1
|
van Oosten L, Altenburg JJ, Nowee G, Kenbeek D, Neef T, Rouw T, Tegelbeckers VIP, van der Heijden J, Mentink S, Willemsen W, Hausjell CS, Kuijpers L, van der Pol L, Roldão A, Correia R, van den Born E, Wijffels RH, Martens DE, van Oers MM, Pijlman GP. Engineered thermoswitch in the baculovirus expression vector system for production of virus-like particle vaccines with minimized baculovirus contaminants. Trends Biotechnol 2025:S0167-7799(25)00131-3. [PMID: 40348731 DOI: 10.1016/j.tibtech.2025.04.004] [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: 10/16/2024] [Revised: 04/03/2025] [Accepted: 04/04/2025] [Indexed: 05/14/2025]
Abstract
The baculovirus expression vector system (BEVS) is commercially used for producing recombinant (glyco)proteins, gene therapy vectors, and virus-like particles (VLPs) in insect cells. A major challenge in VLP downstream processing (DSP) is the removal of similarly sized baculovirus particle contaminants. Herein we engineered a genetic ON/OFF thermoswitch in the baculovirus vector by mutating the essential gp41 gene. This temperature-sensitive (ts) baculovirus expression system (BacFreets) produces high baculovirus titers at 27°C, while contaminating baculovirus production switches off at elevated temperatures. We demonstrate scalable VLP production for chikungunya virus (CHIKV), West Nile virus (WNV), coxsackievirus A6, and foot-and-mouth disease virus (FMDV) at 33°C, concomitant with baculovirus particle reduction up to 99.97%. However, elevated temperature moderately affected the yield of two out of four VLP designs. BacFreets is compatible with commonly used insect cells and scalable infection strategies. BacFreets effectively reduces baculovirus contaminants in the upstream VLP production phase, which will facilitate DSP of human and veterinary VLP-based vaccines.
Collapse
Affiliation(s)
- Linda van Oosten
- Laboratory of Virology, Wageningen University and Research, 6708PB Wageningen, The Netherlands
| | - Jort J Altenburg
- Bioprocess Engineering, Wageningen University and Research, 6708PB Wageningen, The Netherlands
| | - Gwen Nowee
- Laboratory of Virology, Wageningen University and Research, 6708PB Wageningen, The Netherlands
| | - Dennis Kenbeek
- Laboratory of Virology, Wageningen University and Research, 6708PB Wageningen, The Netherlands
| | - Tessa Neef
- Bioprocess Engineering, Wageningen University and Research, 6708PB Wageningen, The Netherlands
| | - Thomas Rouw
- Bioprocess Engineering, Wageningen University and Research, 6708PB Wageningen, The Netherlands
| | - Vivian I P Tegelbeckers
- Bioprocess Engineering, Wageningen University and Research, 6708PB Wageningen, The Netherlands
| | - Jans van der Heijden
- Laboratory of Virology, Wageningen University and Research, 6708PB Wageningen, The Netherlands
| | - Saskia Mentink
- Laboratory of Virology, Wageningen University and Research, 6708PB Wageningen, The Netherlands
| | - Wessel Willemsen
- Laboratory of Virology, Wageningen University and Research, 6708PB Wageningen, The Netherlands
| | - Christina S Hausjell
- Department of Biotechnology, University of Natural Resources and Life Sciences (BOKU) Vienna, 1190 Vienna, Austria
| | | | | | - António Roldão
- IBET, Instituto de Biologia Experimental e Tecnológica, 2780-901 Oeiras, Portugal; ITQB-NOVA, Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, 2780-157 Oeiras, Portugal
| | - Ricardo Correia
- IBET, Instituto de Biologia Experimental e Tecnológica, 2780-901 Oeiras, Portugal; ITQB-NOVA, Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, 2780-157 Oeiras, Portugal
| | | | - René H Wijffels
- Bioprocess Engineering, Wageningen University and Research, 6708PB Wageningen, The Netherlands
| | - Dirk E Martens
- Bioprocess Engineering, Wageningen University and Research, 6708PB Wageningen, The Netherlands
| | - Monique M van Oers
- Laboratory of Virology, Wageningen University and Research, 6708PB Wageningen, The Netherlands
| | - Gorben P Pijlman
- Laboratory of Virology, Wageningen University and Research, 6708PB Wageningen, The Netherlands.
| |
Collapse
|
2
|
Abbo SR, Yan K, Geertsema C, Hick TAH, Altenburg JJ, Nowee G, van Toor C, van Lent JW, Nakayama E, Tang B, Metz SW, Bhowmik R, de Silva AM, Prow NA, Correia R, Alves PM, Roldão A, Martens DE, van Oers MM, Suhrbier A, Pijlman GP. Virus-like particle vaccine with authentic quaternary epitopes protects against Zika virus-induced viremia and testicular damage. J Virol 2025; 99:e0232224. [PMID: 40013767 PMCID: PMC11998496 DOI: 10.1128/jvi.02322-24] [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: 01/07/2025] [Accepted: 01/20/2025] [Indexed: 02/28/2025] Open
Abstract
Zika virus (ZIKV) caused unprecedented outbreaks in South America and the Caribbean in 2015-2016, leading primarily to a series of abnormalities in neonates termed congenital Zika syndrome. The threat of ZIKV reemergence has seen the development of multiple ZIKV vaccines that are at the preclinical stage or in early-stage clinical trials. Herein, we describe a pathway to the development of ZIKV vaccines generated using a baculovirus-insect cell expression system, which is widely applied for the manufacture of biologics for human use. Virus-like particle (VLP) vaccines comprising CprME and subviral particle (SVP) vaccines comprising prME were evaluated for their ability to mediate protection against ZIKV challenge in Ifnar1-/- mice. Initial attempts resulted in VLP and SVP vaccines that failed to present quaternary epitopes and did not provide effective protection. To improve the SVP vaccine, two modifications were introduced: firstly, an alanine to cysteine substitution (A264C) in the E domain II region to promote the formation of stabilized E homodimers and, secondly, the use of Spodoptera frugiperda Sf9 insect cells that had been adapted to grow and produce vaccine at a neutral pH of 7. E homodimers largely retain their pre-fusion conformation at pH 7, which is a requirement for the induction of effective neutralizing antibody responses. The stabilized SVP-A26C vaccine induced high levels of neutralizing antibodies and protected male Ifnar1-/- mice against viremia and testicular damage. Our study reiterates the need to present the immune system with E dimers arranged in authentic quaternary conformations and provides a scalable production method for this novel ZIKV vaccine.IMPORTANCEWe describe the generation of a subviral particle (SVP) vaccine comprising prME proteins of ZIKV, with an envelope protein substitution, A264C, that stabilizes E dimer formation. The SVP vaccine was produced in a novel Sf9 insect cell line adapted to grow in suspension at pH 7. The study highlights the importance of challenge experiments to ascertain whether the responses induced by an experimental vaccine actually mediate protection against virus infection and disease. The study also reiterates the contention that effective flavivirus vaccines need to present the immunogen in an authentic tertiary and quaternary structure with a pre-fusion conformation.
Collapse
Affiliation(s)
- Sandra R. Abbo
- Laboratory of Virology, Wageningen University & Research, Wageningen, the Netherlands
| | - Kexin Yan
- Inflammation Biology Group, QIMR Berghofer Medical Research Institute, Brisbane, Australia
| | - Corinne Geertsema
- Laboratory of Virology, Wageningen University & Research, Wageningen, the Netherlands
| | - Tessy A. H. Hick
- Laboratory of Virology, Wageningen University & Research, Wageningen, the Netherlands
- Bioprocess Engineering, Wageningen University & Research, Wageningen, the Netherlands
| | - Jort J. Altenburg
- Bioprocess Engineering, Wageningen University & Research, Wageningen, the Netherlands
| | - Gwen Nowee
- Laboratory of Virology, Wageningen University & Research, Wageningen, the Netherlands
| | - Chris van Toor
- Laboratory of Virology, Wageningen University & Research, Wageningen, the Netherlands
| | - Jan W. van Lent
- Laboratory of Virology, Wageningen University & Research, Wageningen, the Netherlands
| | - Eri Nakayama
- Inflammation Biology Group, QIMR Berghofer Medical Research Institute, Brisbane, Australia
- Department of Virology I, National Institute of Infectious Diseases, Tokyo, Japan
| | - Bing Tang
- Inflammation Biology Group, QIMR Berghofer Medical Research Institute, Brisbane, Australia
| | - Stefan W. Metz
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Ryan Bhowmik
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Aravinda M. de Silva
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Natalie A. Prow
- Inflammation Biology Group, QIMR Berghofer Medical Research Institute, Brisbane, Australia
| | - Ricardo Correia
- IBET, Instituto de Biologia Experimental e Tecnológica, Oeiras, Portugal
- ITQB NOVA, Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Oeiras, Portugal
| | - Paula M. Alves
- IBET, Instituto de Biologia Experimental e Tecnológica, Oeiras, Portugal
- ITQB NOVA, Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Oeiras, Portugal
| | - António Roldão
- IBET, Instituto de Biologia Experimental e Tecnológica, Oeiras, Portugal
- ITQB NOVA, Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Oeiras, Portugal
| | - Dirk E. Martens
- Bioprocess Engineering, Wageningen University & Research, Wageningen, the Netherlands
| | - Monique M. van Oers
- Laboratory of Virology, Wageningen University & Research, Wageningen, the Netherlands
| | - Andreas Suhrbier
- Inflammation Biology Group, QIMR Berghofer Medical Research Institute, Brisbane, Australia
- GVN Centre of Excellence, Australian Infectious Disease Research Centre, Brisbane, Queensland, Australia
| | - Gorben P. Pijlman
- Laboratory of Virology, Wageningen University & Research, Wageningen, the Netherlands
| |
Collapse
|
3
|
Correia R, Zotler T, Ferraz F, Fernandes B, Graça M, Pijlman GP, Alves PM, Roldão A. Continuous Production of Influenza VLPs Using IC-BEVS and Multi-Stage Bioreactors. Biotechnol Bioeng 2025; 122:846-857. [PMID: 39825519 PMCID: PMC11895415 DOI: 10.1002/bit.28925] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Revised: 12/07/2024] [Accepted: 01/05/2025] [Indexed: 01/20/2025]
Abstract
The insect cell-baculovirus expression vector system (IC-BEVS) has been an asset to produce biologics for over 30 years. With the current trend in biotechnology shifting toward process intensification and integration, developing intensified processes such as continuous production is crucial to hold this platform as a suitable alternative to others. However, the implementation of continuous production has been hindered by the lytic nature of this expression system and the process-detrimental virus passage effect. In this study, we implemented a multi-stage bioreactor setup for continuous production of influenza hemagglutinin-displaying virus-like particles (HA-VLPs) using IC-BEVS. A setup consisting of one Cell Growth Bioreactor simultaneously feeding non-infected insect cells to three parallel Production Bioreactors operated at different residence times (RT) (18, 36, and 54 h) was implemented; Production Bioreactors were continuously harvested. Two insect cell lines (neutral pH-adapted High Five and Sf9) and two recombinant baculovirus (rBAC) constructs (one that originates from a bacmid, rBACbacmid, and another of non-bacteria origin, rBACflashbac) were tested. Combining rBACflashbac with Sf9 cells was the most efficient approach, allowing consistent HA-VLPs titers (34 ± 14 HA titer/mL) and rBAC titers (108-109 pfu/mL) throughout the period of continuous operation (20 days). Cell growth kinetics and viability varied across RT, and higher RT was associated with increased expression of HA-VLPs, independent of the cell line and rBAC used; RT of 54 h allowed to maximize titers. The presence of particles resembling HA-VLPs was confirmed by transmission electron microscopy throughout the continuous operation. This work showcases the implementation of a process for continuous production of a promising class of biotherapeutics (i.e., VLPs), and paves the way for establishing continuous, integrated setups using the IC-BEVS expression system.
Collapse
Affiliation(s)
- Ricardo Correia
- Instituto de Biologia Experimental e Tecnológica (iBET)OeirasPortugal
- Instituto de Tecnologia Química e Biológica António Xavier (ITQB NOVA)Universidade Nova de LisboaOeirasPortugal
| | - Taja Zotler
- Laboratory of VirologyWageningen University & ResearchWageningenthe Netherlands
| | - Francisco Ferraz
- Instituto de Biologia Experimental e Tecnológica (iBET)OeirasPortugal
- Instituto de Tecnologia Química e Biológica António Xavier (ITQB NOVA)Universidade Nova de LisboaOeirasPortugal
| | - Bárbara Fernandes
- Instituto de Biologia Experimental e Tecnológica (iBET)OeirasPortugal
- Instituto de Tecnologia Química e Biológica António Xavier (ITQB NOVA)Universidade Nova de LisboaOeirasPortugal
| | - Miguel Graça
- Instituto de Biologia Experimental e Tecnológica (iBET)OeirasPortugal
- Instituto de Tecnologia Química e Biológica António Xavier (ITQB NOVA)Universidade Nova de LisboaOeirasPortugal
| | - Gorben P. Pijlman
- Laboratory of VirologyWageningen University & ResearchWageningenthe Netherlands
| | - Paula M. Alves
- Instituto de Biologia Experimental e Tecnológica (iBET)OeirasPortugal
- Instituto de Tecnologia Química e Biológica António Xavier (ITQB NOVA)Universidade Nova de LisboaOeirasPortugal
| | - António Roldão
- Instituto de Biologia Experimental e Tecnológica (iBET)OeirasPortugal
- Instituto de Tecnologia Química e Biológica António Xavier (ITQB NOVA)Universidade Nova de LisboaOeirasPortugal
| |
Collapse
|
4
|
Letcher SM, Calkins OP, Clausi HJ, McCreary A, Trimmer BA, Kaplan DL. Establishment & characterization of a non-adherent insect cell line for cultivated meat. Sci Rep 2025; 15:7850. [PMID: 40050299 PMCID: PMC11885424 DOI: 10.1038/s41598-025-86921-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2024] [Accepted: 01/15/2025] [Indexed: 03/09/2025] Open
Abstract
This study presents a blueprint for developing, scaling, and analyzing novel insect cell lines for food. The large-scale production of cultivated meat requires the development and analysis of cell lines that are simple to grow and easy to scale. Insect cells may be a favorable cell source due to their robust growth properties, adaptability to different culture conditions, and resiliency in culture. Cells were isolated from Tobacco hornworm (Manduca sexta) embryos and subsequently adapted to single-cell suspension culture in animal-free growth media. Cells were able to reach relatively high cell densities of over 20 million cells per mL in shake flasks. Cell growth data is presented in various culture vessels and spent media analysis was performed to better understand cell metabolic processes. Finally, a preliminary nutritional profile consisting of proximate, amino acid, mineral, and fatty acid analysis is reported.
Collapse
Affiliation(s)
- Sophia M Letcher
- Department of Biomedical Engineering, Tufts University, Medford, MA, 02155, USA
| | - Olivia P Calkins
- Department of Chemical Engineering, Tufts University, Medford, MA, 02155, USA
| | - Halla J Clausi
- Department of Biomedical Engineering, Tufts University, Medford, MA, 02155, USA
| | - Aidan McCreary
- Department of Chemistry, Tufts University, Medford, MA, 02155, USA
| | - Barry A Trimmer
- Department of Biomedical Engineering, Tufts University, Medford, MA, 02155, USA
- Department of Biology, Tufts University, Medford, MA, 02155, USA
| | - David L Kaplan
- Department of Biomedical Engineering, Tufts University, Medford, MA, 02155, USA.
| |
Collapse
|
5
|
Wang S, Xiao L. Progress in AAV-Mediated In Vivo Gene Therapy and Its Applications in Central Nervous System Diseases. Int J Mol Sci 2025; 26:2213. [PMID: 40076831 PMCID: PMC11899905 DOI: 10.3390/ijms26052213] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2025] [Revised: 02/25/2025] [Accepted: 02/27/2025] [Indexed: 03/14/2025] Open
Abstract
As the blood-brain barrier (BBB) prevents molecules from accessing the central nervous system (CNS), the traditional systemic delivery of chemical drugs limits the development of neurological drugs. However, in recent years, innovative therapeutic strategies have tried to bypass the restriction of traditional drug delivery methods. In vivo gene therapy refers to emerging biopharma vectors that carry the specific genes and target and infect specific tissues; these infected cells and tissues then undergo fundamental changes at the genetic level and produce therapeutic proteins or substances, thus providing therapeutic benefits. Clinical and preclinical trials mainly utilize adeno-associated viruses (AAVs), lentiviruses (LVs), and other viruses as gene vectors for disease investigation. Although LVs have a higher gene-carrying capacity, the vector of choice for many neurological diseases is the AAV vector due to its safety and long-term transgene expression in neurons. Here, we review the basic biology of AAVs and summarize some key issues in recombinant AAV (rAAV) engineering in gene therapy research; then, we summarize recent clinical trials using rAAV treatment for neurological diseases and provide translational perspectives and future challenges on target selection.
Collapse
Affiliation(s)
- Shuming Wang
- Institute for Brain Research and Rehabilitation, Guangdong Key Laboratory of Mental Health and Cognitive Science, Center for Studies of Psychological Application, South China Normal University, Guangzhou 510631, China;
- Key Laboratory of Brain, Cognition and Education Sciences of Ministry of Education, South China Normal University, Guangzhou 510631, China
| | - Lin Xiao
- Institute for Brain Research and Rehabilitation, Guangdong Key Laboratory of Mental Health and Cognitive Science, Center for Studies of Psychological Application, South China Normal University, Guangzhou 510631, China;
- Key Laboratory of Brain, Cognition and Education Sciences of Ministry of Education, South China Normal University, Guangzhou 510631, China
| |
Collapse
|
6
|
Kar S, Mehrotra S, Prajapati VK. From infection to remedy: Harnessing oncolytic viruses in cancer treatment. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2025; 144:213-257. [PMID: 39978967 DOI: 10.1016/bs.apcsb.2024.10.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/22/2025]
Abstract
Oncolytic virus (OV) mediated immunotherapy is one of the recent techniques used to treat higher grade cancers where conventional therapies like chemotherapy, radiation fail. OVs as a therapeutic tool show high efficacy and fewer side effects than conventional methods as supported by multiple preclinical and clinical studies since they are engineered to target tumours. In this chapter, we discuss the modifications in viruses to make them oncolytic, types of strains commonly administered, mechanisms employed by viruses to specifically target and eradicate malignancy and progress achieved as reported in case studies (preclinical and clinical trials). OVs also face some unique challenges with respect to the malignancy being treated and the varied pathogen exposure of the patients, which is also highlighted here. Since pathogen exposure varies according to population dynamics worldwide, chances of generating a non-specific recall response to an OV cannot be negated. Lastly, the future perspectives and ongoing practises of combination therapies are discussed as they provide a leading edge over monotherapies in terms of tumour clearance, blocking metastasis and enhancing patient survival. Efforts undertaken to overcome current challenges are also highlighted.
Collapse
Affiliation(s)
- Sramona Kar
- Department of Biochemistry, University of Delhi South Campus, Benito Juarez Road, Dhaula Kuan, New Delhi, India
| | - Sanjana Mehrotra
- Department of Human Genetics, Guru Nanak Dev University, Amritsar, Punjab, India
| | - Vijay Kumar Prajapati
- Department of Biochemistry, University of Delhi South Campus, Benito Juarez Road, Dhaula Kuan, New Delhi, India.
| |
Collapse
|
7
|
Altenburg JJ, Juarez-Garza BE, van Keimpema J, van Oosten L, Pijlman GP, van Oers MM, Wijffels RH, Martens DE. Process intensification of the baculovirus expression vector system using a perfusion process with a low multiplicity of infection at high cell concentrations. Biotechnol Prog 2025:e3527. [PMID: 39846509 DOI: 10.1002/btpr.3527] [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: 08/23/2024] [Revised: 12/06/2024] [Accepted: 12/10/2024] [Indexed: 01/24/2025]
Abstract
The emergence of new viruses and the spread of existing pathogens necessitate efficient vaccine production methods. The baculovirus expression vector system (BEVS) is an efficient and scalable system for subunit and virus-like particle vaccine production and gene therapy vectors. However, current production processes are often limited to low cell concentrations (1-4 × 106 cells/mL) in fed-batch mode. To improve the volumetric productivity of the BEVS, a medium exchange strategy was investigated. Screening experiments were performed to test baculovirus (expressing green fluorescent protein; GFP) infection and productivity of insect cell cultures infected at high cell concentration (1-2 × 107 cells/mL), showing that infection at high cell concentrations was possible with medium exchange. Next, duplicate perfusion runs with baculovirus infection were performed using a cell concentration upon infection (CCI) of 1.2 × 107 cells/mL and a multiplicity of infection (MOI) of 0.01, reaching a maximum viable cell concentration of 2.8 × 107 cells/mL and a maximum GFP production of 263 mg/L. The volumetric productivity of these perfusion runs was 4.8 times higher than for reference batch processes with a CCI of 3 × 106 cells/mL and an MOI of 1. These results demonstrate that process intensification can be achieved for the BEVS by implementing perfusion, resulting in a higher volumetric productivity.
Collapse
Affiliation(s)
- Jort J Altenburg
- Bioprocess Engineering, Wageningen University & Research, Wageningen, The Netherlands
| | - Brenda E Juarez-Garza
- Bioprocess Engineering, Wageningen University & Research, Wageningen, The Netherlands
| | - Jelle van Keimpema
- Bioprocess Engineering, Wageningen University & Research, Wageningen, The Netherlands
| | - Linda van Oosten
- Laboratory of Virology, Wageningen University & Research, Wageningen, The Netherlands
| | - Gorben P Pijlman
- Laboratory of Virology, Wageningen University & Research, Wageningen, The Netherlands
| | - Monique M van Oers
- Laboratory of Virology, Wageningen University & Research, Wageningen, The Netherlands
| | - René H Wijffels
- Bioprocess Engineering, Wageningen University & Research, Wageningen, The Netherlands
| | - Dirk E Martens
- Bioprocess Engineering, Wageningen University & Research, Wageningen, The Netherlands
| |
Collapse
|
8
|
Ebihara T, Shibuya M, Yamaguchi A, Hino M, Lee JM, Kusakabe T, Mon H. Efficient and accurate BmNPV bacmid editing system by two-step golden gate assembly. J Virol Methods 2024; 330:115029. [PMID: 39243818 DOI: 10.1016/j.jviromet.2024.115029] [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: 06/28/2024] [Revised: 08/31/2024] [Accepted: 09/01/2024] [Indexed: 09/09/2024]
Abstract
The silkworm-baculovirus expression vector system (silkworm-BEVS), using Bombyx mori nucleopolyhedrovirus (BmNPV) and silkworm larvae or pupae, has been used as a cost-effective expression system for the production of various recombinant proteins. Recently, several gene knockouts in baculoviruses have been shown to improve the productivity of recombinant proteins. However, the gene editing of the baculovirus genome (approximately 130 kb) remains challenging and time-consuming. In this study, we sought to further enhance the productivity of the silkworm-BEVS by synthesizing and gene editing the BmNPV bacmid from plasmids containing fragments of BmNPV genomic DNA using a two-step Golden Gate Assembly (GGA). The BmNPV genome, divided into 19 fragments, was amplified by PCR and cloned into the plasmids. From these initial plasmids, four intermediate plasmids containing the BmNPV genomic DNA were constructed by GGA with the type IIS restriction enzyme BsaI. Subsequently, the full-length bacmid was successfully synthesized from the four intermediate plasmids by GGA with another type IIS restriction enzyme PaqCI with a high efficiency of 97.2 %. Furthermore, this methodology enabled the rapid and straightforward generation of the BmNPV bacmid lacking six genes, resulting in the suppression of degradation of recombinant proteins expressed in silkworm pupae. These results indicate that the BmNPV bacmid can be quickly and efficiently edited using only simple cloning techniques and enzymatic reactions, marking a significant advancement in the improvement of the silkworm-BEVS.
Collapse
Affiliation(s)
- Takeru Ebihara
- Laboratory of Insect Genome Science, Kyushu University Graduate School of Bioresource and Bioenvironmental Sciences, Motooka 744, Nishi-ku, Fukuoka 819-0395, Japan
| | - Misaki Shibuya
- Laboratory of Insect Genome Science, Kyushu University Graduate School of Bioresource and Bioenvironmental Sciences, Motooka 744, Nishi-ku, Fukuoka 819-0395, Japan
| | - Ayaka Yamaguchi
- Laboratory of Insect Genome Science, Kyushu University Graduate School of Bioresource and Bioenvironmental Sciences, Motooka 744, Nishi-ku, Fukuoka 819-0395, Japan
| | - Masato Hino
- Laboratory of Silkworm Genetic Resources, Institute of Genetic Resources, Kyushu University Graduate School of BioResources and Bioenvironmental Science, Motooka 744, Nishi-ku, Fukuoka 819-0395, Japan
| | - Jae Man Lee
- Laboratory of Creative Science for Insect Industries, Kyushu University Graduate School of Bioresource and Bioenvironmental Sciences, Motooka 744, Nishi-ku, Fukuoka 819-0395, Japan
| | - Takahiro Kusakabe
- Laboratory of Insect Genome Science, Kyushu University Graduate School of Bioresource and Bioenvironmental Sciences, Motooka 744, Nishi-ku, Fukuoka 819-0395, Japan.
| | - Hiroaki Mon
- Laboratory of Insect Genome Science, Kyushu University Graduate School of Bioresource and Bioenvironmental Sciences, Motooka 744, Nishi-ku, Fukuoka 819-0395, Japan.
| |
Collapse
|
9
|
Miao Q, Nguyen W, Zhu J, Liu G, van Oers MM, Tang B, Yan K, Larcher T, Suhrbier A, Pijlman GP. A getah virus-like-particle vaccine provides complete protection from viremia and arthritis in wild-type mice. Vaccine 2024; 42:126136. [PMID: 39004524 DOI: 10.1016/j.vaccine.2024.07.037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2024] [Revised: 07/08/2024] [Accepted: 07/10/2024] [Indexed: 07/16/2024]
Abstract
Getah virus (GETV) is an emerging mosquito-borne virus with economic impact on the livestock industry in East Asia. In this study, we successfully produced GETV virus-like particles (VLPs) in insect cells using the baculovirus expression vector system. We show that the GETV envelope glycoproteins were successfully expressed at the surface of the insect cell and were glycosylated. VLPs were isolated from the culture fluid as enveloped particles of 60-80 nm in diameter. Two 1 µg vaccinations with this GETV VLP vaccine, without adjuvant, generated neutralizing antibody responses and protected wild-type C57/BL6 mice against GETV viremia and arthritic disease. The GETV VLP vaccine may find application as a horse and/or pig vaccine in the future.
Collapse
Affiliation(s)
- Qiuhong Miao
- Laboratory of Virology, Wageningen University & Research, Wageningen, The Netherlands; Shanghai Veterinary Research Institute Chinese Academy of Agricultural Sciences, China.
| | - Wilson Nguyen
- Inflammation Biology Group, QIMR Berghofer Medical Research Institute, Brisbane, Australia.
| | - Jie Zhu
- Shanghai Veterinary Research Institute Chinese Academy of Agricultural Sciences, China.
| | - Guangqing Liu
- Shanghai Veterinary Research Institute Chinese Academy of Agricultural Sciences, China.
| | - Monique M van Oers
- Laboratory of Virology, Wageningen University & Research, Wageningen, The Netherlands.
| | - Bing Tang
- Inflammation Biology Group, QIMR Berghofer Medical Research Institute, Brisbane, Australia.
| | - Kexin Yan
- Inflammation Biology Group, QIMR Berghofer Medical Research Institute, Brisbane, Australia.
| | | | - Andreas Suhrbier
- Inflammation Biology Group, QIMR Berghofer Medical Research Institute, Brisbane, Australia; GVN Centre of Excellence, Australian Infectious Disease Research Centre, Brisbane, Queensland, Australia.
| | - Gorben P Pijlman
- Laboratory of Virology, Wageningen University & Research, Wageningen, The Netherlands.
| |
Collapse
|
10
|
Azizi-Dargahlou S, Pouresmaeil M, Ahmadabadi M. Tobacco Plant: A Novel and Promising Heterologous Bioreactor for the Production of Recombinant Bovine Chymosin. Mol Biotechnol 2024; 66:2595-2605. [PMID: 38244177 DOI: 10.1007/s12033-023-01043-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2023] [Accepted: 12/18/2023] [Indexed: 01/22/2024]
Abstract
The natural source of chymosin, a key enzyme in the dairy industry, is insufficient for rapidly growing cheese industries. Large-scale production of recombinant proteins in heterologous hosts provides an efficient alternative solution. Here, the codon-optimized synthetic prochymosin gene, which has a CAI index of 0.926, was subcloned from a cloning vector (pUC57-bCYM) into the pBI121 vector, resulting in the construct named pBI121-bCYM. CAI ranges from 0 to 1 and higher CAI improves gene expression in heterologous hosts. The overexpression of the prochymosin gene was under the control of constitutive CaMV 35S promoter and NOS terminator and was transferred into the tobacco via A. tumefaciens strain LBA4404. Explant type, regeneration method, inoculation temperature, cell density (OD600) of Agrobacterium for inoculation, and acetosyringone concentration were leaf explants, direct somatic embryogenesis, 19 °C, 0.1, and 100 µM, respectively. The successful integration and expression of the prochymosin gene, along with the bioactivity of recombinant chymosin, were confirmed by PCR, RT-PCR, and milk coagulation assay, respectively. Overall, this study reports the first successful overexpression of the codon-optimized prochymosin form of the bovine chymosin enzyme in the tobacco via indirect transformation. Production of recombinant bovine chymosin in plants can be an easy-to-scale-up, safe, and inexpensive platform.
Collapse
Affiliation(s)
- Shahnam Azizi-Dargahlou
- Seed and Plant Certification and Registration Institute, Ardabil Agricultural and Natural Resources Research Center, Agricultural Research, Education and Extension Organization (AREEO), Karaj, Iran.
| | - Mahin Pouresmaeil
- Faculty of Agricultural Sciences and Natural Resources, University of Mohaghegh Ardabili, Ardabi, Iran
| | - Mohammad Ahmadabadi
- Department of Biotechnology, Azarbaijan Shahid Madani University, Tabriz, Iran
| |
Collapse
|
11
|
Wang JH, Gessler DJ, Zhan W, Gallagher TL, Gao G. Adeno-associated virus as a delivery vector for gene therapy of human diseases. Signal Transduct Target Ther 2024; 9:78. [PMID: 38565561 PMCID: PMC10987683 DOI: 10.1038/s41392-024-01780-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: 07/05/2023] [Revised: 02/08/2024] [Accepted: 02/19/2024] [Indexed: 04/04/2024] Open
Abstract
Adeno-associated virus (AAV) has emerged as a pivotal delivery tool in clinical gene therapy owing to its minimal pathogenicity and ability to establish long-term gene expression in different tissues. Recombinant AAV (rAAV) has been engineered for enhanced specificity and developed as a tool for treating various diseases. However, as rAAV is being more widely used as a therapy, the increased demand has created challenges for the existing manufacturing methods. Seven rAAV-based gene therapy products have received regulatory approval, but there continue to be concerns about safely using high-dose viral therapies in humans, including immune responses and adverse effects such as genotoxicity, hepatotoxicity, thrombotic microangiopathy, and neurotoxicity. In this review, we explore AAV biology with an emphasis on current vector engineering strategies and manufacturing technologies. We discuss how rAAVs are being employed in ongoing clinical trials for ocular, neurological, metabolic, hematological, neuromuscular, and cardiovascular diseases as well as cancers. We outline immune responses triggered by rAAV, address associated side effects, and discuss strategies to mitigate these reactions. We hope that discussing recent advancements and current challenges in the field will be a helpful guide for researchers and clinicians navigating the ever-evolving landscape of rAAV-based gene therapy.
Collapse
Affiliation(s)
- Jiang-Hui Wang
- Horae Gene Therapy Center, University of Massachusetts Chan Medical School, Worcester, MA, 01605, USA
- Department of Microbiology and Physiological Systems, University of Massachusetts Chan Medical School, Worcester, MA, 01605, USA
- Centre for Eye Research Australia, Royal Victorian Eye and Ear Hospital, East Melbourne, VIC, 3002, Australia
- Ophthalmology, Department of Surgery, University of Melbourne, East Melbourne, VIC, 3002, Australia
| | - Dominic J Gessler
- Horae Gene Therapy Center, University of Massachusetts Chan Medical School, Worcester, MA, 01605, USA
- Department of Microbiology and Physiological Systems, University of Massachusetts Chan Medical School, Worcester, MA, 01605, USA
- Department of Neurological Surgery, University of Massachusetts Chan Medical School, Worcester, MA, 01605, USA
- Department of Neurosurgery, University of Minnesota, Minneapolis, MN, 55455, USA
| | - Wei Zhan
- Horae Gene Therapy Center, University of Massachusetts Chan Medical School, Worcester, MA, 01605, USA
- Department of Microbiology and Physiological Systems, University of Massachusetts Chan Medical School, Worcester, MA, 01605, USA
- Department of Medicine, University of Massachusetts Chan Medical School, Worcester, MA, 01605, USA
- Li Weibo Institute for Rare Diseases Research, University of Massachusetts Chan Medical School, Worcester, MA, 01605, USA
| | - Thomas L Gallagher
- Horae Gene Therapy Center, University of Massachusetts Chan Medical School, Worcester, MA, 01605, USA
| | - Guangping Gao
- Horae Gene Therapy Center, University of Massachusetts Chan Medical School, Worcester, MA, 01605, USA.
- Department of Microbiology and Physiological Systems, University of Massachusetts Chan Medical School, Worcester, MA, 01605, USA.
- Li Weibo Institute for Rare Diseases Research, University of Massachusetts Chan Medical School, Worcester, MA, 01605, USA.
| |
Collapse
|
12
|
Mahdizade Ari M, Dadgar L, Elahi Z, Ghanavati R, Taheri B. Genetically Engineered Microorganisms and Their Impact on Human Health. Int J Clin Pract 2024; 2024:6638269. [PMID: 38495751 PMCID: PMC10944348 DOI: 10.1155/2024/6638269] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/17/2023] [Revised: 11/20/2023] [Accepted: 02/12/2024] [Indexed: 03/19/2024] Open
Abstract
The emergence of antibiotic-resistant strains, the decreased effectiveness of conventional therapies, and the side effects have led researchers to seek a safer, more cost-effective, patient-friendly, and effective method that does not develop antibiotic resistance. With progress in synthetic biology and genetic engineering, genetically engineered microorganisms effective in treatment, prophylaxis, drug delivery, and diagnosis have been developed. The present study reviews the types of genetically engineered bacteria and phages, their impacts on diseases, cancer, and metabolic and inflammatory disorders, the biosynthesis of these modified strains, the route of administration, and their effects on the environment. We conclude that genetically engineered microorganisms can be considered promising candidates for adjunctive treatment of diseases and cancers.
Collapse
Affiliation(s)
- Marzie Mahdizade Ari
- Department of Microbiology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
- Microbial Biotechnology Research Centre, Iran University of Medical Sciences, Tehran, Iran
| | - Leila Dadgar
- Department of Microbiology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
- Microbial Biotechnology Research Centre, Iran University of Medical Sciences, Tehran, Iran
| | - Zahra Elahi
- Department of Microbiology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
- Microbial Biotechnology Research Centre, Iran University of Medical Sciences, Tehran, Iran
| | | | - Behrouz Taheri
- Department of Biotechnology, School of Medicine, Ahvaz Jundishapour University of medical Sciences, Ahvaz, Iran
| |
Collapse
|
13
|
de Jong LA, van Oosten L, Pijlman GP. Scarless Baculovirus Genome Editing Using Lambda-Red Recombineering in E. coli. Methods Mol Biol 2024; 2829:109-126. [PMID: 38951330 DOI: 10.1007/978-1-0716-3961-0_8] [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
Baculoviruses are widely used for their potential as biological pesticide and as platform for the production of recombinant proteins and gene therapy vectors. The Baculovirus Expression Vector System (BEVS) is used for high level of expression of (multiple) proteins in insect cells. Baculovirus recombinants can be quickly constructed by transposition of the gene(s) of interest into a so-called bacmid, which is a baculovirus infectious clone maintained as single-copy, bacterial artificial chromosome in Escherichia coli. A two-step homologous recombineering technique using the lambda-red system in E. coli allows for scarless editing of the bacmid with PCR products based on sequence homology. In the first step, a selection cassette with 50 bp homology arms, typically generated by PCR, is inserted into the designated locus. In the second step, the selection cassette is removed based on a negative selection marker, such as SacB or rpsL. This lambda-red recombineering technique can be used for multiple gene editing purposes, including (large) deletions, insertions, and even single point mutations. Moreover, since there are no remnants of the editing process, successive modifications of the same bacmid are possible. This chapter provides detailed instructions to design and perform two-step homologous recombineering of baculovirus bacmid DNA in E. coli. We present two case studies demonstrating the utility of this technique for creating a deletion mutant of the chitinase and cathepsin genes and for introducing a single point mutation in the baculovirus gene gp41. This scarless genome editing approach can facilitate functional studies of baculovirus genes and improve the production of recombinant proteins using the BEVS.
Collapse
Affiliation(s)
- Linda A de Jong
- Laboratory of Virology, Wageningen University, Wageningen, The Netherlands
| | - Linda van Oosten
- Laboratory of Virology, Wageningen University, Wageningen, The Netherlands
| | - Gorben P Pijlman
- Laboratory of Virology, Wageningen University, Wageningen, The Netherlands.
| |
Collapse
|
14
|
Bruder MR, Aucoin MG. A sensitive assay for scrutiny of Autographa californica multiple nucleopolyhedrovirus genes using CRISPR-Cas9. Appl Microbiol Biotechnol 2023:10.1007/s00253-023-12462-y. [PMID: 37233755 DOI: 10.1007/s00253-023-12462-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Revised: 02/09/2023] [Accepted: 02/27/2023] [Indexed: 05/27/2023]
Abstract
Baculoviruses have very large genomes and previous studies have demonstrated improvements in recombinant protein production and genome stability through the removal of some nonessential sequences. However, recombinant baculovirus expression vectors (rBEVs) in widespread use remain virtually unmodified. Traditional approaches for generating knockout viruses (KOVs) require several experimental steps to remove the target gene prior to the generation of the virus. In order to optimize rBEV genomes by removing nonessential sequences, more efficient techniques for establishing and evaluating KOVs are required. Here, we have developed a sensitive assay utilizing CRISPR-Cas9-mediated gene targeting to examine the phenotypic impact of disruption of endogenous Autographa californica multiple nucleopolyhedrovirus (AcMNPV) genes. For validation, 13 AcMNPV genes were targeted for disruption and evaluated for the production of GFP and progeny virus - traits that are essential for their use as vectors for recombinant protein production. The assay involves transfection of sgRNA into a Cas9-expressing Sf9 cell line followed by infection with a baculovirus vector carrying the gfp gene under the p10 or p6.9 promoters. This assay represents an efficient strategy for scrutinizing AcMNPV gene function through targeted disruption, and represents a valuable tool for developing an optimized rBEV genome. KEY POINTS: [Formula: see text] A method to scrutinize the essentiality of baculovirus genes was developed. [Formula: see text] The method uses Sf9-Cas9 cells, a targeting plasmid carrying a sgRNA, and a rBEV-GFP. [Formula: see text] The method allows scrutiny by only needing to modify the targeting sgRNA plasmid.
Collapse
Affiliation(s)
- Mark R Bruder
- Department of Chemical Engineering, University of Waterloo, 200 University Ave. W., Waterloo, N2L 3G1, Ontario, Canada
| | - Marc G Aucoin
- Department of Chemical Engineering, University of Waterloo, 200 University Ave. W., Waterloo, N2L 3G1, Ontario, Canada.
| |
Collapse
|
15
|
Abbo SR, Nguyen W, Abma-Henkens MHC, van de Kamer D, Savelkoul NHA, Geertsema C, Le TTT, Tang B, Yan K, Dumenil T, van Oers MM, Suhrbier A, Pijlman GP. Comparative Efficacy of Mayaro Virus-Like Particle Vaccines Produced in Insect or Mammalian Cells. J Virol 2023; 97:e0160122. [PMID: 36883812 PMCID: PMC10062127 DOI: 10.1128/jvi.01601-22] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2022] [Accepted: 02/13/2023] [Indexed: 03/09/2023] Open
Abstract
Mayaro virus (MAYV) is a mosquito-transmitted alphavirus that causes often debilitating rheumatic disease in tropical Central and South America. There are currently no licensed vaccines or antiviral drugs available for MAYV disease. Here, we generated Mayaro virus-like particles (VLPs) using the scalable baculovirus-insect cell expression system. High-level secretion of MAYV VLPs in the culture fluid of Sf9 insect cells was achieved, and particles with a diameter of 64 to 70 nm were obtained after purification. We characterize a C57BL/6J adult wild-type mouse model of MAYV infection and disease and used this model to compare the immunogenicity of VLPs from insect cells with that of VLPs produced in mammalian cells. Mice received two intramuscular immunizations with 1 μg of nonadjuvanted MAYV VLPs. Potent neutralizing antibody responses were generated against the vaccine strain, BeH407, with comparable activity seen against a contemporary 2018 isolate from Brazil (BR-18), whereas neutralizing activity against chikungunya virus was marginal. Sequencing of BR-18 illustrated that this virus segregates with genotype D isolates, whereas MAYV BeH407 belongs to genotype L. The mammalian cell-derived VLPs induced higher mean neutralizing antibody titers than those produced in insect cells. Both VLP vaccines completely protected adult wild-type mice against viremia, myositis, tendonitis, and joint inflammation after MAYV challenge. IMPORTANCE Mayaro virus (MAYV) is associated with acute rheumatic disease that can be debilitating and can evolve into months of chronic arthralgia. MAYV is believed to have the potential to emerge as a tropical public health threat, especially if it develops the ability to be efficiently transmitted by urban mosquito vectors, such as Aedes aegypti and/or Aedes albopictus. Here, we describe a scalable virus-like particle vaccine against MAYV that induced neutralizing antibodies against a historical and a contemporary isolate of MAYV and protected mice against infection and disease, providing a potential new intervention for MAYV epidemic preparedness.
Collapse
Affiliation(s)
- Sandra R. Abbo
- Laboratory of Virology, Wageningen University & Research, Wageningen, the Netherlands
| | - Wilson Nguyen
- Inflammation Biology Group, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | | | - Denise van de Kamer
- Laboratory of Virology, Wageningen University & Research, Wageningen, the Netherlands
| | - Niek H. A. Savelkoul
- Laboratory of Virology, Wageningen University & Research, Wageningen, the Netherlands
| | - Corinne Geertsema
- Laboratory of Virology, Wageningen University & Research, Wageningen, the Netherlands
| | - Thuy T. T. Le
- Inflammation Biology Group, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Bing Tang
- Inflammation Biology Group, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Kexin Yan
- Inflammation Biology Group, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Troy Dumenil
- Inflammation Biology Group, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Monique M. van Oers
- Laboratory of Virology, Wageningen University & Research, Wageningen, the Netherlands
| | - Andreas Suhrbier
- Inflammation Biology Group, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
- GVN Center of Excellence, Australian Infectious Disease Research Center, Brisbane, Queensland, Australia
| | - Gorben P. Pijlman
- Laboratory of Virology, Wageningen University & Research, Wageningen, the Netherlands
| |
Collapse
|
16
|
Huang J, Liu H, Xu X. Homologous recombination risk in baculovirus expression vector system. Virus Res 2022; 321:198924. [PMID: 36089109 DOI: 10.1016/j.virusres.2022.198924] [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: 07/28/2022] [Revised: 09/04/2022] [Accepted: 09/07/2022] [Indexed: 12/24/2022]
Abstract
The baculovirus expression vector system (BEVS) is widely used for producing recombinant proteins. To achieve high expression level of recombinant proteins, baculoviral elements, such as enhancers, promoters, signal peptide coding sequences and 3'-UTR, have been extensively employed. There is a recombination risk derived from homologous sequences between viral genome and functional baculovirus-derived elements associated with foreign genes. Although homologous recombination have distinct biological functions, these potential adverse recombination may trigger a DNA fragment being inverted or looped out, resulting in the production of defective viruses and eventual yields declines of recombinant proteins. However, the risk of such homologous recombination has not been systematically assessed. Here, we measured the recombination rate using a promoter-less fluorescent reporter integrated with various lengths homologous of p10 coding region. Homologous fragments longer than 60 bp possess sufficient recombination probability and exerts effect on purity and integrity of virus. Shortening the length of homologous fragments and separating homologous fragments by point mutations can effectively reduce unfavorable recombination. These findings reveal a homologous recombination risk resulted from genome-homologous baculoviral elements and propose reliable strategies reducing recombination rate to facilitate viral stability and integrity in baculovirus expression vector system.
Collapse
Affiliation(s)
- Jianan Huang
- College of Life Sciences, Northwest A&F University, Yangling, Shaanxi, China.
| | - Huanlei Liu
- College of Life Sciences, Northwest A&F University, Yangling, Shaanxi, China.
| | - Xiaodong Xu
- College of Life Sciences, Northwest A&F University, Yangling, Shaanxi, China.
| |
Collapse
|
17
|
Hong M, Li T, Xue W, Zhang S, Cui L, Wang H, Zhang Y, Zhou L, Gu Y, Xia N, Li S. Genetic engineering of baculovirus-insect cell system to improve protein production. Front Bioeng Biotechnol 2022; 10:994743. [PMID: 36204465 PMCID: PMC9530357 DOI: 10.3389/fbioe.2022.994743] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Accepted: 09/05/2022] [Indexed: 11/13/2022] Open
Abstract
The Baculovirus Expression Vector System (BEVS), a mature foreign protein expression platform, has been available for decades, and has been effectively used in vaccine production, gene therapy, and a host of other applications. To date, eleven BEVS-derived products have been approved for use, including four human vaccines [Cervarix against cervical cancer caused by human papillomavirus (HPV), Flublok and Flublok Quadrivalent against seasonal influenza, Nuvaxovid/Covovax against COVID-19], two human therapeutics [Provenge against prostate cancer and Glybera against hereditary lipoprotein lipase deficiency (LPLD)] and five veterinary vaccines (Porcilis Pesti, BAYOVAC CSF E2, Circumvent PCV, Ingelvac CircoFLEX and Porcilis PCV). The BEVS has many advantages, including high safety, ease of operation and adaptable for serum-free culture. It also produces properly folded proteins with correct post-translational modifications, and can accommodate multi-gene- or large gene insertions. However, there remain some challenges with this system, including unstable expression and reduced levels of protein glycosylation. As the demand for biotechnology increases, there has been a concomitant effort into optimizing yield, stability and protein glycosylation through genetic engineering and the manipulation of baculovirus vector and host cells. In this review, we summarize the strategies and technological advances of BEVS in recent years and explore how this will be used to inform the further development and application of this system.
Collapse
Affiliation(s)
- Minqing Hong
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, School of Life Sciences, School of Public Health, Xiamen University, Xiamen, China
- National Institute of Diagnostics and Vaccine Development in Infectious Diseases, Xiamen University, Xiamen, China
- Xiang An Biomedicine Laboratory, Xiamen, China
| | - Tingting Li
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, School of Life Sciences, School of Public Health, Xiamen University, Xiamen, China
- National Institute of Diagnostics and Vaccine Development in Infectious Diseases, Xiamen University, Xiamen, China
- Xiang An Biomedicine Laboratory, Xiamen, China
| | - Wenhui Xue
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, School of Life Sciences, School of Public Health, Xiamen University, Xiamen, China
- National Institute of Diagnostics and Vaccine Development in Infectious Diseases, Xiamen University, Xiamen, China
- Xiang An Biomedicine Laboratory, Xiamen, China
| | - Sibo Zhang
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, School of Life Sciences, School of Public Health, Xiamen University, Xiamen, China
- National Institute of Diagnostics and Vaccine Development in Infectious Diseases, Xiamen University, Xiamen, China
- Xiang An Biomedicine Laboratory, Xiamen, China
| | - Lingyan Cui
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, School of Life Sciences, School of Public Health, Xiamen University, Xiamen, China
- National Institute of Diagnostics and Vaccine Development in Infectious Diseases, Xiamen University, Xiamen, China
- Xiang An Biomedicine Laboratory, Xiamen, China
| | - Hong Wang
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, School of Life Sciences, School of Public Health, Xiamen University, Xiamen, China
- National Institute of Diagnostics and Vaccine Development in Infectious Diseases, Xiamen University, Xiamen, China
- Xiang An Biomedicine Laboratory, Xiamen, China
| | - Yuyun Zhang
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, School of Life Sciences, School of Public Health, Xiamen University, Xiamen, China
- National Institute of Diagnostics and Vaccine Development in Infectious Diseases, Xiamen University, Xiamen, China
- Xiang An Biomedicine Laboratory, Xiamen, China
| | - Lizhi Zhou
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, School of Life Sciences, School of Public Health, Xiamen University, Xiamen, China
- National Institute of Diagnostics and Vaccine Development in Infectious Diseases, Xiamen University, Xiamen, China
- Xiang An Biomedicine Laboratory, Xiamen, China
| | - Ying Gu
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, School of Life Sciences, School of Public Health, Xiamen University, Xiamen, China
- National Institute of Diagnostics and Vaccine Development in Infectious Diseases, Xiamen University, Xiamen, China
- Xiang An Biomedicine Laboratory, Xiamen, China
| | - Ningshao Xia
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, School of Life Sciences, School of Public Health, Xiamen University, Xiamen, China
- National Institute of Diagnostics and Vaccine Development in Infectious Diseases, Xiamen University, Xiamen, China
- Xiang An Biomedicine Laboratory, Xiamen, China
- The Research Unit of Frontier Technology of Structural Vaccinology of Chinese Academy of Medical Sciences, Xiamen, China
| | - Shaowei Li
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, School of Life Sciences, School of Public Health, Xiamen University, Xiamen, China
- National Institute of Diagnostics and Vaccine Development in Infectious Diseases, Xiamen University, Xiamen, China
- Xiang An Biomedicine Laboratory, Xiamen, China
| |
Collapse
|
18
|
Fang Z, Lyu J, Li J, Li C, Zhang Y, Guo Y, Wang Y, Zhang Y, Chen K. Application of bioreactor technology for cell culture-based viral vaccine production: Present status and future prospects. Front Bioeng Biotechnol 2022; 10:921755. [PMID: 36017347 PMCID: PMC9395942 DOI: 10.3389/fbioe.2022.921755] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2022] [Accepted: 07/06/2022] [Indexed: 11/24/2022] Open
Abstract
Bioreactors are widely used in cell culture-based viral vaccine production, especially during the coronavirus disease 2019 (COVID-19) pandemic. In this context, the development and application of bioreactors can provide more efficient and cost-effective vaccine production to meet the global vaccine demand. The production of viral vaccines is inseparable from the development of upstream biological processes. In particular, exploration at the laboratory-scale is urgently required for further development. Therefore, it is necessary to evaluate the existing upstream biological processes, to enable the selection of pilot-scale conditions for academic and industrial scientists to maximize the yield and quality of vaccine development and production. Reviewing methods for optimizing the upstream process of virus vaccine production, this review discusses the bioreactor concepts, significant parameters and operational strategies related to large-scale amplification of virus. On this basis, a comprehensive analysis and evaluation of the various process optimization methods for the production of various viruses (SARS-CoV-2, Influenza virus, Tropical virus, Enterovirus, Rabies virus) in bioreactors is presented. Meanwhile, the types of viral vaccines are briefly introduced, and the established animal cell lines for vaccine production are described. In addition, it is emphasized that the co-development of bioreactor and computational biology is urgently needed to meet the challenges posed by the differences in upstream production scales between the laboratory and industry.
Collapse
Affiliation(s)
- Zhongbiao Fang
- Shulan International Medical College, Zhejiang Shuren University, Hangzhou, China
| | - Jingting Lyu
- Shulan International Medical College, Zhejiang Shuren University, Hangzhou, China
| | - Jianhua Li
- Zhejiang Provincial Center for Disease Control and Prevention, Hangzhou, China
| | - Chaonan Li
- Shulan International Medical College, Zhejiang Shuren University, Hangzhou, China
| | - Yuxuan Zhang
- Shulan International Medical College, Zhejiang Shuren University, Hangzhou, China
| | - Yikai Guo
- Shulan International Medical College, Zhejiang Shuren University, Hangzhou, China
| | - Ying Wang
- Shulan International Medical College, Zhejiang Shuren University, Hangzhou, China
- *Correspondence: Ying Wang, ; Yanjun Zhang, ; Keda Chen,
| | - Yanjun Zhang
- Zhejiang Provincial Center for Disease Control and Prevention, Hangzhou, China
- *Correspondence: Ying Wang, ; Yanjun Zhang, ; Keda Chen,
| | - Keda Chen
- Shulan International Medical College, Zhejiang Shuren University, Hangzhou, China
- *Correspondence: Ying Wang, ; Yanjun Zhang, ; Keda Chen,
| |
Collapse
|
19
|
Azali MA, Mohamed S, Harun A, Hussain FA, Shamsuddin S, Johan MF. Application of Baculovirus Expression Vector system (BEV) for COVID-19 diagnostics and therapeutics: a review. J Genet Eng Biotechnol 2022; 20:98. [PMID: 35792966 PMCID: PMC9259773 DOI: 10.1186/s43141-022-00368-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Accepted: 05/20/2022] [Indexed: 11/10/2022]
Abstract
BACKGROUND The baculovirus expression vector system has been developed for expressing a wide range of proteins, including enzymes, glycoproteins, recombinant viruses, and vaccines. The availability of the SARS-CoV-2 genome sequence has enabled the synthesis of SARS-CoV2 proteins in a baculovirus-insect cell platform for various applications. The most cloned SARS-CoV-2 protein is the spike protein, which plays a critical role in SARS-CoV-2 infection. It is available in its whole length or as subunits like S1 or the receptor-binding domain (RBD). Non-structural proteins (Nsps), another recombinant SARS-CoV-2 protein generated by the baculovirus expression vector system (BEV), are used in the identification of new medications or the repurposing of existing therapies for the treatment of COVID-19. Non-SARS-CoV-2 proteins generated by BEV for SARS-CoV-2 diagnosis or treatment include moloney murine leukemia virus reverse transcriptase (MMLVRT), angiotensin converting enzyme 2 (ACE2), therapeutic proteins, and recombinant antibodies. The recombinant proteins were modified to boost the yield or to stabilize the protein. CONCLUSION This review covers the wide application of the recombinant protein produced using the baculovirus expression technology for COVID-19 research. A lot of improvements have been made to produce functional proteins with high yields. However, there is still room for improvement and there are parts of this field of research that have not been investigated yet.
Collapse
Affiliation(s)
- Muhammad Azharuddin Azali
- Department of Haematology, School of Medical Sciences, Universiti Sains Malaysia, 16150, Kubang Kerian, Kelantan, Malaysia
- School of Agriculture Science and Biotechnology, Faculty of Bioresources and Food Industry, Universiti Sultan Zainal Abidin, 22200, Besut, Terengganu, Malaysia
| | - Salmah Mohamed
- School of Agriculture Science and Biotechnology, Faculty of Bioresources and Food Industry, Universiti Sultan Zainal Abidin, 22200, Besut, Terengganu, Malaysia
| | - Azian Harun
- Department of Medical Microbiology and Parasitology, School of Medical Sciences, Universiti Sains Malaysia, 16150, Kubang Kerian, Kelantan, Malaysia
| | - Faezahtul Arbaeyah Hussain
- Department of Pathology, School of Medical Sciences, Universiti Sains Malaysia, 16150, Kubang Kerian, Kelantan, Malaysia
| | - Shaharum Shamsuddin
- School of Health Sciences, Universiti Sains Malaysia, 16150, Kubang Kerian, Kelantan, Malaysia
| | - Muhammad Farid Johan
- Department of Haematology, School of Medical Sciences, Universiti Sains Malaysia, 16150, Kubang Kerian, Kelantan, Malaysia.
| |
Collapse
|
20
|
Secreted Trimeric Chikungunya Virus Spikes from Insect Cells: Production, Purification, and Glycosylation Status. Processes (Basel) 2022. [DOI: 10.3390/pr10010162] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
Chikungunya virus (CHIKV) is a rapidly emerging mosquito-borne virus that causes a severe febrile illness with long-lasting arthralgia in humans. As there is no vaccine to protect humans and limit CHIKV epidemics, the virus continues to be a global public health concern. The CHIKV envelope glycoproteins E1 and E2 are important immunogens; therefore, the aim of this study is to produce trimeric CHIKV spikes in insect cells using the baculovirus expression system. The CHIKV E1 and E2 ectodomains were covalently coupled by a flexible linker that replaces the 6K transmembrane protein. The C-terminal E1 transmembrane was replaced by a Strep-tag II for the purification of secreted spikes from the culture fluid. After production in Sf9 suspension cells (product yields of 5.8–7.6 mg/L), the CHIKV spikes were purified by Strep-Tactin affinity chromatography, which successfully cleared the co-produced baculoviruses. Bis(sulfosuccinimidyl)suberate cross-linking demonstrated that the spikes are secreted as trimers. PNGase F treatment showed that the spikes are glycosylated. LC–MS/MS-based glycoproteomic analysis confirmed the glycosylation and revealed that the majority are of the mannose- or hybrid-type N-glycans and <2% have complex-type N-glycans. The LC –MS/MS analysis also revealed three O-glycosylation sites in E1. In conclusion, the trimeric, glycosylated CHIKV spikes have been successfully produced in insect cells and are now available for vaccination studies.
Collapse
|
21
|
Two-Component Nanoparticle Vaccine Displaying Glycosylated Spike S1 Domain Induces Neutralizing Antibody Response against SARS-CoV-2 Variants. mBio 2021; 12:e0181321. [PMID: 34634927 PMCID: PMC8510518 DOI: 10.1128/mbio.01813-21] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Vaccines pave the way out of the SARS-CoV-2 pandemic. Besides mRNA and adenoviral vector vaccines, effective protein-based vaccines are needed for immunization against current and emerging variants. We have developed a virus-like particle (VLP)-based vaccine using the baculovirus-insect cell expression system, a robust production platform known for its scalability, low cost, and safety. Baculoviruses were constructed encoding SARS-CoV-2 spike proteins: full-length S, stabilized secreted S, or the S1 domain. Since subunit S only partially protected mice from SARS-CoV-2 challenge, we produced S1 for conjugation to bacteriophage AP205 VLP nanoparticles using tag/catcher technology. The S1 yield in an insect-cell bioreactor was ∼11 mg/liter, and authentic protein folding, efficient glycosylation, partial trimerization, and ACE2 receptor binding was confirmed. Prime-boost immunization of mice with 0.5 μg S1-VLPs showed potent neutralizing antibody responses against Wuhan and UK/B.1.1.7 SARS-CoV-2 variants. This two-component nanoparticle vaccine can now be further developed to help alleviate the burden of COVID-19.
Collapse
|
22
|
Varanda C, Félix MDR, Campos MD, Materatski P. An Overview of the Application of Viruses to Biotechnology. Viruses 2021; 13:2073. [PMID: 34696503 PMCID: PMC8541484 DOI: 10.3390/v13102073] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Accepted: 10/09/2021] [Indexed: 12/23/2022] Open
Abstract
Viruses may cause devastating diseases in several organisms; however, they are simple systems that can be manipulated to be beneficial and useful for many purposes in different areas. In medicine, viruses have been used for a long time in vaccines and are now being used as vectors to carry materials for the treatment of diseases, such as cancer, being able to target specific cells. In agriculture, viruses are being studied to introduce desirable characteristics in plants or render resistance to biotic and abiotic stresses. Viruses have been exploited in nanotechnology for the deposition of specific metals and have been shown to be of great benefit to nanomaterial production. They can also be used for different applications in pharmacology, cosmetics, electronics, and other industries. Thus, viruses are no longer only seen as enemies. They have shown enormous potential, covering several important areas in our lives, and they are making our lives easier and better. Although viruses have already proven their potential, there is still a long road ahead. This prompt us to propose this theme in the Special Issue "The application of viruses to biotechnology". We believe that the articles gathered here highlight recent significant advances in the use of viruses in several fields, contributing to the current knowledge on virus applications.
Collapse
Affiliation(s)
- Carla Varanda
- MED–Mediterranean Institute for Agriculture, Environment and Development, Instituto de Investigação e Formação Avançada, Universidade de Évora, Pólo da Mitra, Ap. 94, 7006-554 Évora, Portugal;
| | - Maria do Rosário Félix
- MED–Mediterranean Institute for Agriculture, Environment and Development & Departamento de Fitotecnia, Escola de Ciências e Tecnologia, Universidade de Évora, Pólo da Mitra, Ap. 94, 7006-554 Évora, Portugal;
| | - Maria Doroteia Campos
- MED–Mediterranean Institute for Agriculture, Environment and Development, Instituto de Investigação e Formação Avançada, Universidade de Évora, Pólo da Mitra, Ap. 94, 7006-554 Évora, Portugal;
| | - Patrick Materatski
- MED–Mediterranean Institute for Agriculture, Environment and Development, Instituto de Investigação e Formação Avançada, Universidade de Évora, Pólo da Mitra, Ap. 94, 7006-554 Évora, Portugal;
| |
Collapse
|