porcine epidemic diarrhea virus/genetics
- Swine
- Herpesvirus 1, Suid/immunology
- Herpesvirus 1, Suid/genetics
- Swine Diseases/prevention & control
- Swine Diseases/virology
- Swine Diseases/immunology
- Antibodies, Viral/blood
- Antibodies, Viral/immunology
- Viral Vaccines/immunology
- Viral Vaccines/genetics
- Epitopes/immunology
- Epitopes/genetics
- Coronavirus Infections/prevention & control
- Coronavirus Infections/veterinary
- Coronavirus Infections/immunology
- Coronavirus Infections/virology
- Antibodies, Neutralizing/blood
- Antibodies, Neutralizing/immunology
- Spike Glycoprotein, Coronavirus/immunology
- Spike Glycoprotein, Coronavirus/genetics
- Pseudorabies/prevention & control
- Pseudorabies/immunology
- Vaccines, Synthetic/immunology
- Vaccines, Synthetic/genetics
- Vaccines, Synthetic/administration & dosage
Collapse
Affiliation(s)
- Xian-Qin Jiao
- Ministry of Education Key Laboratory for Animal Pathogens and Biosafety, College of Veterinary Medicine, Henan Agricultural University, Zhengdong New District Longzi Lake 15#, Zhengzhou 450046, China; (X.-Q.J.); (Y.L.); (X.-M.C.); (C.-Y.W.); (J.-T.C.); (L.-L.Z.)
| | - Ying Liu
- Ministry of Education Key Laboratory for Animal Pathogens and Biosafety, College of Veterinary Medicine, Henan Agricultural University, Zhengdong New District Longzi Lake 15#, Zhengzhou 450046, China; (X.-Q.J.); (Y.L.); (X.-M.C.); (C.-Y.W.); (J.-T.C.); (L.-L.Z.)
| | - Xi-Meng Chen
- Ministry of Education Key Laboratory for Animal Pathogens and Biosafety, College of Veterinary Medicine, Henan Agricultural University, Zhengdong New District Longzi Lake 15#, Zhengzhou 450046, China; (X.-Q.J.); (Y.L.); (X.-M.C.); (C.-Y.W.); (J.-T.C.); (L.-L.Z.)
| | - Cheng-Yuan Wang
- Ministry of Education Key Laboratory for Animal Pathogens and Biosafety, College of Veterinary Medicine, Henan Agricultural University, Zhengdong New District Longzi Lake 15#, Zhengzhou 450046, China; (X.-Q.J.); (Y.L.); (X.-M.C.); (C.-Y.W.); (J.-T.C.); (L.-L.Z.)
| | - Jian-Tao Cui
- Ministry of Education Key Laboratory for Animal Pathogens and Biosafety, College of Veterinary Medicine, Henan Agricultural University, Zhengdong New District Longzi Lake 15#, Zhengzhou 450046, China; (X.-Q.J.); (Y.L.); (X.-M.C.); (C.-Y.W.); (J.-T.C.); (L.-L.Z.)
- Animal Health Supervision Institute, Honghu 433200, China
| | - Lan-Lan Zheng
- Ministry of Education Key Laboratory for Animal Pathogens and Biosafety, College of Veterinary Medicine, Henan Agricultural University, Zhengdong New District Longzi Lake 15#, Zhengzhou 450046, China; (X.-Q.J.); (Y.L.); (X.-M.C.); (C.-Y.W.); (J.-T.C.); (L.-L.Z.)
| | - Shi-Jie Ma
- Ministry of Education Key Laboratory for Animal Pathogens and Biosafety, College of Veterinary Medicine, Henan Agricultural University, Zhengdong New District Longzi Lake 15#, Zhengzhou 450046, China; (X.-Q.J.); (Y.L.); (X.-M.C.); (C.-Y.W.); (J.-T.C.); (L.-L.Z.)
| | - Hong-Ying Chen
- Ministry of Education Key Laboratory for Animal Pathogens and Biosafety, College of Veterinary Medicine, Henan Agricultural University, Zhengdong New District Longzi Lake 15#, Zhengzhou 450046, China; (X.-Q.J.); (Y.L.); (X.-M.C.); (C.-Y.W.); (J.-T.C.); (L.-L.Z.)
| |
Collapse
|
6
|
Lu C, Li H, Chen W, Li H, Ma J, Peng P, Yan Y, Dong W, Jin Y, Pan S, Shang S, Gu J, Zhou J. Immunological characteristics of a recombinant alphaherpesvirus with an envelope-embedded Cap protein of circovirus. Front Immunol 2024; 15:1438371. [PMID: 39081314 PMCID: PMC11286414 DOI: 10.3389/fimmu.2024.1438371] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2024] [Accepted: 06/25/2024] [Indexed: 08/02/2024] Open
Abstract
Introduction Variant pseudorabies virus (PRV) is a newly emerged zoonotic pathogen that can cause human blindness. PRV can take advantage of its large genome and multiple non-essential genes to construct recombinant attenuated vaccines carrying foreign genes. However, a major problem is that the foreign genes in recombinant PRV are only integrated into the genome for independent expression, rather than assembled on the surface of virion. Methods We reported a recombinant PRV with deleted gE/TK genes and an inserted porcine circovirus virus 2 (PCV2) Cap gene into the extracellular domain of the PRV gE gene using the Cre-loxP recombinant system combined with the CRISPR-Cas9 gene editing system. This recombinant PRV (PRV-Cap), with the envelope-embedded Cap protein, exhibits a similar replication ability to its parental virus. Results An immunogenicity assay revealed that PRV-Cap immunized mice have 100% resistance to lethal PRV and PCV2 attacks. Neutralization antibody and ELISPOT detections indicated that PRV-Cap can enhance neutralizing antibodies to PRV and produce IFN-γ secreting T cells specific for both PRV and PCV2. Immunological mechanistic investigation revealed that initial immunization with PRV-Cap stimulates significantly early activation and expansion of CD69+ T cells, promoting the activation of CD4 Tfh cell dependent germinal B cells and producing effectively specific effector memory T and B cells. Booster immunization with PRV-Cap recalled the activation of PRV-specific IFN-γ+IL-2+CD4+ T cells and IFN-γ+TNF-α+CD8+ T cells, as well as PCV2-specific IFN-γ+TNF-α+CD8+ T cells. Conclusion Collectively, our data suggested an immunological mechanism in that the recombinant PRV with envelope-assembled PCV2 Cap protein can serve as an excellent vaccine candidate for combined immunity against PRV and PCV2, and provided a cost-effective method for the production of PRV- PCV2 vaccine.
Collapse
Affiliation(s)
- Chenhe Lu
- MOA Key Laboratory of Animal Virology, Zhejiang University Center for Veterinary Sciences, Hangzhou, China
| | - Haimin Li
- MOA Key Laboratory of Animal Virology, Zhejiang University Center for Veterinary Sciences, Hangzhou, China
| | - Wenjing Chen
- MOA Key Laboratory of Animal Virology, Zhejiang University Center for Veterinary Sciences, Hangzhou, China
| | - Hui Li
- College of Veterinary Medicine, Yangzhou University, Yangzhou, China
| | - Jiayu Ma
- MOA Key Laboratory of Animal Virology, Zhejiang University Center for Veterinary Sciences, Hangzhou, China
| | - Peng Peng
- MOA Key Laboratory of Animal Virology, Zhejiang University Center for Veterinary Sciences, Hangzhou, China
| | - Yan Yan
- MOA Key Laboratory of Animal Virology, Zhejiang University Center for Veterinary Sciences, Hangzhou, China
| | - Weiren Dong
- MOA Key Laboratory of Animal Virology, Zhejiang University Center for Veterinary Sciences, Hangzhou, China
| | - Yulan Jin
- MOA Key Laboratory of Animal Virology, Zhejiang University Center for Veterinary Sciences, Hangzhou, China
| | - Shiyue Pan
- MOA Key Laboratory of Animal Virology, Zhejiang University Center for Veterinary Sciences, Hangzhou, China
| | - Shaobin Shang
- College of Veterinary Medicine, Yangzhou University, Yangzhou, China
| | - Jinyan Gu
- MOA Key Laboratory of Animal Virology, Zhejiang University Center for Veterinary Sciences, Hangzhou, China
| | - Jiyong Zhou
- MOA Key Laboratory of Animal Virology, Zhejiang University Center for Veterinary Sciences, Hangzhou, China
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, First Affiliated Hospital, Zhejiang University, Hangzhou, China
| |
Collapse
|
7
|
Li J, Miller LC, Sang Y. Current Status of Vaccines for Porcine Reproductive and Respiratory Syndrome: Interferon Response, Immunological Overview, and Future Prospects. Vaccines (Basel) 2024; 12:606. [PMID: 38932335 PMCID: PMC11209547 DOI: 10.3390/vaccines12060606] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Revised: 05/26/2024] [Accepted: 05/31/2024] [Indexed: 06/28/2024] Open
Abstract
Porcine reproductive and respiratory syndrome (PRRS) remains a formidable challenge for the global pig industry. Caused by PRRS virus (PRRSV), this disease primarily affects porcine reproductive and respiratory systems, undermining effective host interferon and other immune responses, resulting in vaccine ineffectiveness. In the absence of specific antiviral treatments for PRRSV, vaccines play a crucial role in managing the disease. The current market features a range of vaccine technologies, including live, inactivated, subunit, DNA, and vector vaccines, but only modified live virus (MLV) and killed virus (KV) vaccines are commercially available for PRRS control. Live vaccines are promoted for their enhanced protective effectiveness, although their ability to provide cross-protection is modest. On the other hand, inactivated vaccines are emphasized for their safety profile but are limited in their protective efficacy. This review updates the current knowledge on PRRS vaccines' interactions with the host interferon system, and other immunological aspects, to assess their current status and evaluate advents in PRRSV vaccine development. It presents the strengths and weaknesses of both live attenuated and inactivated vaccines in the prevention and management of PRRS, aiming to inspire the development of innovative strategies and technologies for the next generation of PRRS vaccines.
Collapse
Affiliation(s)
- Jiuyi Li
- Department of Food and Animal Sciences, College of Agriculture, Tennessee State University, 3500 John A Merritt Blvd, Nashville, TN 37209, USA;
| | - Laura C. Miller
- Diagnostic Medicine and Pathobiology, College of Veterinary Medicine, Kansas State University, 1800 Denison Ave, Manhattan, KS 66506, USA;
| | - Yongming Sang
- Department of Food and Animal Sciences, College of Agriculture, Tennessee State University, 3500 John A Merritt Blvd, Nashville, TN 37209, USA;
| |
Collapse
|
8
|
Wang X, Li Y, Dong S, Wang C, Wang Y, Zhang H. Transcriptomic analysis reveals impact of gE/gI/TK deletions on host response to PRV infection. Virol J 2023; 20:303. [PMID: 38115115 PMCID: PMC10731697 DOI: 10.1186/s12985-023-02265-y] [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: 08/17/2023] [Accepted: 12/09/2023] [Indexed: 12/21/2023] Open
Abstract
BACKGROUND Pseudorabies virus (PRV) causes substantial losses in the swine industry worldwide. Attenuated PRV strains with deletions of immunomodulatory genes glycoprotein E (gE), glycoprotein I (gI) and thymidine kinase (TK) are candidate vaccines. However, the effects of gE/gI/TK deletions on PRV-host interactions are not well understood. METHODS To characterize the impact of gE/gI/TK deletions on host cells, we analyzed and compared the transcriptomes of PK15 cells infected with wild-type PRV (SD2017), PRV with gE/gI/TK deletions (SD2017gE/gI/TK) using RNA-sequencing. RESULTS The attenuated SD2017gE/gI/TK strain showed increased expression of inflammatory cytokines and pathways related to immunity compared to wild-type PRV. Cell cycle regulation and metabolic pathways were also perturbed. CONCLUSIONS Deletion of immunomodulatory genes altered PRV interactions with host cells and immune responses. This study provides insights into PRV vaccine design.
Collapse
Affiliation(s)
- Xiaoli Wang
- Department of Preventive Veterinary Medicine, College of Veterinary Medicine, Shandong Agricultural University, Taian, China
| | - Yingguang Li
- Shandong Collaborative Innovation Center for Development of Veterinary Pharmaceuticals, College of Veterinary Medicine, Qingdao Agricultural University, Qingdao, China
| | - Shaoming Dong
- Shandong Collaborative Innovation Center for Development of Veterinary Pharmaceuticals, College of Veterinary Medicine, Qingdao Agricultural University, Qingdao, China
| | - Cong Wang
- China animal husbandry industry Co., Ltd, Beijing, China
| | - Yongming Wang
- Shandong Huahong Biological Engineering Co., Ltd, Binzhou, China
| | - Hongliang Zhang
- Shandong Collaborative Innovation Center for Development of Veterinary Pharmaceuticals, College of Veterinary Medicine, Qingdao Agricultural University, Qingdao, China.
| |
Collapse
|
9
|
Research Progress on Glycoprotein 5 of Porcine Reproductive and Respiratory Syndrome Virus. Animals (Basel) 2023; 13:ani13050813. [PMID: 36899670 PMCID: PMC10000246 DOI: 10.3390/ani13050813] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Revised: 02/14/2023] [Accepted: 02/20/2023] [Indexed: 02/26/2023] Open
Abstract
Porcine reproductive and respiratory syndrome (PRRS) is an acute, febrile, and highly contagious disease caused by the porcine reproductive and respiratory syndrome virus (PRRSV). Glycoprotein 5 (GP5) is a glycosylated envelope protein encoded by the PRRSV ORF5, which has good immunogenicity and can induce the body to produce neutralizing antibodies. Therefore, study of GP5 protein is of great significance in the diagnosis, prevention, and control of PRRSV and the development of new vaccines. We reviewed GP5 protein genetic variation, immune function, interaction with viral protein and host proteins, induction of cell apoptosis, and stimulation of neutralizing antibodies. GP5 protein's influence on virus replication and virulence, as well as its use as a target for viral detection and immunization are reviewed.
Collapse
|
10
|
Parys A, Vandoorn E, Chiers K, Passvogel K, Fuchs W, Mettenleiter TC, Van Reeth K. Exploring Prime-Boost Vaccination Regimens with Different H1N1 Swine Influenza A Virus Strains and Vaccine Platforms. Vaccines (Basel) 2022; 10:1826. [PMID: 36366335 PMCID: PMC9699596 DOI: 10.3390/vaccines10111826] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Revised: 10/26/2022] [Accepted: 10/26/2022] [Indexed: 11/07/2023] Open
Abstract
In a previous vaccination study in pigs, heterologous prime-boost vaccination with whole-inactivated H1N1 virus vaccines (WIV) induced superior antibody responses and protection compared to homologous prime-boost vaccination. However, no pan-H1 antibody response was induced. Therefore, to stimulate both local and systemic immune responses, we first vaccinated pigs intranasally with a pseudorabies vector vaccine expressing the pH1N1 hemagglutinin (prvCA09) followed by a homologous or heterologous WIV booster vaccine. Homologous and heterologous WIV-WIV vaccinated groups and mock-vaccinated or prvCA09 single-vaccinated pigs served as control groups. Five weeks after the second vaccination, pigs were challenged with a homologous pH1N1 or one of two heterologous H1N2 swine influenza A virus strains. A single prvCA09 vaccination resulted in complete protection against homologous challenge, and vector-WIV vaccinated groups were significantly better protected against heterologous challenge compared to the challenge control group or WIV-WIV vaccinated groups. Furthermore, vector-WIV vaccination resulted in broader hemagglutination inhibition antibody responses compared to WIV-WIV vaccination and higher numbers of antibody-secreting cells in peripheral blood, draining lymph nodes and nasal mucosa. However, even though vector-WIV vaccination induced stronger antibody responses and protection, we still failed to induce a pan-H1 antibody response.
Collapse
Affiliation(s)
- Anna Parys
- Laboratory of Virology, Faculty of Veterinary Medicine, Ghent University, 9820 Merelbeke, Belgium
| | - Elien Vandoorn
- Laboratory of Virology, Faculty of Veterinary Medicine, Ghent University, 9820 Merelbeke, Belgium
| | - Koen Chiers
- Laboratory of Veterinary Pathology, Faculty of Veterinary Medicine, Ghent University, 9820 Merelbeke, Belgium
| | - Katharina Passvogel
- Institute of Molecular Virology and Cell Biology, Friedrich-Loeffler-Institut, Federal Research Institute for Animal Health, 17493 Greifswald, Germany
| | - Walter Fuchs
- Institute of Molecular Virology and Cell Biology, Friedrich-Loeffler-Institut, Federal Research Institute for Animal Health, 17493 Greifswald, Germany
| | - Thomas C. Mettenleiter
- Institute of Molecular Virology and Cell Biology, Friedrich-Loeffler-Institut, Federal Research Institute for Animal Health, 17493 Greifswald, Germany
| | - Kristien Van Reeth
- Laboratory of Virology, Faculty of Veterinary Medicine, Ghent University, 9820 Merelbeke, Belgium
| |
Collapse
|
11
|
Li R, Shao G, Xie Z, Hu Z, Feng K, He J, Wang H, Fu J, Zhang X, Xie Q. Construction and Immunogenicity of a Recombinant Pseudorabies Virus Expressing SARS-CoV-2-S and SARS-CoV-2-N. Front Vet Sci 2022; 9:920087. [PMID: 35982925 PMCID: PMC9380597 DOI: 10.3389/fvets.2022.920087] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Accepted: 06/22/2022] [Indexed: 12/05/2022] Open
Abstract
Coronavirus (CoV) is an important pathogen of humans and animals, which can infect humans or animals through the respiratory mucosal route. Syndrome coronavirus 2 (SARS-CoV-2) is quite similar to syndrome coronavirus (SARS-CoV) with the same receptor, angiotensin-converting enzyme 2 (ACE2). The S and N proteins are the most important protective antigens of the SARS-CoV-2. The S protein on the viral membrane mediates the virus attachment with the host cells, and the N protein is the most abundant expression during infection. In this study, the recombinant viruses expressing the S and N proteins of SARS-CoV-2 were successfully constructed by Red/ET recombinant technology using Pseudorabies virus (PRV) strain Bartha-K61 as a vector. Genetic stability and growth kinetics analysis showed that the recombinant viruses rPRV-SARS-CoV-2-S and rPRV-SARS-CoV-2-N had similar genetic stability and proliferation characteristics to the parental PRV. The immunoassay results showed that mice immunized with recombinant viruses could produce total IgG antibodies. Therefore, PRV is feasible and promising as a viral vector to express SARS-CoV-2-S and SARS-CoV-2-N genes. This study can provide a reference for future research on live vector vaccines for domestic animals, pets, and wild animals.
Collapse
Affiliation(s)
- Ruoying Li
- Heyuan Branch, Guangdong Provincial Laboratory of Lingnan Modern Agricultural Science and Technology & Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou, China
- Guangdong Engineering Research Center for Vector Vaccine of Animal Virus, College of Animal Science, South China Agricultural University, Guangzhou, China
- South China Collaborative Innovation Center for Poultry Disease Control and Product Safety, College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Guanming Shao
- Heyuan Branch, Guangdong Provincial Laboratory of Lingnan Modern Agricultural Science and Technology & Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou, China
- Guangdong Engineering Research Center for Vector Vaccine of Animal Virus, College of Animal Science, South China Agricultural University, Guangzhou, China
- South China Collaborative Innovation Center for Poultry Disease Control and Product Safety, College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Zi Xie
- Heyuan Branch, Guangdong Provincial Laboratory of Lingnan Modern Agricultural Science and Technology & Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou, China
- Guangdong Engineering Research Center for Vector Vaccine of Animal Virus, College of Animal Science, South China Agricultural University, Guangzhou, China
- South China Collaborative Innovation Center for Poultry Disease Control and Product Safety, College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Zezhong Hu
- Heyuan Branch, Guangdong Provincial Laboratory of Lingnan Modern Agricultural Science and Technology & Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou, China
- Guangdong Engineering Research Center for Vector Vaccine of Animal Virus, College of Animal Science, South China Agricultural University, Guangzhou, China
- South China Collaborative Innovation Center for Poultry Disease Control and Product Safety, College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Keyu Feng
- Heyuan Branch, Guangdong Provincial Laboratory of Lingnan Modern Agricultural Science and Technology & Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou, China
- Guangdong Engineering Research Center for Vector Vaccine of Animal Virus, College of Animal Science, South China Agricultural University, Guangzhou, China
- South China Collaborative Innovation Center for Poultry Disease Control and Product Safety, College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Jiahui He
- Heyuan Branch, Guangdong Provincial Laboratory of Lingnan Modern Agricultural Science and Technology & Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou, China
- Guangdong Engineering Research Center for Vector Vaccine of Animal Virus, College of Animal Science, South China Agricultural University, Guangzhou, China
- South China Collaborative Innovation Center for Poultry Disease Control and Product Safety, College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Hailong Wang
- State Key Laboratory of Microbial Technology, Institute of Microbial Technology, Helmholtz International Lab for Anti-infectives, Shandong University–Helmholtz Institute of Biotechnology, Shandong University, Jinan, China
| | - Jun Fu
- State Key Laboratory of Microbial Technology, Institute of Microbial Technology, Helmholtz International Lab for Anti-infectives, Shandong University–Helmholtz Institute of Biotechnology, Shandong University, Jinan, China
| | - Xinheng Zhang
- Heyuan Branch, Guangdong Provincial Laboratory of Lingnan Modern Agricultural Science and Technology & Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou, China
- Guangdong Engineering Research Center for Vector Vaccine of Animal Virus, College of Animal Science, South China Agricultural University, Guangzhou, China
- South China Collaborative Innovation Center for Poultry Disease Control and Product Safety, College of Animal Science, South China Agricultural University, Guangzhou, China
- Key Laboratory of Animal Health Aquaculture and Environmental Control, College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Qingmei Xie
- Heyuan Branch, Guangdong Provincial Laboratory of Lingnan Modern Agricultural Science and Technology & Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou, China
- Guangdong Engineering Research Center for Vector Vaccine of Animal Virus, College of Animal Science, South China Agricultural University, Guangzhou, China
- South China Collaborative Innovation Center for Poultry Disease Control and Product Safety, College of Animal Science, South China Agricultural University, Guangzhou, China
- Key Laboratory of Animal Health Aquaculture and Environmental Control, College of Animal Science, South China Agricultural University, Guangzhou, China
- *Correspondence: Qingmei Xie
| |
Collapse
|
12
|
Liu Z, Kong Z, Chen M, Shang Y. Design of live-attenuated animal vaccines based on pseudorabies virus platform. ANIMAL DISEASES 2022. [DOI: 10.1186/s44149-022-00044-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
AbstractPseudorabies virus (PRV) is a double-stranded DNA virus with a genome approximating 150 kb in size. PRV contains many non-essential genes that can be replaced with genes encoding heterogenous antigens without affecting viral propagation. With the ability to induce cellular, humoral and mucosal immune responses in the host, PRV is considered to be an ideal and potential live vector for generation of animal vaccines. In this review, we summarize the advances in attenuated recombinant PRVs and design of PRV-based live vaccines as well as the challenge of vaccine application.
Collapse
|
13
|
Li F, Li B, Niu X, Chen W, Li Y, Wu K, Li X, Ding H, Zhao M, Chen J, Yi L. The Development of Classical Swine Fever Marker Vaccines in Recent Years. Vaccines (Basel) 2022; 10:vaccines10040603. [PMID: 35455351 PMCID: PMC9026404 DOI: 10.3390/vaccines10040603] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Revised: 04/05/2022] [Accepted: 04/10/2022] [Indexed: 02/01/2023] Open
Abstract
Classical swine fever (CSF) is a severe disease that has caused serious economic losses for the global pig industry and is widely prevalent worldwide. In recent decades, CSF has been effectively controlled through compulsory vaccination with a live CSF vaccine (C strain). It has been successfully eradicated in some countries or regions. However, the re-emergence of CSF in Japan and Romania, where it had been eradicated, has brought increased attention to the disease. Because the traditional C-strain vaccine cannot distinguish between vaccinated and infected animals (DIVA), this makes it difficult to fight CSF. The emergence of marker vaccines is considered to be an effective strategy for the decontamination of CSF. This paper summarizes the progress of the new CSF marker vaccine and provides a detailed overview of the vaccine design ideas and immunization effects. It also provides a methodology for the development of a new generation of vaccines for CSF and vaccine development for other significant epidemics.
Collapse
Affiliation(s)
- Fangfang Li
- College of Veterinary Medicine, South China Agricultural University, No. 483 Wushan Road, Tianhe District, Guangzhou 510642, China; (F.L.); (B.L.); (X.N.); (W.C.); (Y.L.); (K.W.); (X.L.); (H.D.); (M.Z.)
- Guangdong Laboratory for Lingnan Modern Agriculture, College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China
- Key Laboratory of Zoonosis Prevention and Control of Guangdong Province, Guangzhou 510642, China
| | - Bingke Li
- College of Veterinary Medicine, South China Agricultural University, No. 483 Wushan Road, Tianhe District, Guangzhou 510642, China; (F.L.); (B.L.); (X.N.); (W.C.); (Y.L.); (K.W.); (X.L.); (H.D.); (M.Z.)
- Guangdong Laboratory for Lingnan Modern Agriculture, College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China
- Key Laboratory of Zoonosis Prevention and Control of Guangdong Province, Guangzhou 510642, China
| | - Xinni Niu
- College of Veterinary Medicine, South China Agricultural University, No. 483 Wushan Road, Tianhe District, Guangzhou 510642, China; (F.L.); (B.L.); (X.N.); (W.C.); (Y.L.); (K.W.); (X.L.); (H.D.); (M.Z.)
- Guangdong Laboratory for Lingnan Modern Agriculture, College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China
- Key Laboratory of Zoonosis Prevention and Control of Guangdong Province, Guangzhou 510642, China
| | - Wenxian Chen
- College of Veterinary Medicine, South China Agricultural University, No. 483 Wushan Road, Tianhe District, Guangzhou 510642, China; (F.L.); (B.L.); (X.N.); (W.C.); (Y.L.); (K.W.); (X.L.); (H.D.); (M.Z.)
- Guangdong Laboratory for Lingnan Modern Agriculture, College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China
- Key Laboratory of Zoonosis Prevention and Control of Guangdong Province, Guangzhou 510642, China
| | - Yuwan Li
- College of Veterinary Medicine, South China Agricultural University, No. 483 Wushan Road, Tianhe District, Guangzhou 510642, China; (F.L.); (B.L.); (X.N.); (W.C.); (Y.L.); (K.W.); (X.L.); (H.D.); (M.Z.)
- Guangdong Laboratory for Lingnan Modern Agriculture, College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China
- Key Laboratory of Zoonosis Prevention and Control of Guangdong Province, Guangzhou 510642, China
| | - Keke Wu
- College of Veterinary Medicine, South China Agricultural University, No. 483 Wushan Road, Tianhe District, Guangzhou 510642, China; (F.L.); (B.L.); (X.N.); (W.C.); (Y.L.); (K.W.); (X.L.); (H.D.); (M.Z.)
- Guangdong Laboratory for Lingnan Modern Agriculture, College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China
- Key Laboratory of Zoonosis Prevention and Control of Guangdong Province, Guangzhou 510642, China
| | - Xiaowen Li
- College of Veterinary Medicine, South China Agricultural University, No. 483 Wushan Road, Tianhe District, Guangzhou 510642, China; (F.L.); (B.L.); (X.N.); (W.C.); (Y.L.); (K.W.); (X.L.); (H.D.); (M.Z.)
- Guangdong Laboratory for Lingnan Modern Agriculture, College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China
- Key Laboratory of Zoonosis Prevention and Control of Guangdong Province, Guangzhou 510642, China
| | - Hongxing Ding
- College of Veterinary Medicine, South China Agricultural University, No. 483 Wushan Road, Tianhe District, Guangzhou 510642, China; (F.L.); (B.L.); (X.N.); (W.C.); (Y.L.); (K.W.); (X.L.); (H.D.); (M.Z.)
- Guangdong Laboratory for Lingnan Modern Agriculture, College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China
- Key Laboratory of Zoonosis Prevention and Control of Guangdong Province, Guangzhou 510642, China
| | - Mingqiu Zhao
- College of Veterinary Medicine, South China Agricultural University, No. 483 Wushan Road, Tianhe District, Guangzhou 510642, China; (F.L.); (B.L.); (X.N.); (W.C.); (Y.L.); (K.W.); (X.L.); (H.D.); (M.Z.)
- Guangdong Laboratory for Lingnan Modern Agriculture, College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China
- Key Laboratory of Zoonosis Prevention and Control of Guangdong Province, Guangzhou 510642, China
| | - Jinding Chen
- College of Veterinary Medicine, South China Agricultural University, No. 483 Wushan Road, Tianhe District, Guangzhou 510642, China; (F.L.); (B.L.); (X.N.); (W.C.); (Y.L.); (K.W.); (X.L.); (H.D.); (M.Z.)
- Guangdong Laboratory for Lingnan Modern Agriculture, College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China
- Key Laboratory of Zoonosis Prevention and Control of Guangdong Province, Guangzhou 510642, China
- Correspondence: (J.C.); (L.Y.); Tel.: +86-20-8528-8017 (J.C.); +86-20-8528-8017 (L.Y.)
| | - Lin Yi
- College of Veterinary Medicine, South China Agricultural University, No. 483 Wushan Road, Tianhe District, Guangzhou 510642, China; (F.L.); (B.L.); (X.N.); (W.C.); (Y.L.); (K.W.); (X.L.); (H.D.); (M.Z.)
- Guangdong Laboratory for Lingnan Modern Agriculture, College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China
- Key Laboratory of Zoonosis Prevention and Control of Guangdong Province, Guangzhou 510642, China
- Correspondence: (J.C.); (L.Y.); Tel.: +86-20-8528-8017 (J.C.); +86-20-8528-8017 (L.Y.)
| |
Collapse
|
14
|
McLean RK, Graham SP. The pig as an amplifying host for new and emerging zoonotic viruses. One Health 2022; 14:100384. [PMID: 35392655 PMCID: PMC8975596 DOI: 10.1016/j.onehlt.2022.100384] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Revised: 03/28/2022] [Accepted: 03/29/2022] [Indexed: 12/23/2022] Open
Abstract
Pig production is a rapidly growing segment of the global livestock sector, especially in Asia and Africa. Expansion and intensification of pig production has resulted in significant changes to traditional pig husbandry practices leading to an environment conducive to increased emergence and spread of infectious diseases. These include a number of zoonotic viruses including influenza, Japanese encephalitis, Nipah and coronaviruses. Pigs are known to independently facilitate the creation of novel reassortant influenza A virus strains, capable of causing pandemics. Moreover, pigs play a role in the amplification of Japanese encephalitis virus, transmitted by mosquito vectors found in areas inhabited by over half the world's human population. Furthermore, pigs acted as an amplifying host in the first and still most severe outbreak of Nipah virus in Malaysia, that necessitated the culling over 1 million pigs. Finally, novel porcine coronaviruses are being discovered in high pig-density countries which have pandemic potential. In this review, we discuss the role that pigs play as intermediate/amplifying hosts for zoonotic viruses with pandemic potential and consider how multivalent vaccination of pigs could in turn safeguard human health.
Collapse
|
15
|
Zhao J, Zhu L, Xu L, Li F, Deng H, Huang Y, Gu S, Sun X, Zhou Y, Xu Z. The Construction and Immunogenicity Analyses of Recombinant Pseudorabies Virus With NADC30-Like Porcine Reproductive and Respiratory Syndrome Virus-Like Particles Co-expression. Front Microbiol 2022; 13:846079. [PMID: 35308386 PMCID: PMC8924499 DOI: 10.3389/fmicb.2022.846079] [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: 12/30/2021] [Accepted: 01/24/2022] [Indexed: 12/15/2022] Open
Abstract
Porcine reproductive and respiratory syndrome (PRRS) and pseudorabies (PR) are highly infectious swine diseases and cause significant financial loss in China. The respiratory system and reproductive system are the main target systems. Previous studies showed that the existing PR virus (PRV) and PRRS virus (PRRSV) commercial vaccines could not provide complete protection against PRV variant strains and NADC30-like PRRSV strains in China. In this study, the PRV variant strain XJ and NADC30-like PRRSV strain CHSCDJY-2019 are used as the parent for constructing a recombinant pseudorabies virus (rPRV)-NC56 with gE/gI/TK gene deletion and co-expressing NADC30-like PRRSV GP5 and M protein. The rPRV-NC56 proliferated stably in BHK-21 cells, and it could stably express GP5 and M protein. Due to the introduction of the self-cleaving 2A peptide, GP5 and M protein were able to express independently and form virus-like particles (VLPs) of PRRSV in rPRV-NC56-infected BHK-21 cells. The rPRV-NC56 is safe for use in mice; it can colonize and express the target protein in mouse lungs for a long time. Vaccination with rPRV-NC56 induces PRV and NADC30-like PRRSV specific humoral and cellular immune responses in mice, and protects 100% of mice from virulent PRV XJ strain. Furthermore, the virus-neutralizing antibody (VNA) elicited by rPRV-NC56 showed significantly lower titer against SCNJ-2016 (HP-PRRSV) than that against CHSCDJY-2019 (NADC30-like PRRSV). Thus, rPRV-NC56 appears to be a promising candidate vaccine against NADC30-like PRRSV and PRV for the control and eradication of the variant PRV and NADC30-like PRRSV.
Collapse
Affiliation(s)
- Jun Zhao
- College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Ling Zhu
- College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China.,Key Laboratory of Animal Disease and Human Health of Sichuan Province, Chengdu, China
| | - Lei Xu
- College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Fengqing Li
- College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Huidan Deng
- College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Yao Huang
- College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Sirui Gu
- College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Xianggang Sun
- College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Yuancheng Zhou
- Animal Breeding and Genetics Key Laboratory of Sichuan Province, Sichuan Animal Science Academy, Chengdu, China
| | - Zhiwen Xu
- College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China.,Key Laboratory of Animal Disease and Human Health of Sichuan Province, Chengdu, China
| |
Collapse
|
16
|
Yuan H, Zheng Y, Yan X, Wang H, Zhang Y, Ma J, Fu J. Direct cloning of a herpesvirus genome for rapid generation of infectious BAC clones. J Adv Res 2022; 43:97-107. [PMID: 36585118 PMCID: PMC9811322 DOI: 10.1016/j.jare.2022.02.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Revised: 02/18/2022] [Accepted: 02/21/2022] [Indexed: 01/07/2023] Open
Abstract
INTRODUCTION The herpesviridae are DNA viruses with large and complicated genomes. The herpesvirus bacterial artificial chromosomes (BACs) have been useful for generating recombinant viruses to study the biology and pathogenesis. However, the conventional method using homologous recombination is not only time consuming but also prone to accumulate attenuating mutations during serial passage of the virus in cells. Elimination of the BAC vector from the recombinant viral genome requires additional step for phenotypically consistence with the original strain. OBJECTIVES To generate a streamlined approach for generating infectious BAC clones of herpesvirus. METHODS The 142-kb pseudorabies virus genome was directly cloned into a bacterial artificial chromosome (BAC) in Escherichia coli by Exonuclease Combined with RecET recombination (ExoCET). Placement of the BAC vector at the terminus of the linear virus genome enabled excision of the BAC backbone from the viral genome by restriction endonuclease for delivery into mammalian cells, with the subsequent rapid rescue of virus that was genetically identical to the original strain. RESULTS This new approach for molecular cloning of the genome from a large DNA virus and isolation of pure virus lacking the BAC vector from transfected mammalian cells bypass the tedious and time-consuming method of multiple rounds of plaque purification. The viral BAC was stable in E. coli, allowing further mutagenesis mediated by the Red system or various site-specific recombination methods. CONCLUSION An efficient method for construction of infectious clones of herpesvirus was established. It is expected to be potentially useful for other viruses with large double-stranded DNA genomes.
Collapse
Affiliation(s)
- Hengxing Yuan
- Shandong University–Helmholtz Institute of Biotechnology, State Key Laboratory of Microbial Technology, Shandong University, Qingdao 266237, China
| | - Yaoyao Zheng
- College of Animal Science, South China Agricultural University, Guangzhou 510642, China
| | - Xiaoling Yan
- College of Animal Science, South China Agricultural University, Guangzhou 510642, China
| | - Hailong Wang
- Shandong University–Helmholtz Institute of Biotechnology, State Key Laboratory of Microbial Technology, Shandong University, Qingdao 266237, China
| | - Youming Zhang
- Shandong University–Helmholtz Institute of Biotechnology, State Key Laboratory of Microbial Technology, Shandong University, Qingdao 266237, China,Corresponding authors.
| | - Jingyun Ma
- College of Animal Science, South China Agricultural University, Guangzhou 510642, China,Corresponding authors.
| | - Jun Fu
- Shandong University–Helmholtz Institute of Biotechnology, State Key Laboratory of Microbial Technology, Shandong University, Qingdao 266237, China,Corresponding authors.
| |
Collapse
|
17
|
Porcine Reproductive and Respiratory Syndrome Virus: Immune Escape and Application of Reverse Genetics in Attenuated Live Vaccine Development. Vaccines (Basel) 2021; 9:vaccines9050480. [PMID: 34068505 PMCID: PMC8150910 DOI: 10.3390/vaccines9050480] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Revised: 04/08/2021] [Accepted: 04/11/2021] [Indexed: 01/16/2023] Open
Abstract
Porcine reproductive and respiratory syndrome virus (PRRSV), an RNA virus widely prevalent in pigs, results in significant economic losses worldwide. PRRSV can escape from the host immune response in several processes. Vaccines, including modified live vaccines and inactivated vaccines, are the best available countermeasures against PRRSV infection. However, challenges still exist as the vaccines are not able to induce broad protection. The reason lies in several facts, mainly the variability of PRRSV and the complexity of the interaction between PRRSV and host immune responses, and overcoming these obstacles will require more exploration. Many novel strategies have been proposed to construct more effective vaccines against this evolving and smart virus. In this review, we will describe the mechanisms of how PRRSV induces weak and delayed immune responses, the current vaccines of PRRSV, and the strategies to develop modified live vaccines using reverse genetics systems.
Collapse
|
18
|
Lv L, Liu X, Jiang C, Wang X, Cao M, Bai J, Jiang P. Pathogenicity and immunogenicity of a gI/gE/TK/UL13-gene-deleted variant pseudorabies virus strain in swine. Vet Microbiol 2021; 258:109104. [PMID: 34004569 DOI: 10.1016/j.vetmic.2021.109104] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Accepted: 05/06/2021] [Indexed: 11/29/2022]
Abstract
Pseudorabies is a highly infectious disease with severe clinical symptoms, causing acute death in infected pigs and leading to substantial economic losses among swine producers. In this study, a vaccine candidate strain in which the protein kinase UL13 gene was deleted was constructed with the CRISPR/Cas9 system based on the recombinant pseudorabies virus (PRV) ZJ01-ΔgI/gE/TK. Pigs immunized with ZJ01-ΔgI/gE/TK or ZJ01-ΔgI/gE/TK/UL13 produced high levels of anti-gB antibodies and virus-neutralizing antibodies. ZJ01-ΔgI/gE/TK/UL13 provided greater protective efficacy against challenge with PRV variant strain ZJ01 than did Bartha-K61 or ZJ01-ΔgI/gE/TK. The pigs vaccinated with ZJ01-ΔgI/gE/TK/UL13 excreted significantly less virus than those vaccinated with Bartha-K61 or ZJ01-ΔgI/gE/TK. The viral loads in the lungs of pigs treated with ZJ01-ΔgI/gE/TK/UL13 were lower than those in pigs treated with ZJ01-ΔgI/gE/TK after challenge with PRV variant strain ZJ01. These data indicated that ZJ01-ΔgI/gE/TK/UL13 had greater protective efficacy and safety than the commercial ZJ01-ΔgI/gE/TK and Bartha-K61 vaccines, and could be developed as a promising vaccine candidate for the prevention and control of this disease.
Collapse
Affiliation(s)
- Lin Lv
- Key Laboratory of Animal Diseases Diagnostic and Immunology, Ministry of Agriculture, MOE International Joint Collaborative Research Laboratory for Animal Health & Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, China
| | - Xing Liu
- Key Laboratory of Animal Diseases Diagnostic and Immunology, Ministry of Agriculture, MOE International Joint Collaborative Research Laboratory for Animal Health & Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, China
| | - Chenlong Jiang
- Key Laboratory of Animal Diseases Diagnostic and Immunology, Ministry of Agriculture, MOE International Joint Collaborative Research Laboratory for Animal Health & Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, China
| | - Xianwei Wang
- Key Laboratory of Animal Diseases Diagnostic and Immunology, Ministry of Agriculture, MOE International Joint Collaborative Research Laboratory for Animal Health & Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, China
| | - Mingzhu Cao
- Key Laboratory of Animal Diseases Diagnostic and Immunology, Ministry of Agriculture, MOE International Joint Collaborative Research Laboratory for Animal Health & Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, China
| | - Juan Bai
- Key Laboratory of Animal Diseases Diagnostic and Immunology, Ministry of Agriculture, MOE International Joint Collaborative Research Laboratory for Animal Health & Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, China.
| | - Ping Jiang
- Key Laboratory of Animal Diseases Diagnostic and Immunology, Ministry of Agriculture, MOE International Joint Collaborative Research Laboratory for Animal Health & Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, China; Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, China.
| |
Collapse
|
19
|
Better immune efficacy triggered by the inactivated gI/gE-deleted pseudorabies virus with the additional insertion of gC gene in mice and weaned pigs. Virus Res 2021; 296:198353. [PMID: 33640358 DOI: 10.1016/j.virusres.2021.198353] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Revised: 02/20/2021] [Accepted: 02/22/2021] [Indexed: 11/21/2022]
Abstract
A new variant of pseudorabies virus (PRV) with high pathogenicity has been prevalent in many swineherds vaccinated with Bartha-K61 in China since 2011. Several gene-deleted vaccine candidates have been developed based on new emerging PRV variants. PRV-AH, a new emerging PRV strain from Anhui Province, was isolated in our laboratory in 2013. In the present study, rPRV-AH-gI-/gE- and rPRV-AH-gI-/gE-/gC+ were generated based on PRV-AH by homologous recombination. The growth kinetics of rPRV-AH-gI-/gE- and rPRV-AH-gI-/gE-/gC+ were similar to their parental strains. Compared with the commercial inactivated vaccine of Ea strain, the immune efficacy of the inactivated vaccine based on recombinant viruses was evaluated in mice and weaned pigs. The result showed that the level of neutralizing antibody in mice immunized with rPRV-AH-gI-/gE-/gC+ was higher compared with those immunized with rPRV-AH-gI-/gE- at a dose of 106 TCID50 at 8 weeks post initial immunization (p < 0.0001). Among the groups immunized at a dose of 105 TCID50, the rPRV-AH-gI-/gE- group showed a survival rate of 37.5 %, while the rPRV-AH-gI-/gE-/gC+ group showed a protection rate of 87.5 % against the PRV-AH challenge. Besides, the rPRV-AH-gI-/gE- and rPRV-AH-gI-/gE-/gC+ group immunized at a dose of 106 TCID50 showed a survival rate of 100 %. Interestingly, compared with the commercial vaccine group, the group of 105 TCID50 rPRV-AH-gI-/gE-/gC+ showed a lower level of neutralizing antibodies (p < 0.0001) but the same protection rate in mice. Moreover, in the pig experiment, the level of neutralizing antibodies in the group vaccinated with inactivated rPRV-AH-gI-/gE-/gC+ was higher than any other groups at 8 weeks post initial immunization (p < 0.05). More importantly, the milder symptoms and pathological lesions occurred in pigs vaccinated with rPRV-AH-gI-/gE-/gC+ after challenge with 106 TCID50 PRV-AH, revealing that additional insertion of gC gene could enhance the protective efficacy in PRV gI/gE-deleted vaccine in pigs. Collectively, these above-mentioned findings suggested that the inactivated vaccine of rPRV-AH-gI-/gE-/gC+ had a better immune efficacy, which could be regarded as a promising inactivated vaccine candidate for PRV control.
Collapse
|
20
|
Abid M, Teklue T, Li Y, Wu H, Wang T, Qiu HJ, Sun Y. Generation and Immunogenicity of a Recombinant Pseudorabies Virus Co-Expressing Classical Swine Fever Virus E2 Protein and Porcine Circovirus Type 2 Capsid Protein Based on Fosmid Library Platform. Pathogens 2019; 8:pathogens8040279. [PMID: 31805703 PMCID: PMC6963705 DOI: 10.3390/pathogens8040279] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2019] [Revised: 10/28/2019] [Accepted: 10/29/2019] [Indexed: 12/19/2022] Open
Abstract
Pseudorabies (PR), classical swine fever (CSF), and porcine circovirus type 2 (PCV2)-associated disease (PCVAD) are economically important infectious diseases of pigs. Co-infections of these diseases often occur in the field, posing significant threat to the swine industry worldwide. gE/gI/TK-gene-deleted vaccines are safe and capable of providing full protection against PR. Classical swine fever virus (CSFV) E2 glycoprotein is mainly used in the development of CSF vaccines. PCV2 capsid (Cap) protein is the major antigen targeted for developing PCV2 subunit vaccines. Multivalent vaccines, and especially virus-vectored vaccines expressing foreign proteins, are attractive strategies to fight co-infections for various swine diseases. The gene-deleted pseudorabies virus (PRV) can be used to develop promising and economical multivalent live virus-vectored vaccines. Herein, we constructed a gE/gI/TK-gene-deleted PRV co-expressing E2 of CSFV and Cap of PCV2 by fosmid library platform established for PRV, and the expression of E2 and Cap proteins was confirmed using immunofluorescence assay and western blotting. The recombinant virus propagated in porcine kidney 15 (PK-15) cells for 20 passages was genetically stable. The evaluation results in rabbits and pigs demonstrate that rPRVTJ-delgE/gI/TK-E2-Cap elicited detectable anti-PRV antibodies, but not anti-PCV2 or anti-CSFV antibodies. These findings provide insights that rPRVTJ-delgE/gI/TK-E2-Cap needs to be optimally engineered as a promising trivalent vaccine candidate against PRV, PCV2 and CSFV co-infections in future.
Collapse
Affiliation(s)
| | | | | | | | | | - Hua-Ji Qiu
- Correspondence: (H.-J.Q.); (Y.S.); Tel.: +86-451-5105-1708
| | - Yuan Sun
- Correspondence: (H.-J.Q.); (Y.S.); Tel.: +86-451-5105-1708
| |
Collapse
|
21
|
Li K, Bai J, Du L, Wang X, Ke C, Yan W, Li C, Ren L, Han H, Zhao Y. Generation of porcine monoclonal antibodies based on single cell technologies. Vet Immunol Immunopathol 2019; 215:109913. [PMID: 31420069 DOI: 10.1016/j.vetimm.2019.109913] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2019] [Revised: 07/17/2019] [Accepted: 07/30/2019] [Indexed: 01/03/2023]
Abstract
The development of a rapid and efficient system to generate porcine monoclonal antibodies (mAbs) is an important step toward the discovery of critical neutralizing targets for designing rational vaccines against porcine viruses. In this study, we established a platform for producing porcine mAbs based on single cell technologies. First, we singled out an optimal donor from 507 pigs based on serum antibody neutralizing activity against porcine reproductive and respiratory syndrome virus (PRRSV). After identifying the contribution of IgG to the neutralizing activity, single CD45R+IgG+Ag+ B cells were sorted from peripheral blood mononuclear cells (PBMCs). Single B cell RT-PCR was performed using primers designed to cover the germline repertoire of the porcine VH/VL gene segments. Paired VH/VLs were cloned into a eukaryotic expression vector and transfected into 293T cells. We demonstrate that full-length porcine mAbs were produced, and antigen-specific mAbs were obtained after further validation. The approach reported in this study can be applied to generate porcine mAbs against any given antigen and may help with the screening of neutralizing antibodies against porcine pathogens.
Collapse
Affiliation(s)
- Kongpan Li
- State Key Laboratory of Agrobiotechnology, College of Biological Sciences, National Engineering Laboratory for Animal Breeding, China Agricultural University, Beijing, People's Republic of China
| | - Jianhui Bai
- State Key Laboratory of Agrobiotechnology, College of Biological Sciences, National Engineering Laboratory for Animal Breeding, China Agricultural University, Beijing, People's Republic of China
| | - Lijuan Du
- State Key Laboratory of Agrobiotechnology, College of Biological Sciences, National Engineering Laboratory for Animal Breeding, China Agricultural University, Beijing, People's Republic of China
| | - Xifeng Wang
- Key Laboratory of Animal Ecology and Conservation Biology, Institute of Zoology, Chinese Academy of Science, Beijing, People's Republic of China
| | - Cuncun Ke
- State Key Laboratory of Agrobiotechnology, College of Biological Sciences, National Engineering Laboratory for Animal Breeding, China Agricultural University, Beijing, People's Republic of China
| | - Wei Yan
- XINDAMUYE Company, Henan, People's Republic of China
| | - Changqing Li
- XINDAMUYE Company, Henan, People's Republic of China
| | - Liming Ren
- State Key Laboratory of Agrobiotechnology, College of Biological Sciences, National Engineering Laboratory for Animal Breeding, China Agricultural University, Beijing, People's Republic of China
| | - Haitang Han
- State Key Laboratory of Agrobiotechnology, College of Biological Sciences, National Engineering Laboratory for Animal Breeding, China Agricultural University, Beijing, People's Republic of China.
| | - Yaofeng Zhao
- State Key Laboratory of Agrobiotechnology, College of Biological Sciences, National Engineering Laboratory for Animal Breeding, China Agricultural University, Beijing, People's Republic of China.
| |
Collapse
|
22
|
Kamel M, El-Sayed A. Utilization of herpesviridae as recombinant viral vectors in vaccine development against animal pathogens. Virus Res 2019; 270:197648. [PMID: 31279828 DOI: 10.1016/j.virusres.2019.197648] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2019] [Revised: 06/27/2019] [Accepted: 06/28/2019] [Indexed: 02/06/2023]
Abstract
Throughout the past few decades, numerous viral species have been generated as vaccine vectors. Every viral vector has its own distinct characteristics. For example, the family herpesviridae encompasses several viruses that have medical and veterinary importance. Attenuated herpesviruses are developed as vectors to convey heterologous immunogens targeting several serious and crucial pathogens. Some of these vectors have already been licensed for use in the veterinary field. One of their prominent features is their capability to accommodate large amount of foreign DNA, and to stimulate both cell-mediated and humoral immune responses. A better understanding of vector-host interaction builds up a robust foundation for the future development of herpesviruses-based vectors. At the time, many molecular tools are applied to enable the generation of herpesvirus-based recombinant vaccine vectors such as BAC technology, homologous and two-step en passant mutagenesis, codon optimization, and the CRISPR/Cas9 system. This review article highlights the most important techniques applied in constructing recombinant herpesviruses vectors, advantages and disadvantages of each recombinant herpesvirus vector, and the most recent research regarding their use to control major animal diseases.
Collapse
Affiliation(s)
- Mohamed Kamel
- Faculty of Veterinary Medicine, Department of Medicine and Infectious Diseases, Cairo University, Giza, Egypt.
| | - Amr El-Sayed
- Faculty of Veterinary Medicine, Department of Medicine and Infectious Diseases, Cairo University, Giza, Egypt
| |
Collapse
|
23
|
Cui J, O’Connell CM, Costa A, Pan Y, Smyth JA, Verardi PH, Burgess DJ, Van Kruiningen HJ, Garmendia AE. A PRRSV GP5-Mosaic vaccine: Protection of pigs from challenge and ex vivo detection of IFNγ responses against several genotype 2 strains. PLoS One 2019; 14:e0208801. [PMID: 30703122 PMCID: PMC6354972 DOI: 10.1371/journal.pone.0208801] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2018] [Accepted: 11/20/2018] [Indexed: 02/07/2023] Open
Abstract
Porcine reproductive and respiratory syndrome virus (PRRSV), is a highly mutable RNA virus that affects swine worldwide and its control is very challenging due to its formidable heterogeneity in the field. In the present study, DNA vaccines constructed with PRRSV GP5-Mosaic sequences were complexed to cationic liposomes and administered to experimental pigs by intradermal and intramuscular injection, followed by three boosters 14, 28 and 42 days later. The GP5-Mosaic vaccine thus formulated was immunogenic and induced protection from challenge in vaccinated pigs comparable to that induced by a wild type (VR2332) GP5 DNA vaccine (GP5-WT). Periodic sampling of blood and testing of vaccine-induced responses followed. Interferon-γ (IFN-γ) mRNA expression by virus-stimulated peripheral blood mononuclear cells (PBMCs) of GP5-Mosaic-vaccinated pigs was significantly higher compared to pigs vaccinated with either GP5-WT or empty vector at 21, 35 and 48 days after vaccination. Cross-reactive cellular responses were also demonstrated in GP5-Mosaic vaccinated pigs after stimulation of PBMCs with divergent strains of PRRSV. Thus, significantly higher levels of IFN-γ mRNA were detected when PBMCs from GP5-Mosaic-vaccinated pigs were stimulated by four Genotype 2 strains (VR2332, NADC9, NADC30 and SDSU73), which have at least 10% difference in GP5 amino acid sequences, while such responses were recorded only upon VR2332 stimulation in GP5-WT-vaccinated pigs. In addition, the levels of virus-specific neutralizing antibodies were higher in GP5-Mosaic or GP5-WT vaccinated pigs than those in vector-control pigs. The experimental pigs vaccinated with either the GP5-Mosaic vaccine or the GP5-WT vaccine were partially protected from challenge with VR2332, as measured by significantly lower viral loads in sera and tissues and lower lung lesion scores than the vector control group. These data demonstrate that the GP5-Mosaic vaccine can induce cross-reactive cellular responses to diverse strains, neutralizing antibodies, and protection in pigs.
Collapse
Affiliation(s)
- Junru Cui
- Department of Pathobiology and Veterinary Science, College of Agriculture, Health and Natural Resources, University of Connecticut, Storrs, Connecticut, United States of America
| | - Caitlin M. O’Connell
- Department of Pathobiology and Veterinary Science, College of Agriculture, Health and Natural Resources, University of Connecticut, Storrs, Connecticut, United States of America
| | - Antonio Costa
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Connecticut, Storrs, Connecticut, United States of America
| | - Yan Pan
- Guangxi Key Laboratory of Animal Vaccines and New Technology, Guangxi Veterinary Research Institute, Nanning, Guangxi, PR China
| | - Joan A. Smyth
- Department of Pathobiology and Veterinary Science, College of Agriculture, Health and Natural Resources, University of Connecticut, Storrs, Connecticut, United States of America
| | - Paulo H. Verardi
- Department of Pathobiology and Veterinary Science, College of Agriculture, Health and Natural Resources, University of Connecticut, Storrs, Connecticut, United States of America
| | - Diane J. Burgess
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Connecticut, Storrs, Connecticut, United States of America
| | - Herbert J. Van Kruiningen
- Department of Pathobiology and Veterinary Science, College of Agriculture, Health and Natural Resources, University of Connecticut, Storrs, Connecticut, United States of America
| | - Antonio E. Garmendia
- Department of Pathobiology and Veterinary Science, College of Agriculture, Health and Natural Resources, University of Connecticut, Storrs, Connecticut, United States of America
- * E-mail:
| |
Collapse
|
24
|
Hübner A, Keil GM, Kabuuka T, Mettenleiter TC, Fuchs W. Efficient transgene insertion in a pseudorabies virus vector by CRISPR/Cas9 and marker rescue-enforced recombination. J Virol Methods 2018; 262:38-47. [PMID: 30248362 DOI: 10.1016/j.jviromet.2018.09.009] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2018] [Revised: 09/19/2018] [Accepted: 09/19/2018] [Indexed: 02/07/2023]
Abstract
For development of vectored vaccines against porcine pathogens the genome of the pseudorabies virus vaccine strain Bartha (PrV-Ba) was previously cloned as an infectious bacterial artificial chromosome (BAC), containing the bacterial replicon and a reporter gene cassette encoding enhanced green fluorescent protein (EGFP) at the nonessential glycoprotein G locus. To facilitate substitution of this insertion, this BAC was now modified by deletion of the adjacent promoter and initiation codon of the essential glycoprotein D (gD) gene of PrV-Ba. Furthermore, rabbit kidney (RK13) cells stably expressing Cas9 nuclease and an EGFP gene-specific guide RNA were prepared to induce site specific cleavage of the BAC DNA. After co-transfection of these cells with the modified BAC and recombination plasmids containing expression cassettes for new transgenes flanked by PrV DNA sequences including the intact 5'-end of the gD gene, >95% of the recombinants exhibited the desired gene substitutions, while no EGFP-expressing progeny virus was detectable. This approach was used for insertion and expression of the open reading frames E199L, CP204L (p30) and KP177R (p22) of African swine fever virus. The studies revealed that codon adaptation significantly enhanced expression of E199L, and that the chimeric CAG promoter increased transgene expression compared to cytomegalovirus immediate-early promoters.
Collapse
Affiliation(s)
- Alexandra Hübner
- Institute of Molecular Virology and Cell Biology, Friedrich-Loeffler-Institut, 17493, Greifswald-Insel Riems, Germany
| | - Günther M Keil
- Institute of Molecular Virology and Cell Biology, Friedrich-Loeffler-Institut, 17493, Greifswald-Insel Riems, Germany
| | - Tonny Kabuuka
- Institute of Molecular Virology and Cell Biology, Friedrich-Loeffler-Institut, 17493, Greifswald-Insel Riems, Germany
| | - Thomas C Mettenleiter
- Institute of Molecular Virology and Cell Biology, Friedrich-Loeffler-Institut, 17493, Greifswald-Insel Riems, Germany
| | - Walter Fuchs
- Institute of Molecular Virology and Cell Biology, Friedrich-Loeffler-Institut, 17493, Greifswald-Insel Riems, Germany.
| |
Collapse
|
25
|
Ding YZ, Lv JL, Zhang ZW, Ma XY, Zhang J, Zhang YG. The program of antiviral agents inhibits virus infection. Arch Microbiol 2018; 200:841-846. [PMID: 29846759 DOI: 10.1007/s00203-018-1525-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2017] [Revised: 09/07/2017] [Accepted: 05/11/2018] [Indexed: 12/15/2022]
Abstract
Virus infections are the root cause of epidemics in the world. Vaccines and antiviral agents have been the two important methods to control viral diseases; in recent times, RNA-mediated therapeutics and prevention have received much attention. In this review, we provide an overview of the current information regarding the use of vaccines, antiviral agents, and RNA-mediated methods in controlling or preventing viral infections. We stress specifically on the potential of existing RNA-mediated methods in clinical applications.
Collapse
Affiliation(s)
- Yao-Zhong Ding
- State Key Laboratory of Veterinary Etiological Biology, National Foot-and-Mouth Disease Reference Laboratory, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, 730046, Gansu, China. .,Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonosis, Yangzhou, 225009, Jiangsu, China.
| | - Jan-Liang Lv
- State Key Laboratory of Veterinary Etiological Biology, National Foot-and-Mouth Disease Reference Laboratory, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, 730046, Gansu, China.,Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonosis, Yangzhou, 225009, Jiangsu, China
| | - Zhong-Wang Zhang
- State Key Laboratory of Veterinary Etiological Biology, National Foot-and-Mouth Disease Reference Laboratory, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, 730046, Gansu, China.,Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonosis, Yangzhou, 225009, Jiangsu, China
| | - Xiao-Yuan Ma
- State Key Laboratory of Veterinary Etiological Biology, National Foot-and-Mouth Disease Reference Laboratory, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, 730046, Gansu, China.,Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonosis, Yangzhou, 225009, Jiangsu, China
| | - Jie Zhang
- State Key Laboratory of Veterinary Etiological Biology, National Foot-and-Mouth Disease Reference Laboratory, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, 730046, Gansu, China.,Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonosis, Yangzhou, 225009, Jiangsu, China
| | - Yong-Guang Zhang
- State Key Laboratory of Veterinary Etiological Biology, National Foot-and-Mouth Disease Reference Laboratory, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, 730046, Gansu, China. .,Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonosis, Yangzhou, 225009, Jiangsu, China.
| |
Collapse
|
26
|
Wang X, Wu CX, Song XR, Chen HC, Liu ZF. Comparison of pseudorabies virus China reference strain with emerging variants reveals independent virus evolution within specific geographic regions. Virology 2017; 506:92-98. [DOI: 10.1016/j.virol.2017.03.013] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2017] [Revised: 03/23/2017] [Accepted: 03/24/2017] [Indexed: 01/01/2023]
|
27
|
Assessment of the efficacy of two novel DNA vaccine formulations against highly pathogenic Porcine Reproductive and Respiratory Syndrome Virus. Sci Rep 2017; 7:41886. [PMID: 28157199 PMCID: PMC5291100 DOI: 10.1038/srep41886] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2016] [Accepted: 01/03/2017] [Indexed: 01/08/2023] Open
Abstract
Since May 2006, a highly pathogenic porcine reproductive and respiratory syndrome virus (HP-PRRSV) has emerged and prevailed in mainland China, affecting over 2 million pigs. Commercial PRRSV killed and modified live vaccines cannot provide complete protection against HP-PRRSV due to genetic variation. Development of more effective vaccines against the emerging HP-PRRSV is urgently required. In our previous studies, two formulations of DNA vaccines (pcDNA3.1-PoIFN-λ1-SynORF5 and BPEI/PLGA-SynORF5) based on the HP-PRRSV were constructed and shown to induce enhanced humoral and cellular immune responses in mice. The objective of this study was to evaluate the immune response induced by these novel formulations in piglets. PcDNA3.1-PoIFN-λ1-SynORF5 and BPEI/PLGA-SynORF5 vaccines induced significantly enhanced GP5-specific antibody and PRRSV-specific neutralizing antibody in pigs compared with the pcDNA3.1-SynORF5 parental construct. Though IFN-γ levels and lymphocyte proliferation responses induced by the two DNA vaccine formulations were comparable to that induced by the pcDNA3.1-SynORF5 construct, each of the novel formulations provided efficient protection against challenge with HP-PRRSV. Non-severe clinical signs and rectal temperatures were observed in pigs immunized with BPEI/PLGA-SynORF5 compared with other groups. Thus, these novel DNA constructs may represent promising candidate vaccines against emerging HP-PRRSV.
Collapse
|
28
|
Du L, Li B, Pang F, Yu Z, Xu X, Fan B, Tan Y, He K, Huang K. Porcine GPX1 enhances GP5-based DNA vaccination against porcine reproductive and respiratory syndrome virus. Vet Immunol Immunopathol 2016; 183:31-39. [PMID: 28063474 DOI: 10.1016/j.vetimm.2016.12.001] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2016] [Revised: 11/12/2016] [Accepted: 12/03/2016] [Indexed: 12/15/2022]
Abstract
Porcine reproductive and respiratory syndrome virus (PRRSV) has been causing huge economic losses to the swine industry worldwide. Commercial PRRSV vaccines including killed and modified live ones are available. However the protective efficacy of these vaccines is incomplete. Thus, it is in urgent need to develop safer and more effective PRRSV vaccines. In this study, we constructed a recombinant plasmid co-expressing porcine glutathione peroxidase-1 (GPX1) and the envelope glycoprotein (GP5) encoding-gene of PRRSV (pcDNA3.1-GPX1-LSynORF5), and investigated the immune response induced following inoculation of mice and piglets. Significantly enhanced GP5-specific antibody, PRRSV-specific neutralizing antibody, IFN-γ level, as well as lymphocyte proliferation response, were induced in mice and pigs immunized with the DNA construct encoding GPX1 and GP5 compared with those inoculated with a construct encoding PRRSV GP5 only (pcDNA3.1-SynORF5). The enhanced cellular immune response in pigs induced by pcDNA3.1-GPX1-LSynORF5 was comparable to that induced by the attenuated virus vaccine JXA1-R, although the humoral immune response induced by the plasmid was much lower than the response induced by JXA1-R. Following the challenge with highly pathogenic PRRSV, less-severe clinical signs and rectal temperatures were observed in pigs immunized with the GPX1-GP5 construct compared with the control group. However, the viraemia of groups immunized with plasmid was more severe than that inoculated with JXA1-R, and it is likely that this could be attributed to the poor humoral response induced by the GPX1-GP5 construct. These results demonstrated that inclusion of GPX1 in a PRRSV DNA vaccine leads to an adjuvant effect, enhancing the humoral and cellular responses following vaccination.
Collapse
Affiliation(s)
- Luping Du
- College of Veterinary Medicine, Nanjing Agricultural University, No. 1 Wei-gang, Nanjing 210095, China; Institute of Veterinary Medicine, Jiangsu Academy of Agricultural Sciences, Key Laboratory of Veterinary Biological Engineering and Technology Ministry of Agriculture, Nanjing, 210014 Jiangsu Province, China; Jiangsu Co-infection Center for Prevention and Control of Important Animal Infectious Disease and Zoonoses, Yangzhou, 225009 Jiangsu Province, China
| | - Bin Li
- Institute of Veterinary Medicine, Jiangsu Academy of Agricultural Sciences, Key Laboratory of Veterinary Biological Engineering and Technology Ministry of Agriculture, Nanjing, 210014 Jiangsu Province, China; Jiangsu Co-infection Center for Prevention and Control of Important Animal Infectious Disease and Zoonoses, Yangzhou, 225009 Jiangsu Province, China
| | - Fengjiao Pang
- College of Veterinary Medicine, Nanjing Agricultural University, No. 1 Wei-gang, Nanjing 210095, China; Institute of Veterinary Medicine, Jiangsu Academy of Agricultural Sciences, Key Laboratory of Veterinary Biological Engineering and Technology Ministry of Agriculture, Nanjing, 210014 Jiangsu Province, China
| | - Zhengyu Yu
- Institute of Veterinary Medicine, Jiangsu Academy of Agricultural Sciences, Key Laboratory of Veterinary Biological Engineering and Technology Ministry of Agriculture, Nanjing, 210014 Jiangsu Province, China; Jiangsu Co-infection Center for Prevention and Control of Important Animal Infectious Disease and Zoonoses, Yangzhou, 225009 Jiangsu Province, China
| | - Xiangwei Xu
- College of Veterinary Medicine, Nanjing Agricultural University, No. 1 Wei-gang, Nanjing 210095, China; Institute of Veterinary Medicine, Jiangsu Academy of Agricultural Sciences, Key Laboratory of Veterinary Biological Engineering and Technology Ministry of Agriculture, Nanjing, 210014 Jiangsu Province, China
| | - Baochao Fan
- Institute of Veterinary Medicine, Jiangsu Academy of Agricultural Sciences, Key Laboratory of Veterinary Biological Engineering and Technology Ministry of Agriculture, Nanjing, 210014 Jiangsu Province, China; Jiangsu Co-infection Center for Prevention and Control of Important Animal Infectious Disease and Zoonoses, Yangzhou, 225009 Jiangsu Province, China
| | - Yeping Tan
- Institute of Veterinary Medicine, Jiangsu Academy of Agricultural Sciences, Key Laboratory of Veterinary Biological Engineering and Technology Ministry of Agriculture, Nanjing, 210014 Jiangsu Province, China; Jiangsu Co-infection Center for Prevention and Control of Important Animal Infectious Disease and Zoonoses, Yangzhou, 225009 Jiangsu Province, China
| | - Kongwang He
- Institute of Veterinary Medicine, Jiangsu Academy of Agricultural Sciences, Key Laboratory of Veterinary Biological Engineering and Technology Ministry of Agriculture, Nanjing, 210014 Jiangsu Province, China; Jiangsu Co-infection Center for Prevention and Control of Important Animal Infectious Disease and Zoonoses, Yangzhou, 225009 Jiangsu Province, China
| | - Kehe Huang
- College of Veterinary Medicine, Nanjing Agricultural University, No. 1 Wei-gang, Nanjing 210095, China.
| |
Collapse
|
29
|
Abstract
The mitochondrial anti-viral signaling protein (MAVS) plays an important role in the type I IFN response. In this study, two feline MAVS transcripts were cloned. Both transcripts have the same open reading frame encoding 523 amino acids. The putative protein shares 76.6 % similarity with canine and exhibits similarity to human, mouse, rat, bovine and porcine, ranging from 46.1 to 65.8 %. Deletion mutant analysis indicated that the transmembrane (TM) domain is necessary for localization in the mitochondrial membrane, and both the caspase activation and recruitment domain and TM domain are indispensible for activating the IFN-β response. Additionally, Sendai virus-induced IFN-β promoter activation was significantly inhibited by siRNA targeting MAVS. Finally, miniMAVS, a second protein encoded by MAVS mRNA, was identified, which interfered with the IFN-β response via the inhibition of NF-κB activation. The identification of MAVS will promote the understanding and control of feline infectious diseases.
Collapse
|
30
|
Lei JL, Xia SL, Wang Y, Du M, Xiang GT, Cong X, Luo Y, Li LF, Zhang L, Yu J, Hu Y, Qiu HJ, Sun Y. Safety and immunogenicity of a gE/gI/TK gene-deleted pseudorabies virus variant expressing the E2 protein of classical swine fever virus in pigs. Immunol Lett 2016; 174:63-71. [PMID: 27113530 DOI: 10.1016/j.imlet.2016.04.014] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2016] [Revised: 04/08/2016] [Accepted: 04/21/2016] [Indexed: 10/21/2022]
Abstract
Classical swine fever (CSF) and pseudorabies (PR) are both major infectious diseases of pigs, causing enormous economic losses to the swine industry in many countries. A marker vaccine that enables differentiation of infected from vaccinated animals (DIVA) is highly desirable for control and eradication of these two diseases in endemic areas. Since late 2011, PR outbreaks have been frequently reported in many Bartha-K61-vaccinated pig farms in China. It has been demonstrated that a pseudorabies virus (PRV) variant with altered antigenicity and increased pathogenicity was responsible for the outbreaks. Previously, we showed that rPRVTJ-delgE/gI/TK, a gE/gI/TK-deleted PRV variant, was safe for susceptible animals and provided a complete protection against lethal PRV variant challenge, indicating that rPRVTJ-delgE/gI/TK can be used as an attractive vaccine vector. To develop a safe bivalent vaccine against CSF and PR, we generated a recombinant virus rPRVTJ-delgE/gI/TK-E2 expressing the E2 protein of classical swine fever virus (CSFV) based on rPRVTJ-delgE/gI/TK and evaluated its safety and immunogenicity in pigs. The results indicated that pigs (n=5) immunized with rPRVTJ-delgE/gI/TK-E2 of different doses did not exhibit clinical signs or viral shedding following immunization, the immunized pigs produced anti-PRV or anti-CSFV neutralizing antibodies and the pigs immunized with 10(6) or 10(5) TCID50 rPRVTJ-delgE/gI/TK-E2 were completely protected against the lethal challenge with either CSFV Shimen strain or variant PRV TJ strain. These findings suggest that rPRVTJ-delgE/gI/TK-E2 is a promising bivalent DIVA vaccine candidate against CSFV and PRV coinfections.
Collapse
Affiliation(s)
- Jian-Lin Lei
- State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin 150001, China; College of Veterinary Medicine, Gansu Agricultural University, Lanzhou 730070, China
| | - Shui-Li Xia
- State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin 150001, China
| | - Yimin Wang
- State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin 150001, China
| | - Mingliang Du
- State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin 150001, China
| | - Guang-Tao Xiang
- State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin 150001, China
| | - Xin Cong
- State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin 150001, China
| | - Yuzi Luo
- State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin 150001, China
| | - Lian-Feng Li
- State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin 150001, China
| | - Lingkai Zhang
- State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin 150001, China
| | - Jiahui Yu
- State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin 150001, China
| | - Yonghao Hu
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou 730070, China.
| | - Hua-Ji Qiu
- State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin 150001, China.
| | - Yuan Sun
- State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin 150001, China.
| |
Collapse
|
31
|
Influence of the amino acid residues at 70 in M protein of porcine reproductive and respiratory syndrome virus on viral neutralization susceptibility to the serum antibody. Virol J 2016; 13:51. [PMID: 27004554 PMCID: PMC4802621 DOI: 10.1186/s12985-016-0505-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2016] [Accepted: 03/14/2016] [Indexed: 01/11/2023] Open
Abstract
Background Porcine reproductive and respiratory syndrome virus (PRRSV) is mainly responsible for the significant economic losses in pig industry in the world. The adaptive immune responses of the host act as an important source of selective pressure in the evolutionary process of the virus. In the previous study, we confirmed that the amino acid (aa) residues at 102 and 104 sites in GP5 played an important role in escaping from the neutralizing antibodies (NAbs) against highly pathogenic PRRSV (HP-PRRSV). In this study, we further analyzed the aa mutants affecting neutralization susceptibility of NAbs in other structure proteins in NAbs resistant variants. Methods Based on the different aa residues of the structural proteins between the resistant virus BB20s and the parent virus BB, 12 recombinant PRRSV strains containing these aa residue substitutions were constructed using reverse genetic techniques. The neutralizing antibody (NA) titers of the recombinant strains were tested on MARC-145 and porcine alveolar macrophages (PAMs). And the NAbs binding abilities of parent and rescued viruses were tested by using ELISA method. Results By using the neutralization assay, it was revealed that the NA titer of N4 serum with rBB/Ms was significantly lower than that with rBB. Meanwhile, NA titer of the serum with rBB20s/M was significantly higher than that with rBB20s. The ELISA binding results showed that rBB/Ms had higher binding inability to N4 than did rBB. And alignment of M protein revealed that the variant aa residue lysine (K) at 70 was also existed in field type 2 and vaccine PRRSV strains. Conclusions The aa residue at 70 in M protein of PRRSV played an important role in regulating neutralization susceptibility to the porcine serum NAbs. It may be helpful for monitoring the antigen variant strains in the field and developing new vaccine against PRRSV in the future. Electronic supplementary material The online version of this article (doi:10.1186/s12985-016-0505-7) contains supplementary material, which is available to authorized users.
Collapse
|
32
|
Eck M, Durán MG, Ricklin ME, Locher S, Sarraseca J, Rodríguez MJ, McCullough KC, Summerfield A, Zimmer G, Ruggli N. Virus replicon particles expressing porcine reproductive and respiratory syndrome virus proteins elicit immune priming but do not confer protection from viremia in pigs. Vet Res 2016; 47:33. [PMID: 26895704 PMCID: PMC4761149 DOI: 10.1186/s13567-016-0318-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2015] [Accepted: 01/29/2016] [Indexed: 01/17/2023] Open
Abstract
Porcine reproductive and respiratory syndrome virus (PRRSV) is the causative agent of one of the most devastating and economically significant viral disease of pigs worldwide. The vaccines currently available on the market elicit only limited protection. Recombinant vesicular stomatitis virus (VSV) replicon particles (VRP) have been used successfully to induce protection against influenza A virus (IAV) in chickens and bluetongue virus in sheep. In this study, VSV VRP expressing the PRRSV envelope proteins GP5, M, GP4, GP3, GP2 and the nucleocapsid protein N, individually or in combination, were generated and evaluated as a potential vector vaccine against PRRSV infection. High level expression of the recombinant PRRSV proteins was demonstrated in cell culture. However, none of the PRRSV antigens expressed from VRP, with the exception of the N protein, did induce any detectable antibody response in pigs before challenge infection with PRRSV. After challenge however, the antibody responses against GP5, GP4 and GP3 appeared in average 2 weeks earlier than in pigs vaccinated with the empty control VRP. No reduction of viremia was observed in the vaccinated group compared with the control group. When pigs were co-vaccinated with VRP expressing IAV antigens and VRP expressing PRRSV glycoproteins, only antibody responses to the IAV antigens were detectable. These data show that the VSV replicon vector can induce immune responses to heterologous proteins in pigs, but that the PRRSV envelope proteins expressed from VSV VRP are poorly immunogenic. Nevertheless, they prime the immune system for significantly earlier B-cell responses following PRRSV challenge infection.
Collapse
Affiliation(s)
- Melanie Eck
- Institute of Virology and Immunology IVI, Sensemattstrasse 293, 3147, Mittelhäusern, Switzerland. .,Graduate School for Cellular and Biomedical Sciences, University of Bern, 3012, Bern, Switzerland.
| | - Margarita García Durán
- Inmunología y Genética aplicada, S.A. (INGENASA), Calle de Los Hermanos García Noblejas 39, 28037, Madrid, Spain.
| | - Meret E Ricklin
- Institute of Virology and Immunology IVI, Sensemattstrasse 293, 3147, Mittelhäusern, Switzerland.
| | - Samira Locher
- Institute of Virology and Immunology IVI, Sensemattstrasse 293, 3147, Mittelhäusern, Switzerland.
| | - Javier Sarraseca
- Inmunología y Genética aplicada, S.A. (INGENASA), Calle de Los Hermanos García Noblejas 39, 28037, Madrid, Spain.
| | - María José Rodríguez
- Inmunología y Genética aplicada, S.A. (INGENASA), Calle de Los Hermanos García Noblejas 39, 28037, Madrid, Spain.
| | - Kenneth C McCullough
- Institute of Virology and Immunology IVI, Sensemattstrasse 293, 3147, Mittelhäusern, Switzerland.
| | - Artur Summerfield
- Institute of Virology and Immunology IVI, Sensemattstrasse 293, 3147, Mittelhäusern, Switzerland. .,Department of Infectious Disease and Pathobiology, Vetsuisse Faculty, University of Bern, Länggassstrasse 122, 3001, Bern, Switzerland.
| | - Gert Zimmer
- Institute of Virology and Immunology IVI, Sensemattstrasse 293, 3147, Mittelhäusern, Switzerland.
| | - Nicolas Ruggli
- Institute of Virology and Immunology IVI, Sensemattstrasse 293, 3147, Mittelhäusern, Switzerland.
| |
Collapse
|
33
|
Abstract
In approaching the development of a veterinary vaccine, researchers must choose from a bewildering array of options that can be combined to enhance benefit. The choice and combination of options is not just driven by efficacy, but also consideration of the cost, practicality, and challenges faced in licensing the product. In this review we set out the different choices faced by veterinary vaccine developers, highlight some issues, and propose some pressing needs to be addressed.
Collapse
Affiliation(s)
- Mark A Chambers
- Department of Pathology and Infectious Diseases, School of Veterinary Medicine, University of Surrey, Guildford, Surrey, GU2 7AL, UK.
- Department of Bacteriology, Animal and Plant Health Agency, Addlestone, Surrey, KT15 3NB, UK.
| | - Simon P Graham
- Department of Pathology and Infectious Diseases, School of Veterinary Medicine, University of Surrey, Guildford, Surrey, GU2 7AL, UK
- The Pirbright Institute, Ash Road, Pirbright, Woking, GU24 0NF, UK
| | - Roberto M La Ragione
- Department of Pathology and Infectious Diseases, School of Veterinary Medicine, University of Surrey, Guildford, Surrey, GU2 7AL, UK
| |
Collapse
|
34
|
Inhibition of highly pathogenic porcine reproductive and respiratory syndrome virus replication by recombinant pseudorabies virus-mediated RNA interference in piglets. Vet Microbiol 2015; 181:212-20. [DOI: 10.1016/j.vetmic.2015.10.016] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2014] [Revised: 08/01/2015] [Accepted: 10/13/2015] [Indexed: 11/20/2022]
|
35
|
Fan B, Liu X, Bai J, Li Y, Zhang Q, Jiang P. The 15N and 46R Residues of Highly Pathogenic Porcine Reproductive and Respiratory Syndrome Virus Nucleocapsid Protein Enhance Regulatory T Lymphocytes Proliferation. PLoS One 2015; 10:e0138772. [PMID: 26397116 PMCID: PMC4580451 DOI: 10.1371/journal.pone.0138772] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2015] [Accepted: 08/09/2015] [Indexed: 12/14/2022] Open
Abstract
Porcine reproductive and respiratory syndrome virus (PRRSV) negatively modulates host immune responses, resulting in persistent infection and immunosuppression. PRRSV infection increases the number of PRRSV-specific regulatory T lymphocytes (Tregs) in infected pigs. However, the target antigens for Tregs proliferation in PRRSV infection have not been fully understood. In this study, we demonstrated that the highly pathogenic PRRSV (HP-PRRSV) induced more CD4+CD25+Foxp3+ Tregs than classical PRRSV (C-PRRSV) strain. Of the recombinant GP5, M and N proteins of HP-PRRSV expressed in baculovirus expression systems, only N protein induced Tregs proliferation. The Tregs assays showed that three amino-acid regions, 15–21, 42–48 and 88–94, in N protein played an important role in induction of Tregs proliferation with synthetic peptides covering the whole length of N protein. By using reverse genetic methods, it was firstly found that the 15N and 46R residues in PRRSV N protein were critical for induction of Tregs proliferation. The phenotype of induced Tregs closely resembled that of transforming-growth-factor-β-secreting T helper 3 Tregs in swine. These data should be useful for understanding the mechanism of immunity to PRRSV and development of infection control strategies in the future.
Collapse
Affiliation(s)
- Baochao Fan
- Key Laboratory of Animal Diseases Diagnostic and Immunology, Ministry of Agriculture, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, China
| | - Xing Liu
- Key Laboratory of Animal Diseases Diagnostic and Immunology, Ministry of Agriculture, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, China
| | - Juan Bai
- Key Laboratory of Animal Diseases Diagnostic and Immunology, Ministry of Agriculture, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, China
| | - Yufeng Li
- Key Laboratory of Animal Diseases Diagnostic and Immunology, Ministry of Agriculture, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, China
| | - Qiaoya Zhang
- Key Laboratory of Animal Diseases Diagnostic and Immunology, Ministry of Agriculture, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, China
| | - Ping Jiang
- Key Laboratory of Animal Diseases Diagnostic and Immunology, Ministry of Agriculture, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, China
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, China
- * E-mail:
| |
Collapse
|
36
|
Generation and Efficacy Evaluation of a Recombinant Pseudorabies Virus Variant Expressing the E2 Protein of Classical Swine Fever Virus in Pigs. CLINICAL AND VACCINE IMMUNOLOGY : CVI 2015; 22:1121-9. [PMID: 26311244 DOI: 10.1128/cvi.00383-15] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/30/2015] [Accepted: 08/19/2015] [Indexed: 01/29/2023]
Abstract
Classical swine fever (CSF) is an economically important infectious disease of pigs caused by classical swine fever virus (CSFV). Pseudorabies (PR), which is caused by pseudorabies virus (PRV), is another important infectious disease of pigs and other animals. Coinfections of pigs with PRV and CSFV occur occasionally in the field. The modified live vaccine Bartha-K61 strain has played an important role in the control of PR in many countries, including China. Since late 2011, however, increasing PR outbreaks caused by an emerging PRV variant have been reported in Bartha-K61-vaccinated swine populations on many farms in China. Previously, we generated a gE/gI-deleted PRV (rPRVTJ-delgE) based on this PRV variant, which was shown to be safe and can provide rapid and complete protection against lethal challenge with the PRV variant in pigs. Here, we generated a new recombinant PRV variant expressing the E2 gene of CSFV (rPRVTJ-delgE/gI-E2) and evaluated its immunogenicity and efficacy in pigs. The results showed that rPRVTJ-delgE/gI-E2 was safe for pigs, induced detectable anti-PRV and anti-CSFV neutralizing antibodies, and provided complete protection against the lethal challenge with either the PRV TJ strain or the CSFV Shimen strain. The data indicate that rPRVTJ-delgE/gI-E2 is a promising candidate bivalent vaccine against PRV and CSFV coinfections.
Collapse
|
37
|
Renukaradhya GJ, Meng XJ, Calvert JG, Roof M, Lager KM. Live porcine reproductive and respiratory syndrome virus vaccines: Current status and future direction. Vaccine 2015; 33:4069-80. [PMID: 26148878 DOI: 10.1016/j.vaccine.2015.06.092] [Citation(s) in RCA: 130] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2014] [Revised: 06/16/2015] [Accepted: 06/23/2015] [Indexed: 10/23/2022]
Abstract
Porcine reproductive and respiratory syndrome (PRRS) caused by PRRS virus (PRRSV) was reported in the late 1980s. PRRS still is a huge economic concern to the global pig industry with a current annual loss estimated at one billion US dollars in North America alone. It has been 20 years since the first modified live-attenuated PRRSV vaccine (PRRSV-MLV) became commercially available. PRRSV-MLVs provide homologous protection and help in reducing shedding of heterologous viruses, but they do not completely protect pigs against heterologous field strains. There have been many advances in understanding the biology and ecology of PRRSV; however, the complexities of virus-host interaction and PRRSV vaccinology are not yet completely understood leaving a significant gap for improving breadth of immunity against diverse PRRS isolates. This review provides insights on immunization efforts using infectious PRRSV-based vaccines since the 1990s, beginning with live PRRSV immunization, development and commercialization of PRRSV-MLV, and strategies to overcome the deficiencies of PRRSV-MLV through use of replicating viral vectors expressing multiple PRRSV membrane proteins. Finally, powerful reverse genetics systems (infectious cDNA clones) generated from more than 20 PRRSV isolates of both genotypes 1 and 2 viruses have provided a great resource for exploring many innovative strategies to improve the safety and cross-protective efficacy of live PRRSV vaccines. Examples include vaccines with diminished ability to down-regulate the immune system, positive and negative marker vaccines, multivalent vaccines incorporating antigens from other porcine pathogens, vaccines that carry their own cytokine adjuvants, and chimeric vaccine viruses with the potential for broad cross-protection against heterologous strains. To combat this devastating pig disease in the future, evaluation and commercialization of such improved live PRRSV vaccines is a shared goal among PRRSV researchers, pork producers and biologics companies.
Collapse
Affiliation(s)
- Gourapura J Renukaradhya
- Food Animal Health Research Program, Ohio Agricultural Research and Development Center, Department of Veterinary Preventive Medicine, The Ohio State University, Wooster, OH, United States.
| | - Xiang-Jin Meng
- Department of Biomedical Sciences and Pathobiology, College of Veterinary Medicine, Virginia Polytechnic Institute and State University, Blacksburg, VA, United States
| | | | - Michael Roof
- Boehringer Ingelheim Vetmedica, Inc., Ames, IA, United States
| | - Kelly M Lager
- Virus and Prion Research Unit, National Animal Disease Center, U.S. Department of Agriculture, Ames, IA, United States.
| |
Collapse
|
38
|
Renukaradhya GJ, Meng XJ, Calvert JG, Roof M, Lager KM. Inactivated and subunit vaccines against porcine reproductive and respiratory syndrome: Current status and future direction. Vaccine 2015; 33:3065-72. [PMID: 25980425 DOI: 10.1016/j.vaccine.2015.04.102] [Citation(s) in RCA: 75] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2015] [Revised: 04/18/2015] [Accepted: 04/30/2015] [Indexed: 02/07/2023]
Abstract
Within a few years of its emergence in the late 1980s, the PRRS virus had spread globally to become the foremost infectious disease concern for the pork industry. Since 1994, modified live-attenuated vaccines against porcine reproductive and respiratory syndrome virus (PRRSV-MLV) have been widely used, but have failed to provide complete protection against emerging and heterologous field strains of the virus. Moreover, like many other MLVs, PRRSV-MLVs have safety concerns including vertical and horizontal transmission of the vaccine virus and several documented incidences of reversion to virulence. Thus, the development of efficacious inactivated vaccines is warranted for the control and eradication of PRRS. Since the early 1990s, researchers have been attempting to develop inactivated PRRSV vaccines, but most of the candidates have failed to elicit protective immunity even against homologous virus challenge. Recent research findings relating to both inactivated and subunit candidate PRRSV vaccines have shown promise, but they need to be pursued further to improve their heterologous efficacy and cost-effectiveness before considering commercialization. In this comprehensive review, we provide information on attempts to develop PRRSV inactivated and subunit vaccines. These includes various virus inactivation strategies, adjuvants, nanoparticle-based vaccine delivery systems, DNA vaccines, and recombinant subunit vaccines produced using baculovirus, plant, and replication-deficient viruses as vector vaccines. Finally, future directions for the development of innovative non-infectious PRRSV vaccines are suggested. Undoubtedly there remains a need for novel PRRSV vaccine strategies targeted to deliver cross-protective, non-infectious vaccines for the control and eradication of PRRS.
Collapse
Affiliation(s)
- Gourapura J Renukaradhya
- Food Animal Health Research Program, Ohio Agricultural Research and Development Center, Department of Veterinary Preventive Medicine, The Ohio State University, Wooster, OH, United States.
| | - Xiang-Jin Meng
- Department of Biomedical Sciences and Pathobiology, College of Veterinary Medicine, Virginia Polytechnic Institute and State University, Blacksburg, VA, United States
| | | | - Michael Roof
- Boehringer Ingelheim Vetmedica, Inc., Ames, IA, United States
| | - Kelly M Lager
- Virology Swine Research Unit, National Animal Disease Center, U.S. Department of Agriculture, Ames, IA, United States.
| |
Collapse
|
39
|
Roques E, Lessard M, Archambault D. The Cholera Toxin B Subunit (CTB) Fused to the Porcine Arterivirus Matrix M and GP5 Envelope Proteins Fails to Enhance the GP5-Specific Antibody Response in Pigs Immunized with Adenovectors. Mol Biotechnol 2015; 57:701-8. [PMID: 25801418 DOI: 10.1007/s12033-015-9861-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
The porcine reproductive and respiratory syndrome virus (PRRSV) is an arterivirus of the Arteriviridae family. As the current commercial vaccines are incompletely protective effective against PRRSV infection, we developed a vaccine strategy using replicating but non-disseminating adenovectors (rAdVs) expressing the PRRSV M matrix protein in fusion with the neutralizing major epitope-carrying GP5 envelope protein (Roques et al. in Vet Res 44:17, 2013). Although production of GP5-specific antibodies (Abs) was observed, no PRRSV-specific neutralizing Abs (NAbs) were induced in pigs given the rAdVs expressing M-GP5 or M-GP5m (GP5m being a mutant form of GP5). Nevertheless, partial protection was observed in the M-GP5m-rAdV-inoculated pigs experimentally infected with PRRSV. Here, we determined the impact of the cholera toxin B subunit (CTB, known for its adjuvant effect) in fusion with the C-terminus of M-GP5m on the Ab response to PRRSV. Three-week-old pigs were immunized twice both intramuscularly and intranasally at 3-week intervals with rAdV-expressing the green fluorescent protein (rAdV-GFP), rAdV-M-GP5m, or rAdV-M-GP5m-CTB. Pigs immunized with rAdV-M-GP5m showed a high level of serum GP5-specific Abs (as determined by an indirect ELISA). In contrast, CTB in fusion with M-GP5m had an unexpected severe negative impact on GP5-specific Ab production. PRRSV-specific NAbs could not be detected in any pigs of all groups.
Collapse
Affiliation(s)
- Elodie Roques
- Department of Biological Sciences, University of Québec at Montréal, Succursale Centre-Ville, P.O. Box 8888, Montreal, QC, H3C 3P8, Canada
| | | | | |
Collapse
|
40
|
Protection of pigs against pandemic swine origin H1N1 influenza A virus infection by hemagglutinin- or neuraminidase-expressing attenuated pseudorabies virus recombinants. Virus Res 2015; 199:20-30. [PMID: 25599604 DOI: 10.1016/j.virusres.2015.01.009] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2014] [Revised: 12/18/2014] [Accepted: 01/10/2015] [Indexed: 11/20/2022]
Abstract
Influenza is an important respiratory disease of pigs, and may lead to novel human pathogens like the 2009 pandemic H1N1 swine-origin influenza virus (SoIV). Therefore, improved influenza vaccines for pigs are required. Recently, we demonstrated that single intranasal immunization with a hemagglutinin (HA)-expressing pseudorabies virus recombinant of vaccine strain Bartha (PrV-Ba) protected pigs from H1N1 SoIV challenge (Klingbeil et al., 2014). Now we investigated enhancement of efficacy by prime-boost vaccination and/or intramuscular administration. Furthermore, a novel PrV-Ba recombinant expressing codon-optimized N1 neuraminidase (NA) was included. In vitro replication of this virus was only slightly affected compared to parental virus. Unlike HA, the abundantly expressed NA was efficiently incorporated into PrV particles. Immunization of pigs with the two PrV recombinants, either singly or in combination, induced B cell proliferation and the expected SoIV-specific antibodies, whose titers increased substantially after boost vaccination. After immunization of animals with either PrV recombinant H1N1 SoIV challenge virus replication was significantly reduced compared to PrV-Ba vaccinated or naïve controls. Protective efficacy of HA-expressing PrV was higher than of NA-expressing PrV, and not significantly enhanced by combination. Despite higher serum antibody titers obtained after intramuscular immunization, transmission of challenge virus to naïve contact animals was only prevented after intranasal prime-boost vaccination with HA-expressing PrV-Ba.
Collapse
|
41
|
Zhu X, Wang D, Zhang H, Zhou Y, Luo R, Chen H, Xiao S, Fang L. Molecular cloning and functional characterization of porcine DEAD (Asp-Glu-Ala-Asp) box polypeptide 41 (DDX41). DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2014; 47:191-196. [PMID: 25086295 DOI: 10.1016/j.dci.2014.07.020] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/25/2014] [Revised: 07/26/2014] [Accepted: 07/26/2014] [Indexed: 06/03/2023]
Abstract
DEAD (Asp-Glu-Ala-Asp) box polypeptide 41 (DDX41), a member of the DEXDc helicase family, was recently identified as an intracellular DNA sensor in mouse myeloid dendritic cells. In this study, porcine DDX41 (poDDX41) was cloned and its role in the type I interferon (IFN) signaling pathway was investigated in porcine kidney (PK-15) cells. Full-length poDDX41 cDNA encodes 622 amino acid residues and contains a DEADc domain and a HELICc domain. poDDX41 mRNA is widely expressed in different tissues, especially the stomach and liver. Overexpression of poDDX41 in PK-15 cells induced IFN-β by activating transcription factors IRF3 and NF-κB. Knockdown of poDDX41 with siRNA significantly reduced IFN-β expression induced by poly(dA:dT), a double-stranded DNA (dsDNA) analogue, or pseudorabies virus, a dsDNA swine virus. Therefore, poDDX41 is involved in the dsDNA- and dsDNA-virus-mediated type I IFN signaling pathway in porcine kidney cells.
Collapse
Affiliation(s)
- Xinyu Zhu
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China
| | - Dang Wang
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China
| | - Huan Zhang
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China
| | - Yanrong Zhou
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China
| | - Rui Luo
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China
| | - Huanchun Chen
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China
| | - Shaobo Xiao
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China
| | - Liurong Fang
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China.
| |
Collapse
|
42
|
An overview of live attenuated recombinant pseudorabies viruses for use as novel vaccines. J Immunol Res 2014; 2014:824630. [PMID: 24995348 PMCID: PMC4068083 DOI: 10.1155/2014/824630] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2013] [Accepted: 03/23/2014] [Indexed: 11/18/2022] Open
Abstract
Pseudorabies virus (PRV) is a double-stranded, DNA-based swine virus with a genome approximating 150 kb in size. PRV has many nonessential genes which can be replaced with genes encoding heterologous antigens but without deleterious effects on virus propagation. Recombinant PRVs expressing both native and foreign antigens are able to stimulate immune responses. In this paper, we review the current status of live attenuated recombinant PRVs and live PRV-based vector vaccines with potential for controlling viral infections in animals.
Collapse
|
43
|
Du Y, Lu Y, Wang X, Qi J, Liu J, Hu Y, Li F, Wu J, Guo L, Liu J, Tao H, Sun W, Chen L, Cong X, Ren S, Shi J, Li J, Wang J, Huang B, Wan R. Highly efficient expression of interleukin-2 under the control of rabbit β-globin intron II gene enhances protective immune responses of porcine reproductive and respiratory syndrome (PRRS) DNA vaccine in pigs. PLoS One 2014; 9:e90326. [PMID: 24603502 PMCID: PMC3946010 DOI: 10.1371/journal.pone.0090326] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2013] [Accepted: 01/31/2014] [Indexed: 01/05/2023] Open
Abstract
Highly pathogenic porcine reproductive and respiratory syndrome virus (HP-PRRSV) had caused catastrophic losses in swine industry in China. The current inactivated vaccine provided only limited protection, and the attenuated live vaccine could protect piglets against the HP-PRRSV but there was a possibility that the attenuated virus returned to high virulence. In this study, the eukaryotic expression vector pVAX1© was modified under the control of rabbit β-globin intron II gene and the modified vector pMVAX1© was constructed. Porcine interleukin-2 (IL-2) and GP3-GP5 fusion protein of HP-PRRSV strain SD-JN were highly expressed by pMVAX1©. Mice inoculated with pMVAX1©-GP35 developed significantly higher PRRSV-specific antibody responses and T cell proliferation than those vaccinated with pVAX1©-GP35. pMVAX1©-GP35 was selected as PRRS DNA vaccine candidate and co-administrated with pVAX1©-IL-2 or pMVAX1©-IL-2 in pigs. pMVAX1©-IL-2+pMVAX1©-GP35 could provide enhanced PRRSV-specific antibody responses, T cell proliferation, Th1-type and Th2-type cytokine responses and CTL responses than pMVAX1©-GP35 and pVAX1©-IL-2+pMVAX1©-GP35. Following homologous challenge with HP-PRRSV strain SD-JN, similar with attenuated PRRS vaccine group, pigs inoculated with pMVAX1©-IL-2+pMVAX1©-GP35 showed no clinical signs, almost no lung lesions and no viremia, as compared to those in pMVAX1©-GP35 and pVAX1©-IL-2+pMVAX1©-GP35 groups. It indicated that pMVAX1©-IL-2 effectively increases humoral and cell mediated immune responses of pMVAX1©-GP35. Co-administration of pMVAX1©-IL-2 and pMVAX1©-GP35 might be attractive candidate vaccines for preventing HP-PRRSV infections.
Collapse
Affiliation(s)
- Yijun Du
- School of Life Sciences, Shandong University, Jinan, China
- Shandong Key Laboratory of Animal Disease Control and Breeding, Institute of Animal Science and Veterinary Medicine, Shandong Academy of Agricultural Sciences, Jinan, China
| | - Yu Lu
- National Research Center of Veterinary Biologicals Engineering and Technology, Jiangsu Academy of Agricultural Sciences, Nanjing, China
| | - Xinglong Wang
- College of Veterinary Medicine, Northwest A&F University, Yangling, China
| | - Jing Qi
- School of Life Sciences, Shandong University, Jinan, China
- Shandong Key Laboratory of Animal Disease Control and Breeding, Institute of Animal Science and Veterinary Medicine, Shandong Academy of Agricultural Sciences, Jinan, China
| | - Jiyu Liu
- Shandong Key Laboratory of Animal Disease Control and Breeding, Institute of Animal Science and Veterinary Medicine, Shandong Academy of Agricultural Sciences, Jinan, China
| | - Yue Hu
- College of Animal Science & Veterinary Medicine, Shandong Agricultural University, Tai'an, China
| | - Feng Li
- Department of Biology and Microbiology, Department of Veterinary and Biomedical Sciences, South Dakota State University, Brookings, South Dakota, United States of America
| | - Jiaqiang Wu
- Shandong Key Laboratory of Animal Disease Control and Breeding, Institute of Animal Science and Veterinary Medicine, Shandong Academy of Agricultural Sciences, Jinan, China
| | - Lihui Guo
- Shandong Key Laboratory of Animal Disease Control and Breeding, Institute of Animal Science and Veterinary Medicine, Shandong Academy of Agricultural Sciences, Jinan, China
| | - Junzhen Liu
- Shandong Key Laboratory of Animal Disease Control and Breeding, Institute of Animal Science and Veterinary Medicine, Shandong Academy of Agricultural Sciences, Jinan, China
| | - Haiying Tao
- Shandong Key Laboratory of Animal Disease Control and Breeding, Institute of Animal Science and Veterinary Medicine, Shandong Academy of Agricultural Sciences, Jinan, China
| | - Wenbo Sun
- Shandong Key Laboratory of Animal Disease Control and Breeding, Institute of Animal Science and Veterinary Medicine, Shandong Academy of Agricultural Sciences, Jinan, China
| | - Lei Chen
- School of Life Sciences, Shandong University, Jinan, China
- Shandong Key Laboratory of Animal Disease Control and Breeding, Institute of Animal Science and Veterinary Medicine, Shandong Academy of Agricultural Sciences, Jinan, China
| | - Xiaoyan Cong
- School of Life Sciences, Shandong University, Jinan, China
- Shandong Key Laboratory of Animal Disease Control and Breeding, Institute of Animal Science and Veterinary Medicine, Shandong Academy of Agricultural Sciences, Jinan, China
| | - Sufang Ren
- Shandong Key Laboratory of Animal Disease Control and Breeding, Institute of Animal Science and Veterinary Medicine, Shandong Academy of Agricultural Sciences, Jinan, China
| | - Jianli Shi
- Shandong Key Laboratory of Animal Disease Control and Breeding, Institute of Animal Science and Veterinary Medicine, Shandong Academy of Agricultural Sciences, Jinan, China
| | - Jun Li
- Shandong Key Laboratory of Animal Disease Control and Breeding, Institute of Animal Science and Veterinary Medicine, Shandong Academy of Agricultural Sciences, Jinan, China
| | - Jinbao Wang
- School of Life Sciences, Shandong University, Jinan, China
- Shandong Key Laboratory of Animal Disease Control and Breeding, Institute of Animal Science and Veterinary Medicine, Shandong Academy of Agricultural Sciences, Jinan, China
- College of Animal Science & Veterinary Medicine, Shandong Agricultural University, Tai'an, China
- * E-mail: (JW); (BH); (RW)
| | - Baohua Huang
- Shandong Key Laboratory of Animal Disease Control and Breeding, Institute of Animal Science and Veterinary Medicine, Shandong Academy of Agricultural Sciences, Jinan, China
- * E-mail: (JW); (BH); (RW)
| | - Renzhong Wan
- College of Animal Science & Veterinary Medicine, Shandong Agricultural University, Tai'an, China
- * E-mail: (JW); (BH); (RW)
| |
Collapse
|
44
|
Klingbeil K, Lange E, Teifke JP, Mettenleiter TC, Fuchs W. Immunization of pigs with an attenuated pseudorabies virus recombinant expressing the haemagglutinin of pandemic swine origin H1N1 influenza A virus. J Gen Virol 2014; 95:948-959. [PMID: 24431235 DOI: 10.1099/vir.0.059253-0] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Pigs can be severely harmed by influenza, and represent important reservoir hosts, in which new human pathogens such as the recent pandemic swine-origin H1N1 influenza A virus can arise by mutation and reassortment of genome segments. To obtain novel, safe influenza vaccines for pigs, and to investigate the antigen-specific immune response, we modified an established live-virus vaccine against Aujeszky's disease of swine, pseudorabies virus (PrV) strain Bartha (PrV-Ba), to serve as vector for the expression of haemagglutinin (HA) of swine-origin H1N1 virus. To facilitate transgene insertion, the genome of PrV-Ba was cloned as a bacterial artificial chromosome. HA expression occurred under control of the human or murine cytomegalovirus immediate early promoters (P-HCMV, P-MCMV), but could be substantially enhanced by synthetic introns and adaptation of the codon usage to that of PrV. However, despite abundant expression, the heterologous glycoprotein was not detectably incorporated into mature PrV particles. Replication of HA-expressing PrV in cell culture was only slightly affected compared to that of the parental virus strain. A single immunization of pigs with the PrV vector expressing the codon-optimized HA gene under control of P-MCMV induced high levels of HA-specific antibodies. The vaccinated animals were protected from clinical signs after challenge with a related swine-origin H1N1 influenza A virus, and challenge virus shedding was significantly reduced.
Collapse
Affiliation(s)
- Katharina Klingbeil
- Institute of Molecular Biology, Friedrich-Loeffler-Institut, Federal Research Institute for Animal Health, Greifswald-Insel Riems, Germany
| | - Elke Lange
- Department of Experimental Animal Facilities and Biorisk Management, Friedrich-Loeffler-Institut, Federal Research Institute for Animal Health, Greifswald-Insel Riems, Germany
| | - Jens P Teifke
- Department of Experimental Animal Facilities and Biorisk Management, Friedrich-Loeffler-Institut, Federal Research Institute for Animal Health, Greifswald-Insel Riems, Germany
| | - Thomas C Mettenleiter
- Institute of Molecular Biology, Friedrich-Loeffler-Institut, Federal Research Institute for Animal Health, Greifswald-Insel Riems, Germany
| | - Walter Fuchs
- Institute of Molecular Biology, Friedrich-Loeffler-Institut, Federal Research Institute for Animal Health, Greifswald-Insel Riems, Germany
| |
Collapse
|
45
|
Chen Q, Liu Q, Liu D, Wang D, Chen H, Xiao S, Fang L. Molecular cloning, functional characterization and antiviral activity of porcine DDX3X. Biochem Biophys Res Commun 2013; 443:1169-75. [PMID: 24380861 DOI: 10.1016/j.bbrc.2013.12.098] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2013] [Accepted: 12/19/2013] [Indexed: 01/18/2023]
Abstract
Human DDX3X is a newly discovered DEAD-box RNA helicase. In addition to involvement of eukaryotic gene expression regulation, human DDX3X has recently been demonstrated to be a critical molecule in innate immune signaling pathways and to contribute to type I interferon (IFN) induction. In the present study, porcine DDX3X was cloned by RT-PCR from PK-15 cells and its function in regulating IFN-β was characterized. The putative porcine DDX3X ORF encodes 662 amino acids possessing several conserved motifs. Sequence alignments indicated that porcine DDX3X has high identity at the amino acid level to those of horse (96.7%), mouse (97.6%), cattle (98.5%), dog (98.6%) and human (98.9%). Ectopic expression of porcine DDX3X significantly activated IFN-β expression, whereas knockdown of porcine DDX3X inhibited dsRNA- or Sendai virus (SeV)-induced IFN-β. Furthermore, porcine DDX3X co-localized with IPS-1, TBK1 and IKKε, and enhanced IFN-β promoter activation induced by these molecules. We also investigated the role of porcine DDX3X during porcine reproductive and respiratory syndrome virus (PRRSV) infection and found that overexpression of DDX3X significantly inhibited PRRSV replication, indicating that DDX3X is a potential antiviral agent.
Collapse
Affiliation(s)
- Quangang Chen
- Division of Animal Infectious Diseases, State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China
| | - Qin Liu
- Division of Animal Infectious Diseases, State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China
| | - Dezhi Liu
- Division of Animal Infectious Diseases, State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China
| | - Dang Wang
- Division of Animal Infectious Diseases, State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China
| | - Huanchun Chen
- Division of Animal Infectious Diseases, State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China
| | - Shaobo Xiao
- Division of Animal Infectious Diseases, State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China
| | - Liurong Fang
- Division of Animal Infectious Diseases, State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China.
| |
Collapse
|
46
|
Construction and immunogenicity of DNA vaccines encoding fusion protein of porcine IFN- λ 1 and GP5 gene of porcine reproductive and respiratory syndrome virus. BIOMED RESEARCH INTERNATIONAL 2013; 2013:318698. [PMID: 24490154 PMCID: PMC3884778 DOI: 10.1155/2013/318698] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/28/2013] [Accepted: 11/29/2013] [Indexed: 01/05/2023]
Abstract
Porcine reproductive and respiratory syndrome virus (PRRSV) has been mainly responsible for the catastrophic economic losses in pig industry worldwide. The commercial vaccines only provide a limited protection against PRRSV infection. Thus, the focus and direction is to develop safer and more effective vaccines in the research field of PRRS. The immune modulators are being considered to enhance the effectiveness of PRRSV vaccines. IFN-λ1 belongs to type III interferon, a new interferon family. IFN-λ1 is an important cytokine with multiple functions in innate and acquired immunity. In this study, porcine IFN-λ1 (PoIFN-λ1) was evaluated for its adjuvant effects on the immunity of a DNA vaccine carrying the GP5 gene of PRRSV. Groups of mice were immunized twice at 2-week interval with 100 μg of the plasmid DNA vaccine pcDNA3.1-SynORF5, pcDNA3.1-PoIFN-λ1-SynORF5, and the blank vector pcDNA3.1, respectively. The results showed that pcDNA3.1-PoIFN-λ1-SynORF5 can significantly enhance GP5-specific ELISA antibody, PRRSV-specific neutralizing antibody, IFN-γ level, and lymphocyte proliferation rather than the responses induced by pcDNA3.1-SynORF5. Therefore, type III interferon PoIFN-λ1 could enhance the immune responses of DNA vaccine of PRRSV, highlighting the potential value of PoIFN-λ1 as a molecular adjuvant in the prevention of PRRSV infection.
Collapse
|
47
|
Roques E, Girard A, St-Louis MC, Massie B, Gagnon CA, Lessard M, Archambault D. Immunogenic and protective properties of GP5 and M structural proteins of porcine reproductive and respiratory syndrome virus expressed from replicating but nondisseminating adenovectors. Vet Res 2013; 44:17. [PMID: 23497101 PMCID: PMC3608016 DOI: 10.1186/1297-9716-44-17] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2012] [Accepted: 02/13/2013] [Indexed: 02/08/2023] Open
Abstract
Porcine reproductive and respiratory syndrome virus (PRRSV) is responsible for significant economic losses in the porcine industry. Currently available commercial vaccines do not allow optimal and safe protection. In this study, replicating but nondisseminating adenovectors (rAdV) were used for the first time in pigs for vaccinal purposes. They were expressing the PRRSV matrix M protein in fusion with either the envelope GP5 wild-type protein (M-GP5) which carries the major neutralizing antibody (NAb)-inducing epitope or a mutant form of GP5 (M-GP5m) developed to theoretically increase the NAb immune response. Three groups of fourteen piglets were immunized both intramuscularly and intranasally at 3-week intervals with rAdV expressing the green fluorescent protein (GFP, used as a negative control), M-GP5 or M-GP5m. Two additional groups of pigs were primed with M-GP5m-expressing rAdV followed by a boost with bacterially-expressed recombinant wild-type GP5 or were immunized twice with a PRRSV inactivated commercial vaccine. The results show that the rAdV expressing the fusion proteins of interest induced systemic and mucosal PRRSV GP5-specific antibody response as determined in an ELISA. Moreover the prime with M-GP5m-expressing rAdV and boost with recombinant GP5 showed the highest antibody response against GP5. Following PRRSV experimental challenge, pigs immunized twice with rAdV expressing either M-GP5 or M-GP5m developed partial protection as shown by a decrease in viremia overtime. The lowest viremia levels and/or percentages of macroscopic lung lesions were obtained in pigs immunized twice with either the rAdV expressing M-GP5m or the PRRSV inactivated commercial vaccine.
Collapse
Affiliation(s)
- Elodie Roques
- Department of Biological Sciences, University of Québec at Montréal, Succursale Centre-Ville, P,O, Box 8888, Montréal, Québec, H3C 3P8, Canada.
| | | | | | | | | | | | | |
Collapse
|
48
|
Wang W, Chen X, Xue C, Du Y, Lv L, Liu Q, Li X, Ma Y, Shen H, Cao Y. Production and immunogenicity of chimeric virus-like particles containing porcine reproductive and respiratory syndrome virus GP5 protein. Vaccine 2012; 30:7072-7. [PMID: 23036496 DOI: 10.1016/j.vaccine.2012.09.048] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2012] [Revised: 09/13/2012] [Accepted: 09/20/2012] [Indexed: 01/28/2023]
Abstract
Porcine reproductive and respiratory syndrome virus (PRRSV) poses a severe threat in swine industry and causes heavy economic losses worldwide. Currently, the available vaccines are the inactivated and attenuated virus vaccines, but the use of PRRSV in their production raises the issue of safety. We developed a chimeric virus-like particles (VLPs) vaccine candidate for PRRSV protection. The chimeric VLPs was composed of M1 protein from H1N1 influenza virus and a fusion protein, denoted as NA/GP5, containing the cytoplasmic and transmembrane domains of H1N1 virus NA protein and PRRSV GP5 protein. Vaccination of BALB/c mice with 10 μg of chimeirc VLPs by intramuscular immunization stimulated antibody responses to GP5 protein, and induced cellular immune response. The data suggested that the chimeric VLP vaccine candidate may provide a new strategy for further development of vaccines against PRRSV infection.
Collapse
Affiliation(s)
- Wei Wang
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou 510006, PR China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Li J, Murtaugh MP. Dissociation of porcine reproductive and respiratory syndrome virus neutralization from antibodies specific to major envelope protein surface epitopes. Virology 2012; 433:367-76. [PMID: 22981434 DOI: 10.1016/j.virol.2012.08.026] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2012] [Revised: 06/09/2012] [Accepted: 08/17/2012] [Indexed: 12/19/2022]
Abstract
Glycoprotein 5 (GP5) and membrane (M) protein are the major proteins in the envelope of porcine reproductive and respiratory syndrome virus (PRRSV). Although viral neutralization epitopes are reported in GP5 and M of type 2 PRRSV, their significance as targets of porcine humoral immunity is not well described. Thus, we constructed recombinant polypeptides containing ectodomain neutralization epitopes to examine their involvement in porcine antibody neutralization and antiviral immunity. PRRSV infection elicited ectodomain-specific antibodies, whose titers did not correlate with the neutralizing antibody (NA) response. Ectodomain-specific antibodies from PRRSV-neutralizing serum bound virus but did not neutralize infectivity. Furthermore, immunization of pigs with ectodomain polypeptides raised specific antibodies and provided partial protection without a detectable NA response. Finally the polypeptides did not block infection of porcine macrophages. These results suggest that the GP5/M ectodomain peptide epitopes are accessible for host antibody recognition, but are not associated with antibody-mediated virus neutralization.
Collapse
Affiliation(s)
- Juan Li
- Department of Veterinary and Biomedical Sciences, University of Minnesota, St. Paul, MN 55108, USA
| | | |
Collapse
|
50
|
Du Y, Qi J, Lu Y, Wu J, Yoo D, Liu X, Zhang X, Li J, Sun W, Cong X, Shi J, Wang J. Evaluation of a DNA vaccine candidate co-expressing GP3 and GP5 of porcine reproductive and respiratory syndrome virus (PRRSV) with interferon α/γ in immediate and long-lasting protection against HP-PRRSV challenge. Virus Genes 2012; 45:474-87. [DOI: 10.1007/s11262-012-0790-1] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2012] [Accepted: 07/16/2012] [Indexed: 12/01/2022]
|