1
|
Zheng H, Zhao H, Xiong H, Awais MM, Zeng S, Sun J. Bioproduction and immunogenic evaluation of SARS-CoV-2 prototype vaccine in silkworm BmN cells. Int J Biol Macromol 2024; 276:134027. [PMID: 39033889 DOI: 10.1016/j.ijbiomac.2024.134027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Revised: 07/17/2024] [Accepted: 07/18/2024] [Indexed: 07/23/2024]
Abstract
COVID-19, caused by the novel coronavirus SARS-CoV-2, has presented a significant challenge to global health, security, and the economy. Vaccination is considered a crucial measure in preventing virus transmission. The silkworm bioreactor has gained widespread usage in antigen presentation, monoclonal antibody preparation, and subunit vaccine development due to its safety, efficiency, convenience, and cost-effectiveness. In this study, we employed silkworm BmN cells and the silkworm MultiBac multigene co-expression system to successfully produce two prototype vaccines: a recombinant baculovirus vector vaccine (NPV) co-displaying the SARS-CoV-2 virus capsid protein and a capsid protein virus-like particle (VLP) vaccine. Following the purification of these vaccines, we immunized BALB/c mice to evaluate their immunogenicity. Our results demonstrated that both VLP and NPV prototype vaccines effectively elicited robust immune responses in mice. However, when equal inoculation doses between groups were compared, the recombinant NPV vaccine exhibited significantly higher serum antibody titers and increased expression of spleen cytokines and lymphocyte immune regulatory factors compared to the VLP group. These results suggested an increased immune efficacy of the recombinant NPV vaccine. Conversely, the VLP prototype vaccine displayed more pronounced effects on lymphocyte cell differentiation induction. This study successfully constructed two distinct morphological recombinant vaccine models and systematically elucidated their differences in humoral immune response and lymphocyte differentiation rate. Furthermore, it has fully harnessed the immense potential of silkworm bioreactors for vaccine research and development, providing valuable technical insights for studying mutated viruses like coronaviruses.
Collapse
Affiliation(s)
- Hao Zheng
- Guangdong Provincial Key Laboratory of Agro-animal Genomics and Molecular Breeding, Subtropical Sericulture and Mulberry Resources Protection and Safety Engineering Research Center, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong 510642, China
| | - Hengfeng Zhao
- Guangdong Provincial Key Laboratory of Agro-animal Genomics and Molecular Breeding, Subtropical Sericulture and Mulberry Resources Protection and Safety Engineering Research Center, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong 510642, China
| | - Haifan Xiong
- Guangdong Provincial Key Laboratory of Agro-animal Genomics and Molecular Breeding, Subtropical Sericulture and Mulberry Resources Protection and Safety Engineering Research Center, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong 510642, China
| | - Mian Muhammad Awais
- Guangdong Provincial Key Laboratory of Agro-animal Genomics and Molecular Breeding, Subtropical Sericulture and Mulberry Resources Protection and Safety Engineering Research Center, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong 510642, China
| | - Songrong Zeng
- Guangdong Provincial Key Laboratory of Utilization and Conservation of Food and Medicinal Resources in Northern Region, Shaoguan University, Shaoguan 512005, China
| | - Jingchen Sun
- Guangdong Provincial Key Laboratory of Agro-animal Genomics and Molecular Breeding, Subtropical Sericulture and Mulberry Resources Protection and Safety Engineering Research Center, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong 510642, China.
| |
Collapse
|
2
|
Zhang X, Zhang F, Chen N, Cui X, Guo X, Sun Z, Guo P, Liao M, Li X. A Rationally Designed H5 Hemagglutinin Subunit Vaccine Provides Broad-Spectrum Protection against Various H5Nx Highly Pathogenic Avian Influenza Viruses in Chickens. Vaccines (Basel) 2024; 12:932. [PMID: 39204055 PMCID: PMC11359994 DOI: 10.3390/vaccines12080932] [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: 07/10/2024] [Revised: 08/14/2024] [Accepted: 08/19/2024] [Indexed: 09/03/2024] Open
Abstract
The evolution of the H5 highly pathogenic avian influenza (HPAI) viruses has led to the emergence of distinct groups with genetically similar clusters of hemagglutinin (HA) sequences. In this study, a consensus H5 HA sequence was cloned into the baculovirus expression system. The HA protein was expressed in baculovirus-infected insect cells and utilized as the antigen for the production of an oil emulsion-based H5 avian influenza vaccine (rBacH5Con5Mut). Twenty-one-day-old SPF chickens were immunized with this vaccine and then challenged at 21 days post-vaccination with clade 2.3.2.1, clade 2.3.4.4, and clade 7.2 of H5 HPAI viruses. The sera of vaccinated chickens exhibited high hemagglutination inhibition (HI) titers against the rBacH5 vaccine antigen, while lower HI titers were observed against the different challenge virus H5 hemagglutinins. Furthermore, the rBacH5Con5Mut vaccine provided 100% protection from mortality and clinical signs. Virus isolation results showed that oropharyngeal and cloacal shedding was prevented in 100% of the vaccinated chickens when challenged with clade 2.3.2.1 and clade 2.3.4.4 H5 viruses. When the rBacH5Con5Mut vaccine candidate was administrated at one day of age, 100% protection was demonstrated against the challenge of clade 2.3.4.4 virus at three weeks of age, indicating the potential of this vaccine for hatchery vaccination. Overall, A single immunization of rBacH5Con5Mut vaccine candidate with a consensus HA antigen can protect chickens against different clades of H5 HPAI viruses throughout the rearing period of broiler chickens without a boost, thus fulfilling the criteria for an efficacious broad-spectrum H5 avian influenza vaccine.
Collapse
Affiliation(s)
- Xuxiao Zhang
- Boehringer Ingelheim Vetmedica (China) Co., Ltd., Taizhou 225300, China; (X.Z.); (F.Z.); (X.G.); (Z.S.); (P.G.)
| | - Fushou Zhang
- Boehringer Ingelheim Vetmedica (China) Co., Ltd., Taizhou 225300, China; (X.Z.); (F.Z.); (X.G.); (Z.S.); (P.G.)
- National and Regional Joint Engineering Laboratory for Medicament of Zoonosis Prevention and Control, College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China
| | - Ning Chen
- Boehringer Ingelheim Vetmedica (China) Co., Ltd., Taizhou 225300, China; (X.Z.); (F.Z.); (X.G.); (Z.S.); (P.G.)
| | - Xiaoping Cui
- Boehringer Ingelheim Animal Health USA Inc., 3239 Satellite Blvd, Duluth, GA 30096, USA;
| | - Xiaoqin Guo
- Boehringer Ingelheim Vetmedica (China) Co., Ltd., Taizhou 225300, China; (X.Z.); (F.Z.); (X.G.); (Z.S.); (P.G.)
| | - Zhi Sun
- Boehringer Ingelheim Vetmedica (China) Co., Ltd., Taizhou 225300, China; (X.Z.); (F.Z.); (X.G.); (Z.S.); (P.G.)
| | - Pengju Guo
- Boehringer Ingelheim Vetmedica (China) Co., Ltd., Taizhou 225300, China; (X.Z.); (F.Z.); (X.G.); (Z.S.); (P.G.)
| | - Ming Liao
- National and Regional Joint Engineering Laboratory for Medicament of Zoonosis Prevention and Control, College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China
- Key Laboratory for Prevention and Control of Avian Influenza and Other Major Poultry Diseases, Ministry of Agriculture and Rural Affairs, Institute of Animal Health, Guangdong Academy of Agricultural Sciences, Guangzhou 510640, China
- College of Animal Science and Technology, Zhongkai University of Agricultural and Engineering, Guangzhou 510550, China
| | - Xin Li
- Boehringer Ingelheim Vetmedica (China) Co., Ltd., Taizhou 225300, China; (X.Z.); (F.Z.); (X.G.); (Z.S.); (P.G.)
| |
Collapse
|
3
|
Stepanova E, Isakova-Sivak I, Mezhenskaya D, Niskanen S, Matyushenko V, Bazhenova E, Rak A, Wong PF, Prokopenko P, Kotomina T, Krutikova E, Legotskiy S, Neterebskii B, Ostroukhova T, Sivak K, Orshanskaya Y, Yakovlev K, Rudenko L. Expression of the SARS-CoV-2 receptor-binding domain by live attenuated influenza vaccine virus as a strategy for designing a bivalent vaccine against COVID-19 and influenza. Virol J 2024; 21:82. [PMID: 38589848 PMCID: PMC11003101 DOI: 10.1186/s12985-024-02350-w] [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: 01/09/2024] [Accepted: 03/22/2024] [Indexed: 04/10/2024] Open
Abstract
Influenza and SARS-CoV-2 are two major respiratory pathogens that cocirculate in humans and cause serious illness with the potential to exacerbate disease in the event of co-infection. To develop a bivalent vaccine, capable of protecting against both infections, we inserted the receptor-binding domain (RBD) of the SARS-CoV-2 spike protein into hemagglutinin (HA) molecule or into the open reading frame of the truncated nonstructural protein 1 (NS1) of live attenuated influenza vaccine (LAIV) virus and assessed phenotypic characteristics of the rescued LAIV-RBD viruses, as well as their immunogenicity in mouse and Syrian hamster animal models. A panel of 9 recombinant LAIV-RBD viruses was rescued using the A/Leningrad/17 backbone. Notably, only two variants with RBD insertions into the HA molecule could express sufficient quantities of RBD protein in infected MDCK cells. Intranasal immunization of mice induced high levels of anti-influenza antibody responses in all chimeric LAIV-RBD viruses, which was comparable to the LAIV virus vector. The RBD-specific antibody responses were most pronounced in the variant expressing RBD194 fragment as a chimeric HA protein. This candidate was further tested in Syrian hamsters and was shown to be immunogenic and capable of protecting animals against both infections.
Collapse
Affiliation(s)
| | | | - Daria Mezhenskaya
- Institute of Experimental Medicine, Saint Petersburg, 197022, Russia
| | - Sergei Niskanen
- Joint-Stock Company «BIOCAD» (JSC «BIOCAD») Saint Petersburg, Intracity Municipality the Settlement of Strelna, the Settlement of Strelna, ul. Svyazi, d. 38, str. 1, pomeshch. 89, Saint Petersburg, 198515, Russia
| | | | | | - Alexandra Rak
- Institute of Experimental Medicine, Saint Petersburg, 197022, Russia
| | - Pei Fong Wong
- Institute of Experimental Medicine, Saint Petersburg, 197022, Russia
| | - Polina Prokopenko
- Institute of Experimental Medicine, Saint Petersburg, 197022, Russia
| | - Tatiana Kotomina
- Institute of Experimental Medicine, Saint Petersburg, 197022, Russia
| | - Elena Krutikova
- Institute of Experimental Medicine, Saint Petersburg, 197022, Russia
| | - Sergei Legotskiy
- Joint-Stock Company «BIOCAD» (JSC «BIOCAD») Saint Petersburg, Intracity Municipality the Settlement of Strelna, the Settlement of Strelna, ul. Svyazi, d. 38, str. 1, pomeshch. 89, Saint Petersburg, 198515, Russia
| | - Bogdan Neterebskii
- Joint-Stock Company «BIOCAD» (JSC «BIOCAD») Saint Petersburg, Intracity Municipality the Settlement of Strelna, the Settlement of Strelna, ul. Svyazi, d. 38, str. 1, pomeshch. 89, Saint Petersburg, 198515, Russia
| | - Tatiana Ostroukhova
- Joint-Stock Company «BIOCAD» (JSC «BIOCAD») Saint Petersburg, Intracity Municipality the Settlement of Strelna, the Settlement of Strelna, ul. Svyazi, d. 38, str. 1, pomeshch. 89, Saint Petersburg, 198515, Russia
| | - Konstantin Sivak
- Smorodintsev Research Institute of Influenza, Saint Petersburg, 197376, Russia
| | - Yana Orshanskaya
- Smorodintsev Research Institute of Influenza, Saint Petersburg, 197376, Russia
| | - Kirill Yakovlev
- Smorodintsev Research Institute of Influenza, Saint Petersburg, 197376, Russia
| | - Larisa Rudenko
- Institute of Experimental Medicine, Saint Petersburg, 197022, Russia
| |
Collapse
|
4
|
Plastine MDP, Amalfi S, López MG, Gravisaco MJ, Taboga O, Alfonso V. Development of a stable Sf9 insect cell line to produce VSV-G pseudotyped baculoviruses. Gene Ther 2024; 31:187-194. [PMID: 38278988 DOI: 10.1038/s41434-024-00442-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Revised: 01/12/2024] [Accepted: 01/16/2024] [Indexed: 01/28/2024]
Abstract
Baculoviruses have shown great potential as gene delivery vectors in mammals, although their effectiveness in transferring genes varies across different cell lines. A widely employed strategy to improve transduction efficiency is the pseudotyping of viral vectors. In this study, we aimed to develop a stable Sf9 insect cell line that inducibly expresses the G-protein of the vesicular stomatitis virus to pseudotype budded baculoviruses. It was obtained by inserting the VSV-G gene under the control of the very strong and infection-inducible pXXL promoter and was subsequently diluted to establish oligoclonal lines, which were selected by the fusogenic properties of VSV-G and its expression levels in infected cells and purified budded virions. Next, to enhance the performance of the cell line, the infection conditions under which functional pseudotyped baculoviruses are obtained were optimized. Finally, different baculoviruses were pseudotyped and the expression of the transgene was quantified in mammalian cells of diverse origins using flow cytometry. The transduction efficiency of pseudotyped baculovirus consistently increased across all tested mammalian cell lines compared with control viruses. These findings demonstrate the feasibility and advantages of improving gene delivery performance without the need to insert the pseudotyping gene into the baculoviral genomes.
Collapse
Affiliation(s)
- María Del Pilar Plastine
- Instituto de Agrobiotecnología y Biología Molecular (IABIMO), Instituto Nacional de Tecnología Agropecuaria (INTA), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), De los Reseros y N. Repetto S/N, Hurlingham, B1686IGC, Buenos Aires, Argentina
| | - Sabrina Amalfi
- Instituto de Agrobiotecnología y Biología Molecular (IABIMO), Instituto Nacional de Tecnología Agropecuaria (INTA), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), De los Reseros y N. Repetto S/N, Hurlingham, B1686IGC, Buenos Aires, Argentina
- Universidad Nacional de Hurlingham, Instituto de Biotecnología, Av. Vergara 2222, Villa Tesei, Hurlingham, B1688GEZ, Buenos Aires, Argentina
| | - María Gabriela López
- Instituto de Agrobiotecnología y Biología Molecular (IABIMO), Instituto Nacional de Tecnología Agropecuaria (INTA), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), De los Reseros y N. Repetto S/N, Hurlingham, B1686IGC, Buenos Aires, Argentina
| | - María José Gravisaco
- Instituto de Agrobiotecnología y Biología Molecular (IABIMO), Instituto Nacional de Tecnología Agropecuaria (INTA), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), De los Reseros y N. Repetto S/N, Hurlingham, B1686IGC, Buenos Aires, Argentina
| | - Oscar Taboga
- Instituto de Agrobiotecnología y Biología Molecular (IABIMO), Instituto Nacional de Tecnología Agropecuaria (INTA), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), De los Reseros y N. Repetto S/N, Hurlingham, B1686IGC, Buenos Aires, Argentina.
| | - Victoria Alfonso
- Instituto de Agrobiotecnología y Biología Molecular (IABIMO), Instituto Nacional de Tecnología Agropecuaria (INTA), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), De los Reseros y N. Repetto S/N, Hurlingham, B1686IGC, Buenos Aires, Argentina.
| |
Collapse
|
5
|
Hsu CY, Jang Y, Huang WR, Wang CY, Wen HW, Tsai PC, Yang CY, Munir M, Liu HJ. Development of Polycistronic Baculovirus Surface Display Vectors to Simultaneously Express Viral Proteins of Porcine Reproductive and Respiratory Syndrome and Analysis of Their Immunogenicity in Swine. Vaccines (Basel) 2023; 11:1666. [PMID: 38005998 PMCID: PMC10674950 DOI: 10.3390/vaccines11111666] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2023] [Revised: 10/21/2023] [Accepted: 10/30/2023] [Indexed: 11/26/2023] Open
Abstract
To simultaneously express and improve expression levels of multiple viral proteins of a porcine reproductive and respiratory syndrome virus (PRRSV), polycistronic baculovirus surface display vectors were constructed and characterized. We engineered polycistronic baculovirus surface display vectors, namely, pBacDual Display EGFP(BacDD)-2GP2-2GP4 and pBacDD-4GP5N34A/N51A (mtGP5), which simultaneously express and display the ectodomain of His-tagged GP2-gp64TM-CTD, His-tagged GP4-gp64TM-CTD, and His-tagged mtGP5-gp64TM-CTD fusion proteins of PRRSV on cell membrane of Sf-9 cells. Specific pathogen-free (SPF) pigs were administered intramuscularly in 2 doses at 21 and 35 days of age with genetic recombinant baculoviruses-infected cells. Our results revealed a high level of ELISA-specific antibodies, neutralizing antibodies, IL-4, and IFN-γ in SPF pigs immunized with the developed PRRSV subunit vaccine. To further assess the co-expression efficiency of different gene combinations, pBacDD-GP2-GP3-2GP4 and pBacDD-2mtGP5-2M constructs were designed for the co-expression of the ectodomain of His-tagged GP2-gp64TM-CTD, His-tagged GP3-gp64TM-CTD, and His-tagged GP4-gp64TM-CTD proteins as well as the ectodomain of His-tagged mtGP5-gp64TM-CTD and His-tagged M-gp64TM-CTD fusion proteins of PRRSV. To develop an ELISA assay for detecting antibodies against PRRSV proteins, the sequences encoding the ectodomain of the GP2, GP3, GP4, mtGP5, and M of PRRSV were amplified and subcloned into the pET32a vector and expressed in E. coli. In this work, the optimum conditions for expressing PRRSV proteins were evaluated, and the results suggested that 4 × 105 of Sf-9 cells supplemented with 7% fetal bovine serum and infected with the recombinant baculoviruses at an MOI of 20 for three days showed a higher expression levels of the protein. Taken together, the polycistronic baculovirus surface display system is a useful tool to increase expression levels of viral proteins and to simultaneously express multiple viral proteins of PRRSV for the preparation of subunit vaccines.
Collapse
Affiliation(s)
- Chao-Yu Hsu
- Department of Medical Research, Tungs’ Taichung Metroharbor Hospital, Taichung 435, Taiwan;
- Ph.D. Program in Translational Medicine, National Chung Hsing University, Taichung 402, Taiwan
| | - Yun Jang
- Institute of Molecular Biology, National Chung Hsing University, Taichung 402, Taiwan; (Y.J.); (W.-R.H.)
| | - Wei-Ru Huang
- Institute of Molecular Biology, National Chung Hsing University, Taichung 402, Taiwan; (Y.J.); (W.-R.H.)
- The iEGG and Animal Biotechnology Center, National Chung Hsing University, Taichung 402, Taiwan
| | - Chi-Young Wang
- Department of Veterinary Medicine, National Chung Hsing University, Taichung 402, Taiwan;
| | - Hsiao-Wei Wen
- Department of Food Science and Biotechnology, National Chung Hsing University, Taichung 402, Taiwan;
| | - Pei-Chien Tsai
- Department of Life Sciences, National Chung Hsing University, Taichung 402, Taiwan;
| | - Cheng-Yao Yang
- Graduate Institute of Veterinary Pathobiology, National Chung Hsing University, Taichung 402, Taiwan;
| | - Muhammad Munir
- Division of Biomedical and Life Sciences, Faculty of Health and Medicine, Lancaster University, Lancaster LA1 4YW, UK;
| | - Hung-Jen Liu
- Ph.D. Program in Translational Medicine, National Chung Hsing University, Taichung 402, Taiwan
- Institute of Molecular Biology, National Chung Hsing University, Taichung 402, Taiwan; (Y.J.); (W.-R.H.)
- The iEGG and Animal Biotechnology Center, National Chung Hsing University, Taichung 402, Taiwan
- Department of Life Sciences, National Chung Hsing University, Taichung 402, Taiwan;
- Rong Hsing Research Center for Translational Medicine, National Chung Hsing University, Taichung 402, Taiwan
| |
Collapse
|
6
|
Ni Z, Yun T, Chen L, Ye W, Hua J, Zhu Y, Liu G, Zhang C. Study on the Protective Immunity Induced by Pseudotyped Baculovirus Expressing the E Protein of Tembusu Virus in Ducklings. Genes (Basel) 2023; 14:1316. [PMID: 37510221 PMCID: PMC10378915 DOI: 10.3390/genes14071316] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2023] [Revised: 06/15/2023] [Accepted: 06/21/2023] [Indexed: 07/30/2023] Open
Abstract
The Duck Tembusu virus (DTMUV), a pathogenic flavivirus, has been causing significant economic losses in the Chinese poultry industry since 2010. This virus can severely decrease egg production and inhibit the growth of laying ducks and ducklings. While many vaccines have been developed to prevent DTMUV infection, fresh outbreaks continue to occur, as few effective vaccines are available. The E glycoprotein of DTMUV is the primary target for inducing protective immunity in the natural host. Therefore, we conducted an investigation and successfully developed a recombinant baculovirus containing the DTMUV E gene. Ducklings were then vaccinated with the purified protein derived from this virus as a potential vaccine candidate. Our findings demonstrated that the E glycoprotein of DTMUV was highly expressed in Sf9 cells. The vaccination of ducklings with the recombinant baculovirus Bac-E resulted in the induction of strong humoral and cellular immune responses. Most significantly, we observed that the vaccine provided 100% protective immunity against lethal challenges with the DTMUV YY5 strain.
Collapse
Affiliation(s)
- Zheng Ni
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-Products, Institute of Animal Husbandry and Veterinary Sciences, Zhejiang Academy of Agricultural Sciences, Hangzhou 310021, China
| | - Tao Yun
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-Products, Institute of Animal Husbandry and Veterinary Sciences, Zhejiang Academy of Agricultural Sciences, Hangzhou 310021, China
| | - Liu Chen
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-Products, Institute of Animal Husbandry and Veterinary Sciences, Zhejiang Academy of Agricultural Sciences, Hangzhou 310021, China
| | - Weicheng Ye
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-Products, Institute of Animal Husbandry and Veterinary Sciences, Zhejiang Academy of Agricultural Sciences, Hangzhou 310021, China
| | - Jionggang Hua
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-Products, Institute of Animal Husbandry and Veterinary Sciences, Zhejiang Academy of Agricultural Sciences, Hangzhou 310021, China
| | - Yinchu Zhu
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-Products, Institute of Animal Husbandry and Veterinary Sciences, Zhejiang Academy of Agricultural Sciences, Hangzhou 310021, China
| | - Guangqing Liu
- Shanghai Veterinary Research Institute, Chinese Academy at Agricultural Sciences, Shanghai 200241, China
| | - Cun Zhang
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-Products, Institute of Animal Husbandry and Veterinary Sciences, Zhejiang Academy of Agricultural Sciences, Hangzhou 310021, China
| |
Collapse
|
7
|
Baculovirus Display of Peptides and Proteins for Medical Applications. Viruses 2023; 15:v15020411. [PMID: 36851625 PMCID: PMC9962271 DOI: 10.3390/v15020411] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Revised: 01/27/2023] [Accepted: 01/30/2023] [Indexed: 02/04/2023] Open
Abstract
Baculoviridae is a large family of arthropod-infective viruses. Recombinant baculoviruses have many applications, the best known is as a system for large scale protein production in combination with insect cell cultures. More recently recombinant baculoviruses have been utilized for the display of proteins of interest with applications in medicine. In the present review we analyze the different strategies for the display of proteins and peptides on the surface of recombinant baculoviruses and provide some examples of the different proteins displayed. We analyze briefly the commercially available systems for recombinant baculovirus production and display and discuss the future of this emerging and powerful technology.
Collapse
|
8
|
Sun L, Yao C, Amanze C, Yin B, Huang J, Hao B. The cytoplasmic tail substitution increases the assembly efficiency of Ebola virus glycoprotein on the budded virus of Bombyx mori nucleopolyhedrovirus. Protein Expr Purif 2022; 200:106156. [PMID: 35987323 DOI: 10.1016/j.pep.2022.106156] [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: 06/24/2022] [Revised: 08/10/2022] [Accepted: 08/10/2022] [Indexed: 11/24/2022]
Abstract
Glycoprotein (GP1,2) of the Ebola virus (EBOV) is the key membrane fusion protein, which is a key candidate protein for vaccine preparations. Previously, GP1,2 was expressed by Bombyx mori nucleopolyhedrovirus (BmNPV) expression vector system; however, few GP1,2 was incorporated into budded virus (BV) of BmNPV. To improve the incorporation efficiency of GP1,2 into the virion, the GP1,2 fusion with the cytoplasmic tail of GP64 of BmNPV was expressed in BmN cells by the BmNPV expression system. The BV was purified by ultracentrifugation, and GP1,2 expression in BV was detected by the antibody. The result indicated that a 532% increase in the relative GP1,2 densitometry signal was observed in constructs utilizing the GP64 C-terminal domain; moreover, the substitution of GP1,2 native signal peptide with GP64 signal peptide increased the incorporation efficiency by 34.6% in the relative GP1,2 densitometry signal. We revealed that the application of the cytoplasmic tail of BmNPV GP64 significantly increased the incorporation rate of GP1,2 into the BV envelope. This study lays a foundation for GP1,2 vaccine development.
Collapse
Affiliation(s)
- Luping Sun
- Jiangsu Key Laboratory of Sericultural Biology and Biotechnology, School of Biotechnology, Jiangsu University of Science and Technology, Zhenjiang, Jiangsu, 212018, China
| | - Congyue Yao
- Jiangsu Key Laboratory of Sericultural Biology and Biotechnology, School of Biotechnology, Jiangsu University of Science and Technology, Zhenjiang, Jiangsu, 212018, China
| | - Charles Amanze
- School of Minerals Processing and Bioengineering, Central South University, Changsha, Hunan, 410083, China
| | - Bo Yin
- National University of Singapore (Suzhou) Research Institute, Suzhou, Jiangsu, China
| | - Jinshan Huang
- Jiangsu Key Laboratory of Sericultural Biology and Biotechnology, School of Biotechnology, Jiangsu University of Science and Technology, Zhenjiang, Jiangsu, 212018, China
| | - Bifang Hao
- Jiangsu Key Laboratory of Sericultural Biology and Biotechnology, School of Biotechnology, Jiangsu University of Science and Technology, Zhenjiang, Jiangsu, 212018, China.
| |
Collapse
|
9
|
Impact of Molecular Modification on the Efficiency of Recombinant Baculovirus Vector Invasion to Mammalian Cells and Its Immunogenicity in Mice. Viruses 2022; 14:v14010140. [PMID: 35062344 PMCID: PMC8779059 DOI: 10.3390/v14010140] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Revised: 01/09/2022] [Accepted: 01/11/2022] [Indexed: 11/21/2022] Open
Abstract
The baculovirus display system (BDS), an excellent eukaryotic surface display technology that offers the advantages of safety, efficiency, and economy, is widely used in biomedicine. A previous study using rBacmid-Δgp64-ires-gp64 expressed in low copy numbers of the gp64 gene achieved high-efficiency expression and co-display of three fluorescent proteins (GFP, YFP, and mCherry). However, low expression of GP64 in recombinant baculoviruses also reduces the efficiency of recombinant baculovirus transduction into mammalian cells. In addition, the baculovirus promoter has no expression activity in mammalian cells and thus cannot meet the application requirements of baculoviral vectors for the BDS. Based on previous research, this study first determined the expression activity of promoters in insect Spodoptera frugiperda 9 cells and mammalian cells and successfully screened the very early promoter pie1 to mediate the co-expression of multiple genes. Second, utilizing the envelope display effect of the INVASIN and VSVG proteins, the efficiency of transduction of recombinant baculovirus particles into non-host cells was significantly improved. Finally, based on the above improvement, a recombinant baculovirus vector displaying four antigen proteins with high efficiency was constructed. Compared with traditional BDSs, the rBacmid-Δgp64 system exhibited increased display efficiency of the target protein by approximately 3-fold and induced an approximately 4-fold increase in the titer of serum antibodies to target antigens in Bal B/c mice. This study systematically explored the application of a new multi-gene co-display technology applicable to multi-vaccine research, and the results provide a foundation for the development of novel BDS technologies.
Collapse
|
10
|
Yoon KW, Chu KB, Kang HJ, Kim MJ, Eom GD, Lee SH, Moon EK, Quan FS. Mucosal Administration of Recombinant Baculovirus Displaying Toxoplasma gondii ROP4 Confers Protection Against T. gondii Challenge Infection in Mice. Front Cell Infect Microbiol 2021; 11:735191. [PMID: 34660343 PMCID: PMC8512701 DOI: 10.3389/fcimb.2021.735191] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Accepted: 09/14/2021] [Indexed: 11/13/2022] Open
Abstract
Pathogens require physical contact with the mucosal surface of the host organism to initiate infection and as such, vaccines eliciting both mucosal and systemic immune responses would be promising. Studies involving the use of recombinant baculoviruses (rBVs) as mucosal vaccines are severely lacking despite their inherently safe nature, especially against pathogens of global importance such as Toxoplasma gondii. Here, we generated rBVs displaying T. gondii rhoptry protein 4 (ROP4) and evaluated their protective efficacy in BALB/c mice following immunization via intranasal (IN) and oral routes. IN immunization with the ROP4-expressing rBVs elicited higher levels of parasite-specific IgA antibody responses compared to oral immunization. Upon challenge infection with a lethal dose of T. gondii ME49, IN immunization elicited significantly higher parasite-specific antibody responses in the mucosal tissues such as intestines, feces, vaginal samples, and brain than oral immunization. Marked increases in IgG and IgA antibody-secreting cell (ASC) responses were observed from intranasally immunized mice. IN immunization elicited significantly enhanced induction of CD4+, CD8+ T cells, and germinal center B (GC B) cell responses from secondary lymphoid organs while limiting the production of the inflammatory cytokines IFN-γ and IL-6 in the brain, all of which contributed to protecting mice against T. gondii lethal challenge infection. Our findings suggest that IN delivery of ROP4 rBVs induced better mucosal and systemic immunity against the lethal T. gondii challenge infection compared to oral immunization.
Collapse
Affiliation(s)
- Keon-Woong Yoon
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul, South Korea
| | - Ki-Back Chu
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul, South Korea
| | - Hae-Ji Kang
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul, South Korea
| | - Min-Ju Kim
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul, South Korea
| | - Gi-Deok Eom
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul, South Korea
| | - Su-Hwa Lee
- Department of Medical Zoology, School of Medicine, Kyung Hee University, Seoul, South Korea
| | - Eun-Kyung Moon
- Department of Medical Zoology, School of Medicine, Kyung Hee University, Seoul, South Korea
| | - Fu-Shi Quan
- Department of Medical Zoology, School of Medicine, Kyung Hee University, Seoul, South Korea
- Medical Research Center for Bioreaction to Reactive Oxygen Species and Biomedical Science Institute, School of Medicine, Graduate School, Kyung Hee University, Seoul, South Korea
| |
Collapse
|
11
|
Hamada M, Yura Y. Efficient Delivery and Replication of Oncolytic Virus for Successful Treatment of Head and Neck Cancer. Int J Mol Sci 2020; 21:E7073. [PMID: 32992948 PMCID: PMC7582277 DOI: 10.3390/ijms21197073] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Revised: 09/18/2020] [Accepted: 09/22/2020] [Indexed: 12/14/2022] Open
Abstract
Head and neck cancer has been treated by a combination of surgery, radiation, and chemotherapy. In recent years, the development of immune checkpoint inhibitors (ICIs) has made immunotherapy a new treatment method. Oncolytic virus (OV) therapy selectively infects tumor cells with a low-pathogenic virus, lyses tumor cells by the cytopathic effects of the virus, and induces anti-tumor immunity to destroy tumors by the action of immune cells. In OV therapy for head and neck squamous cell carcinoma (HNSCC), viruses, such as herpes simplex virus type 1 (HSV-1), vaccinia virus, adenovirus, reovirus, measles virus, and vesicular stomatitis virus (VSV), are mainly used. As the combined use of mutant HSV-1 and ICI was successful for the treatment of melanoma, studies are underway to combine OV therapy with radiation, chemotherapy, and other types of immunotherapy. In such therapy, it is important for the virus to selectively replicate in tumor cells, and to express the viral gene and the introduced foreign gene in the tumor cells. In OV therapy for HNSCC, it may be useful to combine systemic and local treatments that improve the delivery and replication of the inoculated oncolytic virus in the tumor cells.
Collapse
Affiliation(s)
- Masakazu Hamada
- Department of Oral and Maxillofacial Surgery, Osaka University Graduate School of Dentistry, Suita, Osaka 565-0871, Japan;
| | | |
Collapse
|
12
|
Chen YY, Yang WC, Chang YK, Wang CY, Huang WR, Li JY, Chuang KP, Wu HY, Chang CD, Nielsen BL, Liu HJ. Construction of polycistronic baculovirus surface display vectors to express the PCV2 Cap(d41) protein and analysis of its immunogenicity in mice and swine. Vet Res 2020; 51:112. [PMID: 32907618 PMCID: PMC7487469 DOI: 10.1186/s13567-020-00836-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Accepted: 08/27/2020] [Indexed: 11/22/2022] Open
Abstract
To increase expression levels of the PCV2 Cap(d41) protein, novel baculovirus surface display vectors with multiple expression cassettes were constructed to create recombinant baculoviruses BacSC-Cap(d41), BacDD-2Cap(d41), BacDD-3Cap(d41), and BacDD-4Cap(d41). Our results reveal that the recombinant baculovirus BacDD-4Cap(d41) was able to express the highest levels of Cap(d41) protein. Optimum conditions for expressing the PCV2 Cap(d41) protein were determined, and our results show that 107 of Sf-9 infected with the recombinant baculovirus BacDD-4Cap(d41) at an MOI of 5 for 3 days showed the highest level of protein expression. Mice immunized with the 4Cap(d41) vaccine which was prepared from the recombinant baculovirus-infected cells (107) elicited higher ELISA titers compared to the Cap (d41) vaccine. The 4Cap(d41) vaccine could elicit anti-PCV2 neutralizing antibodies and IFN-γ in mice, as confirmed by virus neutralization test and IFN-γ ELISA. Moreover, the swine lymphocyte proliferative responses indicated that the 4Cap(d41) vaccine was able to induce a clear cellular immune response. Flow cytometry analysis showed that the percentage of CD4+ T cells and CD4+/CD8+ ratio was increased significantly in SPF pigs immunized with the 4Cap(d41) vaccine. Importantly, the 4Cap(d41) vaccine induced an IFN-γ response, further confirming that its effect is through cellular immunity in SPF pigs. An in vivo challenge study revealed that the 4Cap(d41) and the commercial vaccine groups significantly reduce the viral load of vaccinated pigs as compared with the CE negative control group. Taken together, we have successfully developed a 4Cap(d41) vaccine that may be a potential subunit vaccine for preventing the disease associated with PCV2 infections.
Collapse
Affiliation(s)
- Ya-Yi Chen
- Department of Stomatology, Tung's Taichung MetroHarbor Hospital, Taichung, Taiwan
- Medical Biotechnology, National Chung Hsing University, Taichung, 402, Taiwan
| | - Wei-Chen Yang
- Institute of Molecular Biology, National Chung Hsing University, Taichung, 402, Taiwan
| | - Yu-Kang Chang
- Department of Medical Research, Tung's Taichung MetroHarbor Hospital, Taichung, Taiwan
- Department of Nursing, Jen-Teh Junior College of Medicine and Management, Hou-Long, Taiwan
| | - Chi-Young Wang
- Department of Veterinary Medicine, National Chung Hsing University, Taichung, 402, Taiwan
| | - Wei-Ru Huang
- Institute of Molecular Biology, National Chung Hsing University, Taichung, 402, Taiwan
- The iEGG and Animal Biotechnology Center, National Chung Hsing University, Taichung, 402, Taiwan
| | - Jyun-Yi Li
- Institute of Molecular Biology, National Chung Hsing University, Taichung, 402, Taiwan
- The iEGG and Animal Biotechnology Center, National Chung Hsing University, Taichung, 402, Taiwan
| | - Kuo-Pin Chuang
- Graduate Institute of Animal Vaccine Technology, National Pingtung University of Science and Technology, Pingtung, 912, Taiwan
| | - Hung-Yi Wu
- Department of Veterinary Medicine, National Pingtung University of Science and Technology, Pingtung, 912, Taiwan
| | - Ching-Dong Chang
- Department of Veterinary Medicine, National Pingtung University of Science and Technology, Pingtung, 912, Taiwan
| | - Brent L Nielsen
- Department of Microbiology and Molecular Biology, Brigham Young University, Provo, UT, USA
| | - Hung-Jen Liu
- Institute of Molecular Biology, National Chung Hsing University, Taichung, 402, Taiwan.
- The iEGG and Animal Biotechnology Center, National Chung Hsing University, Taichung, 402, Taiwan.
- Rong Hsing Research Center for Translational Medicine, National Chung Hsing University, Taichung, 402, Taiwan.
- Translational Medicine, National Chung Hsing University, Taichung, 402, Taiwan.
- Department of Life Sciences, National Chung Hsing University, Taichung, Taiwan.
| |
Collapse
|
13
|
Tao LN, Liu ZH, Xu HL, Lu Y, Liao M, He F. LvYY1 Activates WSSV ie1 Promoter for Enhanced Vaccine Production and Efficacy. Vaccines (Basel) 2020; 8:E510. [PMID: 32911686 PMCID: PMC7563808 DOI: 10.3390/vaccines8030510] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Revised: 08/26/2020] [Accepted: 09/04/2020] [Indexed: 12/14/2022] Open
Abstract
The baculovirus expression vector system (BEVS) has been used as a preferred platform for the production of recombinant protein complexes and efficacious vaccines. However, limited protein yield hinders the application of BEVS. It is well accepted that transcription enhancers are capable of increasing translational efficiency of mRNAs, thereby achieving better protein production. In this study, the ability of LvYY1 as a transcription enhancer was assessed. LvYY1 could interact with the WSSV ie1 promoter via binding to special DNA sites in BEVS. The effects of LvYY1 on protein expression mediated by WSSV ie1 promoter of BEVS was investigated using eGFP as a reporter gene. Enhanced eGFP expression was observed in Sf-9 cells with LvYY1. On this basis, a modified vector combining ie1 promoter and LvYY1 was developed to express either secreting CSFV E2 or baculovirus surface displayed H5 HA of AIVs. Compared to control groups without LvYY1, E2 protein yield increases to 1.6-fold, while H5 production improves as revealed by an upregulated hemagglutination titer of 8-fold at least. Moreover, with LvYY1, H5 displaying baculovirus driven by WSSV ie1 promoter (BV-LvYY1-ie1-HA) sustains the transduction activity in CEF cells. In chicken, BV-LvYY1-ie1-HA elicits a robust immune response against H5 AIVs in the absence of adjuvant, as indicated by specific antibody and cytokine responses. The findings suggest its potential function as both a vectored and subunit vaccine. These results demonstrate that the coexpression with LvYY1 serves as a promising strategy to extensively improve the efficiency of BEVS for efficacious vaccine production.
Collapse
Affiliation(s)
- Li-Na Tao
- Institute of Preventive Veterinary Medicine, College of Animal Sciences, Zhejiang University, Hangzhou 310058, China; (L.-N.T.); (Z.-H.L.); (H.-L.X.); (Y.L.)
| | - Ze-Hui Liu
- Institute of Preventive Veterinary Medicine, College of Animal Sciences, Zhejiang University, Hangzhou 310058, China; (L.-N.T.); (Z.-H.L.); (H.-L.X.); (Y.L.)
| | - Hui-Ling Xu
- Institute of Preventive Veterinary Medicine, College of Animal Sciences, Zhejiang University, Hangzhou 310058, China; (L.-N.T.); (Z.-H.L.); (H.-L.X.); (Y.L.)
| | - Ying Lu
- Institute of Preventive Veterinary Medicine, College of Animal Sciences, Zhejiang University, Hangzhou 310058, China; (L.-N.T.); (Z.-H.L.); (H.-L.X.); (Y.L.)
| | - Min Liao
- Key Laboratory of Animal Virology of Ministry of Agriculture, Zhejiang University, Hangzhou 310058, China;
| | - Fang He
- Institute of Preventive Veterinary Medicine, College of Animal Sciences, Zhejiang University, Hangzhou 310058, China; (L.-N.T.); (Z.-H.L.); (H.-L.X.); (Y.L.)
| |
Collapse
|
14
|
Tung MC, Lu HY, Chang YK, Huang WR, Liao TL, Wu HY, Chang CD, Fan HC, Nielsen BL, Liu HJ. Baculovirus surface display of the HA protein of H5N2 avian influenza virus and its immunogenicity against a lethal challenge with H5N1 virus in chickens. Vet Microbiol 2020; 243:108640. [PMID: 32273019 DOI: 10.1016/j.vetmic.2020.108640] [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: 01/31/2020] [Revised: 03/10/2020] [Accepted: 03/10/2020] [Indexed: 12/14/2022]
Abstract
In the present study, we have generated several H5N2 HA recombinant baculoviruses for production of a HA subunit vaccine against the lethal H5N2 avian influenza virus (AIV). The effective display of functional HA on the cell membrane and baculoviral envelope was examined. Our results reveal that chickens immunized with the chimeric AIV HA protein fused with the baculovirus gp64 cytoplasmic domain (CTD) induced higher HI titer. To further increase the expression level of the H5N2 AIV HA protein, the HA gene of H5N2 AIV was amplified and cloned into three novel baculovirus surface display vectors BacDual DisplayEGFP-2HA, BacDual DisplayEGFP-3HA, BacDual DisplayEGFP-4HA which contains multiple expression cassettes for higher level display of HA proteins on the cell membrane and baculovirus envelope. To determine the optimum conditions for producing HA protein, various MOI, infection times, and shaker times for virus transfection were tested. Our results reveal that the conditions of an MOI of 5, 3 day post infection, and 15 min of shaker time have higher efficiency for HA protein production. Our results reveal that the baculovirus surface display vector pBacDual DisplayEGFP-4HA increases significantly the expression level of the H5N2 AIV HA protein. Chickens that received two doses of BacDual DisplayEGFP-4HA cell lysates formulated with Montanide ISA70 adjuvant elicited efficient immunogenicity and had an average HI titer of 7 log2 at 2 weeks post-vaccination. Challenge studies revealed that vaccinated chickens with HI titers 5 log2 were completely protected against the lethal H5N1 AIV challenge. Furthermore, HI titers could be maintained at 5 log2 for 20 weeks for laying hens. This study suggests that the HA protein expression from the baculovirus surface display system could be a safe and efficacious subunit vaccine for chickens.
Collapse
Affiliation(s)
- Min-Che Tung
- Department of Stomatology & Medical Research, Tung's Taichung Metroharbor Hospital, Taichung, Taiwan
| | - Hsin-Yu Lu
- Institute of Molecular Biology, National Chung Hsing University, Taichung 402, Taiwan
| | - Yu-Kang Chang
- Department of Stomatology & Medical Research, Tung's Taichung Metroharbor Hospital, Taichung, Taiwan; Depertment of Nursing, Jen-Teh Junior College of Medicine and Management, Hou-Loung Town, Taiwan
| | - Wei-Ru Huang
- Institute of Molecular Biology, National Chung Hsing University, Taichung 402, Taiwan
| | - Tsai-Ling Liao
- Department of Medical Research, Taichung Veterans General Hospital, Taichung 407, Taiwan; Rong Hsing Research Center for Translational Medicine, National Chung Hsing University, Taichung 402, Taiwan; Ph. D Program in Translational Medicine, National Chung Hsing University, Taichung 402, Taiwan
| | - Hung-Yi Wu
- Department of Veterinary Medicine, National Pingtung University of Science and Technology, Pingtung 912, Taiwan
| | - Ching-Dong Chang
- Department of Veterinary Medicine, National Pingtung University of Science and Technology, Pingtung 912, Taiwan
| | - Hueng-Chuen Fan
- Department of Stomatology & Medical Research, Tung's Taichung Metroharbor Hospital, Taichung, Taiwan
| | - Brent L Nielsen
- Department of Microbiology and Molecular Biology, Brigham Young University, Provo, UT, USA
| | - Hung-Jen Liu
- Institute of Molecular Biology, National Chung Hsing University, Taichung 402, Taiwan; Rong Hsing Research Center for Translational Medicine, National Chung Hsing University, Taichung 402, Taiwan; Ph. D Program in Translational Medicine, National Chung Hsing University, Taichung 402, Taiwan; The iEGG and Animal Biotechnology Center, National Chung Hsing University, Taichung 402, Taiwan; Department of Life Sciences, National Chung Hsing University, Taichung, Taiwan.
| |
Collapse
|
15
|
Zheng M, Huang J, Tong A, Yang H. Oncolytic Viruses for Cancer Therapy: Barriers and Recent Advances. MOLECULAR THERAPY-ONCOLYTICS 2019; 15:234-247. [PMID: 31872046 PMCID: PMC6911943 DOI: 10.1016/j.omto.2019.10.007] [Citation(s) in RCA: 172] [Impact Index Per Article: 34.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Oncolytic viruses (OVs) are powerful new therapeutic agents in cancer therapy. With the first OV (talimogene laherparepvec [T-vec]) obtaining US Food and Drug Administration approval, interest in OVs has been boosted greatly. Nevertheless, despite extensive research, oncolytic virotherapy has shown limited efficacy against solid tumors. Recent advances in viral retargeting, genetic editing, viral delivery platforms, tracking strategies, OV-based gene therapy, and combination strategies have the potential to broaden the applications of oncolytic virotherapy in oncology. In this review, we present several insights into the limitations and challenges of oncolytic virotherapy, describe the strategies mentioned above, provide a summary of recent preclinical and clinical trials in the field of oncolytic virotherapy, and highlight the need to optimize current strategies to improve clinical outcomes.
Collapse
Affiliation(s)
- Meijun Zheng
- Department of Otolaryngology, Head and Neck Surgery, West China Hospital, West China Medical School, Sichuan University, Chengdu, Sichuan Province, P.R. China
| | - Jianhan Huang
- Department of Neurosurgery, West China Hospital, West China Medical School, Sichuan University, Chengdu, Sichuan Province, P.R. China
| | - Aiping Tong
- State Key Laboratory of Biotherapy, West China Medical School, Sichuan University, Chengdu, Sichuan Province, P.R. China
| | - Hui Yang
- Department of Otolaryngology, Head and Neck Surgery, West China Hospital, West China Medical School, Sichuan University, Chengdu, Sichuan Province, P.R. China
| |
Collapse
|
16
|
Zak AJ, Hill BD, Rizvi SM, Smith MR, Yang M, Wen F. Enhancing the Yield and Quality of Influenza Virus-like Particles (VLPs) Produced in Insect Cells by Inhibiting Cytopathic Effects of Matrix Protein M2. ACS Synth Biol 2019; 8:2303-2314. [PMID: 31487465 DOI: 10.1021/acssynbio.9b00111] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
To provide broader protection and eliminate the need for annual update of influenza vaccines, biomolecular engineering of influenza virus-like particles (VLPs) to display more conserved influenza proteins such as the matrix protein M2 has been explored. However, achieving high surface density of full-length M2 in influenza VLPs has been left unrealized. In this study, we show that the ion channel activity of M2 induces significant cytopathic effects in Spodoptera frugiperda (Sf9) insect cells when expressed using M2-encoding baculovirus. These effects include altered Sf9 cell morphology and reduced baculovirus replication, resulting in impaired influenza protein expression and thus VLP production. On the basis of the function of M2, we hypothesized that blocking its ion channel activity could potentially relieve these cytopathic effects, and thus restore influenza protein expression to improve VLP production. The use of the M2 inhibitor amantadine indeed improves Sf9 cellular expression not only of M2 (∼3-fold), but also of hemagglutinin (HA) (∼7-fold) and of matrix protein M1 (∼3-fold) when coexpressed to produce influenza VLPs. This increased cellular expression of all three influenza proteins further leads to ∼2-fold greater VLP yield. More importantly, the quality of the resulting influenza VLPs is significantly improved, as demonstrated by the ∼2-fold, ∼50-fold, and ∼2-fold increase in the antigen density to approximately 53 HA, 48 M1, and 156 M2 per influenza VLP, respectively. Taken together, this study represents a novel approach to enable the efficient incorporation of full-length M2 while enhancing both the yield and quality of influenza VLPs produced by Sf9 cells.
Collapse
Affiliation(s)
- Andrew J. Zak
- Department of Chemical Engineering, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Brett D. Hill
- Department of Chemical Engineering, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Syed M. Rizvi
- Department of Chemical Engineering, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Mason R. Smith
- Department of Chemical Engineering, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Madeleine Yang
- Department of Chemical Engineering, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Fei Wen
- Department of Chemical Engineering, University of Michigan, Ann Arbor, Michigan 48109, United States
| |
Collapse
|
17
|
Lin CW, Cheng MC, Lin SY, Hung SH, Jhang SY, Chang CW, Chang PC, Hu YC. Hybrid baculovirus-mediated prolonged hemagglutinin expression and secretion in vivo enhances the vaccine efficacy. J Taiwan Inst Chem Eng 2018. [DOI: 10.1016/j.jtice.2018.05.044] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
18
|
Chang CY, Hsu WT, Chao YC, Chang HW. Display of Porcine Epidemic Diarrhea Virus Spike Protein on Baculovirus to Improve Immunogenicity and Protective Efficacy. Viruses 2018; 10:v10070346. [PMID: 29954081 PMCID: PMC6071207 DOI: 10.3390/v10070346] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2018] [Revised: 06/15/2018] [Accepted: 06/15/2018] [Indexed: 01/23/2023] Open
Abstract
A new variant of the porcine epidemic diarrhea virus (PEDV) is an emerging swine disease, killing considerable numbers of neonatal piglets in North America and Asia in recent years. To generate immunogens mimicking the complex spike (S) protein folding with proper posttranslational modification to mount a robust immune response against the highly virulent PEDV, two baculoviruses displaying the full-length S protein (S-Bac) and the S1 protein (S1-Bac) of the virulent Taiwan genotype 2b (G2b) PEDV Pintung 52 (PEDV-PT) strain were constructed. Intramuscular immunizations of mice and piglets with the S-Bac and S1-Bac demonstrated significantly higher levels of systemic anti-PEDV S-specific IgG, as compared with control group. Our results also showed that piglets in the S-Bac group elicited superior PEDV-specific neutralizing antibodies than those of the S1-Bac and control groups. The highly virulent PEDV-PT strain challenge experiment showed that piglets immunized with S-Bac and S1-Bac showed milder clinical symptoms with significantly less fecal viral shedding as compared with non-immunized control piglets. More importantly, piglets immunized with the S-Bac exhibited no to mild clinical signs, with a delayed, minimal viral shedding. Our results demonstrated that the S-Bac could serve as a safe, easy to manipulate, and effective vaccine candidate against the PEDV infection.
Collapse
Affiliation(s)
- Chia-Yu Chang
- School of Veterinary Medicine, National Taiwan University, Taipei 106, Taiwan.
| | - Wei-Ting Hsu
- Institute of Molecular Biology, Academia Sinica, Nankang, Taipei 115, Taiwan.
| | - Yu-Chan Chao
- Institute of Molecular Biology, Academia Sinica, Nankang, Taipei 115, Taiwan.
| | - Hui-Wen Chang
- School of Veterinary Medicine, National Taiwan University, Taipei 106, Taiwan.
- Graduate Institute of Molecular and Comparative Pathobiology, School of Veterinary Medicine, National Taiwan University, Taipei 106, Taiwan.
| |
Collapse
|
19
|
Zheng H, Wang X, Ren F, Zou S, Feng M, Xu L, Yao L, Sun J. Construction of a highly efficient display system for baculovirus and its application on multigene co-display. Mol Genet Genomics 2018; 293:1265-1277. [DOI: 10.1007/s00438-018-1459-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2018] [Accepted: 06/07/2018] [Indexed: 01/05/2023]
|
20
|
Hsieh MS, He JL, Wu TY, Juang RH. A secretary bi-cistronic baculovirus expression system with improved production of the HA1 protein of H6 influenza virus in insect cells and Spodoptera litura larvae. J Immunol Methods 2018; 459:81-89. [PMID: 29894745 PMCID: PMC7094261 DOI: 10.1016/j.jim.2018.06.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2018] [Revised: 04/26/2018] [Accepted: 06/06/2018] [Indexed: 12/14/2022]
Abstract
A bi-cistronic baculovirus expression vector was constructed to facilitate the expression, detection, and isolation of the hemagglutinin (HA) fragment HA1 of H6N1 avian influenza virus (AIV) in an insect and a culture of its cells. In this construct, the GP67sp signal peptide promoted the secretion of the recombinant protein into the culture medium, and improved protein expression and purification. Enhanced green fluorescent protein, co-expressed through an internal ribosome entry site, served as a visible reporter for protein expression detection. The hemolymph of Spodoptera litura larvae infected with the bi-cistronic baculovirus was collected for the purification of the recombinant HA1, which was found to be glycosylated, and monomeric and trimeric forms of the recombinant HA1 were identified. Proteins expressed in both the cell culture and larvae served as effective subunit vaccines for the production of antiserum against HA. The antiserum recognized the H6 subtype of AIV but not the H5 subtype. HA1 of H6N1 influenza virus was expressed in insect and cell culture. The expressed HA1 was glycosylated, and estimated as monomeric and trimeric forms. The expressed HA1 served as an effective subunit vaccine for producing antisera. The antisera specifically recognized influenza H6 subtype but not the H5 subtype.
Collapse
Affiliation(s)
- Ming-Shou Hsieh
- Institute of Biotechnology, National Taiwan University, Taipei 106, Taiwan
| | - Jie-Long He
- Department of Post-Baccalaureate Veterinary Medicine, Asia University, Taichung 413, Taiwan
| | - Tzong-Yuan Wu
- Department of Bioscience Technology, Chung Yuan Christian University, Chungli 320, Taiwan
| | - Rong-Huay Juang
- Institute of Biotechnology, National Taiwan University, Taipei 106, Taiwan; Department of Biochemical Science and Technology, National Taiwan University, Taipei 106, Taiwan.
| |
Collapse
|
21
|
Baliou S, Adamaki M, Kyriakopoulos AM, Spandidos DA, Panayiotidis M, Christodoulou I, Zoumpourlis V. CRISPR therapeutic tools for complex genetic disorders and cancer (Review). Int J Oncol 2018; 53:443-468. [PMID: 29901119 PMCID: PMC6017271 DOI: 10.3892/ijo.2018.4434] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2018] [Accepted: 05/18/2018] [Indexed: 12/13/2022] Open
Abstract
One of the fundamental discoveries in the field of biology is the ability to modulate the genome and to monitor the functional outputs derived from genomic alterations. In order to unravel new therapeutic options, scientists had initially focused on inducing genetic alterations in primary cells, in established cancer cell lines and mouse models using either RNA interference or cDNA overexpression or various programmable nucleases [zinc finger nucleases (ZNF), transcription activator-like effector nucleases (TALEN)]. Even though a huge volume of data was produced, its use was neither cheap nor accurate. Therefore, the clustered regularly interspaced short palindromic repeats (CRISPR) system was evidenced to be the next step in genome engineering tools. CRISPR-associated protein 9 (Cas9)-mediated genetic perturbation is simple, precise and highly efficient, empowering researchers to apply this method to immortalized cancerous cell lines, primary cells derived from mouse and human origins, xenografts, induced pluripotent stem cells, organoid cultures, as well as the generation of genetically engineered animal models. In this review, we assess the development of the CRISPR system and its therapeutic applications to a wide range of complex diseases (particularly distinct tumors), aiming at personalized therapy. Special emphasis is given to organoids and CRISPR screens in the design of innovative therapeutic approaches. Overall, the CRISPR system is regarded as an eminent genome engineering tool in therapeutics. We envision a new era in cancer biology during which the CRISPR-based genome engineering toolbox will serve as the fundamental conduit between the bench and the bedside; nonetheless, certain obstacles need to be addressed, such as the eradication of side-effects, maximization of efficiency, the assurance of delivery and the elimination of immunogenicity.
Collapse
Affiliation(s)
- Stella Baliou
- National Hellenic Research Foundation, 11635 Athens, Greece
| | - Maria Adamaki
- National Hellenic Research Foundation, 11635 Athens, Greece
| | | | - Demetrios A Spandidos
- Laboratory of Clinical Virology, Medical School, University of Crete, Heraklion 71003, Greece
| | - Mihalis Panayiotidis
- Department of Applied Sciences, Northumbria University, Newcastle Upon Tyne, NE1 8ST, UK
| | | | | |
Collapse
|
22
|
Premanand B, Zhong Wee P, Prabakaran M. Baculovirus Surface Display of Immunogenic Proteins for Vaccine Development. Viruses 2018; 10:E298. [PMID: 29857561 PMCID: PMC6024371 DOI: 10.3390/v10060298] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2018] [Revised: 05/22/2018] [Accepted: 05/28/2018] [Indexed: 12/25/2022] Open
Abstract
Vaccination is an efficient way to prevent the occurrence of many infectious diseases in humans. To date, several viral vectors have been utilized for the generation of vaccines. Among them, baculovirus-categorized as a nonhuman viral vector-has been used in wider applications. Its versatile features, like large cloning capacity, nonreplicative nature in mammalian cells, and broad tissue tropism, hold it at an excellent position among vaccine vectors. In addition to ease and safety during swift production, recent key improvements to existing baculovirus vectors (such as inclusion of hybrid promoters, immunostimulatory elements, etc.) have led to significant improvements in immunogenicity and efficacy of surface-displayed antigens. Furthermore, some promising preclinical results have been reported that mirror the scope and practicality of baculovirus as a vaccine vector for human applications in the near future. Herein, this review provides an overview of the induced immune responses by baculovirus surface-displayed vaccines against influenza and other infectious diseases in animal models, and highlights the strategies applied to enhance the protective immune responses against the displayed antigens.
Collapse
Affiliation(s)
- Balraj Premanand
- Temasek Life Sciences Laboratory, 1 Research Link, National University of Singapore, Singapore 117604, Singapore.
| | - Poh Zhong Wee
- Temasek Life Sciences Laboratory, 1 Research Link, National University of Singapore, Singapore 117604, Singapore.
| | - Mookkan Prabakaran
- Temasek Life Sciences Laboratory, 1 Research Link, National University of Singapore, Singapore 117604, Singapore.
| |
Collapse
|
23
|
Molina GN, Tavarone E, Taboga O, Molinari P. Two Distinctive Phenotypes of AcMNPV Display Different Immune Abilities and Intracellular Destiny. PLoS One 2016; 11:e0168939. [PMID: 28033419 PMCID: PMC5199047 DOI: 10.1371/journal.pone.0168939] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2016] [Accepted: 12/08/2016] [Indexed: 11/18/2022] Open
Abstract
The budded phenotype (BV) of the baculovirus AcMNPV has been demonstrated to have strong immunostimulatory properties that are relevant for the development of vaccines and antiviral therapies. Although the occluded phenotype (ODV) shares the main structural proteins and its genome with BV, it has been poorly studied in mammals. In this study, we assessed the capacity of ODV to induce immune responses in mice. In contrast to BVs, ODVs failed to promote the secretion of IFN-gamma, IL-6 and Il-12 and to induce antiviral activity against VSV in the short term. Furthermore, ODVs were unable to induce cellular immunity against a coadministered antigen 7 days after inoculation. By analyzing the interaction of ODVs with BMDCs, we observed that although ODVs entered the cells reaching late and acidic endosomes, they did not induce their maturation. Finally, we also analyzed if BVs and ODVs followed different routes in the cell during the infection. BVs, but not ODVs, colocalized with the protein ovalbumin in compartments with the presence of proteases. The results suggest that structural differences could be responsible for their different destinies in the dendritic cell and this could lead to a different impact on the immune response.
Collapse
Affiliation(s)
- Guido N. Molina
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), CABA, Argentina
- Instituto de Biotecnología, Centro Nacional de Investigaciones Agropecuarias (CNIA), INTA Castelar, Buenos Aires, Argentina
| | - Eugenia Tavarone
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), CABA, Argentina
- Instituto de Biotecnología, Centro Nacional de Investigaciones Agropecuarias (CNIA), INTA Castelar, Buenos Aires, Argentina
| | - Oscar Taboga
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), CABA, Argentina
- Instituto de Biotecnología, Centro Nacional de Investigaciones Agropecuarias (CNIA), INTA Castelar, Buenos Aires, Argentina
- * E-mail:
| | - Paula Molinari
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), CABA, Argentina
- Instituto de Biotecnología, Centro Nacional de Investigaciones Agropecuarias (CNIA), INTA Castelar, Buenos Aires, Argentina
| |
Collapse
|
24
|
Particle and subunit-based hemagglutinin vaccines provide protective efficacy against H1N1 influenza in pigs. Vet Microbiol 2016; 191:35-43. [PMID: 27374905 DOI: 10.1016/j.vetmic.2016.05.011] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2015] [Revised: 05/09/2016] [Accepted: 05/23/2016] [Indexed: 11/21/2022]
Abstract
The increasing diversity of influenza strains circulating in swine herds escalates the potential for the emergence of novel pandemic viruses and highlights the need for swift development of new vaccines. Baculovirus has proven to be a flexible platform for the generation of recombinant forms of hemagglutinin (HA) including subunit, VLP-displayed, and baculovirus-displayed antigens. These presentations have been shown to be efficacious in mouse, chicken, and ferret models but little is known about their immunogenicity in pigs. To assess the utility of these HA presentations in swine, Baculovirus constructs expressing HA fused to swine IgG2a Fc, displayed in a FeLV gag VLP, or displayed in the baculoviral envelope were generated. Vaccines formulated with these antigens wer The e administered to groups of pigs who were subsequently challenged with H1α cluster H1N1 swine influenza virus (SIV) A/Swine/Indiana/1726/88. Our results demonstrate that vaccination with any of these three vaccines elicits robust hemagglutinin inhibition titers in the serum and decreased the severity of SIV-associated lung lesions after challenge when compared to placebo-vaccinated controls. In addition, the number of pigs with virus detected in the lungs and nasal passages was reduced. Taken together, the results demonstrate that these recombinant approaches expressed with the baculovirus expression vector system may be viable options for development of SIV vaccines for swine.
Collapse
|
25
|
Sim SH, Kim JY, Seong BL, Nguyen HH, Chang J. Baculovirus Displaying Hemagglutinin Elicits Broad Cross-Protection against Influenza in Mice. PLoS One 2016; 11:e0152485. [PMID: 27023684 PMCID: PMC4811570 DOI: 10.1371/journal.pone.0152485] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2015] [Accepted: 03/15/2016] [Indexed: 12/26/2022] Open
Abstract
The widespread influenza virus infection further emphasizes the need for novel vaccine strategies that effectively reduce the impact of epidemic as well as pandemic influenza. Conventional influenza vaccines generally induce virus neutralizing antibody responses which are specific for a few antigenically related strains within the same subtype. However, antibodies directed against the conserved stalk domain of HA could neutralize multiple subtypes of influenza virus and thus provide broad-spectrum protection. In this study, we designed and constructed a recombinant baculovirus-based vaccine, rBac-HA virus, that expresses full-length HA of pandemic H1N1 influenza virus (A/California/04/09) on the viral envelope. We demonstrated that repeated intranasal immunizations with rBac-HA virus induced HA stalk-specific antibody responses and protective immunity against homologous as well as heterosubtypic virus challenge. The adoptive transfer experiment shows that the cross-protection is conferred by the immune sera which contain HA stalk-specific antibodies. These results warrant further development of rBac-HA virus as a broad-protective vaccine against influenza. The vaccine induced protection against infection with the same subtype as well as different subtype, promising a potential universal vaccine for broad protection against different subtypes to control influenza outbreaks including pandemic.
Collapse
Affiliation(s)
- Sang-Hee Sim
- Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul, Korea
| | - Joo Young Kim
- Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul, Korea
| | - Baik Lin Seong
- Department of Biotechnology, College of Life Science and Biotechnology, Yonsei University, Seoul, Korea
| | - Huan Huu Nguyen
- Laboratory of Viral Immunology, International Vaccine Institute, Seoul, Korea
| | - Jun Chang
- Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul, Korea
- * E-mail:
| |
Collapse
|
26
|
Ladd Effio C, Hahn T, Seiler J, Oelmeier SA, Asen I, Silberer C, Villain L, Hubbuch J. Modeling and simulation of anion-exchange membrane chromatography for purification of Sf9 insect cell-derived virus-like particles. J Chromatogr A 2015; 1429:142-54. [PMID: 26718185 DOI: 10.1016/j.chroma.2015.12.006] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2015] [Revised: 12/01/2015] [Accepted: 12/03/2015] [Indexed: 11/25/2022]
Abstract
Recombinant protein-based virus-like particles (VLPs) are steadily gaining in importance as innovative vaccines against cancer and infectious diseases. Multiple VLPs are currently evaluated in clinical phases requiring a straightforward and rational process design. To date, there is no generic platform process available for the purification of VLPs. In order to accelerate and simplify VLP downstream processing, there is a demand for novel development approaches, technologies, and purification tools. Membrane adsorbers have been identified as promising stationary phases for the processing of bionanoparticles due to their large pore sizes. In this work, we present the potential of two strategies for designing VLP processes following the basic tenet of 'quality by design': High-throughput experimentation and process modeling of an anion-exchange membrane capture step. Automated membrane screenings allowed the identification of optimal VLP binding conditions yielding a dynamic binding capacity of 5.7 mg/mL for human B19 parvovirus-like particles derived from Spodoptera frugiperda Sf9 insect cells. A mechanistic approach was implemented for radial ion-exchange membrane chromatography using the lumped-rate model and stoichiometric displacement model for the in silico optimization of a VLP capture step. For the first time, process modeling enabled the in silico design of a selective, robust and scalable process with minimal experimental effort for a complex VLP feedstock. The optimized anion-exchange membrane chromatography process resulted in a protein purity of 81.5%, a DNA clearance of 99.2%, and a VLP recovery of 59%.
Collapse
Affiliation(s)
- Christopher Ladd Effio
- Karlsruhe Institute of Technology, Institute of Process Engineering in Life Sciences, Section IV: Biomolecular Separation Engineering, Karlsruhe, Germany
| | - Tobias Hahn
- Karlsruhe Institute of Technology, Institute of Process Engineering in Life Sciences, Section IV: Biomolecular Separation Engineering, Karlsruhe, Germany
| | - Julia Seiler
- Karlsruhe Institute of Technology, Institute of Process Engineering in Life Sciences, Section IV: Biomolecular Separation Engineering, Karlsruhe, Germany
| | - Stefan A Oelmeier
- Karlsruhe Institute of Technology, Institute of Process Engineering in Life Sciences, Section IV: Biomolecular Separation Engineering, Karlsruhe, Germany; Boehringer Ingelheim Pharma GmbH & Co. KG, Germany
| | | | | | | | - Jürgen Hubbuch
- Karlsruhe Institute of Technology, Institute of Process Engineering in Life Sciences, Section IV: Biomolecular Separation Engineering, Karlsruhe, Germany.
| |
Collapse
|
27
|
He F, Leyrer S, Kwang J. Strategies towards universal pandemic influenza vaccines. Expert Rev Vaccines 2015; 15:215-25. [DOI: 10.1586/14760584.2016.1115352] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Affiliation(s)
- Fang He
- Animal Health Biotechnology, Temasek Life Sciences Laboratory, Singapore, Singapore
| | - Sonja Leyrer
- Emergent Product Development Germany GmbH, Munich, Germany
| | - Jimmy Kwang
- Animal Health Biotechnology, Temasek Life Sciences Laboratory, Singapore, Singapore
- Department of Microbiology, Faculty of Medicine, National University of Singapore, Singapore, Singapore
| |
Collapse
|
28
|
Zhang Y, Qiao L, Hu X, Zhao K, Zhang Y, Chai F, Pan Z. Baculovirus vectors expressing F proteins in combination with virus-induced signaling adaptor (VISA) molecules confer protection against respiratory syncytial virus infection. Vaccine 2015; 34:252-260. [PMID: 26643933 DOI: 10.1016/j.vaccine.2015.11.027] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2015] [Revised: 11/04/2015] [Accepted: 11/12/2015] [Indexed: 11/16/2022]
Abstract
Baculovirus has been exploited for use as a novel vaccine vector. To investigate the feasibility and efficacy of recombinant baculoviruses (rBVs) expressing respiratory syncytial virus (RSV) fusion (F) proteins, four constructs (Bac-tF/64, Bac-CF, Bac-CF/tF64 and Bac-CF/tF64-VISA) were generated. Bac-tF64 displays the F ectodomain (tF) on the envelope of rBVs, whereas Bac-CF expresses full-length F protein in transduced mammalian cells. Bac-CF/tF64 not only displays tF on the envelope but also expresses F in cells. Bac-CF/tF64-VISA comprises Bac-CF/tF64 harboring the virus-induced signaling adaptor (VISA) gene. After administration to BALB/c mice, all four vectors elicited RSV neutralizing antibody (Ab), systemic Ab (IgG, IgG1, and IgG2a), and cytokine responses. Compared with Bac-tF64, mice inoculated with Bac-CF and Bac-CF/tF64 exhibited an increased mixed Th1/Th2 cytokine response, increased ratios of IgG2a/IgG1 antibody responses, and reduced immunopathology upon RSV challenge. Intriguingly, co-expression of VISA reduced Th2 cytokine (IL-4, IL-5, and IL-10) production induced by Bac-CF/tF64, thus relieving lung pathology upon a subsequent RSV challenge. Our results indicated that the Bac-CF/tF64 vector incorporated with the VISA molecule may provide an effective vaccine strategy for protection against RSV.
Collapse
Affiliation(s)
- Yuan Zhang
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan 430072, China
| | - Lei Qiao
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan 430072, China
| | - Xiao Hu
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan 430072, China
| | - Kang Zhao
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan 430072, China
| | - Yanwen Zhang
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan 430072, China
| | - Feng Chai
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan 430072, China
| | - Zishu Pan
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan 430072, China.
| |
Collapse
|
29
|
Carnell GW, Ferrara F, Grehan K, Thompson CP, Temperton NJ. Pseudotype-based neutralization assays for influenza: a systematic analysis. Front Immunol 2015; 6:161. [PMID: 25972865 PMCID: PMC4413832 DOI: 10.3389/fimmu.2015.00161] [Citation(s) in RCA: 56] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2015] [Accepted: 03/25/2015] [Indexed: 12/02/2022] Open
Abstract
The use of vaccination against the influenza virus remains the most effective method of mitigating the significant morbidity and mortality caused by this virus. Antibodies elicited by currently licensed influenza vaccines are predominantly hemagglutination-inhibition (HI)-competent antibodies that target the globular head of hemagglutinin (HA) thus inhibiting influenza virus entry into target cells. These antibodies predominantly confer homosubtypic/strain specific protection and only rarely confer heterosubtypic protection. However, recent academia or pharma-led R&D toward the production of a “universal vaccine” has centered on the elicitation of antibodies directed against the stalk of the influenza HA that has been shown to confer broad protection across a range of different subtypes (H1–H16). The accurate and sensitive measurement of antibody responses elicited by these “next-generation” influenza vaccines is, however, hampered by the lack of sensitivity of the traditional influenza serological assays HI, single radial hemolysis, and microneutralization. Assays utilizing pseudotypes, chimeric viruses bearing influenza glycoproteins, have been shown to be highly efficient for the measurement of homosubtypic and heterosubtypic broadly neutralizing antibodies, making them ideal serological tools for the study of cross-protective responses against multiple influenza subtypes with pandemic potential. In this review, we will analyze and compare literature involving the production of influenza pseudotypes with particular emphasis on their use in serum antibody neutralization assays. This will enable us to establish the parameters required for optimization and propose a consensus protocol to be employed for the further deployment of these assays in influenza vaccine immunogenicity studies.
Collapse
Affiliation(s)
- George William Carnell
- Viral Pseudotype Unit, Medway School of Pharmacy, The Universities of Greenwich and Kent at Medway , Chatham Maritime, Kent , UK
| | - Francesca Ferrara
- Viral Pseudotype Unit, Medway School of Pharmacy, The Universities of Greenwich and Kent at Medway , Chatham Maritime, Kent , UK
| | - Keith Grehan
- Viral Pseudotype Unit, Medway School of Pharmacy, The Universities of Greenwich and Kent at Medway , Chatham Maritime, Kent , UK
| | - Craig Peter Thompson
- Viral Pseudotype Unit, Medway School of Pharmacy, The Universities of Greenwich and Kent at Medway , Chatham Maritime, Kent , UK ; Department of Zoology, University of Oxford , Oxford , UK ; The Jenner Institute Laboratories, University of Oxford , Oxford , UK
| | - Nigel James Temperton
- Viral Pseudotype Unit, Medway School of Pharmacy, The Universities of Greenwich and Kent at Medway , Chatham Maritime, Kent , UK
| |
Collapse
|
30
|
Lin SY, Chung YC, Hu YC. Update on baculovirus as an expression and/or delivery vehicle for vaccine antigens. Expert Rev Vaccines 2014; 13:1501-21. [DOI: 10.1586/14760584.2014.951637] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
|
31
|
Immunization with baculovirus displayed H6 hemagglutinin vaccine protects mice against lethal H6 influenza virus challenge. Antiviral Res 2014; 109:42-53. [PMID: 24973759 DOI: 10.1016/j.antiviral.2014.06.002] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2012] [Revised: 05/21/2014] [Accepted: 06/02/2014] [Indexed: 11/22/2022]
Abstract
Low pathogenic influenza viruses of H6 hemagglutinin (HA) subtype have a high prevalence among aquatic and domestic birds and have caused outbreaks in poultry worldwide. The first human infection with wild avian influenza H6N1 virus was reported in Taiwan and these subtype viruses may continue to evolve and accumulate changes which increasing the potential risk of human-to-human transmission. To develop a vaccine against influenza viruses of the H6 subtype, we displayed the HA gene on the baculovirus surface (Bac-HA), and studied its vaccine efficacy against a lethal challenge with mouse-adapted RG-H6(Shorebird) virus carrying the H6 HA gene from A/shorebird/DE/12/2004 (H6N8) virus and 7 genes from A/Puerto Rico/8/1934 (H1N1) virus. Immunization with 256 HA units of Bac-HA via the intranasal route triggered HA-specific serum and mucosal antibodies in mice besides increased HA inhibition titers compared to mice immunized subcutaneously. Moreover, we observed an increase in cellular immune response (IL-4) and improved in vitro neutralization activity in the mice immunized intranasally with live Bac-HA compared to mice immunized with inactivated influenza virus (IV). Interestingly, Bac-HA intranasal immunized mice showed one fold higher neutralization titer against heterologous H6 influenza virus compared to inactivated IV immunized mice. In addition, the live Bac-HA, administered through either immunization route, as well as the adjuvanted inactivated Bac-HA, administered subcutaneously, conferred 100% protection to mice challenged with homologous mouse-adapted RG-H6(Shorebird) virus. The reduction in viral titers and extend of histopathological changes of Bac-HA immunized mice lungs further demonstrated the protective efficacy of Bac-HA. Hence, the recombinant baculovirus subunit vaccine is an alternative candidate against H6 subtypes that could be propagated and administered with minimal biosafety concerns.
Collapse
|
32
|
Hu YC, Yao K, Wu TY. Baculovirus as an expression and/or delivery vehicle for vaccine antigens. Expert Rev Vaccines 2014; 7:363-71. [PMID: 18393606 DOI: 10.1586/14760584.7.3.363] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Affiliation(s)
- Yu-Chen Hu
- Department of Chemical Engineering, National Tsing Hua University, Hsinchu, 300 Taiwan.
| | | | | |
Collapse
|
33
|
Mena JA, Kamen AA. Insect cell technology is a versatile and robust vaccine manufacturing platform. Expert Rev Vaccines 2014; 10:1063-81. [DOI: 10.1586/erv.11.24] [Citation(s) in RCA: 88] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
34
|
Innate immune response induced by baculovirus attenuates transgene expression in mammalian cells. J Virol 2013; 88:2157-67. [PMID: 24335288 DOI: 10.1128/jvi.03055-13] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
The baculovirus Autographa californica nucleopolyhedrovirus (AcNPV) has been widely used to achieve a high level of foreign gene expression in insect cells, as well as for efficient gene transduction into mammalian cells without any replication. In addition to permitting efficient gene delivery, baculovirus has been shown to induce host innate immune responses in various mammalian cells and in mice. In this study, we examined the effects of the innate immune responses on gene expression by recombinant baculoviruses in cultured cells. The reporter gene expression in IRF3-deficient mouse embryonic fibroblasts (MEFs) infected with the recombinant baculovirus was shown to be enhanced in accordance with the suppression of beta interferon (IFN-β) production. Furthermore, efficient gene transduction by the recombinant baculovirus was achieved in MEFs deficient for stimulator of interferon genes (STING), TANK binding kinase 1 (TBK1), IFN regulatory factor 3 (IRF3), or IFN-β promoter stimulator 1 (IPS-1), but not in those deficient for IRF7, MyD88, or Z-DNA binding protein 1 (ZBP1)/DAI. Enhancement of gene expression by the recombinant baculovirus was also observed in human hepatoma cell lines replicating hepatitis C virus (HCV), in which innate immunity was impaired by the cleavage of IPS-1 by the viral protease. In addition, infection with the recombinant baculovirus expressing the BH3-only protein, BIMS, a potent inducer of apoptosis, resulted in a selective cell death in the HCV replicon cells. These results indicate that innate immune responses induced by infection with baculovirus attenuate transgene expression, and this characteristic might be useful for a selective gene transduction into cells with impaired innate immunity arising from infection with various viruses.
Collapse
|
35
|
Kiener TK, Premanand B, Kwang J. Immune responses to baculovirus-displayed enterovirus 71 VP1 antigen. Expert Rev Vaccines 2013; 12:357-64. [PMID: 23560917 DOI: 10.1586/erv.13.18] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
The increased distribution and neurovirulence of enterovirus 71 is an important health threat for young children in Asia Pacific. Vaccine design has concentrated on inactivated virus with the most advanced undergoing Phase III clinical trials. By using a subunit vaccine approach, production costs could be reduced by lowering the need for biocontainment. In addition, novel mutations could be rapidly incorporated to reflect the emergence of new enterovirus 71 subgenogroups. To circumvent the problems associated with conventional subunit vaccines, the antigen can be displayed on a viral vector that conveys stability and facilitates purification. Additional advantages of viral-vectored subunit vaccines are their ability to stimulate the innate immune system by transducing cells and the possibility of oral or nasal delivery, which dispenses with the need for syringes and medical personnel. Baculovirus-displayed VP1 combines all these benefits with protection that is as efficient as inactivated virus.
Collapse
Affiliation(s)
- Tanja K Kiener
- Animal Health Biotechnology, Temasek Life Sciences Laboratory, Singapore
| | | | | |
Collapse
|
36
|
Shen H, Xue C, Lv L, Wang W, Liu Q, Liu K, Chen X, Zheng J, Li X, Cao Y. Assembly and immunological properties of a bivalent virus-like particle (VLP) for avian influenza and Newcastle disease. Virus Res 2013; 178:430-6. [PMID: 24050994 DOI: 10.1016/j.virusres.2013.09.009] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2013] [Revised: 07/25/2013] [Accepted: 09/06/2013] [Indexed: 11/15/2022]
Abstract
Avian influenza virus (AIV) and Newcastle disease virus (NDV) are both important pathogens in poultry worldwide. The protection of poultry from avian influenza and Newcastle disease can be achieved through vaccination. We embarked on the development of a bivalent vaccine that would allow for a single immunization against both avian influenza and Newcastle disease. We constructed a chimeric virus-like particle (VLP) that is composed of the M1 protein and HA protein of avian influenza virus and a chimeric protein containing the cytoplasmic and transmembrane domains of AIV neuraminidase protein (NA) and the ectodomain of the NDV hemagglutinin-neuraminidase (HN) protein (NA/HN). The single immunization of chickens with the chimeric VLP vaccine induced both AIV H5- and NDV-specific antibodies. The HI titers and specific antibodies elicited by the chimeric VLPs were statistically similar to those elicited in animals vaccinated with the corresponding commercial monovalent vaccines. Chickens vaccinated with chimeric VLP vaccine and then challenged with the Newcastle disease F48E9 virus displayed complete protection. Overall, the chimeric VLP vaccine elicits strong immunity and can protect against Newcastle disease virus challenge.
Collapse
Affiliation(s)
- Huifang Shen
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou 510006, PR China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Karuppannan AK, Qiang J, Chang CC, Kwang J. A novel baculovirus vector shows efficient gene delivery of modified porcine reproductive and respiratory syndrome virus antigens and elicits specific immune response. Vaccine 2013; 31:5471-8. [PMID: 24035590 DOI: 10.1016/j.vaccine.2013.08.101] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2013] [Revised: 08/23/2013] [Accepted: 08/29/2013] [Indexed: 01/19/2023]
Abstract
Porcine reproductive and respiratory syndrome (PRRS) is an economically devastating epizootic of porcine species. Current vaccines are inadequate to control the disease burden and outbreaks in the field. We report a novel baculovirus vaccine vector with White spot syndrome virus immediate early 1 shuttle promoter, with strong activity in both insect cells and mammalian cells, for immunization against PRRSV. The insect cell cultured baculovirus vector produces PRRSV envelope glycoproteins ORF2a, ORF3, ORF4 and ORF5, which are similar to the antigens in the infectious PRRS virion, and these antigens are stably incorporated on the surface of the baculovirus. Further, the baculovirus vector efficiently transduces these antigens in cells of porcine origin, thereby simulating a live infection. The baculovirus vectored PRRSV antigens, upon inoculation in mice, elicits robust neutralizing antibodies against the infective PRRS virus. Further, the experiments indicate that hitherto under emphasized ORF2a and ORF4 are important target antigens for neutralizing PRRSV infectivity.
Collapse
Affiliation(s)
- Anbu K Karuppannan
- Centre for Animal Health Studies, Tamil Nadu Veterinary and Animal Sciences University, Madhavaram, Chennai 600051, India
| | | | | | | |
Collapse
|
38
|
Jazayeri SD, Ideris A, Shameli K, Moeini H, Omar AR. Gene expression profiles in primary duodenal chick cells following transfection with avian influenza virus H5 DNA plasmid encapsulated in silver nanoparticles. Int J Nanomedicine 2013; 8:781-90. [PMID: 23459681 PMCID: PMC3582479 DOI: 10.2147/ijn.s39074] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
In order to develop a systemically administered safe and effective nonviral gene delivery system against avian influenza virus (AIV) that induced cytokine expression, the hemagglutinin (H5) gene of AIV, A/Ck/Malaysia/5858/04 (H5N1) and green fluorescent protein were cloned into a coexpression vector pIRES (pIREGFP-H5) and formulated using green synthesis of silver nanoparticles (AgNPs) with poly(ethylene glycol) and transfected into primary duodenal cells taken from 18-day-old specific-pathogen-free chick embryos. The AgNPs were prepared using moderated temperature and characterized for particle size, surface charge, ultraviolet-visible spectra, DNA loading, and stability. AgNPs and AgNP-pIREGFP-H5 were prepared in the size range of 13.9 nm and 25 nm with a positive charge of +78 ± 0.6 mV and +40 ± 6.2 mV, respectively. AgNPs with a positive surface charge could encapsulate pIREGFP-H5 efficiently. The ultraviolet-visible spectra for AgNP-pIREGFP-H5 treated with DNase I showed that the AgNPs were able to encapsulate pIREGFP-H5 efficiently. Polymerase chain reaction showed that AgNP-pIREGFP-H5 entered into primary duodenal cells rapidly, as early as one hour after transfection. Green fluorescent protein expression was observed after 36 hours, peaked at 48 hours, and remained stable for up to 60 hours. In addition, green fluorescent protein expression generally increased with increasing DNA concentration and time. Cells were transfected using Lipocurax in vitro transfection reagent as a positive control. A multiplex quantitative mRNA gene expression assay in the transfected primary duodenal cells via the transfection reagent and AgNPs with pIREGFP-H5 revealed expression of interleukin (IL)-18, IL-15, and IL-12β.
Collapse
|
39
|
Chen CY, Lin SY, Cheng MC, Tsai CP, Hung CL, Lo KW, Hwang Y, Hu YC. Baculovirus vector as an avian influenza vaccine: hemagglutinin expression and presentation augment the vaccine immunogenicity. J Biotechnol 2013; 164:143-50. [PMID: 23313887 DOI: 10.1016/j.jbiotec.2012.12.013] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2012] [Revised: 12/17/2012] [Accepted: 12/21/2012] [Indexed: 12/11/2022]
Abstract
Baculovirus simultaneously displaying and expressing the avian influenza virus (AIV) hemagglutinin (HA) protein can induce potent anti-HA humoral and cellular immune responses. Based on the hypothesis that improving the antigen expression and presentation can further boost the AIV vaccine efficacies, we first constructed a baculoviral vector (Bac-HAW) with HA gene fused with the woodchuck hepatitis virus post-transcriptional regulatory element (WPRE) near its 3' end and expressed under the control of the hybrid CAG promoter. The WPRE fusion improved the HA expression and augmented the humoral and Th1 cellular immune responses after intramuscular administration into BALB/c mice. With Bac-HAW as the backbone, we next constructed Bac-HAMW which harbored the HA gene flanked with the signal sequence (MHCIss) and trafficking domain (MITD) of MHC class I molecule. In comparison with Bac-HAW, Bac-HAMW ameliorated the HA peptide presentation, significantly elevated the HA-specific humoral response (total IgG, IgG2a and hemagglutination inhibition titers) and favorably boosted the Th1 and IFN-γ(+)/CD8(+) T cell responses without extraneous adjuvants. These data collectively confirmed that enhancement of antigen expression and presentation by combining the WPRE and MHCIss/MITD fusion can potentiate the immunogenicity of the baculovirus-based vaccine, and implicates the potential of Bac-HAMW as an appealing AIV vaccine.
Collapse
Affiliation(s)
- Chi-Yuan Chen
- Department of Chemical Engineering, National Tsing Hua University, Hsinchu 30013, Taiwan
| | | | | | | | | | | | | | | |
Collapse
|
40
|
Mooney AJ, Tompkins SM. Experimental vaccines against potentially pandemic and highly pathogenic avian influenza viruses. Future Virol 2013; 8:25-41. [PMID: 23440999 PMCID: PMC3579652 DOI: 10.2217/fvl.12.122] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Influenza A viruses continue to emerge and re-emerge, causing outbreaks, epidemics and occasionally pandemics. While the influenza vaccines licensed for public use are generally effective against seasonal influenza, issues arise with production, immunogenicity, and efficacy in the case of vaccines against pandemic and emerging influenza viruses, and highly pathogenic avian influenza virus in particular. Thus, there is need of improved influenza vaccines and vaccination strategies. This review discusses advances in alternative influenza vaccines, touching briefly on licensed vaccines and vaccine antigens; then reviewing recombinant subunit vaccines, virus-like particle vaccines and DNA vaccines, with the main focus on virus-vectored vaccine approaches.
Collapse
Affiliation(s)
- Alaina J Mooney
- Department of Infectious Diseases, University of Georgia, 111 Carlton St, Athens, GA 30602, USA
| | - S Mark Tompkins
- Department of Infectious Diseases, University of Georgia, 111 Carlton St, Athens, GA 30602, USA
| |
Collapse
|
41
|
Kato T, Suzuki F, Park EY. Display of the human (pro)renin receptor on Bombyx mori nucleopolyhedrovirus (BmNPV) particles using Bm cells. J Biosci Bioeng 2012; 114:564-9. [PMID: 22762973 DOI: 10.1016/j.jbiosc.2012.06.008] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2012] [Revised: 06/07/2012] [Accepted: 06/08/2012] [Indexed: 01/20/2023]
Abstract
The human (pro)renin receptor (hPRR) was displayed on the surface of Bombyx mori nucleopolyhedrovirus (BmNPV) with and without fusion to glycoprotein 64 (GP64) of the BmNPV. hPRR1 is a native hPRR with an additional FLAG peptide sequence inserted between the signal peptide and prorenin-binding domain. hPRR2 has the prorenin-binding domain inserted between amino acid residues (81)Asp and (82)Pro of GP64. hPRR4 has the prorenin-binding domain inserted in (81)Asp and (320)Met of partially deleted GP64. Incorporation of hPRR was confirmed in recombinant BmNPV (rBmNPV) but not in cysteine protease-deleted rBmNPV. hPRR1 was observed in ER, but hPRR2 and hPRR4 were observed around the endoplasmic reticulum (ER) and in its periphery. rBmNPV-hPRR1 and -hPRR2, carrying hPRR1 and hPRR2 respectively, showed binding affinity to human renin, but rBmNPV-hPRR4 did not. The presence of hPRR4 of rBmNPV-hPRR4 was confirmed in western blotting under nonreducing conditions, suggesting that although hPRR4 was incorporated in rBmNPV-hPRR4, it behaved as a non-functional aggregate. This rBmNPV display system can also be used for analyzing a ligand-receptor interaction.
Collapse
Affiliation(s)
- Tatsuya Kato
- Laboratory of Biotechnology, Faculty of Agriculture, Shizuoka University, 836 Ohya, Suruga-ku, Shizuoka 422-8529, Japan
| | | | | |
Collapse
|
42
|
Abstract
Baculovirus is extensively utilized as an excellent tool for production of recombinant protein in insect cells. Baculovirus infects insects in nature and is non-pathogenic to humans. In addition to insect cells, baculovirus is capable of transducing a broad range of animal cells. Due to its biosafety, large cloning capacity, low cytotoxicity, and non-replication nature in the transduced cells as well as the ease of manipulation and production, baculovirus has been utilized as RNA interference mediators, gene delivery vectors, and vaccine vectors for a wide variety of applications. This article focuses on the utilization of baculoviruses as vaccine vectors to prepare antigen or subunit vaccines.
Collapse
Affiliation(s)
- Hsin-Yu Lu
- Institute of Molecular Biology, National Chung Hsing University, Taichung, Taiwan
| | | | | |
Collapse
|
43
|
Kim S, Chang J. Baculovirus-based Vaccine Displaying Respiratory Syncytial Virus Glycoprotein Induces Protective Immunity against RSV Infection without Vaccine-Enhanced Disease. Immune Netw 2012; 12:8-17. [PMID: 22536165 PMCID: PMC3329602 DOI: 10.4110/in.2012.12.1.8] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2011] [Revised: 01/08/2012] [Accepted: 01/08/2012] [Indexed: 02/03/2023] Open
Abstract
Background Respiratory syncytial virus (RSV) is a major cause of severe lower respiratory tract diseases in infancy and early childhood. Despite its importance as a pathogen, there is no licensed vaccine against RSV yet. The attachment glycoprotein (G) of RSV is a potentially important target for protective antiviral immune responses. Recombinant baculovirus has been recently emerged as a new vaccine vector, since it has intrinsic immunostimulatory properties and good bio-safety profile. Methods We have constructed a recombinant baculovirus-based RSV vaccine, Bac-RSV/G, displaying G glycoprotein, and evaluated immunogenicity and protective efficacy by intranasal immunization of BALB/c mice with Bac-RSV/G. Results Bac-RSV/G efficiently provides protective immunity against RSV challenge. Strong serum IgG and mucosal IgA responses were induced by intranasal immunization with Bac-RSV/G. In addition to humoral immunity, G-specific Th17- as well as Th1-type T-cell responses were detected in the lungs of Bac-RSV/G-immune mice upon RSV challenge. Neither lung eosinophilia nor vaccine-induced weight loss was observed upon Bac-RSV/G immunization and subsequent RSV infection. Conclusion Our data demonstrate that intranasal administration of baculovirus-based Bac-RSV/G vaccine is efficient for the induction of protection against RSV and represents a promising prophylactic vaccination regimen.
Collapse
Affiliation(s)
- Sol Kim
- Division of Life & Pharmaceutical Sciences, and Center for Cell Signaling & Drug Discovery Research, Ewha Womans University, Seoul 120-750, Korea
| | | |
Collapse
|
44
|
Sokolenko S, George S, Wagner A, Tuladhar A, Andrich JMS, Aucoin MG. Co-expression vs. co-infection using baculovirus expression vectors in insect cell culture: Benefits and drawbacks. Biotechnol Adv 2012; 30:766-81. [PMID: 22297133 PMCID: PMC7132753 DOI: 10.1016/j.biotechadv.2012.01.009] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2011] [Revised: 01/13/2012] [Accepted: 01/17/2012] [Indexed: 12/12/2022]
Abstract
The baculovirus expression vector system (BEVS) is a versatile and powerful platform for protein expression in insect cells. With the ability to approach similar post-translational modifications as in mammalian cells, the BEVS offers a number of advantages including high levels of expression as well as an inherent safety during manufacture and of the final product. Many BEVS products include proteins and protein complexes that require expression from more than one gene. This review examines the expression strategies that have been used to this end and focuses on the distinguishing features between those that make use of single polycistronic baculovirus (co-expression) and those that use multiple monocistronic baculoviruses (co-infection). Three major areas in which researchers have been able to take advantage of co-expression/co-infection are addressed, including compound structure-function studies, insect cell functionality augmentation, and VLP production. The core of the review discusses the parameters of interest for co-infection and co-expression with time of infection (TOI) and multiplicity of infection (MOI) highlighted for the former and the choice of promoter for the latter. In addition, an overview of modeling approaches is presented, with a suggested trajectory for future exploration. The review concludes with an examination of the gaps that still remain in co-expression/co-infection knowledge and practice.
Collapse
Affiliation(s)
- Stanislav Sokolenko
- Department of Chemical Engineering, Waterloo Institute for Nanotechnology, University of Waterloo, Waterloo, Ontario, Canada N2L 3G1
| | | | | | | | | | | |
Collapse
|
45
|
Xu XG, Wang ZS, Zhang Q, Li ZC, Ding L, Li W, Wu HY, Chang CD, Lee LH, Tong DW, Liu HJ. Baculovirus as a PRRSV and PCV2 bivalent vaccine vector: baculovirus virions displaying simultaneously GP5 glycoprotein of PRRSV and capsid protein of PCV2. J Virol Methods 2011; 179:359-66. [PMID: 22172969 DOI: 10.1016/j.jviromet.2011.11.023] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2011] [Revised: 11/28/2011] [Accepted: 11/28/2011] [Indexed: 11/19/2022]
Abstract
The GP5 glycoprotein of PRRSV is the main target for inducing neutralizing antibodies and protective immunity in the natural host. The capsid (Cap) protein is the major immunogenic protein and associated with the production of PCV2-specific neutralizing antibodies. In the present study, one genetic recombinant baculovirus BacSC-Dual-GP5-Cap was constructed. This virus displays simultaneously histidine-tagged GP5 and Cap proteins with the baculovirus glycoprotein gp64 TM and CTD on the virion surface as well as the surface of the virus-infected cells. After infection, the GP5 and Cap proteins were expressed and anchored simultaneously on the plasma membrane of Sf-9 cells, as revealed by Western blot and confocal microscopy. This report demonstrated first that both GP5 and Cap proteins were displayed successfully on the viral surface, revealed by immunogold electron microscopy. Vaccination of swine with recombinant baculovirus BacSC-Dual-GP5-Cap elicited significantly higher GP5 and Cap ELISA antibody titers in swine than the control groups. Virus neutralization test also showed that serum from the BacSC-Dual-GP5-Cap treated swine had significant levels of virus neutralization titers. Lymphocyte proliferation responses could be induced in swine immunized with BacSC-Dual-GP5-Cap than the control groups. These findings demonstrate that the BacSC-Dual-GP5-Cap bivalent subunit vaccine can be a potential vaccine against PRRSV and PCV2 infections.
Collapse
Affiliation(s)
- Xin-Gang Xu
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi 712100, China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
CD40L-containing virus-like particle as a candidate HIV-1 vaccine targeting dendritic cells. J Acquir Immune Defic Syndr 2011; 56:393-400. [PMID: 21239998 DOI: 10.1097/qai.0b013e31820b844e] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
The central role of dendritic cell (DC) in mounting an immune response to a novel antigen is now well established. We sought to demonstrate the use of a particular vaccine strategy based on directing HIV-1 Gag proteins to DCs in conjunction with an activation signal. CD40L was expressed on the surface of virus-like particles (VLPs) to target HIV-1 Gag antigens to the CD40 receptor on DCs, whereas CD40L-CD40 interaction would also result in cellular activation. Multiple CD40L VLP constructs were made and evaluated in vitro and in vivo. Indeed, one VLP that expressed CD40L to the highest level showed greatest capacity to activate DCs in vitro. Correspondingly, this CD40L-VLP also proved to be most immunogenic in mice in raising both humoral and cellular responses to HIV-1 Gag. Confirmatory studies were performed to demonstrate the increased immunogenicity of CD40L-VLP is no longer observed when tested in CD40-/- mice. Our findings lend support to the belief that vaccine strategies that both target and activate DCs could yield a superior immune response.
Collapse
|
47
|
S SM, Kwang J. Oral vaccination of baculovirus-expressed VP28 displays enhanced protection against White Spot Syndrome Virus in Penaeus monodon. PLoS One 2011; 6:e26428. [PMID: 22069450 PMCID: PMC3206036 DOI: 10.1371/journal.pone.0026428] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2011] [Accepted: 09/26/2011] [Indexed: 12/19/2022] Open
Abstract
White Spot Syndrome Virus (WSSV) is an infectious pathogen of shrimp and other crustaceans, and neither effective vaccines nor adequate treatments are currently available. WSSV is an enveloped dsDNA virus, and one of its major envelope proteins, VP28, plays a pivotal role in WSSV infection. In an attempt to develop a vaccine against WSSV, we inserted the VP28 gene into a baculovirus vector tailored to express VP28 on the baculovirus surface under the WSSV ie1 promoter (Bac-VP28). The Bac-VP28 incorporated abundant quantity (65.3 µg/ml) of VP28. Shrimp were treated by oral and immersion vaccination with either Bac-VP28 or wild-type baculovirus (Bac-wt). The treatment was followed by challenge with WSSV after 3 and 15 days. Bac-VP28 vaccinated shrimp showed significantly higher survival rates (oral: 81.7% and 76.7%; immersion: 75% and 68.4%) than Bac-wt or non-treated shrimp (100% mortality). To verify the protective effects of Bac-VP28, we examined in vivo expression of VP28 by immunohistochemistry and quantified the WSSV copy number by qPCR. In addition to that, we quantified the expression levels shrimp genes LGBP and STAT by real-time RT-PCR from the samples obtained from Bac-VP28 vaccinated shrimp at different duration of vaccine regime. Our findings indicate that oral vaccination of shrimp with Bac-VP28 is an attractive preventative measure against WSSV infection that can be used in the field.
Collapse
Affiliation(s)
- Syed Musthaq S
- Animal Health Biotechnology, Temasek Lifesciences Laboratory, National University of Singapore, Singapore, Singapore
| | - Jimmy Kwang
- Animal Health Biotechnology, Temasek Lifesciences Laboratory, National University of Singapore, Singapore, Singapore
- Department of Microbiology, Faculty of Medicine, National University of Singapore, Singapore, Singapore
- * E-mail:
| |
Collapse
|
48
|
Xu XG, Tong DW, Wang ZS, Zhang Q, Li ZC, Zhang K, Li W, Liu HJ. Baculovirus virions displaying infectious bursal disease virus VP2 protein protect chickens against infectious bursal disease virus infection. Avian Dis 2011; 55:223-9. [PMID: 21793437 DOI: 10.1637/9597-111210-reg.1] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Infectious bursal disease (IBD) is an acute and contagious viral infection of young chickens caused by IBD virus (IBDV). The VP2 protein of IBDV is the only antigen for inducing neutralizing antibodies and protective immunity in the natural host. In the current study, we have succeeded in construction of one recombinant baculovirus BacSC-VP2 expressing His6-tagged VP2 with the baculovirus envelope protein gp64 transmembrane domain (TM) and cytoplasmic domain (CTD). The His6-tagged recombinant VP2 was expressed and anchored on the plasma membrane of Sf-9 cells, as examined by western blot and confocal microscopy. Immunogold electron microscopy demonstrated that the VP2 protein of IBDV was successfully displayed on the viral surface. Vaccination of chickens with the VP2-pseudotyped baculovirus vaccine (BacSC-VP2) elicited significantly higher levels of VP2-specific enzyme-linked immunosorbent assay antibodies and neutralizing antibodies than the control groups. IBDV-specific proliferation of lymphocytes was observed in chickens immunized with the recombinant BacSC-VP2. An in vivo challenge study of the recombinant baculovirus BacSC-VP2 showed effective protection against a very virulent (vv) IBDV infection in chickens. In addition, mortality and gross and histopathological findings in the bursa demonstrated the efficacy of the vaccine in reducing virulence of the disease. These results indicate that the recombinant baculovirus BacSC-VP2 can be a potential vaccine against IBDV infections.
Collapse
Affiliation(s)
- Xin-Gang Xu
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi 712100, People's Republic of China
| | | | | | | | | | | | | | | |
Collapse
|
49
|
Novel post-translational modifications of the hemagglutinin and neuraminidase proteins of avian influenza virus expressed by Kluyveromyces lactis. J Virol Methods 2011; 175:175-81. [DOI: 10.1016/j.jviromet.2011.05.006] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2011] [Revised: 04/21/2011] [Accepted: 05/04/2011] [Indexed: 11/18/2022]
|
50
|
Lin W, Fan H, Cheng X, Ye Y, Chen X, Ren T, Qi W, Liao M. A baculovirus dual expression system-based vaccine confers complete protection against lethal challenge with H9N2 avian influenza virus in mice. Virol J 2011; 8:273. [PMID: 21639929 PMCID: PMC3120790 DOI: 10.1186/1743-422x-8-273] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2011] [Accepted: 06/04/2011] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Avian influenza viruses of H9N2 subtype have become highly prevalent in avian species. Although these viruses generally cause only mild to moderate disease, they can infect a wide variety of species, including chickens, quail, turkeys, ducks, geese, pheasant, partridge, and pigeon, even transmitted to mammalian species, including humans, accelerating the efforts to devise protective strategies against them. RESULTS The results showed that stronger immune responses were induced in a mouse model immunized with BV-Dual-HA than in those vaccinated with a DNA vaccine encoding the same antigen. Moreover, complete protection against lethal challenge with H9N2 virus was observed in mice. CONCLUSION BV-Dual-HA could be utilized as a vaccine candidate against H9N2 virus infection.
Collapse
Affiliation(s)
- Wenyao Lin
- Key Laboratory of Animal Disease Control and Prevention of the Ministry of Agriculture, Guangzhou, China
| | | | | | | | | | | | | | | |
Collapse
|