1
|
Noll JCG, Rani R, Butt SL, Fernandes MHV, do Nascimento GM, Martins M, Caserta LC, Covaleda L, Diel DG. Identification of an Immunodominant B-Cell Epitope in African Swine Fever Virus p30 Protein and Evidence of p30 Antibody-Mediated Antibody Dependent Cellular Cytotoxicity. Viruses 2024; 16:758. [PMID: 38793639 PMCID: PMC11125664 DOI: 10.3390/v16050758] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Revised: 04/12/2024] [Accepted: 04/16/2024] [Indexed: 05/26/2024] Open
Abstract
African Swine Fever Virus (ASFV) is a large dsDNA virus that encodes at least 150 proteins. The complexity of ASFV and lack of knowledge of effector immune functions and protective antigens have hindered the development of safe and effective ASF vaccines. In this study, we constructed four Orf virus recombinant vectors expressing individual ASFV genes B602L, -CP204L, E184L, and -I73R (ORFVΔ121-ASFV-B602L, -CP204L, -E184L, and -I73R). All recombinant viruses expressed the heterologous ASFV proteins in vitro. We then evaluated the immunogenicity of the recombinants by immunizing four-week-old piglets. In two independent animal studies, we observed high antibody titers against ASFV p30, encoded by CP204L gene. Using Pepscan ELISA, we identified a linear B-cell epitope of 12 amino acids in length (Peptide 15) located in an exposed loop region of p30 as an immunodominant ASFV epitope. Additionally, antibodies elicited against ASFV p30 presented antibody-dependent cellular cytotoxicity (ADCC) activity. These results underscore the role of p30 on antibody responses elicited against ASFV and highlight an important functional epitope that contributes to p30-specific antibody responses.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Diego G. Diel
- Department of Population Medicine and Diagnostic Sciences, College of Veterinary Medicine, Cornell University, Ithaca, NY 14853, USA (S.L.B.); (M.H.V.F.); (M.M.); (L.C.C.); (L.C.)
| |
Collapse
|
2
|
do Nascimento GM, de Oliveira PSB, Butt SL, Diel DG. Immunogenicity of chimeric hemagglutinins delivered by an orf virus vector platform against swine influenza virus. Front Immunol 2024; 15:1322879. [PMID: 38482020 PMCID: PMC10933025 DOI: 10.3389/fimmu.2024.1322879] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Accepted: 01/22/2024] [Indexed: 04/05/2024] Open
Abstract
Orf virus (ORFV) is a large DNA virus that can harbor and efficiently deliver viral antigens in swine. Here we used ORFV as a vector platform to deliver chimeric hemagglutinins (HA) of Influenza A virus of swine (IAV-S). Vaccine development against IAV-S faces limitations posed by strain-specific immunity and the antigenic diversity of the IAV-S strains circulating in the field. A promising alternative aiming at re-directing immune responses on conserved epitopes of the stalk segment of the hemagglutinin (HA2) has recently emerged. Sequential immunization with chimeric HAs comprising the same stalk but distinct exotic head domains can potentially induce cross-reactive immune responses against conserved epitopes of the HA2 while breaking the immunodominance of the head domain (HA1). Here, we generated two recombinant ORFVs expressing chimeric HAs encoding the stalk region of a contemporary H1N1 IAV-S strain and exotic heads derived from either H6 or H8 subtypes, ORFVΔ121cH6/1 and ORFVΔ121cH8/1, respectively. The resulting recombinant viruses were able to express the heterologous protein in vitro. Further, the immunogenicity and cross-protection of these vaccine candidates were assessed in swine after sequential intramuscular immunization with OV-cH6/1 and OV-cH8/1, and subsequent challenge with divergent IAV-S strains. Humoral responses showed that vaccinated piglets presented increasing IgG responses in sera. Additionally, cross-reactive IgG and IgA antibody responses elicited by immunization were detected in sera and bronchoalveolar lavage (BAL), respectively, by ELISA against different viral clades and a diverse range of contemporary H1N1 IAV-S strains, indicating induction of humoral and mucosal immunity in vaccinated animals. Importantly, viral shedding was reduced in nasal swabs from vaccinated piglets after intranasal challenge with either Oh07 (gamma clade) or Ca09 (npdm clade) IAV-S strains. These results demonstrated the efficiency of ORFV-based vectors in delivering chimeric IAV-S HA-based vaccine candidates and underline the potential use of chimeric-HAs for prevention and control of influenza in swine.
Collapse
Affiliation(s)
- Gabriela Mansano do Nascimento
- Department of Population Medicine and Diagnostic Sciences, College of Veterinary Medicine, Cornell University, Ithaca, NY, United States
| | - Pablo Sebastian Britto de Oliveira
- Department of Population Medicine and Diagnostic Sciences, College of Veterinary Medicine, Cornell University, Ithaca, NY, United States
- Programa de Pós-graduação em Medicina Veterinária, Universidade Federal de Santa Maria, Santa Maria, Rio Grande do Sul, Brazil
| | - Salman Latif Butt
- Department of Population Medicine and Diagnostic Sciences, College of Veterinary Medicine, Cornell University, Ithaca, NY, United States
| | - Diego G. Diel
- Department of Population Medicine and Diagnostic Sciences, College of Veterinary Medicine, Cornell University, Ithaca, NY, United States
| |
Collapse
|
3
|
Gao K, Li H, Lei X, Sun Z, Zheng T, Chen M, Ning Z. Recombinant Orf virus induced antibody production against capsid protein of porcine circovirus type 3 in mice. Vet Res Commun 2023; 47:2071-2081. [PMID: 37421550 DOI: 10.1007/s11259-023-10169-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Accepted: 07/03/2023] [Indexed: 07/10/2023]
Abstract
The emerging worldwide distributed porcine circovirus type 3 (PCV3) infection poses a serious threat to swine herds. An important means of preventing and controlling PCV3 infection is the development of the vaccine, while, the inability to cultivate in vitro has become the biggest obstacle. Orf virus (ORFV), the prototypic member of the Parapoxviridae, has been proven to be a novel valid vaccine vector for preparing various candidate vaccines. Here, recombinant ORFV expressing capsid protein (Cap) of PCV3 was obtained and proved its favorable immunogenicity inducing antibody against Cap in BALB/c mice. Based on the enhanced green fluorescent protein (EGFP) as a selectable marker, the recombinant rORFVΔ132-PCV3Cap-EGFP was generated. Then, recombinant ORFV expressing Cap only, rORFVΔ132-PCV3Cap, was obtained based on rORFVΔ132-PCV3Cap-EGFP using a double homologous recombination method by screening single non-fluorescent virus plaque. Results of the western blot showed that the Cap can be detected in rORFVΔ132-PCV3Cap infected OFTu cells. The results of immune experiments in BALB/c mice indicated that a specific antibody against Cap of PCV3 in serum was induced by rORFVΔ132-PCV3Cap infection. The results presented here provide a candidate vaccine against PCV3 and a feasible technical platform for vaccine development based on ORFV.
Collapse
Affiliation(s)
- Kuipeng Gao
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, China
| | - Huizi Li
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, China
| | - Xiaoling Lei
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, China
| | - Zhenzhen Sun
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, China
| | - Tingting Zheng
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, China
| | - Ming Chen
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, China
| | - Zhangyong Ning
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, China.
- Maoming Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Maoming, 525000, China.
| |
Collapse
|
4
|
Joshi LR, do Nascimento GM, Diel DG. The transcriptome of the parapoxvirus Orf virus reveals novel promoters for heterologous gene expression by poxvirus vectors. Virology 2023; 587:109864. [PMID: 37595395 DOI: 10.1016/j.virol.2023.109864] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Revised: 07/24/2023] [Accepted: 08/03/2023] [Indexed: 08/20/2023]
Abstract
Orf virus (ORFV) has been used as a vaccine delivery vector for multiple animal species. Several strategies are being used to improve the immunogenicity and efficacy of ORFV vectors, including the use of poxviral promoter(s) with strong early and late activity capable of driving the expression of the heterologous genes for a prolonged time and eliciting a potent immune response. Here, we used RNA-sequencing (RNA-Seq) approach to analyze the transcriptome of ORFV during infection in primary ovine cells. Based on the transcriptional profile of individual ORFV genes, we identified ORFV promoters with strong early and late activity and have shown that they can be used to express heterologous genes in ORFV vectors. Our results show that the intergenic regulatory sequence containing core promoter sequences present upstream of ORF112 (p112) and ORF116 (p116) lead to markedly higher transgene expression than conventional poxviral promoters. Thus, these promoters are valuable alternatives to express transgenes in poxviral vectors.
Collapse
Affiliation(s)
- Lok R Joshi
- Department of Population Medicine and Diagnostic Sciences, College of Veterinary Medicine, Cornell University, Ithaca, USA
| | - Gabriela Mansano do Nascimento
- Department of Population Medicine and Diagnostic Sciences, College of Veterinary Medicine, Cornell University, Ithaca, USA
| | - Diego G Diel
- Department of Population Medicine and Diagnostic Sciences, College of Veterinary Medicine, Cornell University, Ithaca, USA.
| |
Collapse
|
5
|
Shen Z, Liu B, Zhu Z, Du J, Zhou Z, Pan C, Chen Y, Yin C, Luo Y, Li H, Chen X. Construction of a Triple-Gene Deletion Mutant of Orf Virus and Evaluation of Its Safety, Immunogenicity and Protective Efficacy. Vaccines (Basel) 2023; 11:vaccines11050909. [PMID: 37243014 DOI: 10.3390/vaccines11050909] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Revised: 04/17/2023] [Accepted: 04/25/2023] [Indexed: 05/28/2023] Open
Abstract
Contagious ecthyma is a zoonotic disease caused by the orf virus (ORFV). Since there is no specific therapeutic drug available, vaccine immunization is the main tool to prevent and control the disease. Previously, we have reported the construction of a double-gene deletion mutant of ORFV (rGS14ΔCBPΔGIF) and evaluated it as a vaccine candidate. Building on this previous work, the current study reports the construction of a new vaccine candidate, generated by deleting a third gene (gene 121) to generate ORFV rGS14ΔCBPΔGIFΔ121. The in vitro growth characteristics, as well as the in vivo safety, immunogenicity, and protective efficacy, were evaluated. RESULTS: There was a minor difference in viral replication and proliferation between ORFV rGS14ΔCBPΔGIFΔ121 and the other two strains. ORFV rGS14ΔCBPΔGIFΔ121 induced continuous differentiation of PBMC to CD4+T cells, CD8+T cells and CD80+CD86+ cells and caused mainly Th1-like cell-mediated immunity. By comparing the triple-gene deletion mutant with the parental strain and the double-gene deletion mutant, we found that the safety of both the triple-gene deletion mutant and the double-gene deletion mutant could reach 100% in goats, while the safety of parental virus was only 50% after continually observing immunized animals for 14 days. A virulent field strain of ORFV from an ORF scab was used in the challenge experiment by inoculating the virus to the hairless area of the inner thigh of immunized animals. The result showed that the immune protection rate of triple-gene deletion mutant, double-gene mutant, and the parental virus was 100%, 66.7%, and 28.6%, respectively. In conclusion, the safety, immunogenicity, and immune-protectivity of the triple-gene deletion mutant were greatly improved to 100%, making it an excellent vaccine candidate.
Collapse
Affiliation(s)
- Zhanning Shen
- Animal Science and Techology College, Beijing University of Agriculture, Beijing 102208, China
- China Institute of Veterinary Drug Control, Beijing 100081, China
| | - Bo Liu
- China Institute of Veterinary Drug Control, Beijing 100081, China
- International Atomic Energy Agency, Vienna International Centre, P.O. Box 100, A-1400 Vienna, Austria
| | - Zhen Zhu
- China Institute of Veterinary Drug Control, Beijing 100081, China
| | - Jige Du
- China Institute of Veterinary Drug Control, Beijing 100081, China
| | - Zhiyu Zhou
- China Institute of Veterinary Drug Control, Beijing 100081, China
| | - Chenfan Pan
- China Institute of Veterinary Drug Control, Beijing 100081, China
| | - Yong Chen
- China Institute of Veterinary Drug Control, Beijing 100081, China
| | - Chunsheng Yin
- China Institute of Veterinary Drug Control, Beijing 100081, China
| | - Yufeng Luo
- China Institute of Veterinary Drug Control, Beijing 100081, China
| | - Huanrong Li
- Animal Science and Techology College, Beijing University of Agriculture, Beijing 102208, China
| | - Xiaoyun Chen
- China Institute of Veterinary Drug Control, Beijing 100081, China
| |
Collapse
|
6
|
do Nascimento GM, Bugybayeva D, Patil V, Schrock J, Yadagiri G, Renukaradhya GJ, Diel DG. An Orf-Virus (ORFV)-Based Vector Expressing a Consensus H1 Hemagglutinin Provides Protection against Diverse Swine Influenza Viruses. Viruses 2023; 15:994. [PMID: 37112974 PMCID: PMC10147081 DOI: 10.3390/v15040994] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2023] [Revised: 04/12/2023] [Accepted: 04/14/2023] [Indexed: 04/29/2023] Open
Abstract
Influenza A viruses (IAV-S) belonging to the H1 subtype are endemic in swine worldwide. Antigenic drift and antigenic shift lead to a substantial antigenic diversity in circulating IAV-S strains. As a result, the most commonly used vaccines based on whole inactivated viruses (WIVs) provide low protection against divergent H1 strains due to the mismatch between the vaccine virus strain and the circulating one. Here, a consensus coding sequence of the full-length of HA from H1 subtype was generated in silico after alignment of the sequences from IAV-S isolates obtained from public databases and was delivered to pigs using the Orf virus (ORFV) vector platform. The immunogenicity and protective efficacy of the resulting ORFVΔ121conH1 recombinant virus were evaluated against divergent IAV-S strains in piglets. Virus shedding after intranasal/intratracheal challenge with two IAV-S strains was assessed by real-time RT-PCR and virus titration. Viral genome copies and infectious virus load were reduced in nasal secretions of immunized animals. Flow cytometry analysis showed that the frequency of T helper/memory cells, as well as cytotoxic T lymphocytes (CTLs), were significantly higher in the peripheral blood mononuclear cells (PBMCs) of the vaccinated groups compared to unvaccinated animals when they were challenged with a pandemic strain of IAV H1N1 (CA/09). Interestingly, the percentage of T cells was higher in the bronchoalveolar lavage of vaccinated animals in relation to unvaccinated animals in the groups challenged with a H1N1 from the gamma clade (OH/07). In summary, delivery of the consensus HA from the H1 IAV-S subtype by the parapoxvirus ORFV vector decreased shedding of infectious virus and viral load of IAV-S in nasal secretions and induced cellular protective immunity against divergent influenza viruses in swine.
Collapse
Affiliation(s)
- Gabriela Mansano do Nascimento
- Department of Population Medicine and Diagnostic Sciences, Animal Health Diagnostic Center, College of Veterinary Medicine, Cornell University, Ithaca, NY 14850, USA
| | - Dina Bugybayeva
- Department of Animal Sciences, Center for Food Animal Health, College of Food, Agricultural, and Environmental Sciences, The Ohio State University, Wooster, OH 44691, USA
| | - Veerupaxagouda Patil
- Department of Animal Sciences, Center for Food Animal Health, College of Food, Agricultural, and Environmental Sciences, The Ohio State University, Wooster, OH 44691, USA
| | - Jennifer Schrock
- Department of Animal Sciences, Center for Food Animal Health, College of Food, Agricultural, and Environmental Sciences, The Ohio State University, Wooster, OH 44691, USA
| | - Ganesh Yadagiri
- Department of Animal Sciences, Center for Food Animal Health, College of Food, Agricultural, and Environmental Sciences, The Ohio State University, Wooster, OH 44691, USA
| | - Gourapura J. Renukaradhya
- Department of Animal Sciences, Center for Food Animal Health, College of Food, Agricultural, and Environmental Sciences, The Ohio State University, Wooster, OH 44691, USA
| | - Diego G. Diel
- Department of Population Medicine and Diagnostic Sciences, Animal Health Diagnostic Center, College of Veterinary Medicine, Cornell University, Ithaca, NY 14850, USA
| |
Collapse
|
7
|
Natesan K, Isloor S, Vinayagamurthy B, Ramakrishnaiah S, Doddamane R, Fooks AR. Developments in Rabies Vaccines: The Path Traversed from Pasteur to the Modern Era of Immunization. Vaccines (Basel) 2023; 11:vaccines11040756. [PMID: 37112668 PMCID: PMC10147034 DOI: 10.3390/vaccines11040756] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2023] [Revised: 03/27/2023] [Accepted: 03/27/2023] [Indexed: 04/03/2023] Open
Abstract
Rabies is a disease of antiquity and has a history spanning millennia ever since the first interactions between humans and dogs. The alarming fatalities caused by this disease have triggered rabies prevention strategies since the first century BC. There have been numerous attempts over the past 100 years to develop rabies vaccineswith the goal of preventing rabies in both humans and animals. Thepre-Pasteurian vaccinologists, paved the way for the actual history of rabies vaccines with the development of first generation vaccines. Further improvements for less reactive and more immunogenic vaccines have led to the expansion of embryo vaccines, tissue culture vaccines, cell culture vaccines, modified live vaccines, inactivated vaccines, and adjuvanted vaccines. The adventof recombinant technology and reverse genetics have given insight into the rabies viral genome and facilitated genome manipulations, which in turn led to the emergence of next-generation rabies vaccines, such as recombinant vaccines, viral vector vaccines, genetically modified vaccines, and nucleic acid vaccines. These vaccines were very helpful in overcoming the drawbacks of conventional rabies vaccines with increased immunogenicity and clinical efficacies. The path traversed in the development of rabies vaccines from Pasteur to the modern era vaccines, though, faced numerous challenges;these pioneering works have formed the cornerstone for the generation of thecurrent successful vaccines to prevent rabies. In the future, advancements in the scientific technologies and research focus will definitely lay the path for much more sophisticated vaccine candidates for rabies elimination.
Collapse
Affiliation(s)
- Krithiga Natesan
- KVAFSU-CVA Rabies Diagnostic Laboratory, WOAH Reference Laboratory for Rabies, Department of Veterinary Microbiology, Veterinary College, KVAFSU, Hebbal, Bengaluru 560024, Karnataka, India
| | - Shrikrishna Isloor
- KVAFSU-CVA Rabies Diagnostic Laboratory, WOAH Reference Laboratory for Rabies, Department of Veterinary Microbiology, Veterinary College, KVAFSU, Hebbal, Bengaluru 560024, Karnataka, India
- Correspondence: ; Tel.: +91-9449992287
| | | | - Sharada Ramakrishnaiah
- KVAFSU-CVA Rabies Diagnostic Laboratory, WOAH Reference Laboratory for Rabies, Department of Veterinary Microbiology, Veterinary College, KVAFSU, Hebbal, Bengaluru 560024, Karnataka, India
| | - Rathnamma Doddamane
- KVAFSU-CVA Rabies Diagnostic Laboratory, WOAH Reference Laboratory for Rabies, Department of Veterinary Microbiology, Veterinary College, KVAFSU, Hebbal, Bengaluru 560024, Karnataka, India
| | - Anthony R. Fooks
- APHA Weybridge, Woodham Lane, New Haw, Addlestone, Surrey KT15 3NB, UK
| |
Collapse
|
8
|
Yu Y, Zhang F, Duan X, Yang C, Cui Y, Yu L. ORFV can carry TRAP gene expression via intracellular CRISPR/Cas9 gene editing technology. J Virol Methods 2023; 312:114652. [PMID: 36493528 DOI: 10.1016/j.jviromet.2022.114652] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Revised: 11/09/2022] [Accepted: 11/11/2022] [Indexed: 12/12/2022]
Abstract
Orf is an acute and highly contracted human and animal infection caused by orf virus (ORFV), which mainly affects sheep, goats, and other species. Clinically, opportunistic or conditional pathogens such as Staphylococcus aureus (S. aureus) are often detected in cases of orf, which greatly increases the risk of disease progression and clinical death. It has been reported that TRAP gene products of S. aureus can broadly influence bacterial life and pathogenicity in vivo, and introduction of exogenous TRAP genes may help to inhibit the proliferation of bacteria. In order to achieve the combined control of ORFV and S. aureus, a novel approach to design a S. aureus TRAP gene vaccine using a live attenuated ORFV vector is proposed. In this study, CRISPR/Cas9 gene editing technology was used to disable vascular endothelial growth factor E of ORFV (VEGF-v) and introduced TRAP gene into this position. TRAP gene expression was detected in keratinocytes infected with recombinant virus. The construction and experimental verification of recombinant ORFV (ORFV-v/TRAP) will provide a reference for in-depth studies on the prevention and control of mixed infectious disease.
Collapse
Affiliation(s)
- YongZhong Yu
- College of Biological Science and Technology, Heilongjiang Bayi Agricultural University, Daqing 163319, PR China.
| | - Fan Zhang
- College of Biological Science and Technology, Heilongjiang Bayi Agricultural University, Daqing 163319, PR China
| | - Xuyang Duan
- College of Biological Science and Technology, Heilongjiang Bayi Agricultural University, Daqing 163319, PR China.
| | - ChaoQun Yang
- College of Biological Science and Technology, Heilongjiang Bayi Agricultural University, Daqing 163319, PR China
| | - YuDong Cui
- College of Biological Science and Technology, Heilongjiang Bayi Agricultural University, Daqing 163319, PR China
| | - Li Yu
- Division of Livestock Infectious Diseases, State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, No. 427 Maduan Street, Harbin 150001, PR China
| |
Collapse
|
9
|
Lin J, Sun S, Zhao K, Gao F, Wang R, Li Q, Zhou Y, Zhang J, Li Y, Wang X, Du L, Wang S, Li Z, Lu H, Lan Y, Song D, Guo W, Chen Y, Gao F, Zhao Y, Fan R, Guan J, He W. Oncolytic Parapoxvirus induces Gasdermin E-mediated pyroptosis and activates antitumor immunity. Nat Commun 2023; 14:224. [PMID: 36641456 PMCID: PMC9840172 DOI: 10.1038/s41467-023-35917-2] [Citation(s) in RCA: 43] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Accepted: 01/06/2023] [Indexed: 01/15/2023] Open
Abstract
The advantage of oncolytic viruses (OV) in cancer therapy is their dual effect of directly killing tumours while prompting anti-tumour immune response. Oncolytic parapoxvirus ovis (ORFV) and other OVs are thought to induce apoptosis, but apoptosis, being the immunogenically inert compared to other types of cell death, does not explain the highly inflamed microenvironment in OV-challenged tumors. Here we show that ORFV and its recombinant therapeutic derivatives are able to trigger tumor cell pyroptosis via Gasdermin E (GSDME). This effect is especially prominent in GSDME-low tumor cells, in which ORFV-challenge pre-stabilizes GSDME by decreasing its ubiquitination and subsequently initiates pyroptosis. Consistently, GSDME depletion reduces the proportion of intratumoral cytotoxic T lymphocytes, pyroptotic cell death and the success of tumor ORFV virotherapy. In vivo, the OV preferentially accumulates in the tumour upon systemic delivery and elicits pyroptotic tumor killing. Consequentially, ORFV sensitizes immunologically 'cold' tumors to checkpoint blockade. This study thus highlights the critical role of GSDME-mediated pyroptosis in oncolytic ORFV-based antitumor immunity and identifies combinatorial cancer therapy strategies.
Collapse
Affiliation(s)
- Jing Lin
- Key Laboratory of Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Jilin University, 130062, Changchun, China
| | - Shihui Sun
- Key Laboratory of Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Jilin University, 130062, Changchun, China
| | - Kui Zhao
- Key Laboratory of Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Jilin University, 130062, Changchun, China
| | - Fei Gao
- Department of Laboratory Animals, College of Animal Science, Jilin University, 130062, Changchun, China
| | - Renling Wang
- Key Laboratory of Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Jilin University, 130062, Changchun, China
| | - Qi Li
- Key Laboratory of Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Jilin University, 130062, Changchun, China
| | - Yanlong Zhou
- Key Laboratory of Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Jilin University, 130062, Changchun, China
| | - Jing Zhang
- Key Laboratory of Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Jilin University, 130062, Changchun, China
| | - Yue Li
- Key Laboratory of Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Jilin University, 130062, Changchun, China
| | - Xinyue Wang
- Key Laboratory of Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Jilin University, 130062, Changchun, China
| | - Le Du
- Key Laboratory of Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Jilin University, 130062, Changchun, China
| | - Shuai Wang
- Key Laboratory of Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Jilin University, 130062, Changchun, China
| | - Zi Li
- Key Laboratory of Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Jilin University, 130062, Changchun, China
| | - Huijun Lu
- Key Laboratory of Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Jilin University, 130062, Changchun, China
| | - Yungang Lan
- Key Laboratory of Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Jilin University, 130062, Changchun, China
| | - Deguang Song
- Key Laboratory of Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Jilin University, 130062, Changchun, China
| | - Wei Guo
- Department of Hematology, The first hospital of Jilin University, 130021, Changchun, China
| | - Yujia Chen
- Department of Gastrointestinal Surgery, The first hospital of Jilin University, 130021, Changchun, China
| | - Feng Gao
- Key Laboratory of Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Jilin University, 130062, Changchun, China
| | - Yicheng Zhao
- Changchun University of Chinese Medicine, 130017, Changchun, China
| | - Rongrong Fan
- Department of Biosciences and Nutrition, Karolinska Institutet, 14183, Huddinge, Sweden
| | - Jiyu Guan
- Key Laboratory of Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Jilin University, 130062, Changchun, China.
| | - Wenqi He
- Key Laboratory of Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Jilin University, 130062, Changchun, China.
| |
Collapse
|
10
|
Deletion of gene OV132 attenuates Orf virus more effectively than gene OV112. Appl Microbiol Biotechnol 2023; 107:835-851. [PMID: 36484827 PMCID: PMC9734686 DOI: 10.1007/s00253-022-12323-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Revised: 11/27/2022] [Accepted: 11/30/2022] [Indexed: 12/14/2022]
Abstract
Orf virus (ORFV), a Parapoxvirus in Poxviridae, infects sheep and goats resulting in contagious pustular dermatitis. ORFV is regarded as a promising viral vector candidate for vaccine development and oncolytic virotherapy. Owing to their potential clinical application, safety concerns have become increasingly important. Deletion of either the OV132 (encoding vascular endothelial growth factor, VEGF) or OV112 (encoding the chemokine binding protein, CBP) genes reduced ORFV infectivity, which has been independently demonstrated in the NZ2 and NZ7 strains, respectively. This study revealed that the VEGF and CBP gene sequences of the local strain (TW/Hoping) shared a similarity of 47.01% with NZ2 and 90.56% with NZ7. Due to the high sequence divergence of these two immunoregulatory genes among orf viral strains, their contribution to the pathogenicity of Taiwanese ORFV isolates was comparatively characterized. Initially, two ORFV recombinants were generated, in which either the VEGF or CBP gene was deleted and replaced with the reporter gene EGFP. In vitro assays indicated that both the VEGF-deletion mutant ORFV-VEGFΔ-EGFP and the CBP deletion mutant ORFV-CBPΔ-EGFP were attenuated in cells. In particular, ORFV-VEGFΔ-EGFP significantly reduced plaque size and virus yield compared to ORFV-CBPΔ-EGFP and the wild-type control. Similarly, in vivo analysis revealed no virus yield in the goat skin biopsy infected by ORFV-VEGFΔ-EGFP, and significantly reduced the virus yield of ORFV-CBPΔ-EGFP relative to the wild-type control. These results confirmed the loss of virulence of both deletion mutants in the Hoping strain, whereas the VEGF-deletion mutant was more attenuated than the CBP deletion strain in both cell and goat models. KEY POINTS: • VEGF and CBP genes are crucial in ORFV pathogenesis in the TW/Hoping strain • The VEGF-deletion mutant virus was severely attenuated in both cell culture and animal models • Deletion mutant viruses are advantageous vectors for the development of vaccines and therapeutic regimens.
Collapse
|
11
|
Rabies Vaccine: Recent Update and Comprehensive Review of in vitro and in vivo Studies. Process Biochem 2022. [DOI: 10.1016/j.procbio.2022.11.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|
12
|
Joshi LR, Knudsen D, Piñeyro P, Dhakal S, Renukaradhya GJ, Diel DG. Protective Efficacy of an Orf Virus-Vector Encoding the Hemagglutinin and the Nucleoprotein of Influenza A Virus in Swine. Front Immunol 2021; 12:747574. [PMID: 34804030 PMCID: PMC8602839 DOI: 10.3389/fimmu.2021.747574] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Accepted: 09/30/2021] [Indexed: 01/19/2023] Open
Abstract
Swine influenza is a highly contagious respiratory disease of pigs caused by influenza A viruses (IAV-S). IAV-S causes significant economic losses to the swine industry and poses challenges to public health given its zoonotic potential. Thus effective IAV-S vaccines are needed and highly desirable and would benefit both animal and human health. Here, we developed two recombinant orf viruses, expressing the hemagglutinin (HA) gene (OV-HA) or the HA and the nucleoprotein (NP) genes of IAV-S (OV-HA-NP). The immunogenicity and protective efficacy of these two recombinant viruses were evaluated in pigs. Both OV-HA and OV-HA-NP recombinants elicited robust virus neutralizing antibody response in pigs, with higher levels of neutralizing antibodies (NA) being detected in OV-HA-NP-immunized animals pre-challenge infection. Although both recombinant viruses elicited IAV-S-specific T-cell responses, the frequency of IAV-S-specific proliferating CD8+ T cells upon re-stimulation was higher in OV-HA-NP-immunized animals than in the OV-HA group. Importantly, IgG1/IgG2 isotype ELISAs revealed that immunization with OV-HA induced Th2-biased immune responses, whereas immunization with OV-HA-NP virus resulted in a Th1-biased immune response. While pigs immunized with either OV-HA or OV-HA-NP were protected when compared to non-immunized controls, immunization with OV-HA-NP resulted in incremental protection against challenge infection as evidenced by a reduced secondary antibody response (NA and HI antibodies) following IAV-S challenge and reduced virus shedding in nasal secretions (lower viral RNA loads and frequency of animals shedding viral RNA and infectious virus), when compared to animals in the OV-HA group. Interestingly, broader cross neutralization activity was also observed in serum of OV-HA-NP-immunized animals against a panel of contemporary IAV-S isolates representing the major genetic clades circulating in swine. This study demonstrates the potential of ORFV-based vector for control of swine influenza virus in swine.
Collapse
Affiliation(s)
- Lok R Joshi
- Department of Population Medicine and Diagnostic Sciences, Animal Health Diagnostic Center, College of Veterinary Medicine, Cornell University, Ithaca, NY, United States.,Department of Veterinary and Biomedical Sciences, Animal Disease Research And Diagnostic Laboratory, South Dakota State University, Brookings, SD, United States
| | - David Knudsen
- Department of Veterinary and Biomedical Sciences, Animal Disease Research And Diagnostic Laboratory, South Dakota State University, Brookings, SD, United States
| | - Pablo Piñeyro
- Department of Veterinary Diagnostic and Production Animal Medicine, Iowa State University, Ames, IA, United States
| | - Santosh Dhakal
- Department of Veterinary Preventive Medicine, Center for Food Animal Health, Ohio State University, Wooster, OH, United States
| | - Gourapura J Renukaradhya
- Department of Veterinary Preventive Medicine, Center for Food Animal Health, Ohio State University, Wooster, OH, United States
| | - Diego G Diel
- Department of Population Medicine and Diagnostic Sciences, Animal Health Diagnostic Center, College of Veterinary Medicine, Cornell University, Ithaca, NY, United States.,Department of Veterinary and Biomedical Sciences, Animal Disease Research And Diagnostic Laboratory, South Dakota State University, Brookings, SD, United States
| |
Collapse
|
13
|
Bukar AM, Jesse FFA, Abdullah CAC, Noordin MM, Lawan Z, Mangga HK, Balakrishnan KN, Azmi MLM. Immunomodulatory Strategies for Parapoxvirus: Current Status and Future Approaches for the Development of Vaccines against Orf Virus Infection. Vaccines (Basel) 2021; 9:1341. [PMID: 34835272 PMCID: PMC8624149 DOI: 10.3390/vaccines9111341] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Revised: 08/25/2021] [Accepted: 08/25/2021] [Indexed: 11/17/2022] Open
Abstract
Orf virus (ORFV), the prototype species of the parapoxvirus genus, is the causative agent of contagious ecthyma, an extremely devastating skin disease of sheep, goats, and humans that causes enormous economic losses in livestock production. ORFV is known for its ability to repeatedly infect both previously infected and vaccinated sheep due to several immunomodulatory genes encoded by the virus that temporarily suppress host immunity. Therefore, the development of novel, safe and effective vaccines against ORFV infection is an important priority. Although, the commercially licensed live-attenuated vaccines have provided partial protection against ORFV infections, the attenuated viruses have been associated with major safety concerns. In addition to safety issues, the persistent reinfection of vaccinated animals warrants the need to investigate several factors that may affect vaccine efficacy. Perhaps, the reason for the failure of the vaccine is due to the long-term adaptation of the virus in tissue culture. In recent years, the development of vaccines against ORFV infection has achieved great success due to technological advances in recombinant DNA technologies, which have opened a pathway for the development of vaccine candidates that elicit robust immunity. In this review, we present current knowledge on immune responses elicited by ORFV, with particular attention to the effects of the viral immunomodulators on the host immune system. We also discuss the implications of strain variation for the development of rational vaccines. Finally, the review will also aim to demonstrate future strategies for the development of safe and efficient vaccines against ORFV infections.
Collapse
Affiliation(s)
- Alhaji Modu Bukar
- Department of Pathology and Microbiology, Faculty of Veterinary Medicine, Universiti Putra Malaysia, Serdang 43400, Selangor, Malaysia; (M.M.N.); (Z.L.); (H.K.M.); (K.N.B.)
- Department of Science Laboratory Technology, School Agriculture and Applied Sciences, Ramat Polytechnic Maiduguri, Maiduguri 1070, Borno, Nigeria
| | - Faez Firdaus Abdullah Jesse
- Department of Veterinary Clinical Studies, Faculty of Veterinary Medicine, Universiti Putra Malaysia, Serdang 43400, Selangor, Malaysia;
| | | | - Mustapha M. Noordin
- Department of Pathology and Microbiology, Faculty of Veterinary Medicine, Universiti Putra Malaysia, Serdang 43400, Selangor, Malaysia; (M.M.N.); (Z.L.); (H.K.M.); (K.N.B.)
| | - Zaharaddeen Lawan
- Department of Pathology and Microbiology, Faculty of Veterinary Medicine, Universiti Putra Malaysia, Serdang 43400, Selangor, Malaysia; (M.M.N.); (Z.L.); (H.K.M.); (K.N.B.)
| | - Hassana Kyari Mangga
- Department of Pathology and Microbiology, Faculty of Veterinary Medicine, Universiti Putra Malaysia, Serdang 43400, Selangor, Malaysia; (M.M.N.); (Z.L.); (H.K.M.); (K.N.B.)
| | - Krishnan Nair Balakrishnan
- Department of Pathology and Microbiology, Faculty of Veterinary Medicine, Universiti Putra Malaysia, Serdang 43400, Selangor, Malaysia; (M.M.N.); (Z.L.); (H.K.M.); (K.N.B.)
| | - Mohd-Lila Mohd Azmi
- Department of Pathology and Microbiology, Faculty of Veterinary Medicine, Universiti Putra Malaysia, Serdang 43400, Selangor, Malaysia; (M.M.N.); (Z.L.); (H.K.M.); (K.N.B.)
| |
Collapse
|
14
|
Martins M, Rodrigues FS, Joshi LR, Jardim JC, Flores MM, Weiblen R, Flores EF, Diel DG. Orf virus ORFV112, ORFV117 and ORFV127 contribute to ORFV IA82 virulence in sheep. Vet Microbiol 2021; 257:109066. [PMID: 33866062 DOI: 10.1016/j.vetmic.2021.109066] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Accepted: 04/06/2021] [Indexed: 11/17/2022]
Abstract
The parapoxvirus orf virus (ORFV) encodes several immunomodulatory proteins (IMPs) that modulate host innate and pro-inflammatory responses to infection. Using the ORFV IA82 strain as the parental virus, recombinant viruses with individual deletions in the genes encoding the IMPs chemokine binding protein (CBP; ORFV112), inhibitor of granulocyte-monocyte colony-stimulating factor and IL-2 (GIF, ORFV117) and interleukin 10 homologue (vIL-10; ORFV127) were generated and characterized in vitro and in vivo. The replication properties of the individual gene deletion viruses in cell culture was not affected comparing with the parental virus. To investigate the effect of the individual gene deletions in ORFV infection and pathogenesis, groups of four lambs were inoculated with each virus and were monitored thereafter. Lambs inoculated with either recombinant or with the parental ORFV developed characteristic lesions of contagious ecthyma. The onset, nature and severity of the lesions in the oral commissure were similar in all inoculated groups from the onset (3 days post-inoculation [pi]) to the peak of clinical lesions (days 11-13 pi). Nonetheless, from days 11-13 pi onwards, the oral lesions in lambs inoculated with the recombinant viruses regressed faster than the lesions produced by the parental virus. Similarly, the amount of virus shed in the lesions were equivalent among lambs of all groups up to day 15 pi, yet they were significantly higher in the parental virus group from day 16-21 pi. In conclusion, individual deletion of these IMP genes from the ORFV genome resulted in slight reduction in virulence in vivo, as evidenced by a reduction in the duration of the clinical disease and virus shedding.
Collapse
Affiliation(s)
- Mathias Martins
- Department of Veterinary and Biomedical Sciences, South Dakota State University, Brookings, SD, United States; Setor de Virologia, Departamento de Medicina Veterinária Preventiva, Universidade Federal de Santa Maria, Av. Roraima 1000, prédio 63A, Santa Maria, Rio Grande do Sul, 97105-900, Brazil; Laboratório de Virologia, Medicina Veterinária, Programa de Pós-Graduação em Sanidade e Produção Animal, Universidade do Oeste de Santa Catarina, Campus II, Rodovia Rovilho Bortoluzzi, SC 480, Km 3.5, Xanxere, Santa Catarina, 89820-000, Brazil; Department of Population Medicine and Diagnostic Sciences, Animal Health Diagnostic Center, College of Veterinary Medicine, Cornell University, 240 Farrier Rd, Ithaca, NY, 14853, United States; Laboratório de Patologia Veterinária, Departamento de Patologia, Universidade Federal de Santa Maria, Av. Roraima, 1000, Santa Maria, Rio Grande do Sul, 97105-900, Brazil
| | - Fernando S Rodrigues
- Department of Veterinary and Biomedical Sciences, South Dakota State University, Brookings, SD, United States; Laboratório de Patologia Veterinária, Departamento de Patologia, Universidade Federal de Santa Maria, Av. Roraima, 1000, Santa Maria, Rio Grande do Sul, 97105-900, Brazil
| | - Lok R Joshi
- Department of Veterinary and Biomedical Sciences, South Dakota State University, Brookings, SD, United States; Department of Population Medicine and Diagnostic Sciences, Animal Health Diagnostic Center, College of Veterinary Medicine, Cornell University, 240 Farrier Rd, Ithaca, NY, 14853, United States; Laboratório de Patologia Veterinária, Departamento de Patologia, Universidade Federal de Santa Maria, Av. Roraima, 1000, Santa Maria, Rio Grande do Sul, 97105-900, Brazil
| | - José C Jardim
- Department of Veterinary and Biomedical Sciences, South Dakota State University, Brookings, SD, United States; Laboratório de Patologia Veterinária, Departamento de Patologia, Universidade Federal de Santa Maria, Av. Roraima, 1000, Santa Maria, Rio Grande do Sul, 97105-900, Brazil
| | - Mariana M Flores
- Setor de Virologia, Departamento de Medicina Veterinária Preventiva, Universidade Federal de Santa Maria, Av. Roraima 1000, prédio 63A, Santa Maria, Rio Grande do Sul, 97105-900, Brazil; Laboratório de Patologia Veterinária, Departamento de Patologia, Universidade Federal de Santa Maria, Av. Roraima, 1000, Santa Maria, Rio Grande do Sul, 97105-900, Brazil
| | - Rudi Weiblen
- Setor de Virologia, Departamento de Medicina Veterinária Preventiva, Universidade Federal de Santa Maria, Av. Roraima 1000, prédio 63A, Santa Maria, Rio Grande do Sul, 97105-900, Brazil; Laboratório de Patologia Veterinária, Departamento de Patologia, Universidade Federal de Santa Maria, Av. Roraima, 1000, Santa Maria, Rio Grande do Sul, 97105-900, Brazil
| | - Eduardo F Flores
- Setor de Virologia, Departamento de Medicina Veterinária Preventiva, Universidade Federal de Santa Maria, Av. Roraima 1000, prédio 63A, Santa Maria, Rio Grande do Sul, 97105-900, Brazil; Laboratório de Patologia Veterinária, Departamento de Patologia, Universidade Federal de Santa Maria, Av. Roraima, 1000, Santa Maria, Rio Grande do Sul, 97105-900, Brazil.
| | - Diego G Diel
- Department of Veterinary and Biomedical Sciences, South Dakota State University, Brookings, SD, United States; Department of Population Medicine and Diagnostic Sciences, Animal Health Diagnostic Center, College of Veterinary Medicine, Cornell University, 240 Farrier Rd, Ithaca, NY, 14853, United States; Laboratório de Patologia Veterinária, Departamento de Patologia, Universidade Federal de Santa Maria, Av. Roraima, 1000, Santa Maria, Rio Grande do Sul, 97105-900, Brazil.
| |
Collapse
|
15
|
Struzik J, Szulc-Dąbrowska L. NF-κB as an Important Factor in Optimizing Poxvirus-Based Vaccines against Viral Infections. Pathogens 2020; 9:pathogens9121001. [PMID: 33260450 PMCID: PMC7760304 DOI: 10.3390/pathogens9121001] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Revised: 11/24/2020] [Accepted: 11/27/2020] [Indexed: 11/16/2022] Open
Abstract
Poxviruses are large dsDNA viruses that are regarded as good candidates for vaccine vectors. Because the members of the Poxviridae family encode numerous immunomodulatory proteins in their genomes, it is necessary to carry out certain modifications in poxviral candidates for vaccine vectors to improve the vaccine. Currently, several poxvirus-based vaccines targeted at viral infections are under development. One of the important aspects of the influence of poxviruses on the immune system is that they encode a large array of inhibitors of the nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB), which is the key element of both innate and adaptive immunity. Importantly, the NF-κB transcription factor induces the mechanisms associated with adaptive immunological memory involving the activation of effector and memory T cells upon vaccination. Since poxviruses encode various NF-κB inhibitor proteins, before the use of poxviral vaccine vectors, modifications that influence NF-κB activation and consequently affect the immunogenicity of the vaccine should be carried out. This review focuses on NF-κB as an essential factor in the optimization of poxviral vaccines against viral infections.
Collapse
|
16
|
Rall I, Amann R, Malberg S, Herden C, Rubbenstroth D. Recombinant Modified Vaccinia Virus Ankara (MVA) Vaccines Efficiently Protect Cockatiels Against Parrot Bornavirus Infection and Proventricular Dilatation Disease. Viruses 2019; 11:v11121130. [PMID: 31817690 PMCID: PMC6950018 DOI: 10.3390/v11121130] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2019] [Revised: 12/03/2019] [Accepted: 12/04/2019] [Indexed: 01/10/2023] Open
Abstract
Parrot bornaviruses (PaBVs) are the causative agents of proventricular dilatation disease (PDD), a chronic and often fatal neurologic disorder in Psittaciformes. The disease is widely distributed in private parrot collections and threatens breeding populations of endangered species. Thus, immunoprophylaxis strategies are urgently needed. In previous studies we demonstrated a prime-boost vaccination regime using modified vaccinia virus Ankara (MVA) and Newcastle disease virus (NDV) constructs expressing the nucleoprotein and phosphoprotein of PaBV-4 (MVA/PaBV-4 and NDV/PaBV-4, respectively) to protect cockatiels (Nymphicus hollandicus) against experimental challenge infection. Here we investigated the protective effect provided by repeated immunization with either MVA/PaBV-4, NDV/PaBV-4 or Orf virus constructs (ORFV/PaBV-4) individually. While MVA/PaBV-4-vaccinated cockatiels were completely protected against subsequent PaBV-2 challenge infection and PDD-associated lesions, the course of the challenge infection in NDV/PaBV-4- or ORFV/PaBV-4-vaccinated birds did not differ from the unvaccinated control group. We further investigated the effect of vaccination on persistently PaBV-4-infected cockatiels. Remarkably, subsequent immunization with MVA/PaBV-4 and NDV/PaBV-4 neither induced obvious immunopathogenesis exacerbating the disease nor reduced viral loads in the infected birds. In summary, we demonstrated that vaccination with MVA/PaBV-4 alone is sufficient to efficiently prevent PaBV-2 challenge infection in cockatiels, providing a suitable vaccine candidate against avian bornavirus infection and bornavirus-induced PDD.
Collapse
Affiliation(s)
- Isabell Rall
- Institute of Virology, Medical Center–University of Freiburg, Faculty of Medicine, University of Freiburg, Hermann-Herder-Str. 11, D-79104 Freiburg, Germany;
- Department of Immunology, Interfaculty Institute of Cell Biology, Eberhard Karls Universität Tübingen, Auf der Morgenstelle 15, D-72076 Tübingen, Germany;
| | - Ralf Amann
- Department of Immunology, Interfaculty Institute of Cell Biology, Eberhard Karls Universität Tübingen, Auf der Morgenstelle 15, D-72076 Tübingen, Germany;
| | - Sara Malberg
- Institute for Veterinary Pathology, University Justus Liebig Gießen, Frankfurter Str. 96, D-35392 Gießen, Germany; (S.M.); (C.H.)
| | - Christiane Herden
- Institute for Veterinary Pathology, University Justus Liebig Gießen, Frankfurter Str. 96, D-35392 Gießen, Germany; (S.M.); (C.H.)
| | - Dennis Rubbenstroth
- Institute of Virology, Medical Center–University of Freiburg, Faculty of Medicine, University of Freiburg, Hermann-Herder-Str. 11, D-79104 Freiburg, Germany;
- Institute of Diagnostic Virology, Friedrich-Loeffler-Institut, Südufer 10, D-17493 Greifswald–Insel Riems, Germany
- Correspondence: ; Tel.: +49-38351-7-1521
| |
Collapse
|
17
|
Liu F, Li L, Wang Z. An improved system for constructing marker-free recombinant goatpox viruses to express foreign proteins. Res Vet Sci 2019; 126:124-126. [PMID: 31446269 DOI: 10.1016/j.rvsc.2019.08.017] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2019] [Revised: 07/21/2019] [Accepted: 08/12/2019] [Indexed: 01/11/2023]
Abstract
Goatpox virus (GTPV), belonging to the genus Capripoxvirus in the family Poxviridae, causes a contagious disease affecting goats and sheep. Homologous recombination as a conventional method is commonly used to construct recombinant GTPVs but generally with genetic markers, such as enhanced green fluorescent protein (eGFP) and guanine phosphoribosyl transferase (gpt). We have previously constructed a recombinant GTPV, which can efficiently express the EG95 antigen of Echinococcus granulosus, but contains eGFP and gpt markers in viral genome. In this study, our previous GTPV-generating system was modified by reconstruction of a Loxp-containing transfer plasmid for deleting markers using the Cre/Loxp system. Meanwhile, the previous method was significantly improved by introduction of an immortalized goat testis cell line as a substitute for primary cells. Based on the latest system, a marker-free recombinant GTPV was reconstructed for expressing the EG95 antigen, and showed neither a significant difference in replication kinetics from its parental virus nor mutations in the foreign sequence during serial 10 passages in vitro.
Collapse
Affiliation(s)
- Fuxiao Liu
- College of Veterinary Medicine, Qingdao Agricultural University, Qingdao 266109, China
| | - Ling Li
- National Research Center for Exotic Animal Diseases, China Animal Health and Epidemiology Center, No.369 Nanjing Road, Qingdao, Shandong 266032, China
| | - Zhiliang Wang
- National Research Center for Exotic Animal Diseases, China Animal Health and Epidemiology Center, No.369 Nanjing Road, Qingdao, Shandong 266032, China.
| |
Collapse
|
18
|
Pang F, Zhang M, Yang X, Li G, Zhu S, Nie X, Cao R, Yang X, Zhang Z, Huang H, Li B, Wang C, Du L, Wang F. Genome-wide analysis of circular RNAs in goat skin fibroblast cells in response to Orf virus infection. PeerJ 2019; 7:e6267. [PMID: 30697481 PMCID: PMC6346991 DOI: 10.7717/peerj.6267] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2018] [Accepted: 12/11/2018] [Indexed: 12/29/2022] Open
Abstract
Orf, caused by Orf virus (ORFV), is a globally distributed zoonotic disease responsible for serious economic losses in the agricultural sector. However, the mechanism underlying ORFV infection remains largely unknown. Circular RNAs (circRNAs), a novel type of endogenous non-coding RNAs, play important roles in various pathological processes but their involvement in ORFV infection and host response is unclear. In the current study, whole transcriptome sequencing and small RNA sequencing were performed in ORFV-infected goat skin fibroblast cells and uninfected cells. A total of 151 circRNAs, 341 messenger RNAs (mRNAs), and 56 microRNAs (miRNAs) were differently expressed following ORFV infection. Four circRNAs: circRNA1001, circRNA1684, circRNA3127 and circRNA7880 were validated by qRT-PCR and Sanger sequencing. Gene ontology (GO) analysis indicated that host genes of differently expressed circRNAs were significantly enriched in regulation of inflammatory response, epithelial structure maintenance, positive regulation of cell migration, positive regulation of ubiquitin-protein transferase activity, regulation of ion transmembrane transport, etc. The constructed circRNA-miRNA-mRNA network suggested that circRNAs may function as miRNA sponges indirectly regulating gene expression following ORFV infection. Our study presented the first comprehensive profiles of circRNAs in response to ORFV infection, thus providing new clues for the mechanisms of interactions between ORFV and the host.
Collapse
Affiliation(s)
- Feng Pang
- College of Animal Science and Technology, College of Tropical Agriculture and Forestry, Hainan University, Hainan Key Lab of Tropical Animal Reproduction & Breeding and Epidemic Disease Research, Hainan University, Haikou, China
| | - Mengmeng Zhang
- College of Animal Science and Technology, College of Tropical Agriculture and Forestry, Hainan University, Hainan Key Lab of Tropical Animal Reproduction & Breeding and Epidemic Disease Research, Hainan University, Haikou, China
| | - Xiaojian Yang
- College of Animal Science and Technology, College of Tropical Agriculture and Forestry, Hainan University, Hainan Key Lab of Tropical Animal Reproduction & Breeding and Epidemic Disease Research, Hainan University, Haikou, China
| | - Guohua Li
- College of Animal Science and Technology, College of Tropical Agriculture and Forestry, Hainan University, Hainan Key Lab of Tropical Animal Reproduction & Breeding and Epidemic Disease Research, Hainan University, Haikou, China
| | - Shu Zhu
- College of Animal Science and Technology, College of Tropical Agriculture and Forestry, Hainan University, Hainan Key Lab of Tropical Animal Reproduction & Breeding and Epidemic Disease Research, Hainan University, Haikou, China
| | - Xin Nie
- College of Animal Science and Technology, College of Tropical Agriculture and Forestry, Hainan University, Hainan Key Lab of Tropical Animal Reproduction & Breeding and Epidemic Disease Research, Hainan University, Haikou, China
| | - Ruiyong Cao
- College of Animal Science and Technology, College of Tropical Agriculture and Forestry, Hainan University, Hainan Key Lab of Tropical Animal Reproduction & Breeding and Epidemic Disease Research, Hainan University, Haikou, China
| | - Xiaohong Yang
- College of Animal Science and Technology, College of Tropical Agriculture and Forestry, Hainan University, Hainan Key Lab of Tropical Animal Reproduction & Breeding and Epidemic Disease Research, Hainan University, Haikou, China
| | - Zhenxing Zhang
- College of Animal Science and Technology, College of Tropical Agriculture and Forestry, Hainan University, Hainan Key Lab of Tropical Animal Reproduction & Breeding and Epidemic Disease Research, Hainan University, Haikou, China
| | - Haifeng Huang
- College of Animal Science and Technology, College of Tropical Agriculture and Forestry, Hainan University, Hainan Key Lab of Tropical Animal Reproduction & Breeding and Epidemic Disease Research, Hainan University, Haikou, China
| | - Baobao Li
- College of Animal Science and Technology, College of Tropical Agriculture and Forestry, Hainan University, Hainan Key Lab of Tropical Animal Reproduction & Breeding and Epidemic Disease Research, Hainan University, Haikou, China
| | - Chengqiang Wang
- College of Animal Science and Technology, College of Tropical Agriculture and Forestry, Hainan University, Hainan Key Lab of Tropical Animal Reproduction & Breeding and Epidemic Disease Research, Hainan University, Haikou, China
| | - Li Du
- College of Animal Science and Technology, College of Tropical Agriculture and Forestry, Hainan University, Hainan Key Lab of Tropical Animal Reproduction & Breeding and Epidemic Disease Research, Hainan University, Haikou, China
| | - Fengyang Wang
- College of Animal Science and Technology, College of Tropical Agriculture and Forestry, Hainan University, Hainan Key Lab of Tropical Animal Reproduction & Breeding and Epidemic Disease Research, Hainan University, Haikou, China
| |
Collapse
|
19
|
Liu F, Fan X, Li L, Ren W, Han X, Wu X, Wang Z. Development of recombinant goatpox virus expressing Echinococcus granulosus EG95 vaccine antigen. J Virol Methods 2018; 261:28-33. [DOI: 10.1016/j.jviromet.2018.08.002] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2018] [Revised: 08/01/2018] [Accepted: 08/03/2018] [Indexed: 01/22/2023]
|
20
|
Wang R, Wang Y, Liu F, Luo S. Orf virus: A promising new therapeutic agent. Rev Med Virol 2018; 29:e2013. [PMID: 30370570 DOI: 10.1002/rmv.2013] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2018] [Revised: 09/14/2018] [Accepted: 09/20/2018] [Indexed: 12/12/2022]
Abstract
The orf virus (ORFV) is a zoonotic, epitheliotropic, DNA parapoxvirus that infects principally sheep and goats. Exposure of animals to the virus or immunization by an ORFV preparation can accentuate the severity of disease, which has provoked an interest in the underlying cellular, virological, and molecular mechanisms. The identified ORFV virulence genes and the fact that the virus can repeatedly infect a host, owing to its evasive mechanisms, contribute to the development of potent immune modulators in various animal species. ORFV has been developed as a vaccine in veterinary medicine. The unique host immune-evasion ability of ORFV has made it an important candidate for vaccine vectors and biological agents (as an oncolytic virus). Genetic modifications using ORFV to obtain safe and efficient preparations and mechanistic studies are improvements to the currently available methods for disease treatment.
Collapse
Affiliation(s)
- Ruixue Wang
- Department of Laboratory Medicine, School of Stomatology and Medicine, Foshan University, Foshan, Guangdong Province, China.,Department of Basic Medical Sciences, School of Stomatology and Medicine, Foshan University, Foshan, Guangdong Province, China
| | - Yong Wang
- Department of Laboratory Medicine, School of Stomatology and Medicine, Foshan University, Foshan, Guangdong Province, China
| | - Fang Liu
- Department of Basic Medical Sciences, School of Stomatology and Medicine, Foshan University, Foshan, Guangdong Province, China
| | - Shuhong Luo
- Department of Laboratory Medicine, School of Stomatology and Medicine, Foshan University, Foshan, Guangdong Province, China
| |
Collapse
|