Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 5.193
Filter
1.
Methods Mol Biol ; 2829: 3-11, 2024.
Article in English | MEDLINE | ID: mdl-38951323

ABSTRACT

Healthy insect cell cultures are critical for any method described in this book, including making productive baculovirus banks, protein or AAV expression, and determining viral titers. This chapter describes cell maintenance in shake flasks using serum-free conditions and the expansion of virus stocks from a single plaque purified virus. Insect cells can be passaged over multiple generations, but as the cells may undergo changes over multiple passages, limiting the use of your cells to a defined number of passages such as 50 passages is recommendable. Baculovirus stocks once created using serum-free media are not very stable at 4-8 °C. This chapter also includes a simple method to store cells from an early cell passage and your virus stock in liquid nitrogen.


Subject(s)
Baculoviridae , Cell Culture Techniques , Animals , Baculoviridae/genetics , Cell Culture Techniques/methods , Insecta/virology , Insecta/cytology , Cell Line
2.
Methods Mol Biol ; 2829: 21-48, 2024.
Article in English | MEDLINE | ID: mdl-38951325

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-ß 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.


Subject(s)
Baculoviridae , Genetic Vectors , Recombinant Proteins , Baculoviridae/genetics , Animals , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Genetic Vectors/genetics , Sf9 Cells , Gene Expression , Humans , Insecta/genetics , Spodoptera , Cell Line , Homologous Recombination , Cost-Benefit Analysis
3.
Methods Mol Biol ; 2829: 49-66, 2024.
Article in English | MEDLINE | ID: mdl-38951326

ABSTRACT

This chapter outlines the workflow using the ExpiSf™ Expression System designed for high-density infection of suspension ExpiSf9™ cells. The system utilizes a chemically defined, serum-free, protein-free, and animal origin free medium, making it suitable for recombinant protein expression experiments. The ExpiSf™ chemically defined medium allows efficient transfection and baculovirus production directly within the same culture medium. The ExpiSf™ Expression System Starter Kit provides all necessary components, including cells, culture medium, and reagents needed to infect one (1) liter of cell culture. The system's versatility and animal origin free nature make it a valuable tool for various protein expression studies and biotechnological applications.


Subject(s)
Baculoviridae , Recombinant Proteins , Workflow , Animals , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Baculoviridae/genetics , Transfection/methods , Culture Media/chemistry , Cell Culture Techniques/methods , Cell Line , Gene Expression
4.
Methods Mol Biol ; 2829: 13-20, 2024.
Article in English | MEDLINE | ID: mdl-38951324

ABSTRACT

The success of using the insect cell-baculovirus expression technology (BEST) relies on the efficient construction of recombinant baculovirus with genetic stability and high productivity, ideally within a short time period. Generation of recombinant baculoviruses requires the transfection of insect cells, harvesting of recombinant baculovirus pools, isolation of plaques, and the expansion of baculovirus stocks for their use for recombinant protein production. Moreover, many options exist for selecting the genetic elements to be present in the recombinant baculovirus. This chapter describes the most commonly used homologous recombination systems for the production of recombinant baculoviruses, as well as strategies to maximize generation efficiency and recombinant protein or baculovirus production. The key steps for generating baculovirus stocks and troubleshooting strategies are described.


Subject(s)
Baculoviridae , Recombinant Proteins , Baculoviridae/genetics , Animals , Recombinant Proteins/genetics , Genetic Vectors/genetics , Transfection/methods , Homologous Recombination , Sf9 Cells , Cell Line , Spodoptera/virology , Insecta/genetics , Insecta/virology
5.
Methods Mol Biol ; 2829: 91-107, 2024.
Article in English | MEDLINE | ID: mdl-38951329

ABSTRACT

RNA interference (RNAi) serves as an indispensable tool for gene function studies and has been substantiated through extensive research for its practical applications in the baculovirus expression vector system (BEVS). This chapter expands the RNAi toolkit in insect cell culture by including small interfering RNA (siRNA) in the protocol, in addition to the conventional use of double-stranded RNA (dsRNA). This chapter also brings attention to key design and reporting considerations, based on Minimum Information About an RNAi Experiment (MIARE) guidelines. Recommendations regarding online tools for dsRNA and siRNA design are provided, along with guidance on choosing suitable methods for measuring silencing outcomes.


Subject(s)
Baculoviridae , Genetic Vectors , RNA Interference , RNA, Double-Stranded , RNA, Small Interfering , Animals , Baculoviridae/genetics , RNA, Double-Stranded/genetics , RNA, Small Interfering/genetics , Genetic Vectors/genetics , Insecta/genetics , Cell Line , Sf9 Cells
6.
Methods Mol Biol ; 2829: 67-75, 2024.
Article in English | MEDLINE | ID: mdl-38951327

ABSTRACT

This chapter outlines the use of TOPO cloning for streamlined generation of a recombinant plasmid containing your gene of interest for use in the Bac-to-Bac™ Baculovirus Expression System.


Subject(s)
Cloning, Molecular , Plasmids , Plasmids/genetics , Cloning, Molecular/methods , Genetic Vectors/genetics , Baculoviridae/genetics , Chromosomes, Artificial, Bacterial/genetics
7.
Methods Mol Biol ; 2829: 159-173, 2024.
Article in English | MEDLINE | ID: mdl-38951332

ABSTRACT

The baculovirus expression vector system (BEVS) is a powerful platform for protein expression in insect cells. A prevalent application is the expression of complex protein structures consisting of multiple, interacting proteins. Coinfection with multiple baculoviruses allows for production of complex structures, facilitating structure-function studies, allowing augmentation of insect cell functionality, and production of clinically relevant products such as virus-like particles (VLPs) and adeno-associated viral vectors (AAV). Successful coinfections require the generation of robust and well-quantified recombinant baculovirus stocks. Virus production through homologous recombination, combined with rigorous quantification of viral titers, allows for synchronous coinfections producing high end-product titers. In this chapter, we describe the streamlined workflow for generation and quantification of high-quality recombinant baculovirus stocks and successful coinfection as defined by a preponderance of dually infected cells in the insect cell culture.


Subject(s)
Baculoviridae , Genetic Vectors , Recombinant Proteins , Baculoviridae/genetics , Animals , Genetic Vectors/genetics , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Sf9 Cells , Cell Line , Spodoptera/virology
8.
Methods Mol Biol ; 2829: 79-90, 2024.
Article in English | MEDLINE | ID: mdl-38951328

ABSTRACT

Adaptive laboratory evolution (ALE) is a powerful tool for enhancing the fitness of cell lines in specific applications, including recombinant protein production. Through adaptation to nonstandard culture conditions, cells can develop specific traits that make them high producers. Despite being widely used for microorganisms and, to lesser extent, for mammalian cells, ALE has been poorly leveraged for insect cells. Here, we describe a method for adapting insect High Five and Sf9 cells to nonstandard culture conditions via an ALE approach. Aiming to demonstrate the potential of ALE to improve productivity of insect cells, two case studies are demonstrated. In the first, we adapted insect High Five cells from their standard pH (6.2) to neutral pH (7.0); this adaptation allowed to improve production of influenza virus-like particles (VLPs) by threefold, using the transient baculovirus expression vector system. In the second, we adapted insect Sf9 cells from their standard culture temperature (27 °C) to hypothermic growth (22 °C); this adaptation allowed to improve production of influenza VLPs by sixfold, using stable cell lines. These examples demonstrate the potential of ALE for enhancing productivity within distinct insect cell hosts and expression systems by manipulating different culture conditions.


Subject(s)
Recombinant Proteins , Animals , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Cell Line , Sf9 Cells , Baculoviridae/genetics , Cell Culture Techniques/methods , Insecta/genetics , Insecta/cytology , Directed Molecular Evolution/methods , Hydrogen-Ion Concentration , Temperature
9.
Methods Mol Biol ; 2829: 175-183, 2024.
Article in English | MEDLINE | ID: mdl-38951333

ABSTRACT

Monoclonal antibodies have widespread applications in disease treatment and antigen detection. They are traditionally produced using mammalian cell expression system, which is not able to satisfy the increasing demand of these proteins at large scale. Baculovirus expression vector system (BEVS) is an attractive alternative platform for the production of biologically active monoclonal antibodies. In this chapter, we demonstrate the production of an HIV-1 broadly neutralizing antibody b12 in BEVS. The processes including transfer vector construction, recombinant baculovirus generation, and antibody production and detection are described.


Subject(s)
Baculoviridae , Genetic Vectors , Baculoviridae/genetics , Genetic Vectors/genetics , Animals , Humans , Gene Expression , HIV-1/genetics , HIV-1/immunology , Recombinant Proteins/genetics , Recombinant Proteins/biosynthesis , Antibodies, Monoclonal/genetics , Antibodies, Monoclonal/biosynthesis , Antibodies, Monoclonal/immunology , Antibodies, Neutralizing/immunology , Enzyme-Linked Immunosorbent Assay , HIV Antibodies/immunology , HIV Antibodies/genetics , Sf9 Cells
10.
Methods Mol Biol ; 2829: 185-194, 2024.
Article in English | MEDLINE | ID: mdl-38951334

ABSTRACT

Insect cell expression has been successfully used for the production of viral antigens as part of commercial vaccine development. As expression host, insect cells offer advantage over bacterial system by presenting the ability of performing post-translational modifications (PTMs) such as glycosylation and phosphorylation thus preserving the native functionality of the proteins especially for viral antigens. Insect cells have limitation in exactly mimicking some proteins which require complex glycosylation pattern. The recent advancement in insect cell engineering strategies could overcome this limitation to some extent. Moreover, cost efficiency, timelines, safety, and process adoptability make insect cells a preferred platform for production of subunit antigens for human and animal vaccines. In this chapter, we describe the method for producing the SARS-CoV2 spike ectodomain subunit antigen for human vaccine development and the virus like particle (VLP), based on capsid protein of porcine circovirus virus 2 (PCV2d) antigen for animal vaccine development using two different insect cell lines, SF9 & Hi5, respectively. This methodology demonstrates the flexibility and broad applicability of insect cell as expression host.


Subject(s)
Antigens, Viral , Baculoviridae , Spike Glycoprotein, Coronavirus , Animals , Baculoviridae/genetics , Antigens, Viral/genetics , Antigens, Viral/immunology , Sf9 Cells , Humans , Spike Glycoprotein, Coronavirus/genetics , Spike Glycoprotein, Coronavirus/immunology , Spike Glycoprotein, Coronavirus/metabolism , Recombinant Proteins/genetics , Cell Line , SARS-CoV-2/genetics , SARS-CoV-2/immunology , Vaccines, Virus-Like Particle/genetics , Vaccines, Virus-Like Particle/immunology , Vaccines, Virus-Like Particle/biosynthesis , Capsid Proteins/genetics , Capsid Proteins/immunology , Glycosylation , Insecta/genetics , Spodoptera , COVID-19 Vaccines/genetics , COVID-19 Vaccines/immunology
11.
Methods Mol Biol ; 2829: 109-126, 2024.
Article in English | MEDLINE | ID: mdl-38951330

ABSTRACT

Baculoviruses are widely used for their potential as biological pesticide and as platform for the production of recombinant proteins and gene therapy vectors. The Baculovirus Expression Vector System (BEVS) is used for high level of expression of (multiple) proteins in insect cells. Baculovirus recombinants can be quickly constructed by transposition of the gene(s) of interest into a so-called bacmid, which is a baculovirus infectious clone maintained as single-copy, bacterial artificial chromosome in Escherichia coli. A two-step homologous recombineering technique using the lambda-red system in E. coli allows for scarless editing of the bacmid with PCR products based on sequence homology. In the first step, a selection cassette with 50 bp homology arms, typically generated by PCR, is inserted into the designated locus. In the second step, the selection cassette is removed based on a negative selection marker, such as SacB or rpsL. This lambda-red recombineering technique can be used for multiple gene editing purposes, including (large) deletions, insertions, and even single point mutations. Moreover, since there are no remnants of the editing process, successive modifications of the same bacmid are possible. This chapter provides detailed instructions to design and perform two-step homologous recombineering of baculovirus bacmid DNA in E. coli. We present two case studies demonstrating the utility of this technique for creating a deletion mutant of the chitinase and cathepsin genes and for introducing a single point mutation in the baculovirus gene gp41. This scarless genome editing approach can facilitate functional studies of baculovirus genes and improve the production of recombinant proteins using the BEVS.


Subject(s)
Baculoviridae , Escherichia coli , Gene Editing , Genetic Vectors , Gene Editing/methods , Escherichia coli/genetics , Baculoviridae/genetics , Genetic Vectors/genetics , Chromosomes, Artificial, Bacterial/genetics , Genome, Viral , Genetic Engineering/methods , Bacteriophage lambda/genetics , Homologous Recombination
12.
Methods Mol Biol ; 2829: 203-214, 2024.
Article in English | MEDLINE | ID: mdl-38951336

ABSTRACT

The insect cell-baculovirus expression vector (IC-BEV) platform has enabled small research-scale and large commercial-scale production of recombinant proteins and therapeutic biologics including recombinant adeno-associated virus (rAAV)-based gene delivery vectors. The wide use of this platform is comparable with other mammalian cell line-based platforms due to its simplicity, high-yield, comparable quality attributes, and robust bioprocessing features. In this chapter, we describe a rAAV production protocol employing one of the recent modifications of the One-Bac platform that consists of a stable transformed Sf9 cell line carrying AAV Rep2/Cap5 genes that are induced upon infection with a single recombinant baculovirus expression vector harboring the transgene of interest (rAAV genome). The overall protocol consists of essential steps including rBEV working stock preparation, rAAV production, and centrifugation-based clarification of cell culture lysate. The same protocol can also be applied for rAAV vector production using traditional Three-Bac, Two-Bac, and Mono-Bac platforms without requiring significant changes.


Subject(s)
Baculoviridae , Dependovirus , Genetic Vectors , Dependovirus/genetics , Genetic Vectors/genetics , Animals , Sf9 Cells , Baculoviridae/genetics , Humans , Transgenes , Cell Line , Recombinant Proteins/genetics , Recombinant Proteins/biosynthesis
13.
Methods Mol Biol ; 2829: 127-156, 2024.
Article in English | MEDLINE | ID: mdl-38951331

ABSTRACT

The baculovirus expression vector system (BEVS) has now found acceptance in both research laboratories and industry, which can be attributed to many of its key features including the limited host range of the vectors, their non-pathogenicity to humans, and the mammalian-like post-translational modification (PTMs) that can be achieved in insect cells. In fact, this system acts as a middle ground between prokaryotes and higher eukaryotes to produce complex biologics. Still, industrial use of the BEVS lags compared to other platforms. We have postulated that one reason for this has been a lack of genetic tools that can complement the study of baculovirus vectors, while a second reason is the co-production of the baculovirus vector with the desired product. While some genetic enhancements have been made to improve the BEVS as a production platform, the genome remains under-scrutinized. This chapter outlines the methodology for a CRISPR-Cas9-based transfection-infection assay to probe the baculovirus genome for essential/nonessential genes that can potentially maximize foreign gene expression under a promoter of choice.


Subject(s)
Baculoviridae , CRISPR-Cas Systems , Genetic Vectors , Baculoviridae/genetics , Genetic Vectors/genetics , Animals , Genes, Essential , Gene Expression , Transfection/methods , Gene Editing/methods , Sf9 Cells , Humans
14.
Methods Mol Biol ; 2829: 227-235, 2024.
Article in English | MEDLINE | ID: mdl-38951338

ABSTRACT

Virus-like particles (VLPs) of the adeno-associated virus (AAV) can be produced using the baculovirus expression vector system. Insertion of small peptides on the surface of the AAV or AAV VLPs has been used to redirect the AAV to different target tissues and for vaccine development. Usually, the VLPs self-assemble intracellularly, and an extraction step must be performed before purification. Here, we describe the method we have used to extract AAV VLPs from insect cells successfully with peptide insertions on their surface.


Subject(s)
Dependovirus , Peptides , Dependovirus/genetics , Animals , Peptides/chemistry , Peptides/genetics , Genetic Vectors/genetics , Virion/genetics , Baculoviridae/genetics , Sf9 Cells , Humans , Cell Line , Capsid Proteins/genetics , Capsid Proteins/isolation & purification
15.
Methods Mol Biol ; 2829: 247-255, 2024.
Article in English | MEDLINE | ID: mdl-38951340

ABSTRACT

The Baculovirus Expression Vector System (BEVS) is used with cultured insect cells to produce a wide variety of heterologous proteins, which can be secreted into the culture medium during the transient infection process (Smith et al. Mol Cell Biol 12:2156-2165, 1983). When the infection process is complete, centrifugation is often used to separate the desired protein from the spent insect cells. The desired product in the harvested supernatant is contaminated with baculovirus, amino acids, lipids, detergents, oils, lysed cells from the infection process, genomic DNA from the insect cells, and proteases due to the lytic nature of the baculovirus infection process and many other contaminants (Ikonomou et al. Appl Microbiol Biotechnol 62:1-20, 2003). All these contaminants that are present in the centrifuged supernatant with the desired secreted protein make the initial chromatographic capture step critical for effective purification of the desired protein. A purification scheme will be outlined for a slightly acidic secreted protein using cation exchange chromatography (Lundanes et al. Chromatography: basic principles, sample preparations and related methods, 1st edn. Wiley, 2013).


Subject(s)
Baculoviridae , Animals , Baculoviridae/genetics , Baculoviridae/metabolism , Chromatography, Ion Exchange/methods , Recombinant Proteins/isolation & purification , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Insecta/cytology , Sf9 Cells , Genetic Vectors/genetics , Cell Line , Spodoptera
16.
Methods Mol Biol ; 2829: 195-202, 2024.
Article in English | MEDLINE | ID: mdl-38951335

ABSTRACT

The Baculovirus Expression Vector System (BEVS) has revolutionized the field of recombinant protein expression by enabling efficient and high yield production. The platform offers many advantages including manufacturing speed, flexible design, and scalability. In this chapter, we describe the methods including strategies and considerations to successfully optimize and scale-up using BEVS as a tool for production (Fig. 1). As an illustrative case study, we present an example focused on the production of a viral glycoprotein.


Subject(s)
Baculoviridae , Genetic Vectors , Recombinant Proteins , Baculoviridae/genetics , Recombinant Proteins/genetics , Recombinant Proteins/biosynthesis , Genetic Vectors/genetics , Animals , Humans , Sf9 Cells
17.
Methods Mol Biol ; 2829: 271-275, 2024.
Article in English | MEDLINE | ID: mdl-38951343

ABSTRACT

This chapter outlines a rapid detection method to determine the virus titer of your baculovirus stock. Staining of cells with fluorescently labeled gp64 antibody allows for flow cytometer-based quantitation of baculovirus-infected insect cells. In this assay, Sf9 cells are infected with tenfold serial dilutions of the test virus stock, and baculovirus titers are calculated based on the ratio of infected to uninfected cells 13 to 18 h after inoculation.


Subject(s)
Baculoviridae , Flow Cytometry , Flow Cytometry/methods , Baculoviridae/genetics , Animals , Sf9 Cells , Viral Load/methods
18.
Methods Mol Biol ; 2829: 237-246, 2024.
Article in English | MEDLINE | ID: mdl-38951339

ABSTRACT

Virus-like particles (VLP) of the cowpea chlorotic mottle virus (CCMV), a plant virus, have been shown to be safe and noncytotoxic vehicles for delivering various cargos, including nucleic acids and peptides, and as scaffolds for presenting epitopes. Thus, CCMV-VLP have acquired increasing attention to be used in fields such as gene therapy, drug delivery, and vaccine development. Regardless of their production method, most reports purify CCMV-VLP through a series of ultracentrifugation steps using sucrose density gradient ultracentrifugation, which is a complex and time-consuming process. Here, the use of anion exchange chromatography is described as a one-step protocol for purification of CCMV-VLP produced by the insect cell-baculovirus expression vector system (IC-BEVS).


Subject(s)
Bromovirus , Bromovirus/genetics , Animals , Baculoviridae/genetics , Genetic Vectors/genetics , Chromatography, Ion Exchange/methods , Virion/isolation & purification , Virion/genetics , Virion/metabolism
19.
Methods Mol Biol ; 2829: 301-327, 2024.
Article in English | MEDLINE | ID: mdl-38951346

ABSTRACT

Efficient genome editing by using CRISPR technologies requires simultaneous and efficient delivery of multiple genetically encoded components to mammalian cells. Amongst all editing approaches, prime editing (PE) has the unique potential to perform seamless genome rewriting, in the absence of DNA double-strand breaks (DSBs). The cargo capacity required for efficient PE delivery to mammalian cells stands at odd with the limited packaging capacity of traditional viral delivery vectors. By contrast, baculovirus (BV) has a large synthetic DNA capacity and can efficiently transduce mammalian cells. Here we describe a protocol for the assembly of baculovirus vectors for multiplexed prime editing in mammalian cells.


Subject(s)
Baculoviridae , CRISPR-Cas Systems , Gene Editing , Genetic Vectors , Baculoviridae/genetics , Gene Editing/methods , Genetic Vectors/genetics , Humans , Animals , HEK293 Cells
20.
Methods Mol Biol ; 2829: 267-270, 2024.
Article in English | MEDLINE | ID: mdl-38951342

ABSTRACT

There are many methods that can be used to determine the infectious titer of your baculovirus stock. The TCID50 method is a simple end-point dilution method that determines the amount of baculovirus virus needed to produce a cytopathic effect or kill 50% of inoculated insect cells. Serial dilutions of baculovirus stock are added to Sf9 cells cultivated in 96-well plates and 3-5 days after infection, cells are monitored for cell death or cytopathic effect. The titer can then be calculated by the Reed-Muench method as described in this method.


Subject(s)
Baculoviridae , Baculoviridae/genetics , Animals , Sf9 Cells , Cytopathogenic Effect, Viral , Spodoptera/virology , Viral Load/methods , Cell Line
SELECTION OF CITATIONS
SEARCH DETAIL
...