101
|
Hu G, Wang N, Yu W, Wang Z, Zou Y, Zhang Y, Wang A, Deng Z, Yang Y. Generation and immunogenicity of porcine circovirus type 2 chimeric virus-like particles displaying porcine reproductive and respiratory syndrome virus GP5 epitope B. Vaccine 2016; 34:1896-903. [DOI: 10.1016/j.vaccine.2016.02.047] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2015] [Revised: 02/15/2016] [Accepted: 02/17/2016] [Indexed: 10/22/2022]
|
102
|
Peng J, Yuan Y, Du Y, Wu J, Li B, Li J, Yu J, Hu L, Shen S, Wang J, Zhu R. Potentiation of Taishan Pinus massoniana pollen polysaccharide on the immune response and protection elicited by a highly pathogenic porcine reproductive and respiratory syndrome virus glycoprotein 5 subunit in pigs. Mol Cell Probes 2016; 30:83-92. [PMID: 26828953 DOI: 10.1016/j.mcp.2016.01.008] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2015] [Revised: 01/27/2016] [Accepted: 01/27/2016] [Indexed: 01/26/2023]
Affiliation(s)
- Jun Peng
- College of Veterinary Medicine, Shandong Agricultural University, Taian, China; Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, Taian, China
| | - Yanmei Yuan
- College of Veterinary Medicine, Shandong Agricultural University, Taian, China; Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, Taian, China
| | - Yijun Du
- Shandong Key Laboratory of Animal Disease Control and Breeding, Jinan, China; Institute of Animal Science and Veterinary Medicine, Shandong Academy of Agricultural Sciences, Jinan, China
| | - Jiaqiang Wu
- Shandong Key Laboratory of Animal Disease Control and Breeding, Jinan, China; Institute of Animal Science and Veterinary Medicine, Shandong Academy of Agricultural Sciences, Jinan, China
| | - Baoquan Li
- College of Veterinary Medicine, Shandong Agricultural University, Taian, China; Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, Taian, China
| | - Jun Li
- Shandong Key Laboratory of Animal Disease Control and Breeding, Jinan, China; Institute of Animal Science and Veterinary Medicine, Shandong Academy of Agricultural Sciences, Jinan, China
| | - Jiang Yu
- Shandong Key Laboratory of Animal Disease Control and Breeding, Jinan, China; Institute of Animal Science and Veterinary Medicine, Shandong Academy of Agricultural Sciences, Jinan, China
| | - Liping Hu
- Shandong Center for Animal Disease Prevention and Control, Jinan, China
| | - Si Shen
- College of Veterinary Medicine, Shandong Agricultural University, Taian, China; Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, Taian, China
| | - Jinbao Wang
- Shandong Key Laboratory of Animal Disease Control and Breeding, Jinan, China.
| | - Ruiliang Zhu
- College of Veterinary Medicine, Shandong Agricultural University, Taian, China; Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, Taian, China.
| |
Collapse
|
103
|
Lee SC, Choi HW, Nam E, Noh YH, Lee S, Lee YJ, Park GS, Shin JH, Yoon IJ, Kang SY, Lee C. Pathogenicity and genetic characteristics associated with cell adaptation of a virulent porcine reproductive and respiratory syndrome virus nsp2 DEL strain CA-2. Vet Microbiol 2016; 186:174-88. [PMID: 27016772 DOI: 10.1016/j.vetmic.2016.03.002] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2016] [Revised: 03/03/2016] [Accepted: 03/03/2016] [Indexed: 02/06/2023]
Abstract
Porcine reproductive and respiratory syndrome virus (PRRSV) is the most common and world-widespread viral pathogen of swine. We previously reported genomic sequences and pathogenicity of type 2 Korean PRRSV strains belonging to the virulent lineage 1 family, which contain remarkable amino acid deletions in nonstructural protein 2 (nsp2 DEL) compared to VR-2332. Here, a virulent type 2 Korean PRRSV nsp2 DEL strain, CA-2, was serially propagated in MARC-145 cells for up to 100 passages (CA-2-P100). As the passage number increased, the phenotypic characteristics of cell-adapted CA-2 strains were altered, in terms of higher viral titers and larger plaque sizes compared to the parental virus. Pro-inflammatory cytokine genes, including TNF-α, IL-8, MCP-1, and MCP-2, were found to be significantly down-regulated in PAM cells with the CA-2-P100 strain compared to its parental nsp2 DEL virus. Animal inoculation studies demonstrated that the virulence of CA-2-P100 was reduced significantly, with showing normal weight gain, body temperatures, and lung lesions comparable to the control group. Furthermore, high-passage CA-2-P100 showed declined and transient viremia kinetics, as well as delayed and low PRRSV-specific antibody responses in infected pigs. In addition, we determined whole genome sequences of low to high-passage derivatives of CA-2. The nsp2 DEL pattern was conserved for 100 passages, whereas no other deletions or insertions arose during the cell adaptation process. However, CA-2-P100 possessed 54 random nucleotide substitutions that resulted in 27 amino acid changes distributed throughout the genome, suggesting that these genetic drifts provide a possible molecular basis correlated with the cell-adapted features in vitro and the attenuated phenotype in vivo. Taken together, our data indicate that the cell-attenuated CA-2-P100 strain is a promising candidate for developing a safe and effective live PRRSV vaccine.
Collapse
Affiliation(s)
- Seung-Chul Lee
- Choongang Vaccine Laboratory, Daejeon 34055, Republic of Korea; College of Veterinary Medicine, Chungbuk National University, Cheongju 28644, Republic of Korea
| | - Hwan-Won Choi
- Choongang Vaccine Laboratory, Daejeon 34055, Republic of Korea
| | - Eeuri Nam
- Choongang Vaccine Laboratory, Daejeon 34055, Republic of Korea
| | - Yun-Hee Noh
- Choongang Vaccine Laboratory, Daejeon 34055, Republic of Korea
| | - Sunhee Lee
- Animal Virology Laboratory, School of Life Sciences, BK21 Plus KNU Creative BioResearch Group, Kyungpook National University, Daegu 41566, Republic of Korea
| | - Yoo Jin Lee
- Animal Virology Laboratory, School of Life Sciences, BK21 Plus KNU Creative BioResearch Group, Kyungpook National University, Daegu 41566, Republic of Korea
| | - Gun-Seok Park
- School of Applied Biosciences, College of Agriculture and Life Sciences, Kyungpook National University, Daegu 41566, Republic of Korea
| | - Jae-Ho Shin
- School of Applied Biosciences, College of Agriculture and Life Sciences, Kyungpook National University, Daegu 41566, Republic of Korea
| | - In-Joong Yoon
- Choongang Vaccine Laboratory, Daejeon 34055, Republic of Korea
| | - Shien-Young Kang
- College of Veterinary Medicine, Chungbuk National University, Cheongju 28644, Republic of Korea.
| | - Changhee Lee
- Animal Virology Laboratory, School of Life Sciences, BK21 Plus KNU Creative BioResearch Group, Kyungpook National University, Daegu 41566, Republic of Korea.
| |
Collapse
|
104
|
Chen N, Trible BR, Kerrigan MA, Tian K, Rowland RRR. ORF5 of porcine reproductive and respiratory syndrome virus (PRRSV) is a target of diversifying selection as infection progresses from acute infection to virus rebound. INFECTION GENETICS AND EVOLUTION 2016; 40:167-175. [PMID: 26961593 DOI: 10.1016/j.meegid.2016.03.002] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/30/2016] [Revised: 02/28/2016] [Accepted: 03/02/2016] [Indexed: 02/05/2023]
Abstract
Genetic variation in both structural and nonstructural genes is a key factor in the capacity of porcine reproductive and respiratory syndrome virus (PRRSV) to evade host defenses and maintain within animals, farms and metapopulations. However, the exact mechanisms by which genetic variation contribute to immune evasion remain unclear. In a study to understand the role of host genetics in disease resistance, a population of pigs were experimentally infected with a type 2 PRRSV isolate. Four pigs that showed virus rebound at 42days post-infection (dpi) were analyzed by 454 sequencing to characterize the rebound quasispecies. Deep sequencing of variable regions in nsp1, nsp2, ORF3 and ORF5 showed the largest number of nucleotide substitutions at day 28 compared to days 4 and 42 post-infection. Differences were also found in genetic variations when comparing tonsil versus serum. The results of dN/dS ratios showed that the same regions evolved under negative selection. However, eight amino acid sites were identified as possessing significant levels of positive selection, including A27V and N32S substitutions in the GP5 ectodomain region. These changes may alter GP5 peptide signal sequence processing and N-glycosylation, respectively. The results indicate that the greatest genetic diversity occurs during the transition between acute and rebound stages of infection, and the introduction of mutations that may result in a gain of fitness provides a potential mechanism for persistence.
Collapse
Affiliation(s)
- Nanhua Chen
- Department of Diagnostic Medicine and Pathobiology, College of Veterinary Medicine, Kansas State University, Manhattan, KS 66506, United States; College of Veterinary Medicine, Yangzhou University, Yangzhou, Jiangsu 225009, PR China.
| | - Benjamin R Trible
- Department of Diagnostic Medicine and Pathobiology, College of Veterinary Medicine, Kansas State University, Manhattan, KS 66506, United States
| | - Maureen A Kerrigan
- Department of Diagnostic Medicine and Pathobiology, College of Veterinary Medicine, Kansas State University, Manhattan, KS 66506, United States
| | - Kegong Tian
- OIE Porcine Reproductive and Respiratory Syndrome Reference Laboratory, Beijing, PR China
| | - Raymond R R Rowland
- Department of Diagnostic Medicine and Pathobiology, College of Veterinary Medicine, Kansas State University, Manhattan, KS 66506, United States
| |
Collapse
|
105
|
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
|
106
|
Peng J, Yuan Y, Shen S, Niu Z, Du Y, Wu J, Li J, Yu J, Wang T, Wang J. Immunopotentiation of four natural adjuvants co-administered with a highly pathogenic porcine reproductive and respiratory syndrome virus glycoprotein 5 subunit. Virus Genes 2016; 52:261-9. [DOI: 10.1007/s11262-016-1299-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2015] [Accepted: 01/23/2016] [Indexed: 11/28/2022]
|
107
|
Trus I, Frydas IS, Reddy VRAP, Bonckaert C, Li Y, Kvisgaard LK, Larsen LE, Nauwynck HJ. Immunity raised by recent European subtype 1 PRRSV strains allows better replication of East European subtype 3 PRRSV strain Lena than that raised by an older strain. Vet Res 2016; 47:15. [PMID: 26742636 PMCID: PMC4705580 DOI: 10.1186/s13567-015-0292-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2015] [Accepted: 12/10/2015] [Indexed: 11/20/2022] Open
Abstract
Stable spatial distribution of porcine reproductive and respiratory syndrome (PRRSV)-1 subtypes in Europe is accompanied by a strong population immunity induced by local PRRSV strains. In the present study, it was examined if the immunity induced by three West European subtype 1 PRRSV strains (2007 isolate 07V063 and 2013 isolates 13V091 and 13V117) offers protection against the highly virulent East European subtype 3 PRRSV strain Lena. The number of fever days was greater (p < 0.05) in the control group (7.6 ± 1.7 days) compared to the immune groups (07V063-immune: 4.0 ± 1.2 days, 13V091-immune: 4.6 ± 1.1 days, 13V117-immune: 4.0 ± 2.9 days). In all groups, protection was characterized by reduction (p < 0.05) of AUC values of nasal shedding (control: 14.6, 07V063-immune: 3.4, 13V091-immune: 8.9, 13V117-immune: 8.0) and viremia (control: 28.1, 07V063-immune: 5.4, 13V091-immune: 9.0, 13V117-immune: 8.3). Reduction of respiratory disease, nasal shedding (mean AUC and mean peak values) and viremia (mean AUC and mean peak values) was more pronounced in 07V063-immune (p < 0.05) than in 13V091-immune and 13V117-immune animals. Inoculation with subtype 1 PRRSV strains caused priming of the Lena-specific virus neutralization antibody response. Upon challenge with Lena, we observed a very strong serological booster effect for neutralizing antibodies against strains used for the first inoculation. Our results indicate that inoculation with subtype 1 PRRSV strains can partially protect against antigenically divergent subtype 3 strains. The lower protection level elicited by recently isolated subtype 1 PRRSV strains may impair the outcome of the spatial expansion of subtype 3 strains from East Europe to West Europe.
Collapse
Affiliation(s)
- Ivan Trus
- Laboratory of Virology, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium.
| | - Ilias S Frydas
- Laboratory of Virology, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium.
| | - Vishwanatha R A P Reddy
- Laboratory of Virology, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium.
| | - Caroline Bonckaert
- Laboratory of Virology, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium.
| | - Yewei Li
- Laboratory of Virology, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium.
| | - Lise K Kvisgaard
- National Veterinary Institute, Technical University of Denmark, Frederiksberg C, Denmark.
| | - Lars E Larsen
- National Veterinary Institute, Technical University of Denmark, Frederiksberg C, Denmark.
| | - Hans J Nauwynck
- Laboratory of Virology, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium.
| |
Collapse
|
108
|
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
|
109
|
Transdermal delivery of plasmid encoding truncated nucleocapsid protein enhanced PRRSV-specific immune responses. Vaccine 2015; 34:609-615. [PMID: 26724543 DOI: 10.1016/j.vaccine.2015.12.043] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2015] [Revised: 12/11/2015] [Accepted: 12/16/2015] [Indexed: 01/02/2023]
Abstract
BACKGROUND Porcine Reproductive and Respiratory Syndrome virus (PRRSV) induces several immunomodulatory mechanisms that resulted in delayed and ineffective anti-viral immune responses. Recently, it has been shown that intradermal immunization of plasmid encoding truncated nucleocapsid protein (pORF7t) could reduce PRRSV-induced immunomodulatory activities and enhances anti-PRRSV immunity in vaccinated pigs. However, intradermal immunization may not be practical for farm setting. Currently, there are several transdermal delivery systems available in the market, although they were not originally designed for plasmid delivery. OBJECTIVES To investigate the potential use of a transdermal delivery system for delivering of pORF7t and its immunological outcomes. METHOD The immunomodulatory effects induced by transdermal delivery of pORF7t were compared with intradermal immunization in an experimental pig model. In addition, immunomodulatory effects of the DNA vaccine were determined in the fattening pigs kept in a PRRSV-positive farm environment, and in the experimental pigs receiving heterologous prime-boost, pORF7t-modified live vaccine (MLV) immunization. RESULT The patterns of PRRSV-specific cellular responses induced by transdermal and intradermal immunizations of pORF7t were similar. Interestingly, the pigs transdermally immunized with pORF7t exhibited higher number of PRRSV-specific CD8(+)IFN-γ(+) cells. Pigs immunized with pORF7t and kept at PRRSV-positive environment exhibited enhanced PRRSV-specific IFN-γ(+) production, reduced numbers of regulatory T lymphocytes (Tregs) and lower lung scores at the end of the finishing period. In the heterologous prime-boost experiment, priming with pORF7t prior to MLV vaccination resulted in significantly higher numbers of CD3(+)IFN-γ(+) subpopulations, lower numbers of PRRSV-specific CD3(+)IL-10(+) cells and Tregs, and rapid antibody responses in immunized pigs. CONCLUSION Transdermal immunization with pORF7t reduced PRRRSV-induced immunomodulatory effects and enhanced long-term PRRSV-specific cellular responses in vaccinated pigs. Furthermore, heterologous DNA-MLV prime-boost immunization significantly improved the quality of PRRSV-specific cellular and humoral immunity. The information could benefit the future development of PRRSV management and control strategies.
Collapse
|
110
|
Lunney JK, Fang Y, Ladinig A, Chen N, Li Y, Rowland B, Renukaradhya GJ. Porcine Reproductive and Respiratory Syndrome Virus (PRRSV): Pathogenesis and Interaction with the Immune System. Annu Rev Anim Biosci 2015; 4:129-54. [PMID: 26646630 DOI: 10.1146/annurev-animal-022114-111025] [Citation(s) in RCA: 501] [Impact Index Per Article: 50.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
This review addresses important issues of porcine reproductive and respiratory syndrome virus (PRRSV) infection, immunity, pathogenesis, and control. Worldwide, PRRS is the most economically important infectious disease of pigs. We highlight the latest information on viral genome structure, pathogenic mechanisms, and host immunity, with a special focus on immune factors that modulate PRRSV infections during the acute and chronic/persistent disease phases. We address genetic control of host resistance and probe effects of PRRSV infection on reproductive traits. A major goal is to identify cellular/viral targets and pathways for designing more effective vaccines and therapeutics. Based on progress in viral reverse genetics, host transcriptomics and genomics, and vaccinology and adjuvant technologies, we have identified new areas for PRRS control and prevention. Finally, we highlight the gaps in our knowledge base and the need for advanced molecular and immune tools to stimulate PRRS research and field applications.
Collapse
Affiliation(s)
- Joan K Lunney
- Animal Parasitic Diseases Laboratory, BARC ARS USDA, Beltsville, Maryland 20705;
| | - Ying Fang
- College of Veterinary Medicine, Kansas State University, Manhattan, Kansas 66506-5600; , ,
| | - Andrea Ladinig
- University of Veterinary Medicine, Vienna 1210, Austria;
| | - Nanhua Chen
- College of Veterinary Medicine, Kansas State University, Manhattan, Kansas 66506-5600; , , .,College of Veterinary Medicine, Yangzhou University, Yangzhou, Jiangsu 225009, China;
| | - Yanhua Li
- College of Veterinary Medicine, Kansas State University, Manhattan, Kansas 66506-5600; , ,
| | - Bob Rowland
- College of Veterinary Medicine, Kansas State University, Manhattan, Kansas 66506-5600; , ,
| | | |
Collapse
|
111
|
Lyoo KS, Yeom M, Choi JY, Park JH, Yoon SW, Song D. Unusual severe cases of type 1 porcine reproductive and respiratory syndrome virus (PRRSV) infection in conventionally reared pigs in South Korea. BMC Vet Res 2015; 11:272. [PMID: 26497589 PMCID: PMC4619543 DOI: 10.1186/s12917-015-0584-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2015] [Accepted: 10/15/2015] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Porcine reproductive and respiratory syndrome virus (PRRSV) causes a loss of approximately US$ 70 million every year to the South Korean pork industry. There are two distinct genotypes: European (type 1) and North American (type 2). In South Korea, type 1 and type 2 PRRSV are widely distributed and have evolved continuously since the infection was first described. Here, we present two field cases of type 1 PRRSV infection with unusually severe pathogenicity. CASE PRESENTATION The first case farm was a two-site production system comprising farrow-to-grower and grower-to-finish units and was historically free from PRRSV infections. The PRRSV vaccine had not been used in both units. In October 2014, pigs in the grower-to-finish unit experienced severe respiratory distress with the mortality rate reaching to 22%. Despite antibiotic treatment, clinical signs were still noticed in most pigs. The second case farm was also a two-site production system, but had two separate farrow-to-grower units (unit A and unit B). Historically, type 1 PRRSV was continuously present in unit A, but unit B was free from PRRSV. Thus, all grower pigs of unit B were vaccinated before being moved to the grower-to-finish unit. In November 2014, severe respiratory distress was seen in pigs of the grower-to-finish unit. Significant respiratory distress was observed in only the grower herd moved from unit B, and the mortality of those pigs was ~50%. However, no disease was shown in the grower pigs from unit A. CONCLUSIONS To our knowledge, the present study is the first observation of the cases of infection by highly pathogenic type 1 PRRSV in South Korea. The Korean type 1 PRRSV strains have undergone unique evolutionary dynamics for the last decade in this country. Although there are known to be three clusters of Korean type 1 PRRSV, their pathogenicity could not be categorized owing to their high level of genetic diversity. Therefore, further studies are needed to demonstrate the novel classification of Korean type 1 PRRSV strains according to their virulence factors.
Collapse
Affiliation(s)
- Kwang-Soo Lyoo
- Korea Zoonosis Research Institute, Chonbuk National University, Iksan, South Korea.
| | - Minjoo Yeom
- Department of Pharmacy, College of Pharmacy, Korea University, Sejong, South Korea.
| | | | - Jong-Hwan Park
- College of Veterinary Medicine, Jeonnam National University, Gwangju, South Korea.
| | - Sun-Woo Yoon
- Korea Research Institute of Bioscience and Biotechnology, Daejeon, South Korea.
| | - Daesub Song
- Department of Pharmacy, College of Pharmacy, Korea University, Sejong, South Korea.
| |
Collapse
|
112
|
Chen Y, He S, Sun L, Luo Y, Sun Y, Xie J, Zhou P, Su S, Zhang G. Genetic variation, pathogenicity, and immunogenicity of highly pathogenic porcine reproductive and respiratory syndrome virus strain XH-GD at different passage levels. Arch Virol 2015; 161:77-86. [PMID: 26483282 DOI: 10.1007/s00705-015-2597-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2015] [Accepted: 09/02/2015] [Indexed: 02/01/2023]
Abstract
Porcine reproductive and respiratory syndrome (PRRS) is one of the most economically important infectious diseases of swine worldwide. Immunization with an attenuated vaccine is considered an effective method for reducing the economic losses resulting from porcine reproductive and respiratory syndrome virus (PRRSV) infection. Several studies have shown that PRRSV can be attenuated by passage in Marc-145 cells, but it is still not clear whether this attenuation influences the immunogenicity of PRRSV and what the mechanism of attenuation is. In order to study the mechanism of attenuation and immunogenicity of highly pathogenic (HP) PRRSV, the HP-PRRSV strain XH-GD was serially 122 times passaged in Marc-145 cells. Genomic sequence comparisons were made at selected passages. To explore the differences in pathogenicity and immunogenicity at different passages, three passages (P5, P62 and P122) were selected for an animal challenge experiment, which showed that passage in Marc-145 cells resulted in attenuation of the virus. After 122 passages, 35 amino acid changes were observed in the structural proteins and non-structural proteins. The animal challenge experiment showed that pathogenicity decreased with increasing passage number. The N antibody level and specific neutralizing (SN) antibody titers also decreased with increasing passage number in the late stage of the animal experiment. This study indicated that the virulence of XH-GD was decreased by passage in Marc-145 cells and that overattenuation might influence the immunogenicity of virus. These results might contribute to our understanding of the mechanism of attenuation.
Collapse
Affiliation(s)
- Yao Chen
- MOA Key Laboratory of Animal Vaccine Development, Ministry of China, College of Veterinary Medicine, South China Agricultural University, 483 Wushan Road, Tianhe District, Guangzhou, 510642, China
| | - Shuyi He
- MOA Key Laboratory of Animal Vaccine Development, Ministry of China, College of Veterinary Medicine, South China Agricultural University, 483 Wushan Road, Tianhe District, Guangzhou, 510642, China
| | - Long Sun
- MOA Key Laboratory of Animal Vaccine Development, Ministry of China, College of Veterinary Medicine, South China Agricultural University, 483 Wushan Road, Tianhe District, Guangzhou, 510642, China
| | - Yongfeng Luo
- MOA Key Laboratory of Animal Vaccine Development, Ministry of China, College of Veterinary Medicine, South China Agricultural University, 483 Wushan Road, Tianhe District, Guangzhou, 510642, China
| | - Yankuo Sun
- MOA Key Laboratory of Animal Vaccine Development, Ministry of China, College of Veterinary Medicine, South China Agricultural University, 483 Wushan Road, Tianhe District, Guangzhou, 510642, China
| | - Jiexiong Xie
- MOA Key Laboratory of Animal Vaccine Development, Ministry of China, College of Veterinary Medicine, South China Agricultural University, 483 Wushan Road, Tianhe District, Guangzhou, 510642, China
| | - Pei Zhou
- Guangdong Provincial Key Laboratory of Prevention and Control for Severe Clinical Animal Diseases, College of Veterinary Medicine, South China Agricultural University, 483 Wushan Road, Tianhe District, Guangzhou, 510642, China
| | - Shuo Su
- Guangdong Provincial Key Laboratory of Prevention and Control for Severe Clinical Animal Diseases, College of Veterinary Medicine, South China Agricultural University, 483 Wushan Road, Tianhe District, Guangzhou, 510642, China
| | - Guihong Zhang
- MOA Key Laboratory of Animal Vaccine Development, Ministry of China, College of Veterinary Medicine, South China Agricultural University, 483 Wushan Road, Tianhe District, Guangzhou, 510642, China.
| |
Collapse
|
113
|
Li MM, Seelenbinder KM, Ponder MA, Deng L, Rhoads RP, Pelzer KD, Radcliffe JS, Maxwell CV, Ogejo JA, Hanigan MD. Effects of porcine reproductive and respiratory syndrome virus on pig growth, diet utilization efficiency, and gas release from stored manure. J Anim Sci 2015; 93:4424-35. [PMID: 26440342 DOI: 10.2527/jas.2015-8872] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
The objectives of this study were to examine the effects of porcine reproductive and respiratory syndrome virus (PRRSV) infection and vaccination on pig growth, dietary nutrient efficiency of utilization, manure output, and emissions of CO, CH, HS, NO, and NH gases from stored manure. Forty-eight pigs, aged 21 d at the start of the study, were subjected to 1 of 4 treatment combinations arranged in a 2 × 2 factorial design with main factors of PRRSV vaccination and PRRSV infection. Body weight, ADFI, manure output, and nutrient efficiency of utilization were assessed and gas emissions from stored manure were determined daily from 50 to 78 d of age and for 24 d after completion of the animal phase. Infection with PRRSV markedly reduced final BW, ADG, and ADFI ( < 0.01) and reduced efficiencies of ADF and ether extract utilization ( = 0.05 and = 0.02, respectively) regardless of vaccination status. No significant treatment effects were found on manure output, manure pH, efficiencies of lignin utilization, and N retention. Infecting pigs with PRRSV increased daily manure CO emission per pig ( = 0.01). There was an interaction between immunization and infection for NO per pig with manure from uninfected, vaccinated pigs producing as much as the manure from infected, vaccinated pigs whereas there was a difference by PRRSV infection state for nonvaccinated pigs. There were also interactions between treatments for HS and NO emissions per kilogram of manure volatile solids excreted ( = 0.01 and = 0.0001, respectively) with the same pattern as for NO per pig; that is, the vaccinated pigs had similar rates of emission regardless of infection state. Pigs infected with PRRSV increased NO nitrogen per kilogram of total N excreted compared with noninfected groups ( = 0.03). Collectively, these results indicated that PRRSV infection caused decreased growth rates and nutrient utilization efficiency and increased gas emissions from stored manure.
Collapse
|
114
|
A Synthetic Porcine Reproductive and Respiratory Syndrome Virus Strain Confers Unprecedented Levels of Heterologous Protection. J Virol 2015; 89:12070-83. [PMID: 26401031 DOI: 10.1128/jvi.01657-15] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2015] [Accepted: 09/14/2015] [Indexed: 12/16/2022] Open
Abstract
UNLABELLED Current vaccines do not provide sufficient levels of protection against divergent porcine reproductive and respiratory syndrome virus (PRRSV) strains circulating in the field, mainly due to the substantial variation of the viral genome. We describe here a novel approach to generate a PRRSV vaccine candidate that could confer unprecedented levels of heterologous protection against divergent PRRSV isolates. By using a set of 59 nonredundant, full-genome sequences of type 2 PRRSVs, a consensus genome (designated PRRSV-CON) was generated by aligning these 59 PRRSV full-genome sequences, followed by selecting the most common nucleotide found at each position of the alignment. Next, the synthetic PRRSV-CON strain was generated through the use of reverse genetics. PRRSV-CON replicates as efficiently as our prototype PRRSV strain FL12, both in vitro and in vivo. Importantly, when inoculated into pigs, PRRSV-CON confers significantly broader levels of heterologous protection than does wild-type PRRSV. Collectively, our data demonstrate that PRRSV-CON can serve as an excellent candidate for the development of a broadly protective PRRSV vaccine. IMPORTANCE The extraordinary genetic variation of RNA viruses poses a monumental challenge for the development of broadly protective vaccines against these viruses. To minimize the genetic dissimilarity between vaccine immunogens and contemporary circulating viruses, computational strategies have been developed for the generation of artificial immunogen sequences (so-called "centralized" sequences) that have equal genetic distances to the circulating viruses. Thus far, the generation of centralized vaccine immunogens has been carried out at the level of individual viral proteins. We expand this concept to PRRSV, a highly variable RNA virus, by creating a synthetic PRRSV strain based on a centralized PRRSV genome sequence. This study provides the first example of centralizing the whole genome of an RNA virus to improve vaccine coverage. This concept may be significant for the development of vaccines against genetically variable viruses that require active viral replication in order to achieve complete immune protection.
Collapse
|
115
|
Gao L, Wang L, Huang C, Yang L, Guo XK, Yu Z, Liu Y, Yang P, Feng WH. HP-PRRSV is attenuated by de-optimization of codon pair bias in its RNA-dependent RNA polymerase nsp9 gene. Virology 2015; 485:135-44. [PMID: 26247624 DOI: 10.1016/j.virol.2015.07.012] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2015] [Revised: 05/21/2015] [Accepted: 07/16/2015] [Indexed: 10/23/2022]
Abstract
There is an urgent need to develop new vaccines against highly pathogenic PRRS virus (HP-PRRSV) variant in China. The actual use of each codon pairs is more or less frequent than that of the statistical prediction and codon pair bias (CPB) usage affects gene translation. We "shuffled" the existing codons in HP-PRRSV genes GP5, M, nsp2 and nsp9, so that the CPB of these genes could be more negative. De-optimization of nsp9, the RNA-dependent RNA polymerase, significantly decreased PRRSV replication in porcine alveolar macrophages (PAMs). In vitro study showed that HV-nsp9(min) and HV-nsp29(min) were remarkably attenuated in PAMs, and inoculation of pigs with 2 ml⁎10(5.0) TCID50/ml of HV-nsp9(min) or HV-nsp29(min) did not cause PRRS. Importantly, pigs immunized with HV-nsp29(min) were fully protected against different HP-PRRSV strains׳ lethal challenges. Our results imply that the CPB de-optimized HV-nsp29(min) has the potential to be used as a live vaccine candidate against HP-PRRSV.
Collapse
Affiliation(s)
- Li Gao
- China Academy of Medicine Sciences, Peking Union Medical College, Institute of Medicinal Plant Development, Beijing 100193, China
| | - Lianghai Wang
- State Key Laboratory of Agrobiotechnology, Department of Microbiology and Immunology, College of Biological Sciences, China Agricultural University, Beijing 100193, China; Department of Microbiology and Immunology, College of Biological Sciences, China Agricultural University, Beijing 100193, China
| | - Chen Huang
- State Key Laboratory of Agrobiotechnology, Department of Microbiology and Immunology, College of Biological Sciences, China Agricultural University, Beijing 100193, China; Department of Microbiology and Immunology, College of Biological Sciences, China Agricultural University, Beijing 100193, China
| | - Longlong Yang
- The Hamner Institutes for Health Sciences, Research Triangle Park, NC 27709-2137, USA
| | - Xue-Kun Guo
- State Key Laboratory of Agrobiotechnology, Department of Microbiology and Immunology, College of Biological Sciences, China Agricultural University, Beijing 100193, China; Department of Microbiology and Immunology, College of Biological Sciences, China Agricultural University, Beijing 100193, China
| | - Zhibin Yu
- State Key Laboratory of Agrobiotechnology, Department of Microbiology and Immunology, College of Biological Sciences, China Agricultural University, Beijing 100193, China; Department of Microbiology and Immunology, College of Biological Sciences, China Agricultural University, Beijing 100193, China
| | - Yihao Liu
- State Key Laboratory of Agrobiotechnology, Department of Microbiology and Immunology, College of Biological Sciences, China Agricultural University, Beijing 100193, China; Department of Microbiology and Immunology, College of Biological Sciences, China Agricultural University, Beijing 100193, China
| | - Peng Yang
- Beijing Senkang Biotech Development Co., Ltd, Beijing 101400, China
| | - Wen-Hai Feng
- State Key Laboratory of Agrobiotechnology, Department of Microbiology and Immunology, College of Biological Sciences, China Agricultural University, Beijing 100193, China; Department of Microbiology and Immunology, College of Biological Sciences, China Agricultural University, Beijing 100193, China.
| |
Collapse
|
116
|
Piñeyro PE, Kenney SP, Giménez-Lirola LG, Heffron CL, Matzinger SR, Opriessnig T, Meng XJ. Expression of antigenic epitopes of porcine reproductive and respiratory syndrome virus (PRRSV) in a modified live-attenuated porcine circovirus type 2 (PCV2) vaccine virus (PCV1-2a) as a potential bivalent vaccine against both PCV2 and PRRSV. Virus Res 2015; 210:154-64. [PMID: 26239318 DOI: 10.1016/j.virusres.2015.07.027] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2015] [Revised: 07/28/2015] [Accepted: 07/29/2015] [Indexed: 12/01/2022]
Abstract
Co-infection of pigs in the field with porcine circovirus type 2 (PCV2) and porcine reproductive and respiratory syndrome virus (PRRSV) is common and poses a major concern in effective control of PCV2 and PRRSV. We previously demonstrated that insertion of foreign epitope tags in the C-terminus of PCV2 ORF2 produced infectious virions that elicited humoral immune responses against both PCV2 capsid and inserted epitope tags. In this study, we aimed to determine whether the non-pathogenic chimeric virus PCV1-2a, which is the basis for the licensed PCV2 vaccine Fostera PCV, can express PRRSV antigenic epitopes, thus generating dual immunity as a potential bivalent vaccine against both PCV2 and PPRSV. Four different linear B-cell antigenic epitopes of PRRSV were inserted into the C-terminus of the capsid gene of the PCV1-2a vaccine virus. We showed that insertion of 12 (PRRSV-GP2 epitope II, PRRSV-GP3 epitope I, and PRRSV-GP5 epitope I), and 14 (PRRSV-GP5 epitope IV) amino acid residues did not impair the replication of the resulting PCV1-2a-PRRSVEPI chimeric viruses in vitro. The four chimeric PCV1-2a viruses expressing PRRSV B-cell linear epitopes were successfully rescued and characterized. An immunogenicity study in pigs revealed that two of the four chimeric viruses, PCV1-2a-PRRSVEPIGP3IG and PCV1-2a-PRRSVEPIEPIGP5IV, elicited neutralizing antibodies against PRRSV VR2385 as well as PCV2 (strains PCV2a, PCV2b, and mPCV2b). The results have important implications for exploring the potential use of PCV1-2a vaccine virus as a live virus vector to develop bivalent MLVs against both PCV2 and PRRSV.
Collapse
Affiliation(s)
- Pablo E Piñeyro
- Department of Biomedical Sciences & Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Polytechnic Institute and State University, Blacksburg, VA 24060, USA; Department of Veterinary Diagnostic and Production Animal Medicine, Iowa State University College of Veterinary Medicine, Ames, IA 5001, USA
| | - Scott P Kenney
- Department of Biomedical Sciences & Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Polytechnic Institute and State University, Blacksburg, VA 24060, USA
| | - Luis G Giménez-Lirola
- Department of Veterinary Diagnostic and Production Animal Medicine, Iowa State University College of Veterinary Medicine, Ames, IA 5001, USA
| | - C Lynn Heffron
- Department of Biomedical Sciences & Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Polytechnic Institute and State University, Blacksburg, VA 24060, USA
| | - Shannon R Matzinger
- Department of Biomedical Sciences & Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Polytechnic Institute and State University, Blacksburg, VA 24060, USA
| | - Tanja Opriessnig
- Department of Veterinary Diagnostic and Production Animal Medicine, Iowa State University College of Veterinary Medicine, Ames, IA 5001, USA; The Roslin Institute and The Royal (Dick) School of Veterinary Studies, University of Edinburgh, Edinburgh, United Kingdom
| | - Xiang-Jin Meng
- Department of Biomedical Sciences & Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Polytechnic Institute and State University, Blacksburg, VA 24060, USA.
| |
Collapse
|
117
|
Lyoo YS. Porcine reproductive and respiratory syndrome virus vaccine does not fit in classical vaccinology. Clin Exp Vaccine Res 2015; 4:159-65. [PMID: 26273574 PMCID: PMC4524900 DOI: 10.7774/cevr.2015.4.2.159] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2015] [Revised: 06/18/2015] [Accepted: 06/25/2015] [Indexed: 11/15/2022] Open
Abstract
All vaccines are developed to elicit an effective immune response in vaccinated animals such as innate, humoral and cell mediated response to protect animal health. Quality and intensity of the immune responses are differing by characteristics of the vaccine formulation and nature of the infectious agent. Modified live virus vaccines showed advantages over killed vaccines in terms of rapid immune response, duration of the immunity and better cell mediated protection mechanism. The porcine reproductive and respiratory syndrome virus (PRRSV) is relatively newly emerging (1986 in United States, 1990 in Europe) viral pathogen in pigs and tremendous effort has been made to protect pigs from this economically devastating disease such as developing killed, modified live, recombinant protein based and DNA vaccines. However, only cell culture attenuated virus vaccine is practiced with arguably limited efficacy. The PRRSV vaccine did not clear virus from infected pigs nor prevent re-infection of the virus. The vaccine showed very limited innate immune response, low anamnestic immune response and negligible cell mediated immune response. Despite of the current developed scientific technology, there still remain many questions to solve a most important pig disease worldwide.
Collapse
Affiliation(s)
- Young S Lyoo
- Department of Pathology, College of Veterinary Medicine, Konkuk University, Seoul, Korea
| |
Collapse
|
118
|
Loving CL, Osorio FA, Murtaugh MP, Zuckermann FA. Innate and adaptive immunity against Porcine Reproductive and Respiratory Syndrome Virus. Vet Immunol Immunopathol 2015. [PMID: 26209116 PMCID: PMC7112826 DOI: 10.1016/j.vetimm.2015.07.003] [Citation(s) in RCA: 147] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Many highly effective vaccines have been produced against viruses whose virulent infection elicits strong and durable protective immunity. In these cases, characterization of immune effector mechanisms and identification of protective epitopes/immunogens has been informative for the development of successful vaccine programs. Diseases in which the immune system does not rapidly clear the acute infection and/or convalescent immunity does not provide highly effective protection against secondary challenge pose a major hurdle for clinicians and scientists. Porcine reproductive and respiratory syndrome virus (PRRSV) falls primarily into this category, though not entirely. PRRSV causes a prolonged infection, though the host eventually clears the virus. Neutralizing antibodies can provide passive protection when present prior to challenge, though infection can be controlled in the absence of detectable neutralizing antibodies. In addition, primed pigs (through natural exposure or vaccination with a modified-live vaccine) show some protection against secondary challenge. While peripheral PRRSV-specific T cell responses have been examined, their direct contribution to antibody-mediated immunity and viral clearance have not been fully elucidated. The innate immune response following PRRSV infection, particularly the antiviral type I interferon response, is meager, but when provided exogenously, IFN-α enhances PRRSV immunity and viral control. Overall, the quality of immunity induced by natural PRRSV infection is not ideal for informing vaccine development programs. The epitopes necessary for protection may be identified through natural exposure or modified-live vaccines and subsequently applied to vaccine delivery platforms to accelerate induction of protective immunity following vaccination. Collectively, further work to identify protective B and T cell epitopes and mechanisms by which PRRSV eludes innate immunity will enhance our ability to develop more effective methods to control and eliminate PRRS disease.
Collapse
Affiliation(s)
- Crystal L Loving
- USDA-ARS-National Animal Disease Center, Ames, IA, United States.
| | - Fernando A Osorio
- Nebraska Center for Virology and School of Veterinary & Biomedical Sciences, University of Nebraska-Lincoln, United States
| | - Michael P Murtaugh
- Department of Veterinary and Biomedical Sciences, University of Minnesota, St. Paul, MN, United States
| | - Federico A Zuckermann
- Department of Pathobiology, College of Veterinary Medicine, University of Illinois, Urbana-Champaign, IL, United States
| |
Collapse
|
119
|
Dunkelberger J, Boddicker N, Serão N, Young J, Rowland R, Dekkers J. Response of pigs divergently selected for residual feed intake to experimental infection with the PRRS virus. Livest Sci 2015. [DOI: 10.1016/j.livsci.2015.04.014] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
120
|
Suradhat S, Wongyanin P, Kesdangsakonwut S, Teankum K, Lumyai M, Triyarach S, Thanawongnuwech R. A novel DNA vaccine for reduction of PRRSV-induced negative immunomodulatory effects: A proof of concept. Vaccine 2015; 33:3997-4003. [PMID: 26079617 DOI: 10.1016/j.vaccine.2015.06.020] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2015] [Revised: 05/22/2015] [Accepted: 06/02/2015] [Indexed: 12/19/2022]
Abstract
BACKGROUND Viral-induced interleukin (IL)-10 and regulatory T lymphocytes (Tregs) are believed to play a major role in shaping the immunological and clinical outcomes following Porcine Reproductive and Respiratory Syndrome virus (PRRSV) infection. Recently, it has been shown that PRRSV nucleocapsid (N) protein can induce IL-10 production which is essential for induction of PRRSV-specific Tregs. We hypothesized that immunity to N protein should reduce PRRSV-induced negative immunomodulatory effects which will be essential for establishing proper anti-PRRSV immunity in infected pigs. OBJECTIVES To investigate the immunomodulatory effects of DNA vaccine encoding a linearized, truncated form of PRRSV-N protein (pORF7t) which was designed to preferentially induce cell-mediated immunity against PRRSV N protein. METHOD Immunomodulatory effects of the novel DNA vaccine were investigated in an experimental vaccinated-challenged model. In addition, long-term immunomodulatory effects of the DNA vaccine were investigated in vaccinated pigs kept at the PRRSV-positive environment until the end of the fattening period. Pigs were vaccinated either prior to or following natural PRRSV infection. RESULT The results indicated that pORF7t could modulate the anti-PRRSV immune responses and promote the control of viral replication in the vaccinated-challenged pigs. Immunized pigs exhibited increased numbers of PRRSV-specific activated CD4(+)CD25(+) lymphocytes, reduced numbers of PRRSV-specific Tregs, and rapid viral clearance following infection. In a long-term study, regardless of the time of vaccination, DNA vaccine could modulate the host immune responses, resulted in enhanced PRRSV-specific IFN-γ producing cells, and reduced numbers of PRRSV-specific Tregs, without evidence of enhanced antibody responses. No vaccine adverse reaction was observed throughout the study. CONCLUSION This study revealed the novel concept that PRRSV-specific immunity can be modulated by induction of cell-mediated immunity against the nucleocapsid protein. This concept could potentially benefit the development of PRRSV management and control strategies.
Collapse
Affiliation(s)
- Sanipa Suradhat
- Faculty of Veterinary Science, Chulalongkorn University, Pathumwan, Bangkok 10330, Thailand; Center of Excellence in Emerging Infectious Diseases in Animals, Chulalongkorn University (CU-EIDAs), Pathumwan, Bangkok 10330, Thailand.
| | - Piya Wongyanin
- Inter-department of Medical Microbiology, Graduate School, Chulalongkorn University, Pathumwan, Bangkok 10330, Thailand
| | - Sawang Kesdangsakonwut
- Faculty of Veterinary Science, Chulalongkorn University, Pathumwan, Bangkok 10330, Thailand
| | - Komkrich Teankum
- Faculty of Veterinary Science, Chulalongkorn University, Pathumwan, Bangkok 10330, Thailand
| | - Mongkol Lumyai
- Thai-Denmark Swine Breeder Public Company Limited, Bang-na, Bangkok 10260, Thailand
| | - Sittikorn Triyarach
- Thai-Denmark Swine Breeder Public Company Limited, Bang-na, Bangkok 10260, Thailand
| | - Roongroje Thanawongnuwech
- Faculty of Veterinary Science, Chulalongkorn University, Pathumwan, Bangkok 10330, Thailand; Center of Excellence in Emerging Infectious Diseases in Animals, Chulalongkorn University (CU-EIDAs), Pathumwan, Bangkok 10330, Thailand
| |
Collapse
|
121
|
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
|
122
|
Fan B, Liu X, Bai J, Zhang T, Zhang Q, Jiang P. The amino acid residues at 102 and 104 in GP5 of porcine reproductive and respiratory syndrome virus regulate viral neutralization susceptibility to the porcine serum neutralizing antibody. Virus Res 2015; 204:21-30. [PMID: 25907991 DOI: 10.1016/j.virusres.2015.04.015] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2015] [Revised: 04/08/2015] [Accepted: 04/10/2015] [Indexed: 12/29/2022]
Abstract
Porcine reproductive and respiratory syndrome virus (PRRSV) is mainly responsible for the heavy economic losses in pig industry in the world. A number of neutralizing epitopes have been identified in the viral structural proteins GP3, GP4, GP5 and M. In this study, the important amino acid (aa) residues of HP-PRRSV strain BB affecting neutralization susceptibility of antibody were examined using resistant strains generated under neutralizing antibody (NAb) pressure in MARC-145 cells, reverse genetic technique and virus neutralization assay. HP-PRRSV strain BB was passaged under the pressure of porcine NAb serum in vitro. A resistant strain BB34s with 102 and 104 aa substitutions in GP5, which have been predicted to be the positive sites for pressure selection (Delisle et al., 2012), was cloned and identified. To determine the effect of the two aa residues on neutralization, eight recombinant PRRSV strains were generated, and neutralization assay results confirmed that the aa residues 102 and 104 in GP5 played an important role in NAbs against HP-PRRSV in MARC-145 cells and porcine alveolar macrophages. Alignment of GP5 sequences revealed that the variant aa residues at 102 and 104 were frequent among type 2 PRRSV strains. It may be helpful for understanding the mechanism regulating the neutralization susceptibility of PRRSV to the NAbs and monitoring the antigen variant strains in the field.
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
| | - Tingjie Zhang
- 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.
| |
Collapse
|
123
|
Production and evaluation of virus-like particles displaying immunogenic epitopes of porcine reproductive and respiratory syndrome virus (PRRSV). Int J Mol Sci 2015; 16:8382-96. [PMID: 25874763 PMCID: PMC4425087 DOI: 10.3390/ijms16048382] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2015] [Revised: 03/27/2015] [Accepted: 04/01/2015] [Indexed: 12/15/2022] Open
Abstract
Porcine reproductive and respiratory syndrome (PRRS) is the most significant infectious disease currently affecting the swine industry worldwide. Several inactivated and modified live vaccines (MLV) have been developed to curb PRRSV infections. However, the efficacy and safety of these vaccines are unsatisfactory, and hence, there is a strong demand for the development of new PRRS universal vaccines. Virus-like particle (VLP)-based vaccines are gaining increasing acceptance compared to subunit vaccines, as they present the antigens in a more veritable conformation and are readily recognized by the immune system. Hepatitis B virus core antigen (HBcAg) has been successfully used as a carrier for more than 100 viral sequences. In this study, hybrid HBcAg VLPs were generated by fusion of the conserved protective epitopes of PRRSV and expressed in E. coli. An optimized purification protocol was developed to obtain hybrid HBcAg VLP protein from the inclusion bodies. This hybrid HBcAg VLP protein self-assembled to 23-nm VLPs that were shown to block virus infection of susceptible cells when tested on MARC 145 cells. Together with the safety of non-infectious and non-replicable VLPs and the low cost of production through E. coli fermentation, this hybrid VLP could be a promising vaccine candidate for PRRS.
Collapse
|
124
|
|
125
|
Abstract
Porcine reproductive and respiratory disease syndrome (PRRS) is a viral pandemic that especially affects neonates within the “critical window” of immunological development. PRRS was recognized in 1987 and within a few years became pandemic causing an estimated yearly $600,000 economic loss in the USA with comparative losses in most other countries. The causative agent is a single-stranded, positive-sense enveloped arterivirus (PRRSV) that infects macrophages and plasmacytoid dendritic cells. Despite the discovery of PRRSV in 1991 and the publication of >2,000 articles, the control of PRRS is problematic. Despite the large volume of literature on this disease, the cellular and molecular mechanisms describing how PRRSV dysregulates the host immune system are poorly understood. We know that PRRSV suppresses innate immunity and causes abnormal B cell proliferation and repertoire development, often lymphopenia and thymic atrophy. The PRRSV genome is highly diverse, rapidly evolving but amenable to the generation of many mutants and chimeric viruses for experimental studies. PRRSV only replicates in swine which adds to the experimental difficulty since no inbred well-defined animal models are available. In this article, we summarize current knowledge and apply it toward developing a series of provocative and testable hypotheses to explain how PRRSV immunomodulates the porcine immune system with the goal of adding new perspectives on this disease.
Collapse
|
126
|
García-Nicolás O, Rosales RS, Pallarés FJ, Risco D, Quereda JJ, Graham SP, Frossard JP, Morgan SB, Steinbach F, Drew TW, Strickland TS, Salguero FJ. Comparative analysis of cytokine transcript profiles within mediastinal lymph node compartments of pigs after infection with porcine reproductive and respiratory syndrome genotype 1 strains differing in pathogenicity. Vet Res 2015; 46:34. [PMID: 25889072 PMCID: PMC4364558 DOI: 10.1186/s13567-015-0161-8] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2014] [Accepted: 02/06/2015] [Indexed: 01/28/2023] Open
Abstract
Porcine reproductive and respiratory syndrome virus (PRRSV) induces a weak immune response enabling it to persist in different organs of infected pigs. This has been attributed to the ability of PRRSV to influence the induction of cytokine responses. In this study, we investigated the cytokine transcriptional profiles in different compartments of the mediastinal lymph node of pigs infected with three genotype 1 PRRSV strains of differing pathogenicity: the low virulence prototype Lelystad virus (LV), and UK field strain 215–06 and the highly virulent subtype 3 SU1-Bel isolate from Belarus. We have used a combination of laser capture micro-dissection (LCM) followed by real time quantitative PCR (RT-qPCR) and immunohistochemical (IHC) detection of immune cell markers (CD3, CD79a and MAC387) and RT-qPCR quantification of PRRSV and cytokine transcripts. Compared to mock infected pigs, we found a significant downregulation of TNF-α and IFN-α in follicular and interfollicular areas of the mediastinal lymph node from 3 days post-infection (dpi) in animals infected with all three strains. This was accompanied by a transient B cell depletion and T cell and macrophage infiltration in the follicles together with T cell depletion in the interfollicular areas. A delayed upregulation of IFN-γ and IL-23p19 was observed mainly in the follicles. The PRRSV load was higher in all areas and time-points studied in the animals infected with the SU1-Bel strain. This paper describes the first application of LCM to study the cytokine transcript profiles and virus distribution in different compartments of the lymph node of pigs.
Collapse
Affiliation(s)
- Obdulio García-Nicolás
- Department of Anatomy and Comparative Pathology, Faculty of Veterinary Medicine, Murcia University, "Mare Nostrum Excellence Campus - 3738", 30100, Murcia, Spain.
| | - Rubén S Rosales
- Department of Bacteriology, Animal and Plant Health Agency, Addlestone, Surrey, KT15 3NB, UK.
| | - Francisco J Pallarés
- Department of Anatomy and Comparative Pathology, Faculty of Veterinary Medicine, Murcia University, "Mare Nostrum Excellence Campus - 3738", 30100, Murcia, Spain.
| | - David Risco
- Red de Grupos de Investigación Recursos Faunísticos, Facultad de Veterinaria, Universidad de Extremadura, 10003, Cáceres, Spain.
| | - Juan J Quereda
- Spanish National Center of Biotechnology (CSIC), C/Darwin 3, Campus de Cantoblanco, 28049, Madrid, Spain.
| | - Simon P Graham
- Department of Virology, Animal and Plant Health Agency, Addlestone, Surrey, KT15 3NB, UK.
| | - Jean-Pierre Frossard
- Department of Virology, Animal and Plant Health Agency, Addlestone, Surrey, KT15 3NB, UK.
| | - Sophie B Morgan
- Department of Virology, Animal and Plant Health Agency, Addlestone, Surrey, KT15 3NB, UK.
| | - Falko Steinbach
- Department of Virology, Animal and Plant Health Agency, Addlestone, Surrey, KT15 3NB, UK. .,Department of Pathology and Infectious Diseases, School of Veterinary Medicine, University of Surrey, Guildford, GU2 7TE, UK.
| | - Trevor W Drew
- Department of Virology, Animal and Plant Health Agency, Addlestone, Surrey, KT15 3NB, UK.
| | - Tony S Strickland
- Department of Pathology, Animal and Plant Health Agency, Addlestone, KT15 3NB, UK.
| | - Francisco J Salguero
- Department of Pathology, Animal and Plant Health Agency, Addlestone, KT15 3NB, UK. .,Department of Pathology and Infectious Diseases, School of Veterinary Medicine, University of Surrey, Guildford, GU2 7TE, UK.
| |
Collapse
|
127
|
Regulation and evasion of antiviral immune responses by porcine reproductive and respiratory syndrome virus. Virus Res 2014; 202:101-11. [PMID: 25529442 PMCID: PMC7132515 DOI: 10.1016/j.virusres.2014.12.014] [Citation(s) in RCA: 75] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2014] [Revised: 12/10/2014] [Accepted: 12/11/2014] [Indexed: 12/13/2022]
Abstract
Five PRRSV viral proteins are shown to inhibit type I IFN induction and signaling by targeting different intracellular signaling intermediates. PRRSV regulates the expression of IL-10 and TNFα. PRRSV modulates apoptosis during infection. MicroRNAs might play significant roles in subverting immunity for PRRSV. PRRSV escapes from adaptive immunity by impairing antigen presentation, activating Tregs, and ADE.
Virus infection of mammalian cells triggers host innate immune responses to restrict viral replication and induces adaptive immunity for viral elimination. In order to survive and propagate, viruses have evolved sophisticated mechanisms to subvert host defense system by encoding proteins that target key components of the immune signaling pathways. Porcine reproductive and respiratory syndrome virus (PRRSV), a RNA virus, impairs several processes of host immune responses including interfering with interferon production and signaling, modulating cytokine expression, manipulating apoptotic responses and regulating adaptive immunity. In this review, we highlight the molecular mechanisms of how PRRSV interferes with the different steps of initial antiviral host responses to establish persistent infection in pigs. Dissection of the PRRSV–host interaction is the key in understanding PRRSV pathogenesis and will provide a basis for the rational design of vaccines.
Collapse
|
128
|
Gao J, Ji P, Zhang M, Wang X, Li N, Wang C, Xiao S, Mu Y, Zhao Q, Du T, Sun Y, Hiscox JA, Zhang G, Zhou EM. GP5 expression in Marc-145 cells inhibits porcine reproductive and respiratory syndrome virus infection by inducing beta interferon activity. Vet Microbiol 2014; 174:409-418. [DOI: 10.1016/j.vetmic.2014.09.030] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2014] [Revised: 09/24/2014] [Accepted: 09/25/2014] [Indexed: 12/14/2022]
|
129
|
Wang X, Qui L, Dang Y, Xiao S, Zhang S, Yang Z. Linear epitope recognition antibodies strongly respond to the C-terminal domain of HP-PRRSV GP5. Vet Microbiol 2014; 174:565-569. [PMID: 25448446 DOI: 10.1016/j.vetmic.2014.09.004] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2014] [Revised: 08/17/2014] [Accepted: 09/08/2014] [Indexed: 11/25/2022]
Abstract
A total of 155 peptides derived from the highly pathogenic porcine reproductive and respiratory syndrome virus (HP-PRRSV) glycoprotein 5 (GP5) were printed on a chip to reveal the antigen reaction characteristics of the protein. The reactions of these peptides to HP-PRRSV-specific pig serum were scanned and quantified using fluorescence intensity via the PepSlide(®) Analyzer software. The intensity plots showed different reactions in the different sectors of GP5. The highest reaction intensity value reached 3894.5, with a peptide sequence of IVEKGGKVEVEGHLI. Seventeen peptides that showed relatively high reaction levels with HP-PRRSV-specific pig serum were selected as epitope candidates. Furthermore, the antigenic character was predicted using a software and was compared with the peptide scan results. In contrast to the software prediction, the HP-PRRSV-specific antibodies strongly responded to the C-terminal domain of GP5. The acquired data may be useful for understanding the antigenic characteristics of HP-PRRSV GP5.
Collapse
Affiliation(s)
- Xinglong Wang
- College of Veterinary Medicine, Northwest A&F University, No. 22 Xinong road, Yangling 712100, China.
| | - Li Qui
- College of Veterinary Medicine, Northwest A&F University, No. 22 Xinong road, Yangling 712100, China
| | - Yu Dang
- Shaanxi University of Technology, Hanzhong 723001, China
| | - Sha Xiao
- College of Veterinary Medicine, Northwest A&F University, No. 22 Xinong road, Yangling 712100, China
| | - Shuxia Zhang
- College of Veterinary Medicine, Northwest A&F University, No. 22 Xinong road, Yangling 712100, China
| | - Zengqi Yang
- College of Veterinary Medicine, Northwest A&F University, No. 22 Xinong road, Yangling 712100, China.
| |
Collapse
|
130
|
Amadori M, Razzuoli E. Immune Control of PRRS: Lessons to be Learned and Possible Ways Forward. Front Vet Sci 2014; 1:2. [PMID: 26664910 PMCID: PMC4668844 DOI: 10.3389/fvets.2014.00002] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2014] [Accepted: 05/19/2014] [Indexed: 12/29/2022] Open
Abstract
Porcine reproductive and respiratory syndrome (PRRS) is an elusive model of host/virus relationship in which disease is determined by virus pathogenicity, pig breed susceptibility and phenotype, microbial infectious pressure, and environmental conditions. The disease can be controlled by farm management programs, which can be supported by vaccination or conditioning of animals to circulating PRRS virus (PRRSV) strains. Yet, PRRS still represents a cause of heavy losses for the pig industry worldwide. Immunological control strategies are often compounded by poor and late development of adaptive immunity in both vaccinated and infected animals. Also, there is evidence that results of field trials can be worse than those of experimental studies in isolation facilities. Neutralizing antibody (NA) was shown to prevent PRRSV infection. Instead, the role of NA and adaptive immunity on the whole in virus clearance after established PRRSV infections is still contentious. Pigs eventually eliminate PRRSV infection, which may be correlated with an “educated,” innate immune response, which may also develop following vaccination. In addition to vaccination, an immunomodulation strategy for PRRS can be reasonably advocated in pig “problem” farms, where a substantial control of disease prevalence and disease-related losses is badly needed. This is not at odds with vaccination, which should be preferably restricted to PRRSV-free animals bound for PRRSV-infected farm units. Oral, low-dose, interferon-α treatments proved effective on farm for the control of respiratory and reproductive disease outbreaks, whereas the results were less clear in isolation facilities. Having in mind the crucial interaction between PRRSV and bacterial lipopolysaccharides for occurrence of respiratory disease, the strong control actions of low-dose type I interferons on the inflammatory response observed in vitro and in vivo probably underlie the rapid clinical responses observed in field trials.
Collapse
Affiliation(s)
- Massimo Amadori
- Laboratory of Cellular Immunology, Istituto Zooprofilattico Sperimentale della Lombardia e dell'Emilia Romagna , Brescia , Italy
| | - Elisabetta Razzuoli
- Laboratory of Cellular Immunology, Istituto Zooprofilattico Sperimentale della Lombardia e dell'Emilia Romagna , Brescia , Italy
| |
Collapse
|
131
|
Veit M, Matczuk AK, Sinhadri BC, Krause E, Thaa B. Membrane proteins of arterivirus particles: structure, topology, processing and function. Virus Res 2014; 194:16-36. [PMID: 25278143 PMCID: PMC7172906 DOI: 10.1016/j.virusres.2014.09.010] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2014] [Revised: 09/20/2014] [Accepted: 09/23/2014] [Indexed: 01/01/2023]
Abstract
Arteriviruses are important pathogens in veterinary medicine. We review the structure and processing of their membrane proteins. Some features are unique from a cell biological point of view. New data on this topic are also presented. We speculate on the role of the membrane proteins during virus entry and budding.
Arteriviruses, such as equine arteritis virus (EAV) and porcine reproductive and respiratory syndrome virus (PRRSV), are important pathogens in veterinary medicine. Despite their limited genome size, arterivirus particles contain a multitude of membrane proteins, the Gp5/M and the Gp2/3/4 complex, the small and hydrophobic E protein and the ORF5a protein. Their function during virus entry and budding is understood only incompletely. We summarize current knowledge of their primary structure, membrane topology, (co-translational) processing and intracellular targeting to membranes of the exocytic pathway, which are the budding site. We profoundly describe experimental data that led to widely believed conceptions about the function of these proteins and also report new results about processing steps for each glycoprotein. Further, we depict the location and characteristics of epitopes in the membrane proteins since the late appearance of neutralizing antibodies may lead to persistence, a characteristic hallmark of arterivirus infection. Some molecular features of the arteriviral proteins are rare or even unique from a cell biological point of view, particularly the prevention of signal peptide cleavage by co-translational glycosylation, discovered in EAV-Gp3, and the efficient use of overlapping sequons for glycosylation. This article reviews the molecular mechanisms of these cellular processes. Based on this, we present hypotheses on the structure and variability of arteriviral membrane proteins and their role during virus entry and budding.
Collapse
Affiliation(s)
- Michael Veit
- Institut für Virologie, Veterinärmedizin, Freie Universität Berlin, Germany.
| | | | | | - Eberhard Krause
- Leibniz Institute of Molecular Pharmacology (FMP), Berlin, Germany
| | - Bastian Thaa
- Institut für Virologie, Veterinärmedizin, Freie Universität Berlin, Germany
| |
Collapse
|
132
|
Becares M, Sanchez CM, Sola I, Enjuanes L, Zuñiga S. Antigenic structures stably expressed by recombinant TGEV-derived vectors. Virology 2014; 464-465:274-286. [PMID: 25108114 PMCID: PMC7112069 DOI: 10.1016/j.virol.2014.07.027] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2014] [Revised: 06/17/2014] [Accepted: 07/17/2014] [Indexed: 11/21/2022]
Abstract
Coronaviruses (CoVs) are positive-stranded RNA viruses with potential as immunization vectors, expressing high levels of heterologous genes and eliciting both secretory and systemic immune responses. Nevertheless, its high recombination rate may result in the loss of the full-length foreign gene, limiting their use as vectors. Transmissible gastroenteritis virus (TGEV) was engineered to express porcine reproductive and respiratory syndrome virus (PRRSV) small protein domains, as a strategy to improve heterologous gene stability. After serial passage in tissue cultures, stable expression of small PRRSV protein antigenic domains was achieved. Therefore, size reduction of the heterologous genes inserted in CoV-derived vectors led to the stable expression of antigenic domains. Immunization of piglets with these TGEV vectors led to partial protection against a challenge with a virulent PRRSV strain, as immunized animals showed reduced clinical signs and lung damage. Further improvement of TGEV-derived vectors will require the engineering of vectors with decreased recombination rate.
Collapse
Affiliation(s)
- Martina Becares
- Centro Nacional de Biotecnología, CNB-CSIC, Department of Molecular and Cell Biology, Campus Universidad Autónoma de Madrid, Darwin 3, Madrid 28049, Spain
| | - Carlos M Sanchez
- Centro Nacional de Biotecnología, CNB-CSIC, Department of Molecular and Cell Biology, Campus Universidad Autónoma de Madrid, Darwin 3, Madrid 28049, Spain
| | - Isabel Sola
- Centro Nacional de Biotecnología, CNB-CSIC, Department of Molecular and Cell Biology, Campus Universidad Autónoma de Madrid, Darwin 3, Madrid 28049, Spain
| | - Luis Enjuanes
- Centro Nacional de Biotecnología, CNB-CSIC, Department of Molecular and Cell Biology, Campus Universidad Autónoma de Madrid, Darwin 3, Madrid 28049, Spain.
| | - Sonia Zuñiga
- Centro Nacional de Biotecnología, CNB-CSIC, Department of Molecular and Cell Biology, Campus Universidad Autónoma de Madrid, Darwin 3, Madrid 28049, Spain
| |
Collapse
|
133
|
Wu F, Peng K, Tian J, Xu X, Zhou E, Chen H. Immune Response to Fc Tagged GP5 Glycoproteins of Porcine Reproductive and Respiratory Syndrome Virus. Viral Immunol 2014; 27:343-9. [DOI: 10.1089/vim.2014.0041] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Affiliation(s)
- Fang Wu
- College of Life Sciences, Northwest A&F University, Yangling, People's Republic of China
| | - Kefeng Peng
- College of Life Sciences, Northwest A&F University, Yangling, People's Republic of China
| | - Jiao Tian
- College of Life Sciences, Northwest A&F University, Yangling, People's Republic of China
| | - Xiaodong Xu
- College of Life Sciences, Northwest A&F University, Yangling, People's Republic of China
| | - Enmin Zhou
- College of Veterinary Medicine, Northwest A&F University, Yangling, People's Republic of China
| | - Hongying Chen
- College of Life Sciences, Northwest A&F University, Yangling, People's Republic of China
| |
Collapse
|
134
|
García-Nicolás O, Quereda JJ, Gómez-Laguna J, Salguero FJ, Carrasco L, Ramis G, Pallarés FJ. Cytokines transcript levels in lung and lymphoid organs during genotype 1 Porcine Reproductive and Respiratory Syndrome Virus (PRRSV) infection. Vet Immunol Immunopathol 2014; 160:26-40. [DOI: 10.1016/j.vetimm.2014.03.008] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2013] [Revised: 03/10/2014] [Accepted: 03/18/2014] [Indexed: 12/24/2022]
|
135
|
Ren JQ, Sun WC, Lu HJ, Wen SB, Jing J, Yan FL, Liu H, Liu CX, Xiao PP, Chen X, Du SW, Du R, Jin NY. Construction and immunogenicity of a DNA vaccine coexpressing GP3 and GP5 of genotype-I porcine reproductive and respiratory syndrome virus. BMC Vet Res 2014; 10:128. [PMID: 24916952 PMCID: PMC4090398 DOI: 10.1186/1746-6148-10-128] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2013] [Accepted: 05/28/2014] [Indexed: 11/21/2022] Open
Abstract
Background The European (EU) genotype of porcine reproductive and respiratory syndrome virus (Genotype-I PRRSV) has recently emerged in China. The coexistence of Genotype-I and -II PRRSV strains could cause seriously affect PRRSV diagnosis and management. Current vaccines are not able to protect against PRRSV infection completely and have inherent drawbacks. Thus, genetically engineered vaccines, including DNA vaccine and live vector engineered vaccines, have been developed. This study aimed to determine the enhanced immune responses of mice inoculated with a DNA vaccine coexpressing GP3 and GP5 of a Genotype-I PRRSV. Results To evaluate the immunogenicity of GP3 and GP5 proteins from European-type PRRSV, three DNA vaccines, pVAX1-EU-ORF3-ORF5, pVAX1-EU-ORF3 and pVAX1-EU-ORF5, were constructed, which were based on a Genotype-I LV strain (GenBank ID: M96262). BALB/c mice were immunized with the DNA vaccines; delivered in the form of chitosan-DNA nanoparticles. To increase the efficiency of the vaccine, Quil A (Quillaja) was used as an adjuvant. GP3 and GP5-specific antibodies, neutralizing antibodies and cytokines (IL-2, IL-4, IL-10 and IFN gamma) from the immunized mice sera, and other immune parameters, were examined, including T-cell proliferation responses and subgroups of spleen T-lymphocytes. The results showed that ORF3 and ORF5 proteins of Genotype-I PRRSV induced GP3 and GP5-specific antibodies that could neutralize the virus. The levels of Cytokines IL-2, IL-4, IL-10, and IFN–γ of the experimental groups were significantly higher than those of control groups after booster vaccination (P < 0.05). The production of CD3+CD4+ and CD3+CD8+ T lymphocyte was also induced. T lymphocyte proliferation assays showed that the PRRSV LV strain virus could stimulate the proliferation of T lymphocytes in mice in the experimental group. Conclusions Using Quil A as adjuvant, Genotype-I PRRSV GP3 and GP5 proteins produced good immunogenicity and reactivity. More importantly, better PRRSV-specific neutralizing antibody titers and cell-mediated immune responses were observed in mice immunized with the DNA vaccine co-expressing GP3 and GP5 proteins than in mice immunized with a DNA vaccine expressing either protein singly. The results of this study demonstrated that co-immunization with GP3 and GP5 produced a better immune response in mice.
Collapse
Affiliation(s)
| | | | - Hui-Jun Lu
- Institute of Military Veterinary, Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Academy of Military Medical Sciences, Changchun 130122, China.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
136
|
Protective humoral immune response induced by an inactivated porcine reproductive and respiratory syndrome virus expressing the hypo-glycosylated glycoprotein 5. Vaccine 2014; 32:3617-22. [PMID: 24814552 DOI: 10.1016/j.vaccine.2014.04.083] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2013] [Revised: 04/21/2014] [Accepted: 04/23/2014] [Indexed: 11/22/2022]
Abstract
Porcine reproductive and respiratory syndrome (PRRS) causes significant economic losses to the swine industry worldwide. Although inactivated and live vaccines are commercially available for the control of PRRS, both types of vaccine have not always proven successful in terms of generating a protective immune response, particularly in the case of inactivated vaccines. In this study, we tested whether an inactivated vaccine could induce a humoral immune response to PRRS during a homologous challenge. Amino acid substitutions were introduced into glycoprotein (GP) 5 of the FL12 strain of the PRRS virus (PRRSV) using site-directed mutagenesis with a pFL12 infectious clone. The substitutions led to double deglycosylation in the putative glycosylation moieties on GP5. The mutant virus was subsequently inactivated with binary ethylenimine. The efficacy of the inactivated mutant virus was compared with that of the inactivated wild-type PRRSV. Only the inactivated mutant PRRSV induced serum neutralizing antibodies at six weeks post-vaccination. The group that was administered the inactivated mutant virus twice exhibited a significantly increased neutralizing antibody titer after a challenge with the virulent homologous strain and exhibited more rapid clearing of viremia compared to other groups, including the groups that were administered either the inactivated mutant or wild-type virus only once and the group that was administered the inactivated wild-type virus twice. Histopathological examination of lung tissue sections revealed that the group that was administered the inactivated mutant virus twice exhibited significantly thinner alveolar septa, whereas the thickness of the alveolar septa of the other groups were markedly increased due to lymphocyte infiltration. These results indicated that the deglycosylation of GP5 enhanced the immunogenicity of the inactivated mutant PRRSV and that twice administrations of the inactivated mutant virus conferred better protection against the homologous challenge. These findings suggest that the inactivated PRRSV that expresses a hypo-glycosylated GP5 is a potential inactivated vaccine candidate and a valuable tool for controlling PRRS for the swine industry.
Collapse
|
137
|
Efficacy of an attenuated European subtype 1 porcine reproductive and respiratory syndrome virus (PRRSV) vaccine in pigs upon challenge with the East European subtype 3 PRRSV strain Lena. Vaccine 2014; 32:2995-3003. [PMID: 24709589 DOI: 10.1016/j.vaccine.2014.03.077] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2013] [Revised: 03/13/2014] [Accepted: 03/25/2014] [Indexed: 11/21/2022]
Abstract
The efficacy of a commercial attenuated European subtype 1 PRRSV vaccine was evaluated upon challenge with the East European subtype 3 PRRSV strain Lena (83.3% nucleotide identity). Two vaccination experiments were carried out. Four- and seven-week-old pigs were vaccinated with the modified-live vaccine. Upon vaccination, virus specific IPMA antibodies were detected in all vaccinated animals with titers ranging from 10(2.8) to 10(4.6). No virus neutralizing (VN) antibodies were detected after vaccination. Eight (exp. 1) or six (exp. 2) weeks after vaccination, pigs were challenged with 10(6) (exp. 1) resp. 10(5) (exp. 2) TCID50 of the European subtype 3 PRRSV Lena. Upon challenge, non-vaccinated animals showed fever during 5.1 (exp. 1) or 7.7 (exp. 2) days. In vaccinated pigs, the duration of fever was reduced by 1.8 (exp. 1) or 3.5 (exp. 2) days. The modified-live virus vaccine reduced the mean duration of nasal shedding and viremia. In non-vaccinated pigs, virus shedding lasted 5.8 days (exp. 1), resp. 8.3 days (exp. 2). This period was reduced to 3.6 (exp. 1), resp. 3.0 (exp. 2) days in vaccinated animals. Viremia was observed during a shorter period in vaccinated (exp. 1: 7.4 days, exp. 2: 4.8 days) than in non-vaccinated groups (exp. 1: 11.8 days, exp. 2: 12.3 days). Starting from 5 days post challenge, virus titers in nasal secretions and sera were significantly lower in vaccinated animals (P<0.05). Virus-neutralizing antibodies were detected at low titers (≤ 16) after 7 days post challenge in vaccinated animals and 28 days post challenge in control animals. In conclusion, it can be stated that vaccination of pigs with an attenuated European subtype 1 vaccine provides a partial protection against a subsequent exposure to the highly pathogenic East European subtype 3 PRRSV strain Lena.
Collapse
|
138
|
Binjawadagi B, Dwivedi V, Manickam C, Ouyang K, Torrelles JB, Renukaradhya GJ. An innovative approach to induce cross-protective immunity against porcine reproductive and respiratory syndrome virus in the lungs of pigs through adjuvanted nanotechnology-based vaccination. Int J Nanomedicine 2014; 9:1519-35. [PMID: 24711701 PMCID: PMC3969340 DOI: 10.2147/ijn.s59924] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Porcine reproductive and respiratory syndrome (PRRS) is an economically devastating respiratory disease of pigs. The disease is caused by the PRRS virus (PRRSV), an Arterivirus which is a highly mutating RNA virus. Widely used modified live PRRSV vaccines have failed to prevent PRRS outbreaks and reinfections; moreover, safety of the live virus vaccines is questionable. Though poorly immunogenic, inactivated PRRSV vaccine is safe. The PRRSV infects primarily the lung macrophages. Therefore, we attempted to strengthen the immunogenicity of inactivated/killed PRRSV vaccine antigens (KAg), especially in the pig respiratory system, through: 1) entrapping the KAg in biodegradable poly(lactic-co-glycolic acid) nanoparticles (NP-KAg); 2) coupling the NP-KAg with a potent mucosal adjuvant, whole cell lysate of Mycobacterium tuberculosis (M. tb WCL); and 3) delivering the vaccine formulation twice intranasally to growing pigs. We have previously shown that a single dose of NP-KAg partially cleared the challenged heterologous PRRSV. Recently, we reported that NP-KAg coupled with unentrapped M. tb WCL significantly cleared the viremia of challenged heterologous PRRSV. Since PRRSV is primarily a lung disease, our goal in this study was to investigate lung viral load and various immune correlates of protection at the lung mucosal surfaces and its parenchyma in vaccinated heterologous PRRSV-challenged pigs. Our results indicated that out of five different vaccine-adjuvant formulations, the combination of NP-KAg and unentrapped M. tb WCL significantly cleared detectable replicating infective PRRSV with a tenfold reduction in viral RNA load in the lungs, associated with substantially reduced gross and microscopic lung pathology. Immunologically, strong humoral (enhanced virus neutralization titers by high avidity antibodies) and cell-mediated immune responses (augmented population of interferon-γ secreting CD4(+) and CD8(+) lymphocytes and reduced secretion of immunosuppressive cytokines) in the lungs were observed. In conclusion, combination of NP-KAg and soluble M. tb WCL elicits broadly cross-protective anti-PRRSV immunity in the pig respiratory system.
Collapse
Affiliation(s)
- Basavaraj Binjawadagi
- Food Animal Health Research Program, Ohio Agricultural Research and Development Center, Wooster, OH, USA ; Department of Veterinary Preventive Medicine, The Ohio State University, Wooster, OH, USA
| | - Varun Dwivedi
- Food Animal Health Research Program, Ohio Agricultural Research and Development Center, Wooster, OH, USA
| | - Cordelia Manickam
- Food Animal Health Research Program, Ohio Agricultural Research and Development Center, Wooster, OH, USA ; Department of Veterinary Preventive Medicine, The Ohio State University, Wooster, OH, USA
| | - Kang Ouyang
- Food Animal Health Research Program, Ohio Agricultural Research and Development Center, Wooster, OH, USA
| | - Jordi B Torrelles
- Department of Microbial Infection and Immunity, The Ohio State University, Columbus, OH, USA
| | - Gourapura J Renukaradhya
- Food Animal Health Research Program, Ohio Agricultural Research and Development Center, Wooster, OH, USA ; Department of Veterinary Preventive Medicine, The Ohio State University, Wooster, OH, USA
| |
Collapse
|
139
|
Piron R, De Koker S, De Paepe A, Goossens J, Grooten J, Nauwynck H, Depicker A. Boosting in planta production of antigens derived from the porcine reproductive and respiratory syndrome virus (PRRSV) and subsequent evaluation of their immunogenicity. PLoS One 2014; 9:e91386. [PMID: 24614617 PMCID: PMC3948849 DOI: 10.1371/journal.pone.0091386] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2013] [Accepted: 02/10/2014] [Indexed: 12/22/2022] Open
Abstract
Porcine reproductive and respiratory syndrome (PRRS) is a disease of swine, caused by an arterivirus, the PRRS virus (PRRSV). This virus infects pigs worldwide and causes huge economic losses. Due to genetic drift, current vaccines are losing their power. Adaptable vaccines could provide a solution to this problem. This study aims at producing in planta a set of antigens derived from the PRRSV glycoproteins (GPs) to be included in a subunit vaccine. We selected the GP3, GP4 and GP5 and optimized these for production in an Arabidopsis seed platform by removing transmembrane domains (Tm) and/or adding stabilizing protein domains, such as the green fluorescent protein (GFP) and immunoglobulin (IgG) ‘Fragment crystallizable’ (Fc) chains. Accumulation of the GPs with and without Tm was low, reaching no more than 0.10% of total soluble protein (TSP) in homozygous seed. However, addition of stabilizing domains boosted accumulation up to a maximum of 2.74% of TSP when GFP was used, and albeit less effectively, also the Fc chains of the porcine IgG3 and murine IgG2a increased antigen accumulation, to 0.96% and 1.81% of TSP respectively, while the murine IgG3 Fc chain did not. Antigens with Tm were less susceptible to these manipulations to increase yield. All antigens were produced in the endoplasmic reticulum and accordingly, they carried high-mannose N-glycans. The immunogenicity of several of those antigens was assessed and we show that vaccination with purified antigens did elicit the production of antibodies with virus neutralizing activity in mice but not in pigs.
Collapse
Affiliation(s)
- Robin Piron
- Department of Plant Systems Biology, VIB, Ghent, Belgium
- Department of Plant Biotechnology and Bioinformatics, Ghent University, Ghent, Belgium
| | - Stefaan De Koker
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Annelies De Paepe
- Department of Plant Systems Biology, VIB, Ghent, Belgium
- Department of Plant Biotechnology and Bioinformatics, Ghent University, Ghent, Belgium
| | - Julie Goossens
- Department of Bioscience Engineering, VUB, Brussels, Belgium
| | - Johan Grooten
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Hans Nauwynck
- Department of Virology, Parasitology and Immunology, Ghent University, Ghent, Belgium
| | - Ann Depicker
- Department of Plant Systems Biology, VIB, Ghent, Belgium
- Department of Plant Biotechnology and Bioinformatics, Ghent University, Ghent, Belgium
- * E-mail:
| |
Collapse
|
140
|
Comparison of host immune responses to homologous and heterologous type II porcine reproductive and respiratory syndrome virus (PRRSV) challenge in vaccinated and unvaccinated pigs. BIOMED RESEARCH INTERNATIONAL 2014; 2014:416727. [PMID: 24719862 PMCID: PMC3955659 DOI: 10.1155/2014/416727] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/18/2013] [Revised: 01/05/2014] [Accepted: 01/08/2014] [Indexed: 01/08/2023]
Abstract
Porcine reproductive and respiratory syndrome (PRRS) is a high-consequence animal disease with current vaccines providing limited protection from infection due to the high degree of genetic variation of field PRRS virus. Therefore, understanding host immune responses elicited by different PRRSV strains will facilitate the development of more effective vaccines. Using IngelVac modified live PRRSV vaccine (MLV), its parental strain VR-2332, and the heterologous KS-06-72109 strain (a Kansas isolate of PRRSV), we compared immune responses induced by vaccination and/or PRRSV infection. Our results showed that MLV can provide complete protection from homologous virus (VR-2332) and partial protection from heterologous (KS-06) challenge. The protection was associated with the levels of PRRSV neutralizing antibodies at the time of challenge, with vaccinated pigs having higher titers to VR-2332 compared to KS-06 strain. Challenge strain did not alter the cytokine expression profiles in the serum of vaccinated pigs or subpopulations of T cells. However, higher frequencies of IFN-γ-secreting PBMCs were generated from pigs challenged with heterologous PRRSV in a recall response when PBMCs were re-stimulated with PRRSV. Thus, this study indicates that serum neutralizing antibody titers are associated with PRRSV vaccination-induced protection against homologous and heterologous challenge.
Collapse
|
141
|
Anti-idiotypic antibodies reduce efficacy of the attenuated vaccine against highly pathogenic PRRSV challenge. BMC Vet Res 2014; 10:39. [PMID: 24507659 PMCID: PMC3921987 DOI: 10.1186/1746-6148-10-39] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2013] [Accepted: 01/29/2014] [Indexed: 11/10/2022] Open
Abstract
Background The inability of current vaccines to provide effective protection against porcine reproductive and respiratory syndrome virus (PRRSV) infection is not fully understood. One of the reasons might be the presence of anti-idiotypic antibodies (Ab2s) to the envelope glycoprotein GP5 induced by PRRSV infection since our previous studies demonstrated the presence of auto-Ab2s (aAb2s) in pigs infected with PRRSV. To test this hypothesis, PRRSV negative piglets were injected with a monoclonal Ab2 (Mab2-5G2) and aAb2s that are specific for anti-GP5 antibody, vaccinated with the attenuated PRRSV vaccine CH-1R and then challenged with the highly pathogenic PRRSV HuN4 strain. The animals were evaluated for clinical signs, pathological changes of the thymus and lungs, viremia, levels of serum antibodies and cytokines. Results The piglets injected with Mab2-5G2 or aAb2, and who received the attenuated PRRSV vaccine CH-1R before challenge, produced high levels of anti-N antibodies, IL-2 and IL-4, but low levels of neutralizing antibodies. After PRRSV HuN4 challenge, the animals showed obvious clinical signs, including lung lesions, severe thymus atrophy and decreased production of IL-4 and higher level of viremia. Conclusion When anti-GP5 Ab2s are present, the use of attenuated PRRSV vaccine CH-1R against HP-PRRSV infection is not recommended. It can result in poor health status with pneumonia and thymus atrophy.
Collapse
|
142
|
Binjawadagi B, Dwivedi V, Manickam C, Ouyang K, Wu Y, Lee LJ, Torrelles JB, Renukaradhya GJ. Adjuvanted poly(lactic-co-glycolic) acid nanoparticle-entrapped inactivated porcine reproductive and respiratory syndrome virus vaccine elicits cross-protective immune response in pigs. Int J Nanomedicine 2014; 9:679-94. [PMID: 24493925 PMCID: PMC3908835 DOI: 10.2147/ijn.s56127] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Porcine reproductive and respiratory syndrome (PRRS), caused by the PRRS virus (PRRSV), is an economically devastating disease, causing daily losses of approximately $3 million to the US pork industry. Current vaccines have failed to completely prevent PRRS outbreaks. Recently, we have shown that poly(lactic-co-glycolic) acid (PLGA) nanoparticle-entrapped inactivated PRRSV vaccine (NP-KAg) induces a cross-protective immune response in pigs. To further improve its cross-protective efficacy, the NP-KAg vaccine formulation was slightly modified, and pigs were coadministered the vaccine twice intranasally with a potent adjuvant: Mycobacterium tuberculosis whole-cell lysate. In vaccinated virulent heterologous PRRSV-challenged pigs, the immune correlates in the blood were as follows: 1) enhanced PRRSV-specific antibody response with enhanced avidity of both immunoglobulin (Ig)-G and IgA isotypes, associated with augmented virus-neutralizing antibody titers; 2) comparable and increased levels of virus-specific IgG1 and IgG2 antibody subtypes and production of high levels of both T-helper (Th)-1 and Th2 cytokines, indicative of a balanced Th1–Th2 response; 3) suppressed immunosuppressive cytokine response; 4) increased frequency of interferon-γ+ lymphocyte subsets and expanded population of antigen-presenting cells; and most importantly 5) complete clearance of detectable replicating challenged heterologous PRRSV and close to threefold reduction in viral ribonucleic acid load detected in the blood. In conclusion, intranasal delivery of adjuvanted NP-KAg vaccine formulation to growing pigs elicited a broadly cross-protective immune response, showing the potential of this innovative vaccination strategy to prevent PRRS outbreaks in pigs. A similar approach to control other respiratory diseases in food animals and humans appears to be feasible.
Collapse
Affiliation(s)
- Basavaraj Binjawadagi
- Food Animal Health Research Program, Ohio Agricultural Research and Development Center, Wooster, OH, USA ; Department of Veterinary Preventive Medicine, Ohio State University, Wooster, OH, USA
| | - Varun Dwivedi
- Food Animal Health Research Program, Ohio Agricultural Research and Development Center, Wooster, OH, USA
| | - Cordelia Manickam
- Food Animal Health Research Program, Ohio Agricultural Research and Development Center, Wooster, OH, USA ; Department of Veterinary Preventive Medicine, Ohio State University, Wooster, OH, USA
| | - Kang Ouyang
- Food Animal Health Research Program, Ohio Agricultural Research and Development Center, Wooster, OH, USA
| | - Yun Wu
- NanoScale Science and Engineering Center for Affordable Nanoengineering of Polymeric Biomedical Devices, Columbus, OH, USA
| | - Ly James Lee
- NanoScale Science and Engineering Center for Affordable Nanoengineering of Polymeric Biomedical Devices, Columbus, OH, USA
| | - Jordi B Torrelles
- Department of Microbial Infection and Immunity, Ohio State University, Columbus, OH, USA
| | - Gourapura J Renukaradhya
- Food Animal Health Research Program, Ohio Agricultural Research and Development Center, Wooster, OH, USA ; Department of Veterinary Preventive Medicine, Ohio State University, Wooster, OH, USA
| |
Collapse
|
143
|
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
|
144
|
Islam ZU, Bishop SC, Savill NJ, Rowland RRR, Lunney JK, Trible B, Doeschl-Wilson AB. Quantitative analysis of porcine reproductive and respiratory syndrome (PRRS) viremia profiles from experimental infection: a statistical modelling approach. PLoS One 2013; 8:e83567. [PMID: 24358295 PMCID: PMC3866253 DOI: 10.1371/journal.pone.0083567] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2013] [Accepted: 11/13/2013] [Indexed: 11/18/2022] Open
Abstract
Porcine reproductive and respiratory syndrome (PRRS) is one of the most economically significant viral diseases facing the global swine industry. Viremia profiles of PRRS virus challenged pigs reflect the severity and progression of infection within the host and provide crucial information for subsequent control measures. In this study we analyse the largest longitudinal PRRS viremia dataset from an in-vivo experiment. The primary objective was to provide a suitable mathematical description of all viremia profiles with biologically meaningful parameters for quantitative analysis of profile characteristics. The Wood's function, a gamma-type function, and a biphasic extended Wood's function were fit to the individual profiles using Bayesian inference with a likelihood framework. Using maximum likelihood inference and numerous fit criteria, we established that the broad spectrum of viremia trends could be adequately represented by either uni- or biphasic Wood's functions. Three viremic categories emerged: cleared (uni-modal and below detection within 42 days post infection(dpi)), persistent (transient experimental persistence over 42 dpi) and rebound (biphasic within 42 dpi). The convenient biological interpretation of the model parameters estimates, allowed us not only to quantify inter-host variation, but also to establish common viremia curve characteristics and their predictability. Statistical analysis of the profile characteristics revealed that persistent profiles were distinguishable already within the first 21 dpi, whereas it is not possible to predict the onset of viremia rebound. Analysis of the neutralizing antibody(nAb) data indicated that there was a ubiquitous strong response to the homologous PRRSV challenge, but high variability in the range of cross-protection of the nAbs. Persistent pigs were found to have a significantly higher nAb cross-protectivity than pigs that either cleared viremia or experienced rebound within 42 dpi. Our study provides novel insights into the nature and degree of variation of hosts' responses to infection as well as new informative traits for subsequent genomic and modelling studies.
Collapse
Affiliation(s)
- Zeenath U. Islam
- The Roslin Institute & R(D)SVS, University of Edinburgh, Edinburgh, Midlothian, United Kingdom
- * E-mail:
| | - Stephen C. Bishop
- The Roslin Institute & R(D)SVS, University of Edinburgh, Edinburgh, Midlothian, United Kingdom
| | - Nicholas J. Savill
- Institute of Immunology and Infection Research, University of Edinburgh, Edinburgh, United Kingdom
| | - Raymond R. R. Rowland
- Department of Diagnostic Medicine and Pathobiology, College of Veterinary Medicine, Kansas State University, Manhattan, Kansas, United States of America
| | - Joan K. Lunney
- United State Department of Agriculture, Beltsville Agricultural Research Center, Beltsville, Maryland, United States of America
| | - Benjamin Trible
- Department of Diagnostic Medicine and Pathobiology, College of Veterinary Medicine, Kansas State University, Manhattan, Kansas, United States of America
| | | |
Collapse
|
145
|
Wang ZH, Cao XH, Du XG, Feng HB, Zeng XY. Mucosal and systemic immunity in mice after intranasal immunization with recombinant Lactococcus lactis expressing ORF6 of PRRSV. Cell Immunol 2013; 287:69-73. [PMID: 24423464 DOI: 10.1016/j.cellimm.2013.12.004] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2013] [Revised: 11/03/2013] [Accepted: 12/05/2013] [Indexed: 11/18/2022]
Abstract
The purpose of the study was to construct mucosal vaccine of a recombinant Lactococcus lactis expressing PRRSV ORF6 gene and evaluate mucosal and systemic immune response against PRRSV in mice after intranasal immunization. The result show that the vaccine can stimulate mice to produce specific IgG in serum and remarkable special s-IgA in lung lavage fluid, at the same time, the contents of cytokines IL-2 and IFN-γ of the experimental group were significant higher than those of the control group (P < 0.01), however, the contents of cytokines IL-4 was not different to the all groups. In summary, the constructed mucosal vaccine can significantly induce mucosal immune, humoral immunity and cellular immunity involved Th1 type cytokines, which will lay a theoretical foundation on immune mechanism and new efficient vaccines for PRRSV.
Collapse
Affiliation(s)
- Zhen-hua Wang
- Department of Animal and Veterinary Science, Chengdu Vocational College of Agricultural Science and Technology, WenJiang, Sichuan 611130, PR China
| | - Xiao-han Cao
- Isotope Research Laboratory, College of Life and Basic Sciences, Sichuan Agricultural University, Xin Kang Road 46, Ya'an, Sichuan 625014, PR China
| | - Xiao-gang Du
- Isotope Research Laboratory, College of Life and Basic Sciences, Sichuan Agricultural University, Xin Kang Road 46, Ya'an, Sichuan 625014, PR China
| | - Hai-bo Feng
- Department of Veterinary Medicine, Southwest University, Rongchang, Chongqing 402460, PR China
| | - Xian-yin Zeng
- Isotope Research Laboratory, College of Life and Basic Sciences, Sichuan Agricultural University, Xin Kang Road 46, Ya'an, Sichuan 625014, PR China.
| |
Collapse
|
146
|
Evaluation of Different DNA Vaccines against Porcine Reproductive and Respiratory Syndrome (PRRS) in Pigs. Vaccines (Basel) 2013; 1:463-80. [PMID: 26344342 PMCID: PMC4494207 DOI: 10.3390/vaccines1040463] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2013] [Revised: 09/10/2013] [Accepted: 10/09/2013] [Indexed: 01/16/2023] Open
Abstract
In veterinary medicine, there have been different experiences with the plasmid DNA vaccination. In this area and with the hypothesis to demonstrate the effectiveness of different plasmids encoding porcine respiratory and reproductive syndrome (PRRS), five DNA vaccines against PRRS were evaluated for their innocuity and efficacy in pigs. Eighteen animals were divided into five groups which were injected with five (A, B, C, D, E) different DNA vaccines. Albeit, none of the proposed vaccines were able to protect the animals against PRRS virus. Only vaccines A and B were able to reduce the clinical signs of the infection. ELISA IgM were detected 30 days after the first vaccination in the pigs injected by Vaccine A or B. ELISA IgG were detected 90 days after the first vaccination in the pigs injected by Vaccine B or C. Neutralizing antibody were detected Post Challenge Days 61 (PCD) in all groups. In the pigs inoculated with Vaccine C, IFN-g were detected 90 days after first vaccination, and after challenge exposure they increased. In the other groups, the IFN-g were detected after challenge infection. Pigs injected with each of the vaccines A, B, C, D and E showed a significantly higher level of CD4(-)CD8⁺ lymphocytes (p < 0.001) after infection in comparison with their controls.
Collapse
|
147
|
Robinson SR, Abrahante JE, Johnson CR, Murtaugh MP. Purifying selection in porcine reproductive and respiratory syndrome virus ORF5a protein influences variation in envelope glycoprotein 5 glycosylation. INFECTION GENETICS AND EVOLUTION 2013; 20:362-8. [PMID: 24084290 DOI: 10.1016/j.meegid.2013.09.022] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/15/2013] [Revised: 09/05/2013] [Accepted: 09/21/2013] [Indexed: 02/06/2023]
Abstract
Porcine reproductive and respiratory syndrome virus ORF5a protein is encoded in an alternate open reading frame upstream of the major envelope glycoprotein (GP5) in subgenomic mRNA5. Bioinformatic analysis of 3466 type 2 PRRSV sequences showed that the two proteins have co-evolved through a fine balance of purifying codon usage to maintain a conserved RQ-rich motif in ORF5a protein, while eliciting a variable N-linked glycosylation motif in the alternative GP5 reading frame. Conservation of the ORF5a protein RQ-motif also explains an anomalous uracil desert in GP5 hypervariable glycosylation region. The N-terminus of the mature GP5 protein was confirmed to start with amino acid 32, the hypervariable region of the ectodomain. Since GP5 glycosylation variability is assumed to result from immunological selection against neutralizing antibodies, these findings show that an alternative possibility unrelated to immunological selection not only exists, but provides a foundation for investigating previously unsuspected aspects of PRRSV biology. Understanding functional consequences of subtle nucleotide sequence modifications in the region responsible for critical function in ORF5a protein and GP5 glycosylation is essential for rational design of new vaccines against PRRS.
Collapse
Affiliation(s)
- Sally R Robinson
- Department of Veterinary and Biomedical Sciences, University of Minnesota, 1971 Commonwealth Avenue, St. Paul, MN 55108, USA.
| | | | | | | |
Collapse
|
148
|
Geldhof MF, Van Breedam W, De Jong E, Lopez Rodriguez A, Karniychuk UU, Vanhee M, Van Doorsselaere J, Maes D, Nauwynck HJ. Antibody response and maternal immunity upon boosting PRRSV-immune sows with experimental farm-specific and commercial PRRSV vaccines. Vet Microbiol 2013; 167:260-71. [PMID: 24041768 DOI: 10.1016/j.vetmic.2013.08.017] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2013] [Revised: 08/14/2013] [Accepted: 08/19/2013] [Indexed: 11/17/2022]
Abstract
The porcine reproductive and respiratory syndrome virus (PRRSV) causes reproductive failure in sows and respiratory disease in pigs of all ages. Despite the frequent use of vaccines to maintain PRRSV immunity in sows, little is known on how the currently used vaccines affect the immunity against currently circulating and genetically divergent PRRSV variants in PRRSV-immune sows, i.e. sows that have a pre-existing PRRSV-specific immunity due to previous infection with or vaccination against the virus. Therefore, this study aimed to assess the capacity of commercially available attenuated/inactivated PRRSV vaccines and autogenous inactivated PRRSV vaccines - prepared according to a previously optimized in-house protocol - to boost the antibody immunity against currently circulating PRRSV variants in PRRSV-immune sows. PRRSV isolates were obtained from 3 different swine herds experiencing PRRSV-related problems, despite regular vaccination of gilts and sows against the virus. In a first part of the study, the PRRSV-specific antibody response upon booster vaccination with commercial PRRSV vaccines and inactivated farm-specific PRRSV vaccines was evaluated in PRRSV-immune, non-pregnant replacement sows from the 3 herds. A boost in virus-neutralizing antibodies against the farm-specific isolate was observed in all sow groups vaccinated with the corresponding farm-specific inactivated vaccines. Use of the commercial attenuated EU type vaccine boosted neutralizing antibodies against the farm-specific isolate in sows derived from 2 farms, while use of the commercial attenuated NA type vaccine did not boost farm-specific virus-neutralizing antibodies in any of the sow groups. Interestingly, the commercial inactivated EU type vaccine boosted farm-specific virus-neutralizing antibodies in sows from 1 farm. In the second part of the study, a field trial was performed at one of the farms to evaluate the booster effect of an inactivated farm-specific vaccine and a commercial attenuated EU-type vaccine in immune sows at 60 days of gestation. The impact of this vaccination on maternal immunity and on the PRRSV infection pattern in piglets during their first weeks of life was evaluated. Upon vaccination with the farm-specific inactivated vaccine, a significant increase in farm-specific virus-neutralizing antibodies was detected in all sows. Virus-neutralizing antibodies were also transferred to the piglets via colostrum and were detectable in the serum of these animals until 5 weeks after parturition. In contrast, not all sows vaccinated with the commercial attenuated vaccine showed an increase in farm-specific virus-neutralizing antibodies and the piglets of this group generally had lower virus-neutralizing antibody titers. Interestingly, the number of viremic animals (i.e. animals that have infectious virus in their bloodstream) was significantly lower among piglets of both vaccinated groups than among piglets of mock-vaccinated sows and this at least until 9 weeks after parturition. The results of this study indicate that inactivated farm-specific PRRSV vaccines and commercial attenuated vaccines can be useful tools to boost PRRSV-specific (humoral) immunity in sows and reduce viremia in weaned piglets.
Collapse
Affiliation(s)
- Marc F Geldhof
- Department of Virology, Parasitology and Immunology, Faculty of Veterinary Medicine, Ghent University, Salisburylaan 133, 9820 Merelbeke, Belgium.
| | | | | | | | | | | | | | | | | |
Collapse
|
149
|
Abstract
Arteriviruses are positive-stranded RNA viruses that infect mammals. They can cause persistent or asymptomatic infections, but also acute disease associated with a respiratory syndrome, abortion or lethal haemorrhagic fever. During the past two decades, porcine reproductive and respiratory syndrome virus (PRRSV) and, to a lesser extent, equine arteritis virus (EAV) have attracted attention as veterinary pathogens with significant economic impact. Particularly noteworthy were the 'porcine high fever disease' outbreaks in South-East Asia and the emergence of new virulent PRRSV strains in the USA. Recently, the family was expanded with several previously unknown arteriviruses isolated from different African monkey species. At the molecular level, arteriviruses share an intriguing but distant evolutionary relationship with coronaviruses and other members of the order Nidovirales. Nevertheless, several of their characteristics are unique, including virion composition and structure, and the conservation of only a subset of the replicase domains encountered in nidoviruses with larger genomes. During the past 15 years, the advent of reverse genetics systems for EAV and PRRSV has changed and accelerated the structure-function analysis of arterivirus RNA and protein sequences. These systems now also facilitate studies into host immune responses and arterivirus immune evasion and pathogenesis. In this review, we have summarized recent advances in the areas of arterivirus genome expression, RNA and protein functions, virion architecture, virus-host interactions, immunity, and pathogenesis. We have also briefly reviewed the impact of these advances on disease management, the engineering of novel candidate live vaccines and the diagnosis of arterivirus infection.
Collapse
Affiliation(s)
- Eric J Snijder
- Molecular Virology Department, Department of Medical Microbiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Marjolein Kikkert
- Molecular Virology Department, Department of Medical Microbiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Ying Fang
- Department of Diagnostic Medicine and Pathobiology, College of Veterinary Medicine, Kansas State University, Manhattan, Kansas, USA.,Department of Veterinary and Biomedical Sciences, South Dakota State University, Brookings, South Dakota, USA
| |
Collapse
|
150
|
Ooms K, Van Gorp H, Botti S, Van Gaever T, Delputte PL, Nauwynck HJ. Evaluation of viral peptide targeting to porcine sialoadhesin using a porcine reproductive and respiratory syndrome virus vaccination-challenge model. Virus Res 2013; 177:147-55. [PMID: 23932898 DOI: 10.1016/j.virusres.2013.07.019] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2013] [Revised: 07/22/2013] [Accepted: 07/29/2013] [Indexed: 10/26/2022]
Abstract
Targeting antigens to professional antigen presenting cells resident at the sites where effective immune responses are generated is a promising vaccination strategy. As such, targeting sialoadhesin (Sn)-expressing macrophages, abundantly present in spleen and lymph nodes where they appear to be strategically placed for antigen capture and processing, is recently gaining increased attention. Previously, we have shown that humoral immune responses to the model antigen human serum albumin can be enhanced by using a porcine Sn-specific monoclonal antibody to target the model antigen to Sn-expressing macrophages. To date however, no studies have been performed to evaluate whether targeted delivery of a pathogen-derived antigen can enhance the pathogen-specific immune response. Therefore, we selected a linear epitope on glycoprotein 4 of porcine reproductive and respiratory syndrome virus (PRRSV), which is known to be a target of virus-neutralizing antibodies. This paper reports on the targeted delivery of this viral peptide to porcine Sn-expressing macrophages and the evaluation of the subsequent immune response in a vaccination-challenge set-up. Four copies of the selected PRRSV epitope were genetically fused to a previously developed porcine Sn-targeting recombinant antibody or an irrelevant isotype control. Fusion proteins were shown to be efficiently purified from HEK293T cell supernatants and subsequently, only Sn-specific fusion proteins were shown to bind to and to be internalized into Sn-expressing cells. Subsequent immunizations with a single dose of the fusion proteins showed that peptide-specific immune responses and neutralizing antibody responses after PRRSV challenge were enhanced in animals receiving a single 500 μg intramuscular dose of the Sn-targeting fusion protein, although correlations between the two read-outs were hard to effectuate. Furthermore, a minor beneficial effect on viral clearance was observed. Together, these data show that viral peptide targeting to porcine Sn-expressing macrophages can improve the anti-viral immune response, although more research will be needed to further explore vaccination potential.
Collapse
Affiliation(s)
- Karen Ooms
- Laboratory of Virology, Department of Virology, Parasitology and Immunology, Faculty of Veterinary Medicine, Ghent University, Salisburylaan 133, 9820 Merelbeke, Belgium.
| | | | | | | | | | | |
Collapse
|