Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.805
Filter
1.
Arch Virol ; 169(8): 172, 2024 Aug 03.
Article in English | MEDLINE | ID: mdl-39096433

ABSTRACT

Goatpox and sheeppox are highly contagious and economically important viral diseases of small ruminants. Due to the risk they pose to animal health, livestock production, and international trade, capripoxviruses are a considerable threat to the livestock economy. In this study, we expressed two core proteins (A4L and A12L) and one extracellular enveloped virion protein (A33R) of goatpox virus in a baculovirus expression vector system and evaluated their use as diagnostic antigens in ELISA. Full-length A4L, A12L, and A33R genes of the GTPV Uttarkashi strain were amplified, cloned into the pFastBac HT A donor vector, and introduced into DH10Bac cells containing a baculovirus shuttle vector plasmid to generate recombinant bacmids. The recombinant baculoviruses were produced in Sf-21 cells by transfection, and proteins were expressed in TN5 insect cells. The recombinant proteins were analysed by SDS-PAGE and confirmed by western blot, with expected sizes of ~30 kDa, ~31 kDa, and ~32 kDa for A4L, A12L, and A33R, respectively. The recombinant proteins were purified, and the immunoreactivity of the purified proteins was confirmed by western blot using anti-GTPV serum. The antigenic specificity of the expressed proteins as diagnostic antigens was evaluated by testing their reactivity with infected, vaccinated, and negative GTPV/SPPV serum in indirect ELISA, and the A33R-based indirect ELISA was optimized. The diagnostic sensitivity and specificity of the A33R-based indirect ELISA were found to be of 89% and 94% for goats and 98% and 91%, for sheep, respectively. No cross-reactivity was observed with other related viruses. The recombinant-A33R-based indirect ELISA developed in the present study shows that it has potential for the detection of antibodies in GTPV and SPPV infected/vaccinated animals.


Subject(s)
Baculoviridae , Capripoxvirus , Enzyme-Linked Immunosorbent Assay , Goat Diseases , Goats , Viral Envelope Proteins , Capripoxvirus/genetics , Capripoxvirus/isolation & purification , Baculoviridae/genetics , Animals , Goat Diseases/virology , Goat Diseases/diagnosis , Viral Envelope Proteins/genetics , Viral Envelope Proteins/immunology , Goats/virology , Enzyme-Linked Immunosorbent Assay/methods , Poxviridae Infections/diagnosis , Poxviridae Infections/veterinary , Poxviridae Infections/virology , Recombinant Proteins/genetics , Recombinant Proteins/isolation & purification , Recombinant Proteins/immunology , Virion/genetics , Viral Core Proteins/genetics , Viral Core Proteins/immunology , Antibodies, Viral/immunology , Antibodies, Viral/blood , Sf9 Cells , Antigens, Viral/genetics , Antigens, Viral/immunology , Cell Line , Gene Expression
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: 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
4.
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
5.
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
6.
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
7.
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
8.
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
9.
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
10.
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
11.
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
12.
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
13.
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
14.
Methods Mol Biol ; 2829: 217-226, 2024.
Article in English | MEDLINE | ID: mdl-38951337

ABSTRACT

Purification of rAAV is a crucial unit operation of the AAV production process. It enables the capture of AAV and removal of contaminants such as host cell proteins, host cell DNA, and other cell culture-related impurities. Here we describe the purification of rAAV produced in insect cells Sf9/rBEV by immuno-affinity capture chromatography. The method is fully scale-amenable unlike other traditional purification methods based on ultracentrifugation. The method reported herein has two main steps: (1) the clarification of cell lysate by depth filtration and (2) the selective capture and single-step purification of AAV via immune-affinity chromatography. This purification method has been successfully implemented to purify the majority of wild-type AAV serotypes.


Subject(s)
Chromatography, Affinity , Dependovirus , Dependovirus/genetics , Dependovirus/isolation & purification , Animals , Chromatography, Affinity/methods , Sf9 Cells , Genetic Vectors/genetics , Humans , Spodoptera/virology
15.
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
16.
Methods Mol Biol ; 2829: 289-300, 2024.
Article in English | MEDLINE | ID: mdl-38951345

ABSTRACT

Nonviral transfection has been used to express various recombinant proteins, therapeutics, and virus-like particles (VLP) in mammalian and insect cells. Virus-free methods for protein expression require fewer steps for obtaining protein expression by eliminating virus amplification and measuring the infectivity of the virus. The nonviral method uses a nonlytic plasmid to transfect the gene of interest into the insect cells instead of using baculovirus, a lytic system. In this chapter, we describe one of the transfection methods, which uses polyethyleneimine (PEI) as a DNA delivery material into the insect cells to express the recombinant protein in both adherent and suspension cells.


Subject(s)
Polyethyleneimine , Recombinant Proteins , Transfection , Animals , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Transfection/methods , Polyethyleneimine/chemistry , Plasmids/genetics , Insecta/genetics , Sf9 Cells , Cell Line , Gene Expression , Spodoptera
17.
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
18.
Methods Mol Biol ; 2829: 259-265, 2024.
Article in English | MEDLINE | ID: mdl-38951341

ABSTRACT

Plaque assay method enables the quantification of infectious baculovirus when defined as plaque forming units (PFU). It allows to determine the amount of infectious virus needed to infect the cells at a specific multiplicity of infection (MOI). Serial dilutions of baculovirus stock are added to the Sf9 cells monolayer followed by addition of 5% Agarose overlay. Six days after infection clear infection halos are observed using a neutral red solution. Here we describe the quantification of recombinant baculovirus expression vector (rBEV) carrying a transgene in an rAAV expression cassette. Reproducible quantification of PFU is obtained with this method.


Subject(s)
Baculoviridae , Genetic Vectors , Viral Plaque Assay , Baculoviridae/genetics , Sf9 Cells , Viral Plaque Assay/methods , Animals , Genetic Vectors/genetics , Transgenes , Virion/genetics , Dependovirus/genetics , Spodoptera/virology
19.
Methods Mol Biol ; 2824: 121-133, 2024.
Article in English | MEDLINE | ID: mdl-39039410

ABSTRACT

The Rift Valley fever virus (RVFV), transmitted through mosquito bites, leads to severe illness in humans and livestock throughout Africa and the Arabian Peninsula, causing significant morbidity and mortality. As of now, there are no verified and efficacious drugs or licensed vaccines accessible for the prevention or treatment of RVFV infections in both humans and livestock. The mature RVFV virion has two envelope proteins on its surface: glycoprotein N (GN) and glycoprotein C (GC). These proteins play a significant role in facilitating the virus's entry into the host cell, making them prominent targets for entry mechanism research as well as targets for drugs and vaccine development. The initial stage in obtaining atomic-resolution structural and mechanistic information on viral entry as well as developing biochemical and biophysical research tools involves recombinant protein production. In this chapter, we describe a simplified and scalable protocol facilitating the generation of high-quality, high-titer baculovirus virus for expression and purification of RVFV GC, utilizing the baculovirus-mediated expression system in insect cells.


Subject(s)
Baculoviridae , Recombinant Proteins , Rift Valley fever virus , Viral Envelope Proteins , Baculoviridae/genetics , Animals , Viral Envelope Proteins/genetics , Viral Envelope Proteins/isolation & purification , Viral Envelope Proteins/chemistry , Viral Envelope Proteins/metabolism , Recombinant Proteins/genetics , Recombinant Proteins/isolation & purification , Recombinant Proteins/metabolism , Rift Valley fever virus/genetics , Sf9 Cells , Gene Expression , Humans , Genetic Vectors/genetics , Cloning, Molecular/methods
20.
Protein Expr Purif ; 222: 106542, 2024 Oct.
Article in English | MEDLINE | ID: mdl-38969281

ABSTRACT

Human ZC3H11A is an RNA-binding zinc finger protein involved in mRNA export and required for the efficient growth of human nuclear replicating viruses. Its biochemical properties are largely unknown so our goal has been to produce the protein in a pure and stable form suitable for its characterization. This has been challenging since the protein is large (810 amino acids) and with only the N-terminal zinc finger domain (amino acids 1-86) being well structured, the remainder is intrinsically disordered. Our production strategies have encompassed recombinant expression of full-length, truncated and mutated ZC3H11A variants with varying purification tags and fusion proteins in several expression systems, with or without co-expression of chaperones and putative interaction partners. A range of purification schemes have been explored. Initially, only truncated ZC3H11A encompassing the zinc finger domain could successfully be produced in a stable form. It required recombinant expression in insect cells since expression in E. coli gave a protein that aggregated. To reduce problematic nucleic acid contaminations, Cys8, located in one of the zinc fingers, was substituted by Ala and Ser. Interestingly, this did not affect nucleic acid binding, but the full-length protein was stabilised while the truncated version was insoluble. Ultimately, we discovered that when using alkaline buffers (pH 9) for purification, full-length ZC3H11A expressed in Sf9 insect cells was obtained in a stable and >90 % pure form, and as a mixture of monomers, dimers, tetramers and hexamers. Many of the challenges experienced are consistent with its predicted structure and unusual charge distribution.


Subject(s)
Escherichia coli , RNA-Binding Proteins , Humans , RNA-Binding Proteins/genetics , RNA-Binding Proteins/chemistry , RNA-Binding Proteins/metabolism , RNA-Binding Proteins/isolation & purification , Escherichia coli/genetics , Escherichia coli/metabolism , Animals , Zinc Fingers , Recombinant Proteins/genetics , Recombinant Proteins/chemistry , Recombinant Proteins/isolation & purification , Recombinant Proteins/biosynthesis , Recombinant Proteins/metabolism , Sf9 Cells , Protein Stability , Intrinsically Disordered Proteins/genetics , Intrinsically Disordered Proteins/chemistry , Intrinsically Disordered Proteins/isolation & purification , Intrinsically Disordered Proteins/metabolism , Intrinsically Disordered Proteins/biosynthesis
SELECTION OF CITATIONS
SEARCH DETAIL