1
|
Achleitner L, Winter M, Aguilar PP, Lingg N, Jungbauer A, Klausberger M, Satzer P. Robust and resource-efficient production process suitable for large-scale production of baculovirus through high cell density seed train and optimized infection strategy. N Biotechnol 2024; 80:46-55. [PMID: 38302001 DOI: 10.1016/j.nbt.2024.01.002] [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: 10/24/2023] [Revised: 01/08/2024] [Accepted: 01/22/2024] [Indexed: 02/03/2024]
Abstract
The aim of this study was the development of a scalable production process for high titer (108 pfu/mL and above) recombinant baculovirus stocks with low cell line-derived impurities for the production of virus-like particles (VLP). To achieve this, we developed a high cell density (HCD) culture for low footprint cell proliferation, compared different infection strategies at multiplicity of infection (MOI) 0.05 and 0.005, different infection strategies and validated generally applicable harvest criteria of cell viability ≤ 80%. We also investigated online measurable parameters to observe the baculovirus production. The infection strategy employing a very low virus inoculum of MOI 0.005 and a 1:2 dilution with fresh medium one day after infection proved to be the most resource efficient. There, we achieved higher cell-specific titers and lower host cell protein concentrations at harvest than other tested infection strategies with the same MOI, while saving half of the virus stock for infecting the culture compared to other tested infection strategies. HCD culture by daily medium exchange was confirmed as suitable for seed train propagation, infection, and baculovirus production, equally efficient as the conventionally propagated seed train. Online measurable parameters for cell concentration and average cell diameter were found to be effective in monitoring the production process. The study concluded that a more efficient VLP production process in large scale can be achieved using this virus stock production strategy, which could also be extended to produce other proteins or extracellular vesicles with the baculovirus expression system.
Collapse
Affiliation(s)
- Lena Achleitner
- acib - Austrian Centre of Industrial Biotechnology, Muthgasse 11, 1190 Vienna, Austria; Institute of Bioprocess Science and Engineering, Department of Biotechnology, University of Natural Resources and Life Sciences, Vienna (BOKU), Muthgasse 18, 1190 Vienna, Austria
| | - Martina Winter
- acib - Austrian Centre of Industrial Biotechnology, Muthgasse 11, 1190 Vienna, Austria; Institute of Bioprocess Science and Engineering, Department of Biotechnology, University of Natural Resources and Life Sciences, Vienna (BOKU), Muthgasse 18, 1190 Vienna, Austria
| | - Patricia Pereira Aguilar
- acib - Austrian Centre of Industrial Biotechnology, Muthgasse 11, 1190 Vienna, Austria; Institute of Bioprocess Science and Engineering, Department of Biotechnology, University of Natural Resources and Life Sciences, Vienna (BOKU), Muthgasse 18, 1190 Vienna, Austria
| | - Nico Lingg
- acib - Austrian Centre of Industrial Biotechnology, Muthgasse 11, 1190 Vienna, Austria; Institute of Bioprocess Science and Engineering, Department of Biotechnology, University of Natural Resources and Life Sciences, Vienna (BOKU), Muthgasse 18, 1190 Vienna, Austria
| | - Alois Jungbauer
- acib - Austrian Centre of Industrial Biotechnology, Muthgasse 11, 1190 Vienna, Austria; Institute of Bioprocess Science and Engineering, Department of Biotechnology, University of Natural Resources and Life Sciences, Vienna (BOKU), Muthgasse 18, 1190 Vienna, Austria
| | - Miriam Klausberger
- Institute of Molecular Biotechnology, Department of Biotechnology, University of Natural Resources and Life Sciences, Vienna (BOKU), Muthgasse 18, 1190 Vienna, Austria
| | - Peter Satzer
- acib - Austrian Centre of Industrial Biotechnology, Muthgasse 11, 1190 Vienna, Austria; Institute of Bioprocess Science and Engineering, Department of Biotechnology, University of Natural Resources and Life Sciences, Vienna (BOKU), Muthgasse 18, 1190 Vienna, Austria.
| |
Collapse
|
2
|
Schönherr R, Boger J, Lahey-Rudolph JM, Harms M, Kaiser J, Nachtschatt S, Wobbe M, Duden R, König P, Bourenkov G, Schneider TR, Redecke L. A streamlined approach to structure elucidation using in cellulo crystallized recombinant proteins, InCellCryst. Nat Commun 2024; 15:1709. [PMID: 38402242 PMCID: PMC10894269 DOI: 10.1038/s41467-024-45985-7] [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: 06/02/2023] [Accepted: 02/02/2024] [Indexed: 02/26/2024] Open
Abstract
With the advent of serial X-ray crystallography on microfocus beamlines at free-electron laser and synchrotron facilities, the demand for protein microcrystals has significantly risen in recent years. However, by in vitro crystallization extensive efforts are usually required to purify proteins and produce sufficiently homogeneous microcrystals. Here, we present InCellCryst, an advanced pipeline for producing homogeneous microcrystals directly within living insect cells. Our baculovirus-based cloning system enables the production of crystals from completely native proteins as well as the screening of different cellular compartments to maximize chances for protein crystallization. By optimizing cloning procedures, recombinant virus production, crystallization and crystal detection, X-ray diffraction data can be collected 24 days after the start of target gene cloning. Furthermore, improved strategies for serial synchrotron diffraction data collection directly from crystals within living cells abolish the need to purify the recombinant protein or the associated microcrystals.
Collapse
Affiliation(s)
- Robert Schönherr
- Institute of Biochemistry, University of Lübeck, Lübeck, Germany
| | - Juliane Boger
- Institute of Biochemistry, University of Lübeck, Lübeck, Germany
| | - J Mia Lahey-Rudolph
- Institute of Biochemistry, University of Lübeck, Lübeck, Germany
- Center for Free-Electron Laser Science (CFEL), Hamburg, Germany
- X-ray technology lab, TH Lübeck - University of Applied Sciences Lübeck, Lübeck, Germany
| | - Mareike Harms
- Institute of Biochemistry, University of Lübeck, Lübeck, Germany
| | | | | | - Marla Wobbe
- Institute of Biochemistry, University of Lübeck, Lübeck, Germany
| | - Rainer Duden
- Institute of Biology, University of Lübeck, Lübeck, Germany
| | - Peter König
- Institute of Anatomy, University of Lübeck, Lübeck, Germany
- Airway Research Center North (ARCN), University of Lübeck, German Center for Lung Research (DZL), Lübeck, Germany
| | - Gleb Bourenkov
- European Molecular Biology Laboratory, Hamburg Unit c/o Deutsches Elektronen-Synchrotron DESY, Hamburg, Germany
| | - Thomas R Schneider
- European Molecular Biology Laboratory, Hamburg Unit c/o Deutsches Elektronen-Synchrotron DESY, Hamburg, Germany
| | - Lars Redecke
- Institute of Biochemistry, University of Lübeck, Lübeck, Germany.
- Deutsches Elektronen-Synchrotron DESY, Hamburg, Germany.
| |
Collapse
|
3
|
Loup-Forest J, Matuchet M, Schnitzler C, Pichard S, Poterszman A. A Time and Cost-Effective Pipeline for Expression Screening and Protein Production in Insect Cells Based on the HR-Bac Toolbox to Generate Recombinant Baculoviruses. Methods Mol Biol 2024; 2829:21-48. [PMID: 38951325 DOI: 10.1007/978-1-0716-3961-0_3] [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
The baculovirus expression vector system (BEVS) is recognized as a powerful platform for producing challenging proteins and multiprotein complexes both in academia and industry. Since a baculovirus was first used to produce heterologous human IFN-β protein in insect cells, the BEVS has continuously been developed and its applications expanded. We have recently established a multigene expression toolbox (HR-bac) composed of a set of engineered bacmids expressing a fluorescent marker to monitor virus propagation and a library of transfer vectors. Unlike platforms that rely on Tn7-medidated transposition for the construction of baculoviruses, HR-bac relies on homologous recombination, which allows to evaluate expression constructs in 2 weeks and is thus perfectly adapted to parallel expression screening. In this chapter, we detail our standard operating procedures for the preparation of the reagents, the construction and evaluation of baculoviruses, and the optimization of protein production for both intracellularly expressed and secreted proteins.
Collapse
Affiliation(s)
- Jules Loup-Forest
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Integrated Structural Biology Department, Center for Integrated Structural Biology (CBI), Illkirch, France
- Centre National de la Recherche Scientifique, UMR7104, Institut National de la Santé et de la Recherche Médicale, U1258, Illkirch, France
- Université de Strasbourg, Equipe labellisée Ligue Contre le Cancer, BP, Illkirch, France
| | - Manon Matuchet
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Integrated Structural Biology Department, Center for Integrated Structural Biology (CBI), Illkirch, France
- Centre National de la Recherche Scientifique, UMR7104, Institut National de la Santé et de la Recherche Médicale, U1258, Illkirch, France
- Université de Strasbourg, Equipe labellisée Ligue Contre le Cancer, BP, Illkirch, France
| | - Charlotte Schnitzler
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Integrated Structural Biology Department, Center for Integrated Structural Biology (CBI), Illkirch, France
- Centre National de la Recherche Scientifique, UMR7104, Institut National de la Santé et de la Recherche Médicale, U1258, Illkirch, France
- Université de Strasbourg, Equipe labellisée Ligue Contre le Cancer, BP, Illkirch, France
| | - Simon Pichard
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Integrated Structural Biology Department, Center for Integrated Structural Biology (CBI), Illkirch, France
- Centre National de la Recherche Scientifique, UMR7104, Institut National de la Santé et de la Recherche Médicale, U1258, Illkirch, France
- Université de Strasbourg, Equipe labellisée Ligue Contre le Cancer, BP, Illkirch, France
| | - Arnaud Poterszman
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Integrated Structural Biology Department, Center for Integrated Structural Biology (CBI), Illkirch, France.
- Centre National de la Recherche Scientifique, UMR7104, Institut National de la Santé et de la Recherche Médicale, U1258, Illkirch, France.
- Université de Strasbourg, Equipe labellisée Ligue Contre le Cancer, BP, Illkirch, France.
| |
Collapse
|
4
|
Eltokhi A, Catterall WA, Gamal El-Din TM. Cell-cycle arrest at the G1/S boundary enhances transient voltage-gated ion channel expression in human and insect cells. CELL REPORTS METHODS 2023; 3:100559. [PMID: 37751687 PMCID: PMC10545908 DOI: 10.1016/j.crmeth.2023.100559] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Revised: 06/01/2023] [Accepted: 07/20/2023] [Indexed: 09/28/2023]
Abstract
Heterologous expression of recombinant ion channel subunits in cell lines is often limited by the presence of a low number of channels at the cell surface level. Here, we introduce a combination of two techniques: viral expression using the baculovirus system plus cell-cycle arrest at the G1/S boundary using either thymidine or hydroxyurea. This method achieved a manifold increase in the peak current density of expressed ion channels compared with the classical liposome-mediated transfection methods. The enhanced ionic current was accompanied by an increase in the density of gating charges, confirming that the increased yield of protein and ionic current reflects the functional localization of channels in the plasma membrane. This modified method of viral expression coordinated with the cell cycle arrest will pave the way to better decipher the structure and function of ion channels and their association with ion channelopathies.
Collapse
Affiliation(s)
- Ahmed Eltokhi
- Department of Pharmacology, University of Washington, Seattle, WA 98195-7280, USA.
| | - William A Catterall
- Department of Pharmacology, University of Washington, Seattle, WA 98195-7280, USA
| | - Tamer M Gamal El-Din
- Department of Pharmacology, University of Washington, Seattle, WA 98195-7280, USA.
| |
Collapse
|
5
|
de Mello RG, Bernardino TC, Guardalini LGO, Astray RM, Antoniazzi MM, Jared SGS, Núñez EGF, Jorge SAC. Zika virus-like particles (VLPs) produced in insect cells. Front Pharmacol 2023; 14:1181566. [PMID: 37377933 PMCID: PMC10291072 DOI: 10.3389/fphar.2023.1181566] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Accepted: 05/30/2023] [Indexed: 06/29/2023] Open
Abstract
Introdutcion: The Zika virus (ZIKV) infections are a healthcare concern mostly in the Americas, Africa, and Asia but have increased its endemicity area beyond these geographical regions. Due to the advances in infections by Zika virus, it is imperative to develop diagnostic and preventive tools against this viral agent. Virus-like particles (VLPs) appear as a suitable approach for use as antiviral vaccines. Methods: In this work, a methodology was established to produce virus-like particles containing the structural proteins, C, prM, and E of Zika virus produced in insect cells using the gene expression system derived from baculovirus. The vector pFast- CprME -ZIKV was constructed containing the gene sequences of Zika virus structural proteins and it was used to generate the recombinant bacmids (Bac- CprME -ZIKV) through transformation into DH10BacTM cells. The Bac- CprME -ZIKV was transfected in Spodoptera frugiperda (Sf9) insect cells and batches of BV- CprME -ZIKV were obtained by infection assays using a multiplicity of infection of 2. The Sf9 cells were infected, and the supernatant was collected 96 h post-infection. The expression of the CprME -ZIKV protein on the cell surface could be observed by immunochemical assays. To concentrate and purify virus-like particles, the sucrose and iodixanol gradients were evaluated, and the correct CprME -ZIKV proteins' conformation was evaluated by the Western blot assay. The virus-like particles were also analyzed and characterized by transmission electron microscopy. Results and discussion: Spherical structures like the native Zika virus from 50 to 65 nm containing the CprME -ZIKV proteins on their surface were observed in micrographs. The results obtained can be useful in the development path for a vaccine candidate against Zika virus.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Eutimio Gustavo Fernández Núñez
- Grupo de Engenharia de Bioprocessos, Escola de Artes, Ciências e Humanidades (EACH), Universidade de São Paulo, São Paulo, SP, Brazil
| | | |
Collapse
|
6
|
Hausjell CS, Ernst W, Grünwald-Gruber C, Arcalis E, Grabherr R. Quantitative proteomic analysis of extracellular vesicles in response to baculovirus infection of a Trichoplusia ni cell line. PLoS One 2023; 18:e0281060. [PMID: 36716331 PMCID: PMC9886248 DOI: 10.1371/journal.pone.0281060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Accepted: 01/16/2023] [Indexed: 02/01/2023] Open
Abstract
Due to its outstanding suitability to produce complex biopharmaceutical products including virus-like particles and subunit vaccines, the baculovirus/insect cell expression system has developed into a highly popular production platform in the biotechnological industry. For high productivity, virus-cell communication and an efficient spreading of the viral infection are crucial, and, in this context, extracellular vesicles (EVs) might play a significant role. EVs are small particles, utilized by cells to transfer biologically active compounds such as proteins, lipids as well as nucleic acids to recipient cells for intracellular communication. Studies in mammalian cells showed that the release of EVs is altered in response to infection with many viruses, ultimately either limiting or fostering infection spreading. In this study we isolated and characterized EVs, from both uninfected and baculovirus infected Tnms42 insect cells. Via quantitative proteomic analysis we identified more than 3000 T. ni proteins in Tnms42 cell derived EVs, of which more than 400 were significantly differentially abundant upon baculovirus infection. Subsequent gene set enrichment analysis revealed a depletion of proteins related to the extracellular matrix in EVs from infected cultures. Our findings show a significant change of EV protein cargo upon baculovirus infection, suggesting a major role of EVs as stress markers. Our study might serve in designing new tools for process monitoring and control to further improve biopharmaceutical production within the baculovirus/insect cell expression system.
Collapse
Affiliation(s)
- Christina Sophie Hausjell
- Department of Biotechnology, Institute of Molecular Biotechnology, University of Natural Resources and Life Sciences, Vienna, Austria
| | - Wolfgang Ernst
- Department of Biotechnology, Institute of Molecular Biotechnology, University of Natural Resources and Life Sciences, Vienna, Austria
| | - Clemens Grünwald-Gruber
- Core Facility Mass Spectrometry, University of Natural Resources and Life Sciences, Vienna, Austria
| | - Elsa Arcalis
- Department of Applied Genetics and Cell Biology, Institute of Plant Biotechnology and Cell Biology, University of Natural Resources and Life Sciences, Vienna, Austria
| | - Reingard Grabherr
- Department of Biotechnology, Institute of Molecular Biotechnology, University of Natural Resources and Life Sciences, Vienna, Austria
| |
Collapse
|
7
|
Zhang X, He A, Zong Y, Tian H, Zhang Z, Zhao K, Xu X, Chen H. Improvement of protein production in baculovirus expression vector system by removing a total of 10 kb of nonessential fragments from Autographa californica multiple nucleopolyhedrovirus genome. Front Microbiol 2023; 14:1171500. [PMID: 37125202 PMCID: PMC10133524 DOI: 10.3389/fmicb.2023.1171500] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Accepted: 03/24/2023] [Indexed: 05/02/2023] Open
Abstract
Baculovirus expression vector system (BEVS) is a powerful and versatile platform for recombinant protein production in insect cells. As the most frequently used baculovirus, Autographa californica multiple nucleopolyhedrovirus (AcMNPV) encodes 155 open reading frames (ORFs), including a considerable number of non-essential genes for the virus replication in cell culture. Studies have shown that protein production in BEVS can be improved by removing some viral dispensable genes, and these AcMNPV vectors also offer the possibility of accommodating larger exogenous gene fragments. In this study, we, respectively, deleted 14 DNA fragments from AcMNPV genome, each of them containing at least two contiguous genes that were known nonessential for viral replication in cell culture or functionally unknown. The effects of these fragment-deletions on virus replication and exogenous protein production were examined. The results showed that 11 of the 14 fragments, containing 43 genes, were dispensable for the virus replication in cultured cells. By detecting the expression of intracellularly expressed and secreted reporter proteins, we demonstrated that nine of the fragment-deletions benefited protein production in Sf9 cells and/or in High Five cells. After combining the deletion of some dispensable fragments, we obtained two AcMNPV vectors shortened by more than 10 kb but displayed an improved capacity for recombinant protein production. The deletion strategies used in this study has the potential to further improve the BEVS.
Collapse
|
8
|
Choque-Guevara R, Poma-Acevedo A, Montesinos-Millán R, Rios-Matos D, Gutiérrez-Manchay K, Montalvan-Avalos A, Quiñones-Garcia S, Cauti-Mendoza MDG, Agurto-Arteaga A, Ramirez-Ortiz I, Criollo-Orozco M, Huaccachi-Gonzales E, Romero YK, Perez-Martinez N, Isasi-Rivas G, Sernaque-Aguilar Y, Villanueva-Pérez D, Ygnacio F, Vallejos-Sánchez K, Fernández-Sánchez M, Guevara-Sarmiento LA, Fernández-Díaz M, Zimic M. Squalene in oil-based adjuvant improves the immunogenicity of SARS-CoV-2 RBD and confirms safety in animal models. PLoS One 2022; 17:e0269823. [PMID: 35998134 PMCID: PMC9397949 DOI: 10.1371/journal.pone.0269823] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Accepted: 05/30/2022] [Indexed: 01/09/2023] Open
Abstract
COVID-19 pandemic has accelerated the development of vaccines against its etiologic agent, SARS-CoV-2. However, the emergence of new variants of the virus lead to the generation of new alternatives to improve the current sub-unit vaccines in development. In the present report, the immunogenicity of the Spike RBD of SARS-CoV-2 formulated with an oil-in-water emulsion and a water-in-oil emulsion with squalene was evaluated in mice and hamsters. The RBD protein was expressed in insect cells and purified by chromatography until >95% purity. The protein was shown to have the appropriate folding as determined by ELISA and flow cytometry binding assays to its receptor, as well as by its detection by hamster immune anti-S1 sera under non-reducing conditions. In immunization assays, although the cellular immune response elicited by both adjuvants were similar, the formulation based in water-in-oil emulsion and squalene generated an earlier humoral response as determined by ELISA. Similarly, this formulation was able to stimulate neutralizing antibodies in hamsters. The vaccine candidate was shown to be safe, as demonstrated by the histopathological analysis in lungs, liver and kidney. These results have shown the potential of this formulation vaccine to be evaluated in a challenge against SARS-CoV-2 and determine its ability to confer protection.
Collapse
Affiliation(s)
| | | | | | - Dora Rios-Matos
- Laboratorios de investigación y desarrollo, FARVET SAC, Chincha, Ica, Perú
| | | | | | - Stefany Quiñones-Garcia
- Laboratorios de investigación y desarrollo, FARVET SAC, Chincha, Ica, Perú
- Laboratorio de Bioinformática, Biología Molecular y Desarrollos Tecnológicos, Laboratorios de Investigación y Desarrollo, Facultad de Ciencias y Filosofía, Universidad Peruana Cayetano Heredia, Lima, Peru
| | - Maria de Grecia Cauti-Mendoza
- Laboratorios de investigación y desarrollo, FARVET SAC, Chincha, Ica, Perú
- Laboratorio de Bioinformática, Biología Molecular y Desarrollos Tecnológicos, Laboratorios de Investigación y Desarrollo, Facultad de Ciencias y Filosofía, Universidad Peruana Cayetano Heredia, Lima, Peru
| | | | | | | | | | - Yomara K. Romero
- Laboratorio de Bioinformática, Biología Molecular y Desarrollos Tecnológicos, Laboratorios de Investigación y Desarrollo, Facultad de Ciencias y Filosofía, Universidad Peruana Cayetano Heredia, Lima, Peru
| | | | - Gisela Isasi-Rivas
- Laboratorios de investigación y desarrollo, FARVET SAC, Chincha, Ica, Perú
| | | | | | - Freddy Ygnacio
- Laboratorios de investigación y desarrollo, FARVET SAC, Chincha, Ica, Perú
| | - Katherine Vallejos-Sánchez
- Laboratorio de Bioinformática, Biología Molecular y Desarrollos Tecnológicos, Laboratorios de Investigación y Desarrollo, Facultad de Ciencias y Filosofía, Universidad Peruana Cayetano Heredia, Lima, Peru
| | | | | | | | - Mirko Zimic
- Laboratorios de investigación y desarrollo, FARVET SAC, Chincha, Ica, Perú
- Laboratorio de Bioinformática, Biología Molecular y Desarrollos Tecnológicos, Laboratorios de Investigación y Desarrollo, Facultad de Ciencias y Filosofía, Universidad Peruana Cayetano Heredia, Lima, Peru
- * E-mail:
| | | |
Collapse
|
9
|
Arya SK, Goodman CL, Stanley D, Palli SR. A database of crop pest cell lines. In Vitro Cell Dev Biol Anim 2022; 58:719-757. [PMID: 35994130 DOI: 10.1007/s11626-022-00710-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Accepted: 07/13/2022] [Indexed: 11/27/2022]
Abstract
We have developed an online database describing the known cell lines from Coleoptera, Diptera, Hemiptera, Hymenoptera, and Lepidoptera that were developed from agricultural pests. Cell line information has been primarily obtained from previous compilations of insect cell lines. We conducted in-depth Internet literature searches and drew on Internet sources such as the Cellosaurus database (https://web.expasy.org/cellosaurus/), and inventories from cell line depositories. Here, we report on a new database of insect cell lines, which covers 719 cell lines from 86 species. We have not included cell lines developed from Drosophila because they are already known from published databases, such as https://dgrc.bio.indiana.edu/cells/Catalog. We provide the designation, tissue and species of origin, cell line developer, unique characteristics, its use in various applications, publications, and patents, and, when known, insect virus susceptibility. This information has been assembled and organized into a searchable database available at the link https://entomology.ca.uky.edu/aginsectcellsdatabase which will be updated on an ongoing basis.
Collapse
Affiliation(s)
- Surjeet Kumar Arya
- Department of Entomology, College of Agriculture, Food and Environment, University of Kentucky, Lexington, KY, 40546, USA
| | - Cynthia L Goodman
- Biological Control of Insects Research Laboratory, United States Department of Agriculture, Agricultural Research Service, Columbia, Missouri, 65203, USA
| | - David Stanley
- Biological Control of Insects Research Laboratory, United States Department of Agriculture, Agricultural Research Service, Columbia, Missouri, 65203, USA
| | - Subba Reddy Palli
- Department of Entomology, College of Agriculture, Food and Environment, University of Kentucky, Lexington, KY, 40546, USA.
| |
Collapse
|
10
|
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: 3.0] [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
|
11
|
Correia R, Fernandes B, Castro R, Nagaoka H, Takashima E, Tsuboi T, Fukushima A, Viebig NK, Depraetere H, Alves PM, Roldão A. Asexual Blood-Stage Malaria Vaccine Candidate PfRipr5: Enhanced Production in Insect Cells. Front Bioeng Biotechnol 2022; 10:908509. [PMID: 35845392 PMCID: PMC9280424 DOI: 10.3389/fbioe.2022.908509] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Accepted: 05/09/2022] [Indexed: 01/02/2023] Open
Abstract
The malaria asexual blood-stage antigen PfRipr and its most immunogenic fragment PfRipr5 have recently risen as promising vaccine candidates against this infectious disease. Continued development of high-yielding, scalable production platforms is essential to advance the malaria vaccine research. Insect cells have supplied the production of numerous vaccine antigens in a fast and cost-effective manner; improving this platform further could prove key to its wider use. In this study, insect (Sf9 and High Five) and human (HEK293) cell hosts as well as process-optimizing strategies (new baculovirus construct designs and a culture temperature shift to hypothermic conditions) were employed to improve the production of the malaria asexual blood-stage vaccine candidate PfRipr5. Protein expression was maximized using High Five cells at CCI of 2 × 106 cell/mL and MOI of 0.1 pfu/cell (production yield = 0.49 mg/ml), with high-purity PfRipr5 binding to a conformational anti-PfRipr monoclonal antibody known to hold GIA activity and parasite PfRipr staining capacity. Further improvements in the PfRipr5 expression were achieved by designing novel expression vector sequences and performing a culture temperature shift to hypothermic culture conditions. Addition of one alanine (A) amino acid residue adjacent to the signal peptide cleavage site and a glycine-serine linker (GGSGG) between the PfRipr5 sequence and the purification tag (His6) induced a 2.2-fold increase in the expression of secreted PfRipr5 over using the expression vector with none of these additions. Performing a culture temperature shift from the standard 27–22°C at the time of infection improved the PfRipr5 expression by up to 1.7 fold. Notably, a synergistic effect was attained when combining both strategies, enabling to increase production yield post-purification by 5.2 fold, with similar protein quality (i.e., purity and binding to anti-PfRipr monoclonal antibody). This work highlights the potential of insect cells to produce the PfRipr5 malaria vaccine candidate and the importance of optimizing the expression vector and culture conditions to boost the expression of secreted proteins.
Collapse
Affiliation(s)
- 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
| | - Bárbara Fernandes
- 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
| | - Rute Castro
- IBET, Instituto de Biologia Experimental e Tecnológica, Oeiras, Portugal
| | - Hikaru Nagaoka
- Division of Malaria Research, Proteo-Science Center, Ehime University, Matsuyama, Japan
| | - Eizo Takashima
- Division of Malaria Research, Proteo-Science Center, Ehime University, Matsuyama, Japan
| | - Takafumi Tsuboi
- Division of Malaria Research, Proteo-Science Center, Ehime University, Matsuyama, Japan
| | | | - Nicola K. Viebig
- European Vaccine Initiative, UniversitätsKlinikum Heidelberg, Heidelberg, Germany
| | - Hilde Depraetere
- European Vaccine Initiative, UniversitätsKlinikum Heidelberg, Heidelberg, Germany
| | - 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
- *Correspondence: António Roldão,
| |
Collapse
|
12
|
Snead K, Wall V, Ambrose H, Esposito D, Drew M. Polycistronic baculovirus expression of SUGT1 enables high-yield production of recombinant leucine-rich repeat proteins and protein complexes. Protein Expr Purif 2022; 193:106061. [PMID: 35131438 PMCID: PMC8881745 DOI: 10.1016/j.pep.2022.106061] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Revised: 01/31/2022] [Accepted: 02/01/2022] [Indexed: 12/12/2022]
Abstract
The SHOC2-MRAS-PPP1CA (SMP) complex is a holoenzyme that plays a vital role in the MAP kinase signaling pathway. Previous attempts to produce this challenging three-protein complex have relied on co-infection with multiple viruses and the use of affinity tags to attempt to isolate functional recombinant protein complexes. Leucine-rich repeat containing proteins have been historically challenging to express, and we hypothesized that co-expression of appropriate chaperones may be necessary for optimal production. We describe here how the SUGT1 chaperone can, in conjunction with polycistronic protein expression in baculovirus-infected insect cells, dramatically enhance production yield and quality of recombinant SHOC2, the SMP complex, and other leucine-rich repeat proteins.
Collapse
Affiliation(s)
- Kelly Snead
- Protein Expression Laboratory, NCI RAS Initiative, Cancer Research Technology Program, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - Vanessa Wall
- Protein Expression Laboratory, NCI RAS Initiative, Cancer Research Technology Program, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - Hannah Ambrose
- Protein Expression Laboratory, NCI RAS Initiative, Cancer Research Technology Program, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - Dominic Esposito
- Protein Expression Laboratory, NCI RAS Initiative, Cancer Research Technology Program, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - Matthew Drew
- Protein Expression Laboratory, NCI RAS Initiative, Cancer Research Technology Program, Frederick National Laboratory for Cancer Research, Frederick, MD, USA.
| |
Collapse
|
13
|
Sullivan E, Sung PY, Wu W, Berry N, Kempster S, Ferguson D, Almond N, Jones IM, Roy P. SARS-CoV-2 Virus-like Particles Produced by a Single Recombinant Baculovirus Generate Anti-S Antibody and Protect against Variant Challenge. Viruses 2022; 14:914. [PMID: 35632656 PMCID: PMC9143203 DOI: 10.3390/v14050914] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Revised: 04/23/2022] [Accepted: 04/26/2022] [Indexed: 01/27/2023] Open
Abstract
Coronavirus Disease 2019 (COVID-19), caused by infection with Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2), has highlighted the need for the rapid generation of efficient vaccines for emerging disease. Virus-like particles, VLPs, are an established vaccine technology that produces virus-like mimics, based on expression of the structural proteins of a target virus. SARS-CoV-2 is a coronavirus where the basis of VLP formation has been shown to be the co-expression of the spike, membrane and envelope structural proteins. Here we describe the generation of SARS-CoV-2 VLPs by the co-expression of the salient structural proteins in insect cells using the established baculovirus expression system. VLPs were heterologous ~100 nm diameter enveloped particles with a distinct fringe that reacted strongly with SARS-CoV-2 convalescent sera. In a Syrian hamster challenge model, non-adjuvanted VLPs induced neutralizing antibodies to the VLP-associated Wuhan S protein and reduced virus shedding and protected against disease associated weight loss following a virulent challenge with SARS-CoV-2 (B.1.1.7 variant). Immunized animals showed reduced lung pathology and lower challenge virus replication than the non-immunized controls. Our data suggest SARS-CoV-2 VLPs offer an efficient vaccine that mitigates against virus load and prevents severe disease.
Collapse
Affiliation(s)
- Edward Sullivan
- Department of Pathogen Molecular Biology, London School of Hygiene and Tropical Medicine, London WC1E 7HT, UK; (E.S.); (P.-Y.S.); (W.W.)
| | - Po-Yu Sung
- Department of Pathogen Molecular Biology, London School of Hygiene and Tropical Medicine, London WC1E 7HT, UK; (E.S.); (P.-Y.S.); (W.W.)
| | - Weining Wu
- Department of Pathogen Molecular Biology, London School of Hygiene and Tropical Medicine, London WC1E 7HT, UK; (E.S.); (P.-Y.S.); (W.W.)
| | - Neil Berry
- Division of Infectious Disease Diagnostics, National Institute for Biological Standards and Control, Potters Bar EN6 3QG, UK; (N.B.); (S.K.); (D.F.); (N.A.)
| | - Sarah Kempster
- Division of Infectious Disease Diagnostics, National Institute for Biological Standards and Control, Potters Bar EN6 3QG, UK; (N.B.); (S.K.); (D.F.); (N.A.)
| | - Deborah Ferguson
- Division of Infectious Disease Diagnostics, National Institute for Biological Standards and Control, Potters Bar EN6 3QG, UK; (N.B.); (S.K.); (D.F.); (N.A.)
| | - Neil Almond
- Division of Infectious Disease Diagnostics, National Institute for Biological Standards and Control, Potters Bar EN6 3QG, UK; (N.B.); (S.K.); (D.F.); (N.A.)
| | - Ian M. Jones
- School of Biological Sciences, University of Reading, Reading RG6 6AH, UK;
| | - Polly Roy
- Department of Pathogen Molecular Biology, London School of Hygiene and Tropical Medicine, London WC1E 7HT, UK; (E.S.); (P.-Y.S.); (W.W.)
| |
Collapse
|
14
|
Ma C, Zhang X, Li X, Ding W, Feng Y. An embryonic cell line from the American cockroach Periplaneta americana L. (Blattaria: Blattidae) exhibits susceptibility to AcMNPV. In Vitro Cell Dev Biol Anim 2022; 58:278-288. [PMID: 35460045 DOI: 10.1007/s11626-021-00628-9] [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: 03/25/2021] [Accepted: 10/13/2021] [Indexed: 11/05/2022]
Abstract
Although the baculovirus expression vector system (BEVS) is widely used in the production of recombinant proteins, only a few lepidopteran insect cell lines have been successfully used so far. This study aimed at evaluating the characteristics of an embryonic cell line from the American cockroach Periplaneta americana L., RIRI-PA1, and determining whether it could be used in recombinant protein expression. Wild type Autographa californica multiple nucleopolyhedrovirus (AcMNPV-wt) and green fluorescent protein (GFP)-replicating recombinant baculoviruses (AcMNPV-GFP) were used to infect RIRI-PA1 respectively, demonstrating that RIRI-PA1 cells could be infected by AcMNPV and express recombinant proteins. Within 24 h of infection with AcMNPV-GFP, the GFP expression was higher than that in Sf21 cells. Furthermore, the infection of RIRI-PA1 cells increased gradually (multiplicity of infection, 10) within 24 h, while in Sf21 cells, the infection only began to increase within 48 h. However, after exposure for 96-168 h, the virus progeny and recombinant protein production of RIRI-PA1 cells was lower than those of Sf21 cells. Western blotting revealed that RIRI-PA1 cells could express recombinant GFP, and the protein expression level positively correlated with the multiplicity of infection. In conclusion, this is the first report that a cell line from P. americana has shown susceptibility to infection from a baculovirus and likewise express recombinant protein. Although the yield of recombinant GFP was not as high as that of Sf21 cells, the results nonetheless showed that RIRI-PA1 had an infection rate advantage in the short term (within 24 h of infection), which is of great value for further development and utilization.
Collapse
Affiliation(s)
- Chenjing Ma
- Key Laboratory of Breeding and Utilization of Resource Insects of National Forestry and Grassland Administration, Institute of Highland Forest Science, Chinese Academy of Forestry, Kunming, Yunnan Province, 650224, China
| | - Xin Zhang
- Key Laboratory of Breeding and Utilization of Resource Insects of National Forestry and Grassland Administration, Institute of Highland Forest Science, Chinese Academy of Forestry, Kunming, Yunnan Province, 650224, China.
| | - Xian Li
- Key Laboratory of Breeding and Utilization of Resource Insects of National Forestry and Grassland Administration, Institute of Highland Forest Science, Chinese Academy of Forestry, Kunming, Yunnan Province, 650224, China
| | - Weifeng Ding
- Key Laboratory of Breeding and Utilization of Resource Insects of National Forestry and Grassland Administration, Institute of Highland Forest Science, Chinese Academy of Forestry, Kunming, Yunnan Province, 650224, China
| | - Ying Feng
- Key Laboratory of Breeding and Utilization of Resource Insects of National Forestry and Grassland Administration, Institute of Highland Forest Science, Chinese Academy of Forestry, Kunming, Yunnan Province, 650224, China
| |
Collapse
|
15
|
Pichard S, Troffer-Charlier N, Kolb-Cheynel I, Poussin-Courmontagne P, Abdulrahman W, Birck C, Cura V, Poterszman A. Insect Cells-Baculovirus System for the Production of Difficult to Express Proteins: From Expression Screening for Soluble Constructs to Protein Quality Control. METHODS IN MOLECULAR BIOLOGY (CLIFTON, N.J.) 2022; 2406:281-317. [PMID: 35089564 DOI: 10.1007/978-1-0716-1859-2_17] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Rapid preparation of proteins for functional and structural analysis is a major challenge both in academia and industry. The number potential targets continuously increases and many are difficult to express proteins which, when produced in bacteria, result in insoluble and/or misfolded recombinant proteins, protein aggregates, or unusable low protein yield. We focus here on the baculovirus expression vector system which is now commonly used for heterologous production of human targets. This chapter describes simple and cost-effective protocols that enable iterative cycles of construct design, expression screening and optimization of protein production. We detail time- and cost-effective methods for generation of baculoviruses by homologous recombination and titer evaluation. Handling of insect cell cultures and preparation of bacmid for cotransfection are also presented.
Collapse
Affiliation(s)
- Simon Pichard
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Center for Integrated Structural Biology (CBI), Centre National de la Recherche Scientifique (CNRS), Institut National de la Santé et de la Recherche Médicale (INSERM), Université de Strasbourg, Illkirch, France
| | - Nathalie Troffer-Charlier
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Center for Integrated Structural Biology (CBI), Centre National de la Recherche Scientifique (CNRS), Institut National de la Santé et de la Recherche Médicale (INSERM), Université de Strasbourg, Illkirch, France
| | - Isabelle Kolb-Cheynel
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Center for Integrated Structural Biology (CBI), Centre National de la Recherche Scientifique (CNRS), Institut National de la Santé et de la Recherche Médicale (INSERM), Université de Strasbourg, Illkirch, France
| | - Pierre Poussin-Courmontagne
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Center for Integrated Structural Biology (CBI), Centre National de la Recherche Scientifique (CNRS), Institut National de la Santé et de la Recherche Médicale (INSERM), Université de Strasbourg, Illkirch, France
| | | | - Catherine Birck
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Center for Integrated Structural Biology (CBI), Centre National de la Recherche Scientifique (CNRS), Institut National de la Santé et de la Recherche Médicale (INSERM), Université de Strasbourg, Illkirch, France
| | - Vincent Cura
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Center for Integrated Structural Biology (CBI), Centre National de la Recherche Scientifique (CNRS), Institut National de la Santé et de la Recherche Médicale (INSERM), Université de Strasbourg, Illkirch, France
| | - Arnaud Poterszman
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Center for Integrated Structural Biology (CBI), Centre National de la Recherche Scientifique (CNRS), Institut National de la Santé et de la Recherche Médicale (INSERM), Université de Strasbourg, Illkirch, France.
| |
Collapse
|
16
|
Rivera-de-Torre E, Rimbault C, Jenkins TP, Sørensen CV, Damsbo A, Saez NJ, Duhoo Y, Hackney CM, Ellgaard L, Laustsen AH. Strategies for Heterologous Expression, Synthesis, and Purification of Animal Venom Toxins. Front Bioeng Biotechnol 2022; 9:811905. [PMID: 35127675 PMCID: PMC8811309 DOI: 10.3389/fbioe.2021.811905] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Accepted: 12/24/2021] [Indexed: 11/13/2022] Open
Abstract
Animal venoms are complex mixtures containing peptides and proteins known as toxins, which are responsible for the deleterious effect of envenomations. Across the animal Kingdom, toxin diversity is enormous, and the ability to understand the biochemical mechanisms governing toxicity is not only relevant for the development of better envenomation therapies, but also for exploiting toxin bioactivities for therapeutic or biotechnological purposes. Most of toxinology research has relied on obtaining the toxins from crude venoms; however, some toxins are difficult to obtain because the venomous animal is endangered, does not thrive in captivity, produces only a small amount of venom, is difficult to milk, or only produces low amounts of the toxin of interest. Heterologous expression of toxins enables the production of sufficient amounts to unlock the biotechnological potential of these bioactive proteins. Moreover, heterologous expression ensures homogeneity, avoids cross-contamination with other venom components, and circumvents the use of crude venom. Heterologous expression is also not only restricted to natural toxins, but allows for the design of toxins with special properties or can take advantage of the increasing amount of transcriptomics and genomics data, enabling the expression of dormant toxin genes. The main challenge when producing toxins is obtaining properly folded proteins with a correct disulfide pattern that ensures the activity of the toxin of interest. This review presents the strategies that can be used to express toxins in bacteria, yeast, insect cells, or mammalian cells, as well as synthetic approaches that do not involve cells, such as cell-free biosynthesis and peptide synthesis. This is accompanied by an overview of the main advantages and drawbacks of these different systems for producing toxins, as well as a discussion of the biosafety considerations that need to be made when working with highly bioactive proteins.
Collapse
Affiliation(s)
- Esperanza Rivera-de-Torre
- Department of Biotechnology and Biomedicine, Technical University of Denmark, Kongens Lyngby, Denmark
- *Correspondence: Esperanza Rivera-de-Torre, ; Andreas H. Laustsen,
| | - Charlotte Rimbault
- Department of Biotechnology and Biomedicine, Technical University of Denmark, Kongens Lyngby, Denmark
| | - Timothy P. Jenkins
- Department of Biotechnology and Biomedicine, Technical University of Denmark, Kongens Lyngby, Denmark
| | - Christoffer V. Sørensen
- Department of Biotechnology and Biomedicine, Technical University of Denmark, Kongens Lyngby, Denmark
| | - Anna Damsbo
- Department of Biotechnology and Biomedicine, Technical University of Denmark, Kongens Lyngby, Denmark
| | - Natalie J. Saez
- Institute for Molecular Bioscience, The University of Queensland, St Lucia, QLD, Australia
| | - Yoan Duhoo
- Institute for Molecular Bioscience, The University of Queensland, St Lucia, QLD, Australia
| | - Celeste Menuet Hackney
- Department of Biology, Linderstrøm-Lang Centre for Protein Science, University of Copenhagen, Copenhagen, Denmark
| | - Lars Ellgaard
- Department of Biology, Linderstrøm-Lang Centre for Protein Science, University of Copenhagen, Copenhagen, Denmark
| | - Andreas H. Laustsen
- Department of Biotechnology and Biomedicine, Technical University of Denmark, Kongens Lyngby, Denmark
- *Correspondence: Esperanza Rivera-de-Torre, ; Andreas H. Laustsen,
| |
Collapse
|
17
|
Xu W, Du S, Li T, Wu S, Jin N, Ren L, Li C. Generation and Evaluation of Recombinant Baculovirus Coexpressing GP5 and M Proteins of Porcine Reproductive and Respiratory Syndrome Virus Type 1. Viral Immunol 2021; 34:697-707. [PMID: 34935524 DOI: 10.1089/vim.2021.0018] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Porcine reproductive and respiratory syndrome virus (PRRSV) is the pathogen of the porcine reproductive and respiratory syndrome, which is one of the most economically devastating diseases of the swine industry. However, whether the inactivated vaccine and modified live attenuated vaccines are effective in disease control is still controversial. Although several groups developed PRRSV virus-like particles (VLPs) as a vaccine against PRRSV, all these VLP-based vaccines targeted PRRSV-2, but not PRRSV-1 or both. Therefore, it is urgent to produce VLPs against PRRSV-1. In this study, we rescued recombinant baculovirus expressing GP5 and M proteins of PRRSV-1 through the Bac-to-Bac® baculovirus expression system. Thereafter, PRRSV VLP was obtained efficiently in the recombinant baculovirus-infected High Five insect cells. Moreover, the PRRSV VLP and PRRSV VLP+A5 could efficiently trigger specific humoral immune responses and B cellular immune responses through intranasal immunization. The combination of PRRSV VLP and A5 adjuvant could improve the level of the immune response. The PRRSV-1 VLPs generated in this study have greater potential for vaccine development to control PRRSV-1 infection.
Collapse
Affiliation(s)
- Wang Xu
- College of Veterinary Medicine, Northwest A&F University, Yangling, China.,Research Unit of Key Technologies for Prevention and Control of Virus Zoonoses, Chinese Academy of Medical Sciences, Military Veterinary Institute, Academy of Military Medical Sciences, Changchun, China
| | - Shouwen Du
- Research Unit of Key Technologies for Prevention and Control of Virus Zoonoses, Chinese Academy of Medical Sciences, Military Veterinary Institute, Academy of Military Medical Sciences, Changchun, China.,Department of Infectious Diseases, Shenzhen People's Hospital, Second Clinical Hospital of Jinan University, Shenzhen, China
| | - Tiyuan Li
- Research Unit of Key Technologies for Prevention and Control of Virus Zoonoses, Chinese Academy of Medical Sciences, Military Veterinary Institute, Academy of Military Medical Sciences, Changchun, China
| | - Shipin Wu
- Research Unit of Key Technologies for Prevention and Control of Virus Zoonoses, Chinese Academy of Medical Sciences, Military Veterinary Institute, Academy of Military Medical Sciences, Changchun, China
| | - Ningyi Jin
- Research Unit of Key Technologies for Prevention and Control of Virus Zoonoses, Chinese Academy of Medical Sciences, Military Veterinary Institute, Academy of Military Medical Sciences, Changchun, China.,Jiangsu Co-Innovation Center for the Prevention and Control of Important Animal Infectious Disease and Zoonosis, Yangzhou University, Yangzhou, China
| | - Linzhu Ren
- Key Lab for Zoonoses Research, Ministry of Education, College of Animal Sciences, Jilin University, Changchun, China
| | - Chang Li
- Research Unit of Key Technologies for Prevention and Control of Virus Zoonoses, Chinese Academy of Medical Sciences, Military Veterinary Institute, Academy of Military Medical Sciences, Changchun, China
| |
Collapse
|
18
|
Membrane Protein Production and Purification from Escherichia coli and Sf9 Insect Cells. Methods Mol Biol 2021. [PMID: 33582985 DOI: 10.1007/978-1-0716-0724-4_1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/06/2023]
Abstract
A major obstacle to studying membrane proteins by biophysical techniques is the difficulty in producing sufficient amounts of materials for functional and structural studies. To overexpress the target membrane protein heterologously, especially an eukaryotic protein, a key step is to find the optimal host expression system and perform subsequent expression optimization. In this chapter, we describe protocols for screening membrane protein production using bacterial and insect cells, solubilization screening, large-scale production, and commonly used affinity chromatography purification methods. We discuss general optimization conditions, such as promoters and tags, and describe current techniques that can be used in any laboratory without specialized expensive equipment. Especially for insect cells, GFP fusions are particularly useful for localization and in-gel fluorescence detection of the proteins on SDS-PAGE. We give detailed protocols that can be used to screen the best expression and purification conditions for membrane protein study.
Collapse
|
19
|
Joshi PRH, Venereo-Sanchez A, Chahal PS, Kamen AA. Advancements in molecular design and bioprocessing of recombinant adeno-associated virus gene delivery vectors using the insect-cell baculovirus expression platform. Biotechnol J 2021; 16:e2000021. [PMID: 33277815 DOI: 10.1002/biot.202000021] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2020] [Revised: 11/27/2020] [Indexed: 01/23/2023]
Abstract
Despite rapid progress in the field, scalable high-yield production of adeno-associated virus (AAV) is still one of the critical bottlenecks the manufacturing sector is facing. The insect cell-baculovirus expression vector system (IC-BEVS) has emerged as a mainstream platform for the scalable production of recombinant proteins with clinically approved products for human use. In this review, we provide a detailed overview of the advancements in IC-BEVS for rAAV production. Since the first report of baculovirus-induced production of rAAV vector in insect cells in 2002, this platform has undergone significant improvements, including enhanced stability of Bac-vector expression and a reduced number of baculovirus-coinfections. The latter streamlining strategy led to the eventual development of the Two-Bac, One-Bac, and Mono-Bac systems. The one baculovirus system consisting of an inducible packaging insect cell line was further improved to enhance the AAV vector quality and potency. In parallel, the implementation of advanced manufacturing approaches and control of critical processing parameters have demonstrated promising results with process validation in large-scale bioreactor runs. Moreover, optimization of the molecular design of vectors to enable higher cell-specific yields of functional AAV particles combined with bioprocess intensification strategies may also contribute to addressing current and future manufacturing challenges.
Collapse
Affiliation(s)
- Pranav R H Joshi
- Department of Bioengineering, McGill University, Montréal, Quebec, Canada
| | | | - Parminder S Chahal
- Human Health Therapeutics Portfolio, National Research Council of Canada, Montréal, Quebec, Canada
| | - Amine A Kamen
- Department of Bioengineering, McGill University, Montréal, Quebec, Canada
| |
Collapse
|
20
|
Gröhn S, Heinimäki S, Tamminen K, Blazevic V. Expression of influenza A virus-derived peptides on a rotavirus VP6-based delivery platform. Arch Virol 2021; 166:213-217. [PMID: 33067651 PMCID: PMC7567002 DOI: 10.1007/s00705-020-04847-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2020] [Accepted: 09/09/2020] [Indexed: 12/11/2022]
Abstract
Recombinant protein technology enables the engineering of modern vaccines composed of a carrier protein displaying poorly immunogenic heterologous antigens. One promising carrier is based on the rotavirus inner-capsid VP6 protein. We explored different VP6 insertion sites for the presentation of two peptides (23 and 140 amino acids) derived from the M2 and HA genes of influenza A virus. Both termini and three surface loops of VP6 were successfully exploited as genetic fusion sites, as demonstrated by the expression of the fusion proteins. However, further studies are needed to assess the morphology and immunogenicity of these constructs.
Collapse
Affiliation(s)
- Stina Gröhn
- Faculty of Medicine and Health Technology, Vaccine Development and Immunology/Vaccine Research Center, Tampere University, Arvo Ylpön katu 34, FI-33520 Tampere, Finland
| | - Suvi Heinimäki
- Faculty of Medicine and Health Technology, Vaccine Development and Immunology/Vaccine Research Center, Tampere University, Arvo Ylpön katu 34, FI-33520 Tampere, Finland
| | - Kirsi Tamminen
- Faculty of Medicine and Health Technology, Vaccine Development and Immunology/Vaccine Research Center, Tampere University, Arvo Ylpön katu 34, FI-33520 Tampere, Finland
| | - Vesna Blazevic
- Faculty of Medicine and Health Technology, Vaccine Development and Immunology/Vaccine Research Center, Tampere University, Arvo Ylpön katu 34, FI-33520 Tampere, Finland
| |
Collapse
|
21
|
Villar-Fernández MA, Cardoso da Silva R, Firlej M, Pan D, Weir E, Sarembe A, Raina VB, Bange T, Weir JR, Vader G. Biochemical and functional characterization of a meiosis-specific Pch2/ORC AAA+ assembly. Life Sci Alliance 2020; 3:3/11/e201900630. [PMID: 32826290 PMCID: PMC7442955 DOI: 10.26508/lsa.201900630] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Revised: 08/13/2020] [Accepted: 08/13/2020] [Indexed: 12/14/2022] Open
Abstract
The AAA+ protein Pch2 forms a biochemical complex with Orc1/ORC to suppress DNA break formation in the meiotic G2/prophase. Pch2 is a meiosis-specific AAA+ protein that controls several important chromosomal processes. We previously demonstrated that Orc1, a subunit of the ORC, functionally interacts with budding yeast Pch2. The ORC (Orc1-6) AAA+ complex loads the AAA+ MCM helicase to origins of replication, but whether and how ORC collaborates with Pch2 remains unclear. Here, we show that a Pch2 hexamer directly associates with ORC during the meiotic G2/prophase. Biochemical analysis suggests that Pch2 uses its non-enzymatic NH2-terminal domain and AAA+ core and likely engages the interface of ORC that also binds to Cdc6, a factor crucial for ORC-MCM binding. Canonical ORC function requires association with origins, but we show here that despite causing efficient removal of Orc1 from origins, nuclear depletion of Orc2 and Orc5 does not trigger Pch2/Orc1-like meiotic phenotypes. This suggests that the function for Orc1/Pch2 in meiosis can be executed without efficient association of ORC with origins of replication. In conclusion, we uncover distinct functionalities for Orc1/ORC that drive the establishment of a non-canonical, meiosis-specific AAA+ assembly with Pch2.
Collapse
Affiliation(s)
- María Ascensión Villar-Fernández
- Department of Mechanistic Cell Biology, Max Planck Institute of Molecular Physiology, Dortmund, Germany.,International Max Planck Research School in Chemical and Molecular Biology, Max Planck Institute of Molecular Physiology, Dortmund, Germany
| | - Richard Cardoso da Silva
- Department of Mechanistic Cell Biology, Max Planck Institute of Molecular Physiology, Dortmund, Germany
| | | | - Dongqing Pan
- Department of Mechanistic Cell Biology, Max Planck Institute of Molecular Physiology, Dortmund, Germany
| | - Elisabeth Weir
- Department of Mechanistic Cell Biology, Max Planck Institute of Molecular Physiology, Dortmund, Germany
| | - Annika Sarembe
- Department of Mechanistic Cell Biology, Max Planck Institute of Molecular Physiology, Dortmund, Germany
| | - Vivek B Raina
- Department of Mechanistic Cell Biology, Max Planck Institute of Molecular Physiology, Dortmund, Germany.,International Max Planck Research School in Chemical and Molecular Biology, Max Planck Institute of Molecular Physiology, Dortmund, Germany
| | - Tanja Bange
- Department of Mechanistic Cell Biology, Max Planck Institute of Molecular Physiology, Dortmund, Germany
| | - John R Weir
- Department of Mechanistic Cell Biology, Max Planck Institute of Molecular Physiology, Dortmund, Germany.,Friedrich Miescher Laboratory, Tübingen, Germany
| | - Gerben Vader
- Department of Mechanistic Cell Biology, Max Planck Institute of Molecular Physiology, Dortmund, Germany
| |
Collapse
|
22
|
Maghodia AB, Geisler C, Jarvis DL. A new nodavirus-negative Trichoplusia ni cell line for baculovirus-mediated protein production. Biotechnol Bioeng 2020; 117:3248-3264. [PMID: 32662870 DOI: 10.1002/bit.27494] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Revised: 05/21/2020] [Accepted: 07/12/2020] [Indexed: 12/22/2022]
Abstract
Cell lines derived from Trichoplusia ni (Tn) are widely used as hosts in the baculovirus-insect cell system (BICS). One advantage of Tn cell lines is they can produce recombinant proteins at higher levels than cell lines derived from other insects. However, Tn cell lines are persistently infected with an alphanodavirus, Tn5 cell-line virus (TnCLV), which reduces their utility as a host for the BICS. Several groups have isolated TnCLV-negative Tn cell lines, but none were thoroughly characterized and shown to be free of other adventitious viruses. Thus, we isolated and extensively characterized a new TnCLV-negative line, Tn-nodavirus-negative (Tn-NVN). Tn-NVN cells have no detectable TnCLV, no other previously identified viral contaminants of lepidopteran insect cell lines, and no sequences associated with any replicating virus or other viral adventitious agents. Tn-NVN cells tested negative for >60 species of Mycoplasma, Acholeplasma, Spiroplasma, and Ureaplasma. Finally, Tn-NVN cells grow well as a single-cell suspension culture in serum-free medium, produce recombinant proteins at levels similar to High Five™ cells, and do not produce recombinant glycoproteins with immunogenic core α1,3-fucosylation. Thus, Tn-NVN is a new, well-characterized TnCLV-negative cell line with several other features enhancing its utility as a host for the BICS.
Collapse
Affiliation(s)
| | | | - Donald L Jarvis
- GlycoBac, LLC, Laramie, Wyoming.,Department of Molecular Biology, University of Wyoming, Laramie, Wyoming
| |
Collapse
|
23
|
Puente-Massaguer E, Saccardo P, Ferrer-Miralles N, Lecina M, Gòdia F. Coupling Microscopy and Flow Cytometry for a Comprehensive Characterization of Nanoparticle Production in Insect Cells. Cytometry A 2020; 97:921-932. [PMID: 32515126 DOI: 10.1002/cyto.a.24033] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2019] [Revised: 03/27/2020] [Accepted: 04/01/2020] [Indexed: 01/01/2023]
Abstract
Advancements in the field of characterization techniques have broadened the opportunities to deepen into nanoparticle production bioprocesses. Gag-based virus-like particles (VLPs) have shown their potential as candidates for recombinant vaccine development. However, comprehensive characterization of the production process is still a requirement to meet the desired critical quality attributes. In this work, the production process of Gag VLPs by baculovirus (BV) infection in the reference High Five and Sf9 insect cell lines is characterized in detail. To this end, the Gag polyprotein was fused in frame to the enhanced green fluorescent protein (eGFP) to favor process evaluation with multiple analytical tools. Tracking of the infection process using confocal microscopy and flow cytometry revealed a pronounced increase in the complexity of High Five over Sf9 cells. Cryogenic transmission electron microscopy (cryo-TEM) characterization determined that changes in cell complexity could be attributed to the presence of occlusion-derived BV in High Five cells, whereas Sf9 cells evidenced a larger proportion of the budded virus phenotype (23-fold). Initial evaluation of the VLP production process using spectrofluorometry showed that higher levels of the Gag-eGFP polyprotein were obtained in High Five cells (3.6-fold). However, comparative analysis based on nanoparticle quantification by flow virometry and nanoparticle tracking analysis (NTA) proved that Sf9 cells were 1.7- and 1.5-fold more productive in terms of assembled VLPs, respectively. Finally, analytical ultracentrifugation coupled to flow virometry evidenced a larger sedimentation coefficient of High Five-derived VLPs, indicating a possible interaction with other cellular compounds. Taken together, these results highlight the combined use of microscopy and flow cytometry techniques to improve vaccine development processes using the insect cell/BV expression vector system. © 2020 International Society for Advancement of Cytometry.
Collapse
Affiliation(s)
- Eduard Puente-Massaguer
- Departament d'Enginyeria Química, Biològica i Ambiental, Escola d'Enginyeria, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Barcelona, Spain
| | - Paolo Saccardo
- Plataforma de Producción de Proteínas, CIBER de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Barcelona, Spain
| | - Neus Ferrer-Miralles
- Plataforma de Producción de Proteínas, CIBER de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Barcelona, Spain
| | - Martí Lecina
- IQS School of Engineering, Universitat Ramón Llull, Barcelona, Spain
| | - Francesc Gòdia
- Departament d'Enginyeria Química, Biològica i Ambiental, Escola d'Enginyeria, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Barcelona, Spain
| |
Collapse
|
24
|
Rowland RJ, Wu L, Liu F, Davies GJ. A baculoviral system for the production of human β-glucocerebrosidase enables atomic resolution analysis. Acta Crystallogr D Struct Biol 2020; 76:565-580. [PMID: 32496218 PMCID: PMC7271948 DOI: 10.1107/s205979832000501x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Accepted: 04/09/2020] [Indexed: 11/18/2022] Open
Abstract
The lysosomal glycoside hydrolase β-glucocerebrosidase (GBA; sometimes called GBA1 or GCase) catalyses the hydrolysis of glycosphingolipids. Inherited deficiencies in GBA cause the lysosomal storage disorder Gaucher disease (GD). Consequently, GBA is of considerable medical interest, with continuous advances in the development of inhibitors, chaperones and activity-based probes. The development of new GBA inhibitors requires a source of active protein; however, the majority of structural and mechanistic studies of GBA today rely on clinical enzyme-replacement therapy (ERT) formulations, which are incredibly costly and are often difficult to obtain in adequate supply. Here, the production of active crystallizable GBA in insect cells using a baculovirus expression system is reported, providing a nonclinical source of recombinant GBA with comparable activity and biophysical properties to ERT preparations. Furthermore, a novel crystal form of GBA is described which diffracts to give a 0.98 Å resolution unliganded structure. A structure in complex with the inactivator 2,4-dinitrophenyl-2-deoxy-2-fluoro-β-D-glucopyranoside was also obtained, demonstrating the ability of this GBA formulation to be used in ligand-binding studies. In light of its purity, stability and activity, the GBA production protocol described here should circumvent the need for ERT formulations for structural and biochemical studies and serve to support GD research.
Collapse
Affiliation(s)
- Rhianna J. Rowland
- Department of Chemistry, York Structural Biology Laboratory, University of York, Heslington, York YO10 5DD, United Kingdom
| | - Liang Wu
- Department of Chemistry, York Structural Biology Laboratory, University of York, Heslington, York YO10 5DD, United Kingdom
| | - Feng Liu
- Department of Chemistry, University of British Columbia, Vancouver, British Columbia V6T 1Z1, Canada
| | - Gideon J. Davies
- Department of Chemistry, York Structural Biology Laboratory, University of York, Heslington, York YO10 5DD, United Kingdom
| |
Collapse
|
25
|
Puente-Massaguer E, Lecina M, Gòdia F. Integrating nanoparticle quantification and statistical design of experiments for efficient HIV-1 virus-like particle production in High Five cells. Appl Microbiol Biotechnol 2020; 104:1569-1582. [PMID: 31907573 PMCID: PMC7224031 DOI: 10.1007/s00253-019-10319-x] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2019] [Revised: 12/04/2019] [Accepted: 12/13/2019] [Indexed: 02/06/2023]
Abstract
The nature of enveloped virus-like particles (VLPs) has triggered high interest in their application to different research fields, including vaccine development. The baculovirus expression vector system (BEVS) has been used as an efficient platform for obtaining large amounts of these complex nanoparticles. To date, most of the studies dealing with VLP production by recombinant baculovirus infection utilize indirect detection or quantification techniques that hinder the appropriate characterization of the process and product. Here, we propose the application of cutting-edge quantification methodologies in combination with advanced statistical designs to exploit the full potential of the High Five/BEVS as a platform to produce HIV-1 Gag VLPs. The synergies between CCI, MOI, and TOH were studied using a response surface methodology approach on four different response functions: baculovirus infection, VLP production, VLP assembly, and VLP productivity. TOH and MOI proved to be the major influencing factors in contrast with previous reported data. Interestingly, a remarkable competition between Gag VLP production and non-assembled Gag was detected. Also, the use of nanoparticle tracking analysis and flow virometry revealed the existence of remarkable quantities of extracellular vesicles. The different responses of the study were combined to determine two global optimum conditions, one aiming to maximize the VLP titer (quantity) and the second aiming to find a compromise between VLP yield and the ratio of assembled VLPs (quality). This study provides a valuable approach to optimize VLP production and demonstrates that the High Five/BEVS can support mass production of Gag VLPs and potentially other complex nanoparticles.
Collapse
Affiliation(s)
- Eduard Puente-Massaguer
- Departament d'Enginyeria Química, Biològica i Ambiental, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Barcelona, Spain.
| | - Martí Lecina
- IQS School of Engineering, Universitat Ramón Llull, Barcelona, Spain
| | - Francesc Gòdia
- Departament d'Enginyeria Química, Biològica i Ambiental, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Barcelona, Spain
| |
Collapse
|
26
|
Durous L, Rosa-Calatrava M, Petiot E. Advances in influenza virus-like particles bioprocesses. Expert Rev Vaccines 2019; 18:1285-1300. [DOI: 10.1080/14760584.2019.1704262] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Affiliation(s)
- Laurent Durous
- Virologie et Pathologie Humaine - VirPath team - Centre International de Recherche en Infectiologie (CIRI), INSERM U1111, CNRS UMR5308, ENS Lyon, Université Claude Bernard Lyon 1, Université de Lyon, Lyon, France
| | - Manuel Rosa-Calatrava
- Virologie et Pathologie Humaine - VirPath team - Centre International de Recherche en Infectiologie (CIRI), INSERM U1111, CNRS UMR5308, ENS Lyon, Université Claude Bernard Lyon 1, Université de Lyon, Lyon, France
- VirNext, Faculté de Médecine RTH Laennec, Université Claude Bernard Lyon 1, Université de Lyon, Lyon, France
| | - Emma Petiot
- Virologie et Pathologie Humaine - VirPath team - Centre International de Recherche en Infectiologie (CIRI), INSERM U1111, CNRS UMR5308, ENS Lyon, Université Claude Bernard Lyon 1, Université de Lyon, Lyon, France
| |
Collapse
|
27
|
An illustration of optimal selected glycosidase for N-glycoproteins deglycosylation and crystallization. Int J Biol Macromol 2019; 122:265-271. [DOI: 10.1016/j.ijbiomac.2018.10.138] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2018] [Revised: 10/02/2018] [Accepted: 10/18/2018] [Indexed: 01/11/2023]
|
28
|
Aksular M, Calvo-Pinilla E, Marín-López A, Ortego J, Chambers AC, King LA, Castillo-Olivares J. A single dose of African horse sickness virus (AHSV) VP2 based vaccines provides complete clinical protection in a mouse model. Vaccine 2018; 36:7003-7010. [PMID: 30309744 PMCID: PMC6219453 DOI: 10.1016/j.vaccine.2018.09.065] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2018] [Revised: 09/06/2018] [Accepted: 09/27/2018] [Indexed: 11/24/2022]
Abstract
Baculovirus-expressed AHS-VP2 and MVA-VP2 vaccines were evaluated in mice. Clinical protection was complete in mice receiving one or two doses of MVA-VP2. Clinical protection complete after two doses of baculovirus-expressed VP2. Significant reduction of viraemia in all vaccinated groups. Significant levels of immunity were achieved with one dose of either vaccine.
African horse sickness is a severe, often fatal, arboviral disease of equids. The control of African horse sickness virus (AHSV) in endemic countries is based currently on the use of live attenuated vaccines despite some biosafety concerns derived from its biological properties. Thus, experimental vaccination platforms have been developed over the years in order to avoid the biosafety concerns associated with the use of attenuated vaccines. Various studies showed that baculovirus-expressed AHSV-VP2 or modified Vaccinia Ankara virus expressing AHSV-VP2 (MVA-VP2) induced virus neutralising antibodies and protective immunity in small animals and horses. AHSV is an antigenically diverse pathogen and immunity against AHS is serotype-specific. Therefore, AHS vaccines for use in endemic countries need to induce an immune response capable of protecting against all existing serotypes. For this reason, current live attenuated vaccines are administered as polyvalent preparations comprising combinations of AHSV attenuated strains of different serotypes. Previous studies have shown that it is possible to induce cross-reactive virus neutralising antibodies against different serotypes of AHSV by using polyvalent vaccines comprising combinations of either different serotype-specific VP2 proteins, or MVA-VP2 viruses. However, these strategies could be difficult to implement if induction of protective immunity is highly dependent on using a two-dose vaccination regime for each serotype the vaccine intends to protect against. In our study, we have tested the protective capacity of MVA-VP2 and baculovirus-expressed VP2 vaccines when a single dose was used. Groups of interferon alpha receptor knock-out mice were inoculated with either MVA-VP2 or baculovirus-expressed VP2 vaccines using one dose or the standard two-dose vaccination regime. After vaccination, all four vaccinated groups were challenged with AHSV and clinical responses, lethality and viraemia compared between the groups. Our results show that complete clinical protection was achieved after a single vaccination with either MVA-VP2 or baculovirus sub-unit VP2 vaccines.
Collapse
Affiliation(s)
- Mine Aksular
- Department of Biological & Medical Sciences, Oxford Brookes University, Oxford OX3 0BP, UK; Oxford Expression Technologies Ltd, Gipsy Lane, Oxford OX3 0BP, UK; The Pirbright Institute, Ash Road, Woking, Surrey GU24 0NB, UK
| | | | | | | | - Adam C Chambers
- Oxford Expression Technologies Ltd, Gipsy Lane, Oxford OX3 0BP, UK
| | - Linda A King
- Department of Biological & Medical Sciences, Oxford Brookes University, Oxford OX3 0BP, UK
| | - Javier Castillo-Olivares
- Department of Biological & Medical Sciences, Oxford Brookes University, Oxford OX3 0BP, UK; The Pirbright Institute, Ash Road, Woking, Surrey GU24 0NB, UK; Department of Veterinary Medicine, University of Cambridge, Madingley Road, CB3 0ES Cambridge, UK.
| |
Collapse
|
29
|
Weidner T, Druzinec D, Mühlmann M, Buchholz R, Czermak P. The components of shear stress affecting insect cells used with the baculovirus expression vector system. ACTA ACUST UNITED AC 2018; 72:429-439. [PMID: 28822988 DOI: 10.1515/znc-2017-0066] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2017] [Accepted: 07/17/2017] [Indexed: 02/07/2023]
Abstract
Insect-based expression platforms such as the baculovirus expression vector system (BEVS) are widely used for the laboratory- and industrial-scale production of recombinant proteins. Thereby, major drawbacks to gain high-quality proteins are the lytic infection cycle and the shear sensitivity of infected insect cells due to turbulence and aeration. Smaller bubbles were formerly assumed to be more harmful than larger ones, but we found that cell damage is also dependent on the concentration of protective agents such as Pluronic®. At the appropriate concentration, Pluronic forms a layer around air bubbles and hinders the attachment of cells, thus limiting the damage. In this context, we used microaeration to vary bubble sizes and confirmed that size is not the most important factor, but the total gas surface area in the reactor is. If the surface area exceeds a certain threshold, the concentration of Pluronic is no longer sufficient for cell protection. To investigate the significance of shear forces, a second study was carried out in which infected insect cells were cultivated in a hollow fiber module to protect them from shear forces. Both model studies revealed important aspects of the design and scale-up of BEVS processes for the production of recombinant proteins.
Collapse
|
30
|
Koczka K, Peters P, Ernst W, Himmelbauer H, Nika L, Grabherr R. Comparative transcriptome analysis of a Trichoplusia ni cell line reveals distinct host responses to intracellular and secreted protein products expressed by recombinant baculoviruses. J Biotechnol 2018; 270:61-69. [PMID: 29432775 DOI: 10.1016/j.jbiotec.2018.02.001] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2017] [Revised: 01/12/2018] [Accepted: 02/02/2018] [Indexed: 12/17/2022]
Abstract
The baculovirus insect cell expression system has become a firmly established production platform in biotechnology. Various complex proteins, multi-subunit particles including veterinary and human vaccines are manufactured with this system on a commercial scale. Apart from baculovirus infected Spodoptera frugiperda (Sf9) cells, the Trichoplusia ni (HighFive) cell line is alternatively used as host organism. In this study, we explored the protein production capabilities of Tnms42 insect cells, a new derivative of HighFive, which is free of latent nodavirus infection. As a model system, a cytosolic (mCherry) and a secreted (hemagglutinin) protein were overexpressed in Tnms42 cells. The response of the host cells was followed in a time course experiment over the infection cycle by comparative transcriptome analysis (RNA-seq). As expected, the baculovirus infection per se had a massive impact on the host cell transcriptome, which was observed by the huge total number of differentially expressed transcripts (>14,000). Despite this severe overall cellular reaction, a specific response could be clearly attributed to the overexpression of secreted hemagglutinin, revealing limits in the secretory capacity of the host cell. About 400 significantly regulated transcripts were identified and assigned to biochemical pathways and gene ontology (GO) categories, all related to protein processing, folding and response to unfolded protein. The identification of relevant target genes will serve to design specific virus engineering concepts for improving the yield of proteins that are dependent on the secretory pathway.
Collapse
Affiliation(s)
- Krisztina Koczka
- Department of Biotechnology, University of Natural Resources and Life Sciences, Muthgasse 18, 1190 Vienna, Austria; acib - Austrian Centre of Industrial Biotechnology, Muthgasse 11, 1190 Vienna, Austria.
| | - Philipp Peters
- Department of Biotechnology, University of Natural Resources and Life Sciences, Muthgasse 18, 1190 Vienna, Austria; acib - Austrian Centre of Industrial Biotechnology, Muthgasse 11, 1190 Vienna, Austria.
| | - Wolfgang Ernst
- Department of Biotechnology, University of Natural Resources and Life Sciences, Muthgasse 18, 1190 Vienna, Austria; acib - Austrian Centre of Industrial Biotechnology, Muthgasse 11, 1190 Vienna, Austria.
| | - Heinz Himmelbauer
- Department of Biotechnology, University of Natural Resources and Life Sciences, Muthgasse 18, 1190 Vienna, Austria; acib - Austrian Centre of Industrial Biotechnology, Muthgasse 11, 1190 Vienna, Austria.
| | - Lisa Nika
- Department of Biotechnology, University of Natural Resources and Life Sciences, Muthgasse 18, 1190 Vienna, Austria.
| | - Reingard Grabherr
- Department of Biotechnology, University of Natural Resources and Life Sciences, Muthgasse 18, 1190 Vienna, Austria; acib - Austrian Centre of Industrial Biotechnology, Muthgasse 11, 1190 Vienna, Austria.
| |
Collapse
|
31
|
Clarke EC, Collar AL, Ye C, Caì Y, Anaya E, Rinaldi D, Martinez B, Yarborough S, Merle C, Theisen M, Wada J, Kuhn JH, Bradfute SB. Production and Purification of Filovirus Glycoproteins in Insect and Mammalian Cell Lines. Sci Rep 2017; 7:15091. [PMID: 29118454 PMCID: PMC5678155 DOI: 10.1038/s41598-017-15416-3] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2017] [Accepted: 10/26/2017] [Indexed: 01/10/2023] Open
Abstract
Filoviruses are highly virulent pathogens capable of causing severe disease. The glycoproteins of filoviruses are the only virally expressed proteins on the virion surface and are required for receptor binding. As such, they are the main candidate vaccine antigen. Despite their virulence, most filoviruses are not comprehensively characterized, and relatively few commercially produced reagents are available for their study. Here, we describe two methods for production and purification of filovirus glycoproteins in insect and mammalian cell lines. Considerations of expression vector choice, modifications to sequence, troubleshooting of purification method, and glycosylation differences are all important for successful expression of filovirus glycoproteins in cell lines. Given the scarcity of commercially available filovirus glycoproteins, we hope our experiences with possible difficulties in purification of the proteins will facilitate other researchers to produce and purify filovirus glycoproteins rapidly.
Collapse
Affiliation(s)
- Elizabeth C Clarke
- Center for Global Health, Division of Infectious Diseases, Department of Internal Medicine, University of New Mexico, Albuquerque, New Mexico, 87131, USA
| | - Amanda L Collar
- Center for Global Health, Division of Infectious Diseases, Department of Internal Medicine, University of New Mexico, Albuquerque, New Mexico, 87131, USA
| | - Chunyan Ye
- Center for Global Health, Division of Infectious Diseases, Department of Internal Medicine, University of New Mexico, Albuquerque, New Mexico, 87131, USA
| | - Yíngyún Caì
- Integrated Research Facility at Fort Detrick, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Frederick, Maryland, 21702, USA
| | - Eduardo Anaya
- Department of Pathology, University of New Mexico, Albuquerque, New Mexico, 87131, USA
| | - Derek Rinaldi
- Department of Pathology, University of New Mexico, Albuquerque, New Mexico, 87131, USA
| | - Britney Martinez
- Undergraduate Pipeline Network, University of New Mexico, Albuquerque, New Mexico, 87131, USA
| | - Sarah Yarborough
- Undergraduate Pipeline Network, University of New Mexico, Albuquerque, New Mexico, 87131, USA
| | | | | | - Jiro Wada
- Integrated Research Facility at Fort Detrick, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Frederick, Maryland, 21702, USA
| | - Jens H Kuhn
- Integrated Research Facility at Fort Detrick, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Frederick, Maryland, 21702, USA
| | - Steven B Bradfute
- Center for Global Health, Division of Infectious Diseases, Department of Internal Medicine, University of New Mexico, Albuquerque, New Mexico, 87131, USA.
| |
Collapse
|
32
|
Shang H, Garretson TA, Kumar CS, Dieter RF, Cheng XW. Improved pFastBac™ donor plasmid vectors for higher protein production using the Bac-to-Bac® baculovirus expression vector system. J Biotechnol 2017. [DOI: 10.1016/j.jbiotec.2017.06.397] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
|
33
|
Hashimoto Y, Macri D, Srivastava I, McPherson C, Felberbaum R, Post P, Cox M. Complete study demonstrating the absence of rhabdovirus in a distinct Sf9 cell line. PLoS One 2017; 12:e0175633. [PMID: 28423032 PMCID: PMC5397025 DOI: 10.1371/journal.pone.0175633] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2016] [Accepted: 03/29/2017] [Indexed: 12/26/2022] Open
Abstract
A putative novel rhabdovirus (SfRV) was previously identified in a Spodoptera frugiperda cell line (Sf9 cells [ATCC CRL-1711 lot 58078522]) by next generation sequencing and extensive bioinformatic analysis. We performed an extensive analysis of our Sf9 cell bank (ATCC CRL-1711 lot 5814 [Sf9L5814]) to determine whether this virus was already present in cells obtained from ATCC in 1987. Inverse PCR of DNA isolated from Sf9 L5814 cellular DNA revealed integration of SfRV sequences in the cellular genome. RT-PCR of total RNA showed a deletion of 320 nucleotides in the SfRV RNA that includes the transcriptional motifs for genes X and L. Concentrated cell culture supernatant was analyzed by sucrose density gradient centrifugation and revealed a single band at a density of 1.14 g/ml. This fraction was further analysed by electron microscopy and showed amorphous and particulate debris that did not resemble a rhabdovirus in morphology or size. SDS-PAGE analysis confirmed that the protein composition did not contain the typical five rhabdovirus structural proteins and LC-MS/MS analysis revealed primarily of exosomal marker proteins, the SfRV N protein, and truncated forms of SfRV N, P, and G proteins. The SfRV L gene fragment RNA sequence was recovered from the supernatant after ultracentrifugation of the 1.14 g/ml fraction treated with diethyl ether suggesting that the SfRV L gene fragment sequence is not associated with a diethyl ether resistant nucleocapsid. Interestingly, the 1.14 g/ml fraction was able to transfer baculovirus DNA into Sf9L5814 cells, consistent with the presence of functional exosomes. Our results demonstrate the absence of viral particles in ATCC CRL-1711 lot 5814 Sf9 cells in contrast to a previous study that suggested the presence of infectious rhabdoviral particles in Sf9 cells from a different lot. This study highlights how cell lines with different lineages may present different virosomes and therefore no general conclusions can be drawn across Sf9 cells from different laboratories.
Collapse
Affiliation(s)
- Yoshifumi Hashimoto
- Protein Sciences Corporation, Meriden, Connecticut, Unites States of America
| | - Daniel Macri
- Protein Sciences Corporation, Meriden, Connecticut, Unites States of America
| | - Indresh Srivastava
- Protein Sciences Corporation, Meriden, Connecticut, Unites States of America
| | - Clifton McPherson
- Protein Sciences Corporation, Meriden, Connecticut, Unites States of America
| | - Rachael Felberbaum
- Protein Sciences Corporation, Meriden, Connecticut, Unites States of America
| | - Penny Post
- Protein Sciences Corporation, Meriden, Connecticut, Unites States of America
| | - Manon Cox
- Protein Sciences Corporation, Meriden, Connecticut, Unites States of America
| |
Collapse
|
34
|
Minimizing fucosylation in insect cell-derived glycoproteins reduces binding to IgE antibodies from the sera of patients with allergy. Biotechnol J 2016. [DOI: 10.1002/biot.201300061] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|
35
|
Monteiro F, Bernal V, Alves PM. The role of host cell physiology in the productivity of the baculovirus-insect cell system: Fluxome analysis of Trichoplusia ni
and Spodoptera frugiperda
cell lines. Biotechnol Bioeng 2016; 114:674-684. [DOI: 10.1002/bit.26089] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2016] [Revised: 06/30/2016] [Accepted: 08/25/2016] [Indexed: 02/04/2023]
Affiliation(s)
- Francisca Monteiro
- iBET; Instituto de Biologia Experimental e Tecnológica; Oeiras Portugal
- Instituto; de Tecnologia Química e Biológica; Universidade Nova de Lisboa; Oeiras Portugal
| | - Vicente Bernal
- Departamento de Biología Molecular B e Inmunología, Facultad de Química, Campus Internacional de Excelencia “Mare Nostrum”; Universidad de Murcia; Murcia Spain
| | - Paula M. Alves
- iBET; Instituto de Biologia Experimental e Tecnológica; Oeiras Portugal
- Instituto; de Tecnologia Química e Biológica; Universidade Nova de Lisboa; Oeiras Portugal
| |
Collapse
|
36
|
Kwang TW, Zeng X, Wang S. Manufacturing of AcMNPV baculovirus vectors to enable gene therapy trials. Mol Ther Methods Clin Dev 2016; 3:15050. [PMID: 26858963 PMCID: PMC4729316 DOI: 10.1038/mtm.2015.50] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2015] [Revised: 11/11/2015] [Accepted: 11/11/2015] [Indexed: 12/17/2022]
Abstract
Over the past two decades, baculoviruses have become workhorse research tools for transient transgene expression. Although they have not yet been used directly as a gene therapy vector in the clinical setting, numerous preclinical studies have suggested the highly promising potential of baculovirus as a delivery vector for a variety of therapeutic applications including vaccination, tissue engineering, and cancer treatment. As such, there is growing interest in using baculoviruses as human gene therapy vectors, which has led to advances in baculovirus bioprocessing methods. This review provides an overview of the current approaches for scaled-up amplification, concentration, purification, and formulation of AcMNPV baculoviruses, and highlights the key regulatory requirements that must be met before gene therapy clinical trials can be initiated.
Collapse
Affiliation(s)
| | | | - Shu Wang
- Department of Biological Sciences, National University of Singapore, Singapore
- Institute of Bioengineering and Nanotechnology, Singapore
| |
Collapse
|
37
|
A simple plasmid-based transient gene expression method using High Five cells. J Biotechnol 2015; 216:67-75. [DOI: 10.1016/j.jbiotec.2015.10.007] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2015] [Revised: 10/05/2015] [Accepted: 10/09/2015] [Indexed: 12/12/2022]
|
38
|
Yu Q, Xiong Y, Gao H, Liu J, Chen Z, Wang Q, Wen D. Comparative proteomics analysis of Spodoptera frugiperda cells during Autographa californica multiple nucleopolyhedrovirus infection. Virol J 2015; 12:115. [PMID: 26239618 PMCID: PMC4524103 DOI: 10.1186/s12985-015-0346-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2015] [Accepted: 07/21/2015] [Indexed: 11/21/2022] Open
Abstract
Background Increasing evidence sugggest that in addition of balculovirus controling insect host, host cells also responds to balculovirus infection. However, compared to existing knowledge on virus gene, host cell responses are relatively poorly understood. Methods In this study, Spodoptera frugiperda (Sf9) cells were infected with Autographa californica multiple nucleopolyhedrovirus (AcMNPV). The protein composition and protein changes of Spodoptera frugiperda (Sf9) cells of different infection stages were analysed by isobaric tag for relative and absolute quantification (iTRAQ) techniques. Results A total of 4004 Sf9 proteins were identified by iTRAQ and 413 proteins were found as more than 1.5-fold changes in abundance. The 413 proteins were categorised according to GO classification for insects and were categorised into: biological process, molecular function and cellular component. Conclusions The determination of the protein changes in infected Sf9 cells would help to better understanding of host cell responses and facilitate better design of this virus-host cell interaction in pest insect control and other related fields. Electronic supplementary material The online version of this article (doi:10.1186/s12985-015-0346-9) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Qian Yu
- College of Food Science and Technology, Zhongkai University of Agriculture and Engineering, No. 501 Zhongkai Road, Haizhu District, Guangdong, 510225, People's Republic of China.
| | - Youhua Xiong
- College of Food Science and Technology, Zhongkai University of Agriculture and Engineering, No. 501 Zhongkai Road, Haizhu District, Guangdong, 510225, People's Republic of China.
| | - Hang Gao
- College of Food Science and Technology, Zhongkai University of Agriculture and Engineering, No. 501 Zhongkai Road, Haizhu District, Guangdong, 510225, People's Republic of China.
| | - Jianliang Liu
- College of Food Science and Technology, Zhongkai University of Agriculture and Engineering, No. 501 Zhongkai Road, Haizhu District, Guangdong, 510225, People's Republic of China.
| | - Zhiqiang Chen
- College of Food Science and Technology, Zhongkai University of Agriculture and Engineering, No. 501 Zhongkai Road, Haizhu District, Guangdong, 510225, People's Republic of China.
| | - Qin Wang
- College of Food Science and Technology, Zhongkai University of Agriculture and Engineering, No. 501 Zhongkai Road, Haizhu District, Guangdong, 510225, People's Republic of China.
| | - Dongling Wen
- College of Food Science and Technology, Zhongkai University of Agriculture and Engineering, No. 501 Zhongkai Road, Haizhu District, Guangdong, 510225, People's Republic of China.
| |
Collapse
|
39
|
Qiao L, Lee KJ, Ko K. Characterization of the Glycan Structures of Glycoprotein GA733-Fc Expressed in a Baculovirus-Insect Cell System. B KOREAN CHEM SOC 2015. [DOI: 10.1002/bkcs.10035] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Affiliation(s)
- Lu Qiao
- Department of Medicine, Therapeutic Protein Engineering Lab, College of Medicine; Chung-Ang University; Seoul 156-756 Korea
| | - Kyung Jin Lee
- Department of Medicine, Therapeutic Protein Engineering Lab, College of Medicine; Chung-Ang University; Seoul 156-756 Korea
| | - Kisung Ko
- Department of Medicine, Therapeutic Protein Engineering Lab, College of Medicine; Chung-Ang University; Seoul 156-756 Korea
| |
Collapse
|
40
|
Palmberger D, Ashjaei K, Strell S, Hoffmann-Sommergruber K, Grabherr R. Minimizing fucosylation in insect cell-derived glycoproteins reduces binding to IgE antibodies from the sera of patients with allergy. Biotechnol J 2014; 9:1206-14. [PMID: 24753388 DOI: 10.1002/biot.201400061] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2014] [Revised: 03/13/2014] [Accepted: 03/21/2014] [Indexed: 11/10/2022]
Abstract
The baculovirus/insect cell system has proven to be a very powerful tool for the expression of several therapeutics. Nevertheless, these products sometimes suffer from reduced biological activity and unwanted side effects. Several studies have demonstrated that glycosylation can greatly influence the structure, function, half-life, antigenicity and immunogenicity of various glycoproteins. Yet, the glycosylation pattern of insect cell-derived products is not favorable for many applications. Especially, the presence of core α1,3-linked fucose bears the risk of causing immediate hypersensitivity reactions in patients with allergy. In this study, we evaluated the impact of fucose residues on the allergenic potential of an insect cell-expressed vaccine candidate. In order to block the GDP-L-fucose de novo synthesis pathway, we integrated the Pseudomonas aeruginosa GDP-6-deoxy-D-lyxo-4-hexulose reductase (RMD) gene into a baculovirus backbone. This virus was then used for the expression of soluble influenza A virus hemagglutinin (HA). Expression studies showed that the co-expression of RMD did not influence the overall level of recombinant protein secretion. We confirmed the result of our strategy by analyzing PNGase A-released N-glycans using MALDI-TOF-MS. In order to evaluate the biological impact of defucosylation of influenza HA we tested the binding activity of IgE derived from the sera of patients with allergy to the purified antigen. The non-fucosylated HA showed a 10-fold decrease in IgE binding levels as compared to wildtype variants.
Collapse
Affiliation(s)
- Dieter Palmberger
- Vienna Institute of BioTechnology - VIBT, University of Natural Resources and Life Sciences, Vienna, Austria
| | | | | | | | | |
Collapse
|