1
|
Puente-Massaguer E, Gòdia F. Recombinant Protein Production in Suspension Mammalian Cells Using the BacMam Baculovirus Expression System. Methods Mol Biol 2024; 2829:329-339. [PMID: 38951347 DOI: 10.1007/978-1-0716-3961-0_25] [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] [Indexed: 07/03/2024]
Abstract
Mammalian cell lines are one of the best options when it comes to the production of complex proteins requiring specific glycosylation patterns. Plasmid DNA transfection and stable cell lines are frequently used for recombinant protein production, but they are expensive at large scale or can become time-consuming, respectively. The BacMam baculovirus (BV) is a safe and cost-effective platform to produce recombinant proteins in mammalian cells. The process of generating BacMam BVs is straightforward and similar to the generation of "insect" BVs, with different commercially available platforms. Although there are several protocols that describe recombinant protein expression with the BacMam BV in adherent cell lines, limited information is available on suspension cells. Therefore, it is of relevance to define the conditions to produce recombinant proteins in suspension cell cultures with BacMam BVs that facilitate bioprocess transfer to larger volumes. Here, we describe a method to generate a high titer BacMam BV stock and produce recombinant proteins in suspension HEK293 cells.
Collapse
Affiliation(s)
- Eduard Puente-Massaguer
- Department of Chemical, Biological and Environmental Engineering, Universitat Autònoma de Barcelona, Barcelona, Spain.
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| | - Francesc Gòdia
- Department of Chemical, Biological and Environmental Engineering, Universitat Autònoma de Barcelona, Barcelona, Spain
| |
Collapse
|
2
|
Zhu M, Shen Z, Gu Y, Tong X, Zhang Y, Pan J, Feng Y, Hu X, Wang Y, Cao G, Xue R, Gong C. A recombinant baculovirus vector vaccine (BacMCP) against the infectious spleen and kidney necrosis virus (ISKNV). JOURNAL OF FISH DISEASES 2023; 46:165-176. [PMID: 36423261 DOI: 10.1111/jfd.13731] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Revised: 11/05/2022] [Accepted: 11/12/2022] [Indexed: 06/16/2023]
Abstract
The infectious spleen and kidney necrosis virus (ISKNV) is a highly lethal virus, which has brought significant losses to aquaculture. Therefore, a new vaccine against ISKNV with high efficiency, safety and convenience must be developed. While baculoviruses are more commonly used as protein expression systems for vaccine antigen production, this paper used baculovirus technology to develop a live-vector vaccine, BacMCP, which contains the coding sequence of the major capsid protein (MCP) (GenBank accession no. AF371960) of ISKNV and is driven by a CMV promoter. Real-time PCR and immunofluorescence showed that the MCP gene was successfully delivered to and expressed in fish cells and tissues inoculated with BacMCP. Immune-related gene (IgM, TGF-β, IL-1, IL-8, TNF-α) expression was induced in BacMCP-treated groups of largemouth bass compared with control groups. Specific antibodies could be detected in the serum of BacMCP injection-vaccinated largemouth bass by ELISA. After injection or immersion vaccination with BacMCP for 21 days, largemouth bass were infected with ISKNV. The immune effect of the injected immunization on fish in different sizes was evaluated. The vaccine efficacy of injection-vaccinated bass was 100% in small bass and 85.7% in large bass. The vaccine efficacy of immersion-vaccinated small bass was 77.3%. This study suggested that BacMCP can be used as a vector-based vaccine candidate to prevent the diseases caused by ISKNV infection.
Collapse
Affiliation(s)
- Min Zhu
- School of Biology and Basic Medical Sciences, Soochow University, Suzhou, China
- Agricultural Biotechnology Research Institute, Agricultural biotechnology and Ecological Research Institute, Soochow University, Suzhou, China
| | - Zeen Shen
- School of Biology and Basic Medical Sciences, Soochow University, Suzhou, China
| | - Yuchao Gu
- School of Biology and Basic Medical Sciences, Soochow University, Suzhou, China
| | - Xinyu Tong
- School of Biology and Basic Medical Sciences, Soochow University, Suzhou, China
| | - Yaxin Zhang
- School of Biology and Basic Medical Sciences, Soochow University, Suzhou, China
| | - Jun Pan
- School of Biology and Basic Medical Sciences, Soochow University, Suzhou, China
| | - Yongjie Feng
- School of Biology and Basic Medical Sciences, Soochow University, Suzhou, China
- Agricultural Biotechnology Research Institute, Agricultural biotechnology and Ecological Research Institute, Soochow University, Suzhou, China
| | - Xiaolong Hu
- School of Biology and Basic Medical Sciences, Soochow University, Suzhou, China
- Agricultural Biotechnology Research Institute, Agricultural biotechnology and Ecological Research Institute, Soochow University, Suzhou, China
| | - Yujun Wang
- Guangxi Key Laboratory of Beibu Gulf Marine Biodiversity Conservation, Beibu Gulf University, Qinzhou, China
| | - Guangli Cao
- School of Biology and Basic Medical Sciences, Soochow University, Suzhou, China
- Agricultural Biotechnology Research Institute, Agricultural biotechnology and Ecological Research Institute, Soochow University, Suzhou, China
| | - Renyu Xue
- School of Biology and Basic Medical Sciences, Soochow University, Suzhou, China
- Agricultural Biotechnology Research Institute, Agricultural biotechnology and Ecological Research Institute, Soochow University, Suzhou, China
| | - Chengliang Gong
- School of Biology and Basic Medical Sciences, Soochow University, Suzhou, China
- Agricultural Biotechnology Research Institute, Agricultural biotechnology and Ecological Research Institute, Soochow University, Suzhou, China
| |
Collapse
|
3
|
Wang Q, Fang J, Pan Q, Wang Y, Xue T, Li L, Chen T. Efficient and Stable Delivery of Multiple Genes to Fish Cells by a Modified Recombinant Baculovirus System. Int J Mol Sci 2018; 19:ijms19123767. [PMID: 30486430 PMCID: PMC6321422 DOI: 10.3390/ijms19123767] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2018] [Revised: 11/23/2018] [Accepted: 11/23/2018] [Indexed: 12/26/2022] Open
Abstract
The recombinant baculovirus has been widely used as an efficient tool to mediate gene delivery into mammalian cells but has barely been used in fish cells. In the present study, we constructed a recombinant baculovirus containing the dual-promoter cytomegalovirus (CMV) and white spot syndrome virus (WSSV) immediate-early gene 1 (ie1) (WSSV ie1), followed by a puromycin⁻green fluorescent protein (Puro-GFP, pf) or puromycin⁻red fluorescent protein (Puro-RFP, pr) cassette, which simultaneously allowed for easy observation, rapid titer determination, drug selection, and exogenous gene expression. This recombinant baculovirus was successfully transduced into fish cells, including Mylopharyngodon piceus bladder (MPB), fin (MPF), and kidney (MPK); Oryzias latipes spermatogonia (SG3); and Danio rerio embryonic fibroblast (ZF4) cells. Stable transgenic cell lines were generated after drug selection, which was further verified by Western blot. A cell monoclonal formation assay proved the stable heredity of transgenic MPB cells. In addition, a recombinant baculovirus containing a pr cassette and four transcription factors for induced pluripotent stem cells (iPSC) was constructed and transduced into ZF4 cells, and these exogenous genes were simultaneously delivered and transcribed efficiently in drug-selected ZF4 cells, proving the practicability of this modified recombinant baculovirus system. We also proved that the WSSV ie1 promoter had robust activity in fish cells in vitro and in vivo. Taken together, this modified recombinant baculovirus can be a favorable transgenic tool to obtain transient or stable transgenic fish cells.
Collapse
Affiliation(s)
- Qian Wang
- Key Laboratory of Freshwater Animal Breeding, Ministry of Agriculture, College of Fisheries, Huazhong Agricultural University, Wuhan 430070, China.
| | - Jian Fang
- Key Laboratory of Freshwater Animal Breeding, Ministry of Agriculture, College of Fisheries, Huazhong Agricultural University, Wuhan 430070, China.
| | - Qihua Pan
- Key Laboratory of Freshwater Animal Breeding, Ministry of Agriculture, College of Fisheries, Huazhong Agricultural University, Wuhan 430070, China.
| | - Yizhou Wang
- Key Laboratory of Freshwater Animal Breeding, Ministry of Agriculture, College of Fisheries, Huazhong Agricultural University, Wuhan 430070, China.
| | - Ting Xue
- Key Laboratory of Freshwater Animal Breeding, Ministry of Agriculture, College of Fisheries, Huazhong Agricultural University, Wuhan 430070, China.
| | - Lingyu Li
- Key Laboratory of Freshwater Animal Breeding, Ministry of Agriculture, College of Fisheries, Huazhong Agricultural University, Wuhan 430070, China.
| | - Tiansheng Chen
- Key Laboratory of Freshwater Animal Breeding, Ministry of Agriculture, College of Fisheries, Huazhong Agricultural University, Wuhan 430070, China.
- Collaborative Innovation Center for Efficient and Health Production of Fisheries in Hunan Province, Changde 41500, China.
- Hubei Engineering Technology Research Center for Aquatic Animal Diseases Control and Prevention, Wuhan 430070, China.
| |
Collapse
|
4
|
El-Mogy MA, Abdalla MA, Misic V, Haj-Ahmad Y. Effect of adenovirus infection on transgene expression under the adenoviral MLP/TPL and the CMVie promoter/enhancer in CHO cells. J Genet Eng Biotechnol 2017; 15:211-217. [PMID: 30647657 PMCID: PMC6296590 DOI: 10.1016/j.jgeb.2017.04.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2015] [Revised: 02/27/2017] [Accepted: 04/05/2017] [Indexed: 11/19/2022]
Abstract
The adenovirus major late promoter (MLP) and its translational regulator - the tripartite leader (TPL) sequence - can actively drive efficient gene expression during adenoviral infection. However, both elements have not been widely tested in transgene expression outside of the adenovirus genome context. In this study, we tested whether the combination of MLP and TPL would enhance transgene expression beyond that of the most widely used promoter in transgene expression in mammalian cells, the cytomegalovirus immediate early (CMVie) promoter/enhancer. The activity of these two regulatory elements was compared in Chinese hamster ovary (CHO) cells. Although transient expression was significantly higher under the control of the CMVie promoter/enhance compared to the MLP/TPL, this difference was greater at the level of transcription (30 folds) than translation (11 folds). Even with adenovirus infection to provide additional elements (in trans), CMVie promoter/enhancer exhibited significantly higher activity relative to MLP/TPL. Interestingly, the CMVie promoter/enhancer was 1.9 folds more active in adenovirus-infected cells than in non-infected cells. Our study shows that the MLP-TPL drives lower transgene expression than the CMVie promoter/enhancer particularly at the transcription level. The data also highlight the utility of the TPL sequence at the translation level and/or possible overwhelming of the cellular translational machinery by the high transcription activity of the CMVie promoter/enhancer. In addition, here we present data that show stimulation of the CMVie promoter/enhancer by adenovirus infection, which may prove interesting in future work to test the combination of CMVie/TPL sequence, and additional adenovirus elements, for transgene expression.
Collapse
Key Words
- Adenovirus
- CHO, Chinese hamster ovary
- CMVie promoter/enhancer
- CMVie, cytomegalovirus immediate early
- GFP, green fluorescence proteins
- IRES, internal ribosome entry site
- MLP
- MLP, major late promoter
- MOI, multiplicity of infection
- PFU, plaque forming unit
- RFU, relative fluorescence units
- TPL
- TPL, tripartite leader
- Transgene expression
- qPCR, Quantitative PCR
Collapse
Affiliation(s)
- Mohamed A. El-Mogy
- Molecular Biology Department, National Research Centre, Dokki, Giza, Egypt
- Department of Biological Sciences, Brock University, St. Catharines, ON, Canada
- Corresponding author at: Molecular Biology Department, National Research Centre, Dokki, Giza, Egypt.
| | - Moemen A.K. Abdalla
- Department of Biochemistry, Faculty of Science, Alexandria University, Alexandria, Egypt
| | - Vanja Misic
- Department of Biological Sciences, Brock University, St. Catharines, ON, Canada
| | - Yousef Haj-Ahmad
- Department of Biological Sciences, Brock University, St. Catharines, ON, Canada
| |
Collapse
|
5
|
Zhang Z, Zhang J, Zhang J, Li Q, Miao P, Liu J, Li S, Huang J, Liao M, Fan H. Coimmunization with recombinant epitope-expressing baculovirus enhances protective effects of inactivated H5N1 vaccine against heterologous virus. Vet Microbiol 2017; 203:143-148. [DOI: 10.1016/j.vetmic.2017.03.004] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2016] [Revised: 03/01/2017] [Accepted: 03/02/2017] [Indexed: 12/13/2022]
|
6
|
Puthumana J, Philip R, Bright Singh IS. Transgene expression in Penaeus monodon cells: evaluation of recombinant baculoviral vectors with shrimp specific hybrid promoters. Cytotechnology 2016; 68:1147-59. [PMID: 25982944 PMCID: PMC4960163 DOI: 10.1007/s10616-015-9872-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2014] [Accepted: 03/27/2015] [Indexed: 12/30/2022] Open
Abstract
It has been realized that shrimp cell immortalization may not be accomplished without in vitro transformation by expressing immortalizing gene in cells. In this process, efficiency of transgene expression is confined to the ability of vectors to transmit gene of interests to the genome. Over the years, unavailability of such vectors has been hampering application of such a strategy in shrimp cells. We report the use of recombinant baculovirus mediated transduction using hybrid promoter system for transgene expression in lymphoid cells of Penaeus monodon. Two recombinant baculovirus vectors with shrimp viral promoters (WSSV-Ie1 and IHHNV-P2) were constructed (BacIe1-GFP and BacP2-GFP) and green fluorescent protein (GFP) used as the transgene. The GFP expression in cells under the control of hybrid promoters, PH-Ie1 or PH-P2, were analyzed and confirmed in shrimp cells. The results indicate that the recombinant baculovirus with shrimp specific viral promoters (hybrid) can be employed for delivery of foreign genes to shrimp cells for in vitro transformation.
Collapse
Affiliation(s)
- Jayesh Puthumana
- National Centre for Aquatic Animal Health, Cochin University of Science and Technology, Fine Arts Avenue, Kochi, 682016, Kerala, India
| | - Rosamma Philip
- Department of Marine Biology, Microbiology and Biochemistry, Cochin University of Science and Technology, Fine Arts Avenue, Kochi, 682016, Kerala, India
| | - I S Bright Singh
- National Centre for Aquatic Animal Health, Cochin University of Science and Technology, Fine Arts Avenue, Kochi, 682016, Kerala, India.
| |
Collapse
|
7
|
Baculovirus-mediated GCRV vp7 and vp6 genes expression in silkworm and grass carp. Mol Biol Rep 2016; 43:509-15. [PMID: 27085857 DOI: 10.1007/s11033-016-3984-0] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2015] [Accepted: 04/11/2016] [Indexed: 01/28/2023]
Abstract
Grass carp hemorrhagic disease is a common fish disease and often results in significant economic losses in grass carp aquaculture in China. This study was aimed to develop a novel oral vaccine against grass carp reovirus (GCRV). GCRV vp6 and vp7 genes with β-actin promoter of Megalobrama amblycephala and polyhedrin promoter (Ph10) of baculovirus, respectively, were cloned into plasmid pFast™-Dual to construct a vector pFast-PHVP7-AVP6, which was used to generate a recombinant baculovirus BacFish-vp6/vp7 via Bac-to-Bac system. The VP7 expression was analyzed from freeze-dried powder of the BacFish-vp6/vp7-infected silkworm pupae by western blotting, and VP6 expression was analyzed from orally vaccinated fish with the freeze-dried powder by RT-PCR. The VP6 expression was also analyzed from both CIK cells transduced with BacFish-vp6/vp7 and tissues of vaccinated fish by immunofluorescence analysis. Recombinant VP7 could be detected from the BacFish-vp6/vp7-infected silkworm pupae. Pathological changes were not observed in CIK cells transduced with BacFish-vp6/vp7, and VP6 expression was found in CIK cells. When the grass carps were orally administrated with the freeze-dried powder, vp6 gene transcription was found in blood of the vaccinated fishes and VP6 protein was observed in liver and kidney of the vaccinated fish by immunofluorescence analysis. These results indicated that vp7 gene was expressed in the BacFish-vp6/vp7-infected silkworm and vp6 gene was expressed in orally vaccinated fish with freeze-dried powder of the BacFish-vp6/vp7-infected silkworm pupae, suggesting the possibility to use the powder as an orally administrated vaccine.
Collapse
|
8
|
Makkonen KE, Airenne K, Ylä-Herttulala S. Baculovirus-mediated gene delivery and RNAi applications. Viruses 2015; 7:2099-125. [PMID: 25912715 PMCID: PMC4411692 DOI: 10.3390/v7042099] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2015] [Revised: 04/02/2015] [Accepted: 04/16/2015] [Indexed: 12/11/2022] Open
Abstract
Baculoviruses are widely encountered in nature and a great deal of data is available about their safety and biology. Recently, these versatile, insect-specific viruses have demonstrated their usefulness in various biotechnological applications including protein production and gene transfer. Multiple in vitro and in vivo studies exist and support their use as gene delivery vehicles in vertebrate cells. Recently, baculoviruses have also demonstrated high potential in RNAi applications in which several advantages of the virus make it a promising tool for RNA gene transfer with high safety and wide tropism.
Collapse
Affiliation(s)
- Kaisa-Emilia Makkonen
- Virtanen Institute, Department of Biotechnology and Molecular Medicine, University of Eastern Finland, Kuopio 70211 Finland.
| | - Kari Airenne
- Virtanen Institute, Department of Biotechnology and Molecular Medicine, University of Eastern Finland, Kuopio 70211 Finland.
| | - Seppo Ylä-Herttulala
- Virtanen Institute, Department of Biotechnology and Molecular Medicine, University of Eastern Finland, Kuopio 70211 Finland.
- Gene Therapy Unit, Kuopio University Hospital, Kuopio 70211, Finland.
- Science Service Center, Kuopio University Hospital, Kuopio 70211, Finland.
| |
Collapse
|
9
|
Zhang J, Chen XW, Tong TZ, Ye Y, Liao M, Fan HY. BacMam virus-based surface display of the infectious bronchitis virus (IBV) S1 glycoprotein confers strong protection against virulent IBV challenge in chickens. Vaccine 2013; 32:664-70. [PMID: 24342247 DOI: 10.1016/j.vaccine.2013.12.006] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2013] [Revised: 11/27/2013] [Accepted: 12/02/2013] [Indexed: 12/01/2022]
Abstract
Avian infectious bronchitis virus (IBV) is associated with production inefficiencies in domestic fowl, and causes massive economic losses to the poultry industry worldwide. Progress has been made in designing novel and efficient candidate vaccines to control IBV infection. BacMam virus, a modified baculovirus mediating transgene expression under the control of a mammalian promoter, has emerged as a versatile and safe vector during vaccine development. In previous work, we generated the BacMam virus Ac-CMV-S1, which expressed the S1 glycoprotein of IBV-M41. We showed that Ac-CMV-S1 induced excellent cellular immunity, but did not confer adequate protection in chickens compared with the conventional inactivated vaccine. In the current study, we generated an improved BacMam virus, BV-Dual-S1. This virus displayed the S1 glycoprotein on the baculovirus envelope, and was capable of expressing it in mammalian cells. BV-Dual-S1 elicited stronger humoral and cell-mediated immune responses, and showed greater capacity for induction of cytotoxic T lymphocyte responses, compared with Ac-CMV-S1 in specific pathogen-free chickens. A significant difference was not observed for protection rates between chickens immunized with BV-Dual-S1 (83%) or inactivated vaccine (89%) following challenge with virulent IBV-M41. Our findings show that the protective efficacy of BV-Dual-S1 could be significantly enhanced by baculovirus display technology. BacMam virus-based surface display strategies could serve as effective tools in designing vaccines against IB and other infectious diseases.
Collapse
Affiliation(s)
- Jie Zhang
- Key Laboratory of Animal Vaccine Development, Ministry of Agriculture, Guangzhou 510642, China; College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China
| | - Xiao-Wei Chen
- Key Laboratory of Animal Vaccine Development, Ministry of Agriculture, Guangzhou 510642, China; College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China
| | - Tie-Zhu Tong
- Huizhou Entry-Exit Inspection and Quarantine Bureau, Huizhou 516001, China
| | - Yu Ye
- Key Laboratory of Animal Vaccine Development, Ministry of Agriculture, Guangzhou 510642, China; College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China
| | - Ming Liao
- Key Laboratory of Animal Vaccine Development, Ministry of Agriculture, Guangzhou 510642, China; College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China.
| | - Hui-Ying Fan
- Key Laboratory of Animal Vaccine Development, Ministry of Agriculture, Guangzhou 510642, China; College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China.
| |
Collapse
|
10
|
Hu ZP, Yin J, Zhang YY, Jia SY, Chen ZJ, Zhong J. Characterization of the immune responses elicited by baculovirus-based vector vaccines against influenza virus hemagglutinin. Acta Pharmacol Sin 2012; 33:783-90. [PMID: 22562016 PMCID: PMC4010374 DOI: 10.1038/aps.2012.23] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
AIM To compare the specific immune responses elicited by different baculovirus vectors in immunized mice. METHODS We constructed and characterized two recombinant baculoviruses carrying the expression cassette for the H5N1 influenza virus hemagglutinin (HA) gene driven by either an insect cell promoter (vAc-HA) or a dual-promoter active both in insect and mammalian cells (vAc-HA-DUAL). Virus without the HA gene (vAc-EGFP) was used as a control. These viruses were used to immunize mice subcutaneously and intraperitoneally. The production of total and specific antibodies was determined by ELISA and competitive ELISA. Cytokine production by the spleen cells of immunized mice was studied using the ELISPOT assay. RESULTS Both the vAc-HA and vAc-HA-DUAL vectors expressed HA proteins in insect Sf9 cells, and HA antigen was present in progeny virions. The vAc-HA-DUAL vector also mediated HA expression in virus-transduced mammalian cell lines (BHK and A547). Both vAc-HA and vAc-HA-DUAL exhibited higher transduction efficiencies than vAc-EGFP in mammalian cells, as shown by the expression of the reporter gene egfp. Additionally, both vAc-HA and vAc-HA-DUAL induced high levels of HA-specific antibody production in immunized mice; vAc-HA-DUAL was more efficient in inducing IFN-γ and IL-2 upon stimulation with specific antigen, whereas vAc-HA was more efficient in inducing IL-4 and IL-6. CONCLUSION Baculovirus vectors elicited efficient, specific immune responses in immunized mice. The vector displaying the HA antigen on the virion surface (vAc-HA) elicited a Th2-biased immune response, whereas the vector displaying HA and mediating HA expression in the cell (vAc-HA-DUAL) elicited a Th1-biased immune response.
Collapse
|
11
|
Chen CY, Lin CY, Chen GY, Hu YC. Baculovirus as a gene delivery vector: recent understandings of molecular alterations in transduced cells and latest applications. Biotechnol Adv 2011; 29:618-31. [PMID: 21550393 PMCID: PMC7126054 DOI: 10.1016/j.biotechadv.2011.04.004] [Citation(s) in RCA: 107] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2011] [Revised: 04/15/2011] [Accepted: 04/15/2011] [Indexed: 12/13/2022]
Abstract
Baculovirus infects insects in nature and is non-pathogenic to humans, but can transduce a broad range of mammalian and avian cells. Thanks to the 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 gained explosive popularity as a gene delivery vector for a wide variety of applications. This article extensively reviews the recent understandings of the molecular mechanisms pertinent to baculovirus entry and cellular responses, and covers the latest advances in the vector improvements and applications, with special emphasis on antiviral therapy, cancer therapy, regenerative medicine and vaccine.
Collapse
Affiliation(s)
- Chi-Yuan Chen
- Department of Chemical Engineering, National Tsing Hua University, Hsinchu 30013, Taiwan
| | | | | | | |
Collapse
|
12
|
Efficient gene delivery into fish cells by an improved recombinant baculovirus. J Virol Methods 2011; 173:294-9. [PMID: 21354209 DOI: 10.1016/j.jviromet.2011.02.022] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2010] [Revised: 02/14/2011] [Accepted: 02/17/2011] [Indexed: 01/17/2023]
Abstract
Studies on transduction of mammalian cells have shown that baculovirus can be used as an effective vector for gene delivery. However, previous studies have found the gene delivery efficiency to be very low in differentiated fish cells. In this study, an improved recombinant baculovirus, containing cytomegalovirus immediate-early (CMV-IE) promoter and an enhanced green fluorescent protein (EGFP) gene as the reporter gene, was constructed. The transduction efficiency of recombinant baculovirus in several fish cell lines was measured by flow cytometry (FCM) and the persistence of EGFP was monitored by fluorescence microscopy. The results demonstrated that baculovirus can mediate the high efficiency of gene delivery into differentiated fish cells. Furthermore, it was found that growth medium is superior to PBS as the surrounding solution to enhance the transduction efficiency in some fish cells. In addition, transgene expression can persist for a lengthy period in fish cells, and attained highest level several days later after transduction. This study suggest that the improved recombinant baculovirus can be an excellent gene delivery vector for fish cells and also providing new insights into the transduction of vertebrate cells with baculovirus.
Collapse
|
13
|
Paul A, Jardin BA, Kulamarva A, Malhotra M, Elias CB, Prakash S. Recombinant baculovirus as a highly potent vector for gene therapy of human colorectal carcinoma: molecular cloning, expression, and in vitro characterization. Mol Biotechnol 2010; 45:129-39. [PMID: 20143184 DOI: 10.1007/s12033-010-9248-7] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Present therapeutic strategies for most cancers are restricted mainly to the primary tumors and are also not very effective in controlling metastatic states. Alternatively, gene therapy can be a potential option for treating such cancers. Currently mammalian viral-based cancer gene therapy is the most popular approach, but the efficacy has been shown to be quite low in clinical trials. In this study, for the first time, the insect cell-specific baculovirus Autographa californica multiple nucleopolyhedrovirus (AcMNPV) has been evaluated as a vector for gene delivery to colorectal cancer cells. Experiments involving factorial design were employed to study the individual and combined effects of different parameters such as multiplicity of infection (MOI), viral incubation time and epigenetic factors on transduction efficiency. The results demonstrate that baculovirus gene delivery system holds immense potential for development of a new generation of highly effective virotherapy for colorectal, as well as other major carcinomas (breast, pancreas, and brain), and offers significant benefits to traditional animal virus-based vectors with respect to safety concerns.
Collapse
Affiliation(s)
- Arghya Paul
- Biomedical Technology and Cell Therapy Research Laboratory, Department of Biomedical Engineering and Artificial Cells and Organs Research Centre, Faculty of Medicine, McGill University, 3775 University Street, Montreal, QC H3A 2B4, Canada
| | | | | | | | | | | |
Collapse
|
14
|
Reisinger H, Steinfellner W, Katinger H, Kunert R. Serum-free transfection of CHO cells with chemically defined transfection systems and investigation of their potential for transient and stable transfection. Cytotechnology 2009; 60:115-23. [PMID: 19760126 DOI: 10.1007/s10616-009-9224-x] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2008] [Accepted: 08/20/2009] [Indexed: 11/24/2022] Open
Abstract
The generation of transgenic cell lines is acquired by facilitating the uptake and integration of DNA. Unfortunately, most of the systems generating stable expression systems are cost and time-consuming and transient expression is optimized to generate milligram amounts of the recombinant protein. Therefore we improved and compared two transfection systems, one based on cationic liposomes consisting of DOTAP/DOPE and the second one on polyethylenimine (PEI). Both systems have been used as chemically defined transfection systems in combination with serum-free cultivated host cell line. At first we had determined the toxicity and ideal ratio of DNA to PEI followed by determination of the optimal transfection conditions in order to achieve maximum transfection efficiency. We then directly compared DOTAP/DOPE and PEI in transient transfection experiments using enhanced green fluorescence protein (EGFP) and a human monoclonal antibody, mAb 2F5, as a model protein. The results which were achieved in case of EGFP were more than 15% transfectants at a viability of 85%. Despite the fact that expression of the mAb was found negligible we used both techniques to generate stable mAb 2F5 expressing cell lines that underwent several cycles of screening and amplification with methotrexate, and resulted in cell lines with similar volumetric production titers. These experiments serve to demonstrate the potential of stable cell lines even in case where the transient systems did not show satisfying results.
Collapse
Affiliation(s)
- Hannes Reisinger
- Department of Biotechnology, University of Natural Resources and Applied Life Sciences Vienna, Muthgasse 18, Vienna, 1190, Austria,
| | | | | | | |
Collapse
|
15
|
Lemos MAN, Santos ASD, Astray RM, Pereira CA, Jorge SAC. Rabies virus glycoprotein expression in Drosophila S2 cells. I: design of expression/selection vectors, subpopulations selection and influence of sodium butyrate and culture medium on protein expression. J Biotechnol 2009; 143:103-10. [PMID: 19615415 DOI: 10.1016/j.jbiotec.2009.07.003] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2009] [Revised: 06/30/2009] [Accepted: 07/08/2009] [Indexed: 11/30/2022]
Abstract
The cDNA encoding the rabies virus glycoprotein (RVGP) gene was cloned in expression plasmids under the control of the inductive metallothionein promoter. They were designed in order to bear or not a secretion signal (i) and a cDNA coding for the selection hygromycin. These vectors were transfected into S2 cells, cell populations selected and subpopulations were then obtained by reselection with hygromycin. Cell cultures were examined for kinetics of cell growth, detection of RVGP mRNA and expression of RVGP. All cell populations were shown to express the RVGP mRNA upon induction. S2MtRVGPHy cell population, transfected with one vector that contains RGPV gene and selection gene, was shown to express higher amounts of RVGP as evaluated by flow cytometry ( approximately 52%) and ELISA (0.64 microg/10(7)cells at day 7). Subpopulation selection allowed a higher RVGP expression, specially for the S2MtRVGPHy(+) (5.5 microg/10(7)cells at day 7). NaBu treatment leading to lower cell growth and higher RVGP expression allowed an even higher RVGP synthesis by S2MtRVGPHy(+) (8.4 microg/10(7)cells at day 7). SF900II medium leading to a higher S2MtRVGPHy(+)cell growth allowed a higher final RVGP synthesis in this cell culture. RVGP synthesis may be optimized by the expression/selection vectors design, cell subpopulations selection, chromatin exposure and culture medium employed.
Collapse
|
16
|
Yu IL, Lin YC, Robinson JH, Lung O. Transduction of vertebrate cells with Spodoptera exigua multiple nucleopolyhedrovirus F protein-pseudotyped gp64-null Autographa californica multiple nucleopolyhedrovirus. J Gen Virol 2009; 90:2282-7. [PMID: 19474242 DOI: 10.1099/vir.0.012138-0] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Budded virions of the baculovirus Autographa californica multiple nucleopolyhedrovirus (AcMNPV) can enter a variety of non-host cells. The capacity of GP64, AcMNPV's endogenous envelope fusion protein, and SeF, the fusion protein from a gp64(-) baculovirus, to mediate baculovirus entry into vertebrate cells was examined by comparing the transduction efficiencies of engineered AcMNPV variants with either of the two envelope proteins into 17 vertebrate cell lines. At an m.o.i. of 500, GP64-expressing viruses transduced all cell lines with varying efficiencies. Transduction efficiencies of SeF-pseudotyped gp64-null AcMNPV into all cell lines were lower than those of GP64-expressing viruses, and were undetectable in seven cell lines. At an m.o.i. of 50, transduction of all mammalian cell lines transducible by the SeF-pseudotyped gp64-null AcMNPV at an m.o.i. of 500 was no longer detectable. An amplifiable SeF-pseudotyped gp64-null AcMNPV vector with greatly reduced tropism for vertebrate cells may have applications in engineering AcMNPV for targeted transduction.
Collapse
Affiliation(s)
- Ian-Ling Yu
- Department of Biological Sciences, University of Lethbridge, Lethbridge, AB T1K 3M4, Canada
| | | | | | | |
Collapse
|
17
|
Antigen delivery systems for veterinary vaccine development. Viral-vector based delivery systems. Vaccine 2009; 26:6508-28. [PMID: 18838097 PMCID: PMC7131726 DOI: 10.1016/j.vaccine.2008.09.044] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2008] [Revised: 08/21/2008] [Accepted: 09/16/2008] [Indexed: 11/30/2022]
Abstract
The recent advances in molecular genetics, pathogenesis and immunology have provided an optimal framework for developing novel approaches in the rational design of vaccines effective against viral epizootic diseases. This paper reviews most of the viral-vector based antigen delivery systems (ADSs) recently developed for vaccine testing in veterinary species, including attenuated virus and DNA and RNA viral vectors. Besides their usefulness in vaccinology, these ADSs constitute invaluable tools to researchers for understanding the nature of protective responses in different species, opening the possibility of modulating or potentiating relevant immune mechanisms involved in protection.
Collapse
|
18
|
He F, Ho Y, Yu L, Kwang J. WSSV ie1 promoter is more efficient than CMV promoter to express H5 hemagglutinin from influenza virus in baculovirus as a chicken vaccine. BMC Microbiol 2008; 8:238. [PMID: 19116038 PMCID: PMC2631607 DOI: 10.1186/1471-2180-8-238] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2007] [Accepted: 12/31/2008] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND The worldwide outbreak of influenza A (H5N1) viruses among poultry species and humans highlighted the need to develop efficacious and safe vaccines based on efficient and scaleable production. RESULTS White spot syndrome virus (WSSV) immediate-early promoter one (ie1) was shown to be a stronger promoter for gene expression in insect cells compared with Cytomegalovirus immediate-early (CMV) promoter in luciferase assays. In an attempt to improve expression efficiency, a recombinant baculovirus was constructed expressing hemagglutinin (HA) of H5N1 influenza virus under the control of WSSV ie1 promoter. HA expression in SF9 cells increased significantly with baculovirus under WSSV ie1 promoter, compared with CMV promoter based on HA contents and hemagglutination activity. Further, immunization with baculovirus under WSSV ie1 promoter in chickens elicited higher level anti-HA antibodies compared to CMV promoter, as indicated in hemagglutination inhibition, virus neutralization and enzyme-linked immunosorbent assays. By immunohistochemistry, strong HA antigen expression was observed in different chicken organs with vaccination of WSSV ie1 promoter controlled baculovirus, confirming higher efficiency in HA expression by WSSV ie1 promoter. CONCLUSION The production of H5 HA by baculovirus was enhanced with WSSV ie1 promoter, especially compared with CMV promoter. This contributed to effective elicitation of HA-specific antibody in vaccinated chickens. This study provides an alternative choice for baculovirus based vaccine production.
Collapse
Affiliation(s)
- Fang He
- Animal Health Biotechnology, Temasek Life Sciences Laboratory, National University of Singapore, Singapore.
| | | | | | | |
Collapse
|
19
|
Yang DG, Chung YC, Lai YK, Lai CW, Liu HJ, Hu YC. Avian influenza virus hemagglutinin display on baculovirus envelope: cytoplasmic domain affects virus properties and vaccine potential. Mol Ther 2007; 15:989-96. [PMID: 17375072 DOI: 10.1038/mt.sj.6300131] [Citation(s) in RCA: 81] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Hemagglutinin (HA) is the major immunogen on the envelope of avian influenza virus (AIV). Therefore we constructed two recombinant baculoviruses: Bac-HA, expressing histidine-tagged HA with the cytoplasmic domain (CTD) derived from HA, and Bac-HA64, expressing histidine-tagged HA with the CTD derived from baculovirus envelope protein gp64. After infection, HA with either CTD was expressed and anchored on the plasma membrane of Sf-9 cells, as revealed by confocal microscopy. Immunogold electron microscopy demonstrated that both Bac-HA and Bac-HA64 displayed HA on the viral surface. However, analyses of purified viruses revealed that significantly more HA was incorporated into Bac-HA64 than into Bac-HA. In comparison with Bac-HA, Bac-HA64 significantly improved the gene delivery and transgene expression in mammalian cells, as determined by quantitative real-time polymerase chain reaction and flow cytometry. Bac-HA64 elicited significantly higher hemagglutination inhibition titers in mouse models than Bac-HA and the negative controls. These data collectively confirmed that the gp64 CTD, in comparison with HA CTD, resulted in more efficient HA incorporation into baculovirus, more efficient transgene delivery and expression, and elevated immunogenicity. This is the first report demonstrating the potential of HA-pseudotyped baculovirus as an avian influenza vaccine and that the choice of CTD tremendously affects baculovirus properties and vaccine efficacy.
Collapse
Affiliation(s)
- Ding-Gang Yang
- Department of Chemical Engineering, National Tsing Hua, University, Hsinchu, Taiwan
| | | | | | | | | | | |
Collapse
|
20
|
Santos MG, Jorge SAC, Brillet K, Pereira CA. Improving heterologous protein expression in transfected Drosophila S2 cells as assessed by EGFP expression. Cytotechnology 2007; 54:15-24. [PMID: 19003014 DOI: 10.1007/s10616-007-9060-9] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2006] [Accepted: 02/13/2007] [Indexed: 10/23/2022] Open
Abstract
Drosophila melanogaster S2 cells were co-transfected with plasmid vectors containing the enhanced green fluorescent protein gene (EGFP), under the control of metallothionein promoter (pMt), and the hygromycin selection gene, in view of establishing parameters for optimized gene expression. A protocol of transfection was worked out, leading after hygromycin selection, to approximately 90% of S2MtEGFP fluorescent cells at day 5 after copper sulfate (CuSO(4)) induction. As analyzed by confocal microscopy, S2MtEGFP cell cultures were shown to be quite heterogeneous regarding the intensity and cell localization of fluorescence among the EGFP expressing cells. Spectrofluorimetry kinetic studies of CuSO(4) induced S2MtEGFP cells showed the EGFP expression at 510 nm as soon as 5 h after induction, the fluorescence increasing progressively from this time to attain values of 4.6 x 10(5) counts/s after 72 h of induction. Induction with 700 muM of CuSO(4) performed at the exponential phase of the S2MtEGFP culture (10(6) cells/mL) led to a better performance in terms of cell growth, percent of fluorescent cells and culture intensity of fluorescence. Sodium butyrate (NaBu) treatment of CuSO(4) induced S2MtEGFP cell cultures, although leading to a loss of cell culture viability, increased the percent of EGFP expressing cells and sharply enhanced the cell culture fluorescence intensity. The present study established parameters for improving heterologous protein expression in stably transfected Drosophila S2 cells, as assessed by the EGFP expression.
Collapse
Affiliation(s)
- Mariza G Santos
- Laboratório de Imunologia Viral, Instituto Butantan, Sao Paulo, Brazil
| | | | | | | |
Collapse
|