101
|
Belforte FS, Targovnik AM, González-Lebrero RM, Osorio Larroche C, Citterio CE, González-Sarmiento R, Miranda MV, Targovnik HM, Rivolta CM. Kinetic characterization of human thyroperoxidase. Normal and pathological enzyme expression in Baculovirus system: a molecular model of functional expression. Mol Cell Endocrinol 2015; 404:9-15. [PMID: 25576858 DOI: 10.1016/j.mce.2014.12.026] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/22/2014] [Revised: 12/25/2014] [Accepted: 12/30/2014] [Indexed: 11/18/2022]
Abstract
BACKGROUND Human thyroperoxidase (hTPO) is a membrane-bound glycoprotein located at the apical membrane of the thyroid follicular cells which catalyzes iodide oxidation and organification in the thyroglobulin (TG) tyrosine residues, leading to the thyroid hormone synthesis by coupling of iodotyrosine residues. Mutations in hTPO gene are the main cause of iodine organification defects (IOD) in infants. METHODS We investigated the functional impact of hTPO gene missense mutations previously identified in our laboratory (p.C808R, p.G387R and p.P499L). In order to obtain the whole wild-type (WT) coding sequence of hTPO, sequential cloning strategy in pGEMT vector was carried out. Then, site-directed mutagenesis was performed. WT and mutant hTPOs were cloned into the pAcGP67B transfer vector and the recombinant proteins were expressed in Baculovirus System, purified and characterized by SDS-PAGE and Western blot. Moreover, we report for the first time the kinetic constants of hTPO, of both WT and mutant enzymes. RESULTS The functional evaluation of the recombinant hTPOs showed decreased activity in the three mutants with respect to WT. Regarding to the affinity for the substrate, the mutants showed higher Km values with respect to the WT. Additionally, the three mutants showed lower reaction efficiencies (Vmax/Km) with respect to WT hTPO. CONCLUSIONS We optimize the expression and purification of recombinant hTPOs using the Baculovirus System and we report for the first time the kinetic characterization of hTPOs.
Collapse
Affiliation(s)
- Fiorella S Belforte
- Laboratorio de Genética y Biología Molecular, Instituto de Inmunología, Genética y Metabolismo (INIGEM, CONICET-UBA), Hospital de Clínicas "José de San Martín", C1120AAR Buenos Aires, Argentina; Cátedra de Genética y Biología Molecular, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, C1113AAD Buenos Aires, Argentina
| | - Alexandra M Targovnik
- Instituto de Nanobiotecnología (NANOBIOTEC, CONICET-UBA), Cátedra de Biotecnología, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, C1113AAD Buenos Aires, Argentina
| | - Rodolfo M González-Lebrero
- Instituto de Química y Fisicoquímica Biológicas "Prof. Alejandro C. Paladini" (IQUIFIB, CONICET-UBA) and Departamento de Química Biológica, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, C1113AAD Buenos Aires, Argentina
| | - Carolina Osorio Larroche
- Laboratorio de Genética y Biología Molecular, Instituto de Inmunología, Genética y Metabolismo (INIGEM, CONICET-UBA), Hospital de Clínicas "José de San Martín", C1120AAR Buenos Aires, Argentina; Cátedra de Genética y Biología Molecular, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, C1113AAD Buenos Aires, Argentina
| | - Cintia E Citterio
- Laboratorio de Genética y Biología Molecular, Instituto de Inmunología, Genética y Metabolismo (INIGEM, CONICET-UBA), Hospital de Clínicas "José de San Martín", C1120AAR Buenos Aires, Argentina; Cátedra de Genética y Biología Molecular, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, C1113AAD Buenos Aires, Argentina
| | - Rogelio González-Sarmiento
- Unidad de Medicina Molecular-Departamento de Medicina, IBMCC and IBSAL, Universidad de Salamanca-CSIC, 37007 Salamanca, España
| | - María V Miranda
- Instituto de Nanobiotecnología (NANOBIOTEC, CONICET-UBA), Cátedra de Biotecnología, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, C1113AAD Buenos Aires, Argentina
| | - Héctor M Targovnik
- Laboratorio de Genética y Biología Molecular, Instituto de Inmunología, Genética y Metabolismo (INIGEM, CONICET-UBA), Hospital de Clínicas "José de San Martín", C1120AAR Buenos Aires, Argentina; Cátedra de Genética y Biología Molecular, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, C1113AAD Buenos Aires, Argentina
| | - Carina M Rivolta
- Laboratorio de Genética y Biología Molecular, Instituto de Inmunología, Genética y Metabolismo (INIGEM, CONICET-UBA), Hospital de Clínicas "José de San Martín", C1120AAR Buenos Aires, Argentina; Cátedra de Genética y Biología Molecular, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, C1113AAD Buenos Aires, Argentina.
| |
Collapse
|
102
|
Liu X, Yang Y, Zhang W, Sun Y, Peng F, Jeffrey L, Harvey L, McNeil B, Bai Z. Expression of recombinant protein using Corynebacterium Glutamicum: progress, challenges and applications. Crit Rev Biotechnol 2015; 36:652-64. [PMID: 25714007 DOI: 10.3109/07388551.2015.1004519] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Corynebacterium glutamicum (C. glutamicum) is a highly promising alternative prokaryotic host for recombinant protein expression, as it possesses several significant advantages over Escherichia coli (E. coli), the currently leading bacterial protein expression system. During the past decades, several experimental techniques and vector components for genetic manipulation of C. glutamicum have been developed and validated, including strong promoters for tightly regulating target gene expression, various types of plasmid vectors, protein secretion systems and methods of genetically modifying the host strain genome to improve protein production potential. This review critically discusses current progress in establishing C. glutamicum as a host for recombinant protein expression, and examines, in depth, some successful case studies of actual application of this expression system. The established "expression tool box" for developing novel constructs based on C. glutamicum as a host are also evaluated. Finally, the existing issues and solutions in process development with C. glutamicum as a host are specifically addressed.
Collapse
Affiliation(s)
- Xiuxia Liu
- a National Engineering Laboratory of Cereal Fermentation Technology , School of Biotechnology, JiangNan University , Wuxi , China and
| | - Yankun Yang
- a National Engineering Laboratory of Cereal Fermentation Technology , School of Biotechnology, JiangNan University , Wuxi , China and
| | - Wei Zhang
- a National Engineering Laboratory of Cereal Fermentation Technology , School of Biotechnology, JiangNan University , Wuxi , China and
| | - Yang Sun
- a National Engineering Laboratory of Cereal Fermentation Technology , School of Biotechnology, JiangNan University , Wuxi , China and
| | - Feng Peng
- a National Engineering Laboratory of Cereal Fermentation Technology , School of Biotechnology, JiangNan University , Wuxi , China and
| | - Laura Jeffrey
- b Institute of Pharmacy & Biomedical Sciences, Strathclyde University , Glasgow , UK
| | - Linda Harvey
- b Institute of Pharmacy & Biomedical Sciences, Strathclyde University , Glasgow , UK
| | - Brian McNeil
- b Institute of Pharmacy & Biomedical Sciences, Strathclyde University , Glasgow , UK
| | - Zhonghu Bai
- a National Engineering Laboratory of Cereal Fermentation Technology , School of Biotechnology, JiangNan University , Wuxi , China and
| |
Collapse
|
103
|
Pijlman GP. Enveloped virus-like particles as vaccines against pathogenic arboviruses. Biotechnol J 2015; 10:659-70. [DOI: 10.1002/biot.201400427] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2014] [Revised: 11/27/2014] [Accepted: 12/22/2014] [Indexed: 12/26/2022]
|
104
|
Pecora A, Malacari DA, Pérez Aguirreburualde MS, Bellido D, Escribano JM, Dus Santos MJ, Wigdorovitz A. Development of an enhanced bovine viral diarrhea virus subunit vaccine based on E2 glycoprotein fused to a single chain antibody which targets to antigen-presenting cells. Rev Argent Microbiol 2015; 47:4-8. [PMID: 25697468 DOI: 10.1016/j.ram.2014.12.004] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2014] [Revised: 12/22/2014] [Accepted: 12/26/2014] [Indexed: 11/28/2022] Open
Abstract
Bovine viral diarrhea virus (BVDV) is an important cause of economic losses worldwide. E2 is an immunodominant protein and a promising candidate to develop subunit vaccines. To improve its immunogenicity, a truncated E2 (tE2) was fused to a single chain antibody named APCH, which targets to antigen-presenting cells. APCH-tE2 and tE2 proteins were expressed in the baculovirus system and their immunogenicity was firstly compared in guinea pigs. APCH-tE2 vaccine was the best one to evoke a humoral response, and for this reason, it was selected for a cattle vaccination experiment. All the bovines immunized with 1.5 μg of APCH-tE2 developed high levels of neutralizing antibodies against BVDV up to a year post-immunization, demonstrating its significant potential as a subunit vaccine. This novel vaccine is undergoing scale-up and was transferred to the private sector. Nowadays, it is being evaluated for registration as the first Argentinean subunit vaccine for cattle.
Collapse
Affiliation(s)
- Andrea Pecora
- Instituto de Virología, CICVyA, INTA Castelar, Buenos Aires, Argentina.
| | - Darío A Malacari
- Instituto de Virología, CICVyA, INTA Castelar, Buenos Aires, Argentina
| | | | | | | | | | | |
Collapse
|
105
|
|
106
|
Palomares LA, Realpe M, Ramírez OT. An Overview of Cell Culture Engineering for the Insect Cell-Baculovirus Expression Vector System (BEVS). CELL ENGINEERING 2015. [DOI: 10.1007/978-3-319-10320-4_15] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
107
|
Palmberger D, Rendic D. SweetBac: Applying MultiBac Technology Towards Flexible Modification of Insect Cell Glycosylation. Methods Mol Biol 2015; 1321:153-169. [PMID: 26082221 DOI: 10.1007/978-1-4939-2760-9_11] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/04/2023]
Abstract
Observed different glycosylation patterns of glycoconjugates (recombinantly) produced in various eukaryotic organisms are a direct consequence of differences in numerous proteins involved in biosynthesis of the relevant glycan chains in these species. The need for efficient, robust and flexible methods for recombinant expression of proteins is met by the recently described MultiBac technology, an advanced and optimized baculovirus-based system for simultaneous recombinant protein expression in insect cells. A derivative of MultiBac technology, the SweetBac system aims at the modification of the glycosylation potential of insect cells as expression hosts. The application of SweetBac, including the methods needed to investigate the glycosylation pattern of the purified recombinant protein, is described in this chapter.
Collapse
Affiliation(s)
- Dieter Palmberger
- Department of Biotechnology, University of Natural Resources and Life Sciences, Vienna, Austria
| | | |
Collapse
|
108
|
Transfiguracion J, Manceur AP, Petiot E, Thompson CM, Kamen AA. Particle quantification of influenza viruses by high performance liquid chromatography. Vaccine 2015; 33:78-84. [DOI: 10.1016/j.vaccine.2014.11.027] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2014] [Revised: 11/08/2014] [Accepted: 11/15/2014] [Indexed: 01/02/2023]
|
109
|
Hennig R, Rapp E, Kottler R, Cajic S, Borowiak M, Reichl U. N-Glycosylation Fingerprinting of Viral Glycoproteins by xCGE-LIF. Methods Mol Biol 2015; 1331:123-43. [PMID: 26169738 DOI: 10.1007/978-1-4939-2874-3_8] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
The ongoing threat of pathogens, increasing resistance against antibiotics, and the risk of fast spreading of infectious diseases in a global community resulted in an intensified development of vaccines. Antigens used for vaccination comprise a wide variety of macromolecules including glycoproteins, lipopolysaccharides, and complex carbohydrates. For all of these antigens the sugar composition plays a crucial role for immunogenicity and protective efficacy of the vaccine. Here, we provide a protocol for N-glycosylation fingerprinting utilizing high performance multiplexed capillary gel electrophoresis with laser-induced fluorescence detection (xCGE-LIF) technology. The method described, enables to analyze the N-glycosylation of specific proteins out of a complex sample or even the total of all N-glycans contained in such a sample. The protocol is exemplarily demonstrated for N-glycosylation fingerprinting of cell culture-derived influenza A and B viruses and their major antigens, the membrane glycoproteins hemagglutinin and neuraminidase.
Collapse
Affiliation(s)
- René Hennig
- Max Planck Institute for Dynamics of Complex Technical Systems, Sandtorstrasse1, 39106, Magdeburg, Germany
| | | | | | | | | | | |
Collapse
|
110
|
Kaiser SC, Kraume M, Eibl D, Eibl R. Single-Use Bioreactors for Animal and Human Cells. CELL ENGINEERING 2015. [DOI: 10.1007/978-3-319-10320-4_14] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
|
111
|
Dragosits M, Yan S, Razzazi-Fazeli E, Wilson IBH, Rendic D. Enzymatic properties and subtle differences in the substrate specificity of phylogenetically distinct invertebrate N-glycan processing hexosaminidases. Glycobiology 2014; 25:448-64. [PMID: 25488985 PMCID: PMC4339880 DOI: 10.1093/glycob/cwu132] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Fused lobes (FDL) hexosaminidases are the most recently genetically defined glycosidases involved in the biosynthesis of N-glycans in invertebrates, and their narrow specificity is essential for the generation of paucimannosidic N-glycans in insects. In this study, we explored the potential of FDL hexosaminidases in the utilization of different artificial and natural substrates, both as purified, native compounds or generated in vitro using various relevant glycosyltransferases. In addition to the already-known FDL enzyme from Drosophila melanogaster, we now have identified and characterized the Apis mellifera FDL homolog. The enzymatic properties of the soluble forms of the affinity-purified insect FDL enzymes, expressed in both yeast and insect cells, were compared with those of the phylogenetically distinct recombinant Caenorhabditis elegans FDL-like enzymes and the N-acetylgalactosamine (GalNAc)-specific Caenorhabditis hexosaminidase HEX-4. In tests with a range of substrates, including natural N-glycans, we show that the invertebrate FDL(-like) enzymes are highly specific for N-acetylglucosamine attached to the α1,3-mannose, but under extreme conditions also remove other terminal GalNAc and N-acetylglucosamine residues. Recombinant FDL also proved useful in the analysis of complex mixtures of N-glycans originating from wild-type and mutant Caenorhabditis strains, thereby aiding isomeric definition of paucimannosidic and hybrid N-glycans in this organism. Furthermore, differences in activity and specificity were shown for two site-directed mutants of Drosophila FDL, compatible with the high structural similarity of chitinolytic and N-glycan degrading exohexosaminidases in insects. Our studies are another indication for the variety of structural and function aspects in the GH20 hexosaminidase family important for both catabolism and biosynthesis of glycoconjugates in eukaryotes.
Collapse
Affiliation(s)
- Martin Dragosits
- Department of Chemistry, University of Natural Resources and Life Sciences, Vienna VetCore Facility for Research, University of Veterinary Medicine, Vienna, Austria
| | - Shi Yan
- Department of Chemistry, University of Natural Resources and Life Sciences, Vienna
| | | | - Iain B H Wilson
- Department of Chemistry, University of Natural Resources and Life Sciences, Vienna
| | - Dubravko Rendic
- Department of Chemistry, University of Natural Resources and Life Sciences, Vienna
| |
Collapse
|
112
|
Contreras-Gómez A, Sánchez-Mirón A, García-Camacho F, Molina-Grima E, Chisti Y. Protein production using the baculovirus-insect cell expression system. Biotechnol Prog 2014; 30:1-18. [PMID: 24265112 DOI: 10.1002/btpr.1842] [Citation(s) in RCA: 103] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2013] [Revised: 11/12/2013] [Accepted: 11/12/2013] [Indexed: 12/21/2022]
Abstract
The baculovirus-insect cell expression system is widely used in producing recombinant proteins. This review is focused on the use of this expression system in developing bioprocesses for producing proteins of interest. The issues addressed include: the baculovirus biology and genetic manipulation to improve protein expression and quality; the suppression of proteolysis associated with the viral enzymes; the engineering of the insect cell lines for improved capability in glycosylation and folding of the expressed proteins; the impact of baculovirus on the host cell and its implications for protein production; the effects of the growth medium on metabolism of the host cell; the bioreactors and the associated operational aspects; and downstream processing of the product. All these factors strongly affect the production of recombinant proteins. The current state of knowledge is reviewed.
Collapse
|
113
|
Huber VC. Influenza vaccines: from whole virus preparations to recombinant protein technology. Expert Rev Vaccines 2014; 13:31-42. [PMID: 24192014 DOI: 10.1586/14760584.2014.852476] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
Vaccination against influenza represents our most effective form of prevention. Historical approaches toward vaccine creation and production have yielded highly effective vaccines that are safe and immunogenic. Despite their effectiveness, these historical approaches do not allow for the incorporation of changes into the vaccine in a timely manner. In 2013, a recombinant protein-based vaccine that induces immunity toward the influenza virus hemagglutinin was approved for use in the USA. This vaccine represents the first approved vaccine formulation that does not require an influenza virus intermediate for production. This review presents a brief history of influenza vaccines, with insight into the potential future application of vaccines generated using recombinant technology.
Collapse
Affiliation(s)
- Victor C Huber
- Division of Basic Biomedical Sciences, University of South Dakota, 414 E Clark Street, Vermillion, SD 57069, USA
| |
Collapse
|
114
|
Lin SY, Chung YC, Hu YC. Update on baculovirus as an expression and/or delivery vehicle for vaccine antigens. Expert Rev Vaccines 2014; 13:1501-21. [DOI: 10.1586/14760584.2014.951637] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
|
115
|
Wang YT, Chueh LL, Wan CH. An eight-year epidemiologic study based on baculovirus-expressed type-specific spike proteins for the differentiation of type I and II feline coronavirus infections. BMC Vet Res 2014; 10:186. [PMID: 25123112 PMCID: PMC4236817 DOI: 10.1186/s12917-014-0186-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2014] [Accepted: 08/11/2014] [Indexed: 01/19/2023] Open
Abstract
Background Feline infectious peritonitis (FIP) is a fatal disease caused by feline coronavirus (FCoV). FCoVs are divided into two serotypes with markedly different infection rates among cat populations around the world. A baculovirus-expressed type-specific domain of the spike proteins of FCoV was used to survey the infection of the two viruses over the past eight years in Taiwan. Results An immunofluorescence assay based on cells infected with the recombinant viruses that was capable of distinguishing between the two types of viral infection was established. A total of 833 cases from a teaching hospital was surveyed for prevalence of different FCoV infections. Infection of the type I FCoV was dominant, with a seropositive rate of 70.4%, whereas 3.5% of cats were infected with the type II FCoV. In most cases, results derived from serotyping and genotyping were highly agreeable. However, 16.7% (4/24) FIP cats and 9.8% (6/61) clinically healthy cats were found to possess antibodies against both viruses. Moreover, most of the cats (84.6%, 22/26) infected with a genotypic untypable virus bearing a type I FCoV antibody. Conclusion A relatively simple serotyping method to distinguish between two types of FCoV infection was developed. Based on this method, two types of FCoV infection in Taiwan was first carried out. Type I FCoV was found to be predominant compared with type II virus. Results derived from serotyping and genotyping support our current understanding of evolution of disease-related FCoV and transmission of FIP.
Collapse
Affiliation(s)
| | - Ling-Ling Chueh
- Graduate Institute of Veterinary Medicine, School of Veterinary Medicine, National Taiwan University, Taipei 10617, Taiwan.
| | | |
Collapse
|
116
|
Self-assembly and release of peste des petits ruminants virus-like particles in an insect cell-baculovirus system and their immunogenicity in mice and goats. PLoS One 2014; 9:e104791. [PMID: 25117931 PMCID: PMC4130610 DOI: 10.1371/journal.pone.0104791] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2014] [Accepted: 07/14/2014] [Indexed: 11/19/2022] Open
Abstract
Peste des petits ruminants (PPR) is an acute, febrile, viral disease of small ruminants that has a significant economic impact. For many viral diseases, vaccination with virus-like particles (VLPs) has shown considerable promise as a prophylactic approach; however, the processes of assembly and release of peste des petits ruminants virus (PPRV) VLPs are not well characterized, and their immunogenicity in the host is unknown. In this study, VLPs of PPRV were generated in a baculovirus system through simultaneous expression of PPRV matrix (M) protein and hemaglutin in (H) or fusion (F) protein. The released VLPs showed morphology similar to that of the native virus particles. Subcutaneous injection of these VLPs (PPRV-H, PPRV-F) into mice and goats elicited PPRV-specific IgG production, increased the levels of virus neutralizing antibodies, and promoted lymphocyte proliferation. Without adjuvants, the immune response induced by the PPRV-H VLPs was comparable to that obtained using equivalent amounts of PPRV vaccine. Thus, our results demonstrated that VLPs containing PPRV M protein and H or F protein are potential “differentiating infected from vaccinated animals” (DIVA) vaccine candidates for the surveillance and eradication of PPR.
Collapse
|
117
|
Gómez-Sebastián S, López-Vidal J, Escribano JM. Significant productivity improvement of the baculovirus expression vector system by engineering a novel expression cassette. PLoS One 2014; 9:e96562. [PMID: 24824596 PMCID: PMC4019511 DOI: 10.1371/journal.pone.0096562] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2014] [Accepted: 04/08/2014] [Indexed: 12/21/2022] Open
Abstract
Here we describe the development of a baculovirus vector expression cassette containing rearranged baculovirus-derived genetic regulatory elements. This newly designed expression cassette conferred significant production improvements to the baculovirus expression vector system (BEVS), including prolonged cell integrity after infection, improved protein integrity, and around 4-fold increase in recombinant protein production yields in insect cells with respect to a standard baculovirus vector. The expression cassette consisted of a cDNA encoding for the baculovirus transactivation factors IE1 and IE0, expressed under the control of the polyhedrin promoter, and a homologous repeated transcription enhancer sequence operatively cis-linked to the p10 promoter or to chimeric promoters containing p10. The prolonged cell integrity observed in cells infected by baculoviruses harbouring the novel expression cassette reduced the characteristic proteolysis and aberrant forms frequently found in baculovirus-derived recombinant proteins. The new expression cassette developed here has the potential to significantly improve the productivity of the BEVS.
Collapse
Affiliation(s)
| | | | - José M. Escribano
- Department of Biotechnology, Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria (INIA), Madrid, Spain
| |
Collapse
|
118
|
Elmgren L, Li X, Wilson C, Ball R, Wang J, Cichutek K, Pfleiderer M, Kato A, Cavaleri M, Southern J, Jivapaisarnpong T, Minor P, Griffiths E, Sohn Y, Wood D. A global regulatory science agenda for vaccines. Vaccine 2014; 31 Suppl 2:B163-75. [PMID: 23598478 DOI: 10.1016/j.vaccine.2012.10.117] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2012] [Revised: 10/22/2012] [Accepted: 10/31/2012] [Indexed: 02/06/2023]
Abstract
The Decade of Vaccines Collaboration and development of the Global Vaccine Action Plan provides a catalyst and unique opportunity for regulators worldwide to develop and propose a global regulatory science agenda for vaccines. Regulatory oversight is critical to allow access to vaccines that are safe, effective, and of assured quality. Methods used by regulators need to constantly evolve so that scientific and technological advances are applied to address challenges such as new products and technologies, and also to provide an increased understanding of benefits and risks of existing products. Regulatory science builds on high-quality basic research, and encompasses at least two broad categories. First, there is laboratory-based regulatory science. Illustrative examples include development of correlates of immunity; or correlates of safety; or of improved product characterization and potency assays. Included in such science would be tools to standardize assays used for regulatory purposes. Second, there is science to develop regulatory processes. Illustrative examples include adaptive clinical trial designs; or tools to analyze the benefit-risk decision-making process of regulators; or novel pharmacovigilance methodologies. Included in such science would be initiatives to standardize regulatory processes (e.g., definitions of terms for adverse events [AEs] following immunization). The aim of a global regulatory science agenda is to transform current national efforts, mainly by well-resourced regulatory agencies, into a coordinated action plan to support global immunization goals. This article provides examples of how regulatory science has, in the past, contributed to improved access to vaccines, and identifies gaps that could be addressed through a global regulatory science agenda. The article also identifies challenges to implementing a regulatory science agenda and proposes strategies and actions to fill these gaps. A global regulatory science agenda will enable regulators, academics, and other stakeholders to converge around transformative actions for innovation in the regulatory process to support global immunization goals.
Collapse
|
119
|
Wagner JM, Pajerowski JD, Daniels CL, McHugh PM, Flynn JA, Balliet JW, Casimiro DR, Subramanian S. Enhanced production of Chikungunya virus-like particles using a high-pH adapted spodoptera frugiperda insect cell line. PLoS One 2014; 9:e94401. [PMID: 24713807 PMCID: PMC3979839 DOI: 10.1371/journal.pone.0094401] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2014] [Accepted: 03/14/2014] [Indexed: 11/18/2022] Open
Abstract
Chikungunya virus-like particles (VLPs) have potential to be used as a prophylactic vaccine based on testing in multiple animal models and are currently being evaluated for human use in a Phase I clinical trial. The current method for producing these enveloped alphavirus VLPs by transient gene expression in mammalian cells presents challenges for scalable and robust industrial manufacturing, so the insect cell baculovirus expression vector system was evaluated as an alternative expression technology. Subsequent to recombinant baculovirus infection of Sf21 cells in standard culture media (pH 6.2–6.4), properly processed Chikungunya structural proteins were detected and assembled capsids were observed. However, an increase in culture pH to 6.6–6.8 was necessary to produce detectable concentrations of assembled VLPs. Since this elevated production pH exceeds the optimum for growth medium stability and Sf21 culture, medium modifications were made and a novel insect cell variant (SfBasic) was derived by exposure of Sf21 to elevated culture pH for a prolonged period of time. The high-pH adapted SfBasic insect cell line described herein is capable of maintaining normal cell growth into the typical mammalian cell culture pH range of 7.0–7.2 and produces 11-fold higher Chikungunya VLP yields relative to the parental Sf21 cell line. After scale-up into stirred tank bioreactors, SfBasic derived VLPs were chromatographically purified and shown to be similar in size and structure to a VLP standard derived from transient gene expression in HEK293 cells. Total serum anti-Chikungunya IgG and neutralizing titers from guinea pigs vaccinated with SfBasic derived VLPs or HEK293 derived VLPs were not significantly different with respect to production method, suggesting that this adapted insect cell line and production process could be useful for manufacturing Chikungunya VLPs for use as a vaccine. The adaptation of Sf21 to produce high levels of recombinant protein and VLPs in an elevated pH range may also have applications for other pH-sensitive protein or VLP targets.
Collapse
Affiliation(s)
- James M. Wagner
- Vaccine Research and Development, Merck Research Laboratories, Merck & Co., Inc., West Point, Pennsylvania, United States of America
| | - J. David Pajerowski
- Vaccine Research and Development, Merck Research Laboratories, Merck & Co., Inc., West Point, Pennsylvania, United States of America
| | - Christopher L. Daniels
- Vaccine Research and Development, Merck Research Laboratories, Merck & Co., Inc., West Point, Pennsylvania, United States of America
| | - Patrick M. McHugh
- Vaccine Research and Development, Merck Research Laboratories, Merck & Co., Inc., West Point, Pennsylvania, United States of America
| | - Jessica A. Flynn
- Vaccine Research and Development, Merck Research Laboratories, Merck & Co., Inc., West Point, Pennsylvania, United States of America
| | - John W. Balliet
- Vaccine Research and Development, Merck Research Laboratories, Merck & Co., Inc., West Point, Pennsylvania, United States of America
| | - Danilo R. Casimiro
- Vaccine Research and Development, Merck Research Laboratories, Merck & Co., Inc., West Point, Pennsylvania, United States of America
| | - Shyamsundar Subramanian
- Vaccine Research and Development, Merck Research Laboratories, Merck & Co., Inc., West Point, Pennsylvania, United States of America
- * E-mail:
| |
Collapse
|
120
|
Abstract
The challenges in successful vaccination against influenza using conventional approaches lie in their variable efficacy in different age populations, the antigenic variability of the circulating virus, and the production and manufacturing limitations to ensure safe, timely, and adequate supply of vaccine. The conventional influenza vaccine platform is based on stimulating immunity against the major neutralizing antibody target, hemagglutinin (HA), by virus attenuation or inactivation. Improvements to this conventional system have focused primarily on improving production and immunogenicity. Cell culture, reverse genetics, and baculovirus expression technology allow for safe and scalable production, while adjuvants, dose variation, and alternate routes of delivery aim to improve vaccine immunogenicity. Fundamentally different approaches that are currently under development hope to signal new generations of influenza vaccines. Such approaches target nonvariable regions of antigenic proteins, with the idea of stimulating cross-protective antibodies and thus creating a "universal" influenza vaccine. While such approaches have obvious benefits, there are many hurdles yet to clear. Here, we discuss the process and challenges of the current influenza vaccine platform as well as new approaches that are being investigated based on the same antigenic target and newer technologies based on different antigenic targets.
Collapse
|
121
|
Yamaji H. Suitability and perspectives on using recombinant insect cells for the production of virus-like particles. Appl Microbiol Biotechnol 2014; 98:1963-70. [PMID: 24407451 DOI: 10.1007/s00253-013-5474-9] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2013] [Revised: 12/12/2013] [Accepted: 12/14/2013] [Indexed: 10/25/2022]
Abstract
Virus-like particles (VLPs) can be produced in recombinant protein production systems by expressing viral surface proteins that spontaneously assemble into particulate structures similar to authentic viral or subviral particles. VLPs serve as excellent platforms for the development of safe and effective vaccines and diagnostic antigens. Among various recombinant protein production systems, the baculovirus-insect cell system has been used extensively for the production of a wide variety of VLPs. This system is already employed for the manufacture of a licensed human papillomavirus-like particle vaccine. However, the baculovirus-insect cell system has several inherent limitations including contamination of VLPs with progeny baculovirus particles. Stably transformed insect cells have emerged as attractive alternatives to the baculovirus-insect cell system. Different types of VLPs, with or without an envelope and composed of either single or multiple structural proteins, have been produced in stably transformed insect cells. VLPs produced by stably transformed insect cells have successfully elicited immune responses in vivo. In some cases, the yield of VLPs attained with recombinant insect cells was comparable to, or higher than, that obtained by baculovirus-infected insect cells. Recombinant insect cells offer a promising approach to the development and production of VLPs.
Collapse
Affiliation(s)
- Hideki Yamaji
- Department of Chemical Science and Engineering, Graduate School of Engineering, Kobe University, 1-1 Rokkodai, Nada, Kobe, 657-8501, Japan,
| |
Collapse
|
122
|
Thomas S, Luxon BA. Vaccines based on structure-based design provide protection against infectious diseases. Expert Rev Vaccines 2014; 12:1301-11. [DOI: 10.1586/14760584.2013.840092] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
123
|
Enhancing the work of the Department of Health and Human Services national vaccine program in global immunization: recommendations of the National Vaccine Advisory Committee: approved by the National Vaccine Advisory Committee on September 12, 2013. Public Health Rep 2014; 129 Suppl 3:12-85. [PMID: 25100887 PMCID: PMC4121882 DOI: 10.1177/00333549141295s305] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
|
124
|
|
125
|
Gutiérrez-Granados S, Cervera L, Gòdia F, Carrillo J, Segura MM. Development and validation of a quantitation assay for fluorescently tagged HIV-1 virus-like particles. J Virol Methods 2013; 193:85-95. [DOI: 10.1016/j.jviromet.2013.05.010] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2013] [Revised: 04/24/2013] [Accepted: 05/08/2013] [Indexed: 02/03/2023]
|
126
|
Yang LPH. Recombinant trivalent influenza vaccine (flublok(®)): a review of its use in the prevention of seasonal influenza in adults. Drugs 2013; 73:1357-66. [PMID: 23928902 DOI: 10.1007/s40265-013-0103-6] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Flublok(®) is a trivalent influenza vaccine manufactured using the baculovirus-insect cell system, which has a number of differences compared with the traditional embryonated chicken egg system. Flublok(®) is the first vaccine containing recombinant protein to be approved for the prevention of seasonal influenza, and is indicated in the USA in adults aged 18-49 years. In a large, placebo-controlled, phase III trial in individuals aged 18-49 years, Flublok(®) met the US FDA requirements for immunogenicity, effectiveness and safety, despite a high incidence of antigenic mismatch between confirmed cases of influenza and the vaccine component strains. Flublok(®) was also compared with Fluzone(®) in two noninferiority trials in older adults aged 50-64 or ≥65 years; although noninferiority was shown for a number of endpoints in these trials, Flublok(®) is not currently approved in these age groups. Flublok(®) is a useful and generally well-tolerated vaccination option for the prevention of seasonal influenza in adults aged 18-49 years, including those with egg allergy.
Collapse
Affiliation(s)
- Lily P H Yang
- Adis, 41 Centorian Drive, Private Bag 65901, Mairangi Bay, North Shore, 0754 Auckland, New Zealand.
| |
Collapse
|
127
|
Au S, Wu W, Panté N. Baculovirus nuclear import: open, nuclear pore complex (NPC) sesame. Viruses 2013; 5:1885-900. [PMID: 23881277 PMCID: PMC3738967 DOI: 10.3390/v5071885] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2013] [Revised: 07/17/2013] [Accepted: 07/17/2013] [Indexed: 01/16/2023] Open
Abstract
Baculoviruses are one of the largest viruses that replicate in the nucleus of their host cells. During infection, the rod-shape, 250-nm long nucleocapsid delivers its genome into the nucleus. Electron microscopy evidence suggests that baculoviruses, specifically the Alphabaculoviruses (nucleopolyhedroviruses) and the Betabaculoviruses (granuloviruses), have evolved two very distinct modes for doing this. Here we review historical and current experimental results of baculovirus nuclear import studies, with an emphasis on electron microscopy studies employing the prototypical baculovirus Autographa californica multiple nucleopolyhedrovirus infecting cultured cells. We also discuss the implications of recent studies towards theories of nuclear transport mechanisms.
Collapse
Affiliation(s)
| | | | - Nelly Panté
- Author to whom correspondence should be addressed; E-Mail: ; Tel.: +1-604-822-3369; Fax: +1-604-822-2416
| |
Collapse
|
128
|
|
129
|
Yamaji H, Konishi E. Production of Japanese encephalitis virus-like particles in insect cells. Bioengineered 2013; 4:438-42. [PMID: 23639981 DOI: 10.4161/bioe.24514] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Virus-like particles (VLPs) are composed of one or several recombinant viral surface proteins that spontaneously assemble into particulate structures without the incorporation of virus DNA or RNA. The baculovirus-insect cell system has been used extensively for the production of recombinant virus proteins including VLPs. While the baculovirus-insect cell system directs the transient expression of recombinant proteins in a batch culture, stably transformed insect cells allow constitutive production. In our recent study, a secretory form of Japanese encephalitis (JE) VLPs was successfully produced by Trichoplusia ni BTI-TN-5B1-4 (High Five) cells engineered to coexpress the JE virus (JEV) premembrane (prM) and envelope (E) proteins. A higher yield of E protein was attained with recombinant High Five cells than with the baculovirus-insect cell system. This study demonstrated that recombinant insect cells offer a promising approach to the high-level production of VLPs for use as vaccines and diagnostic antigens.
Collapse
Affiliation(s)
- Hideki Yamaji
- Department of Chemical Science and Engineering; Graduate School of Engineering; Kobe University; Kobe, Japan; BIKEN Endowed Department of Dengue Vaccine Development; Faculty of Tropical Medicine; Mahidol University; Bangkok, Thailand
| | - Eiji Konishi
- BIKEN Endowed Department of Dengue Vaccine Development; Faculty of Tropical Medicine; Mahidol University; Bangkok, Thailand
| |
Collapse
|
130
|
Hyper-enhanced production of foreign recombinant protein by fusion with the partial polyhedrin of nucleopolyhedrovirus. PLoS One 2013; 8:e60835. [PMID: 23593321 PMCID: PMC3621880 DOI: 10.1371/journal.pone.0060835] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2012] [Accepted: 03/03/2013] [Indexed: 11/19/2022] Open
Abstract
To enhance the production efficiency of foreign protein in baculovirus expression systems, the effects of polyhedrin fragments were investigated by fusion expressing them with the enhanced green fluorescent protein (EGFP). Recombinant viruses were generated to express EGFP fused with polyhedrin fragments based on the previously reported minimal region for self-assembly and the KRKK nuclear localization signal (NLS). Fusion expressions with polyhedrin amino acids 19 to 110 and 32 to 110 lead to localization of recombinant protein into the nucleus and mediate its assembly. The marked increase of EGFP by these fusion expressions was confirmed through protein and fluorescence intensity analyses. The importance of nuclear localization for enhanced production was shown by the mutation of the NLS within the fused polyhedrin fragment. In addition, when the polyhedrin fragment fused with EGFP was not localized in the nucleus, some fragments increased the production of protein. Among these fragments, some degradation of only the fused polyhedrin was observed in the fusion of amino acids 19 to 85 and 32 to 85. The fusion of amino acids 32 to 85 may be more useful for the enhanced and intact production of recombinant protein. The production of E2 protein, which is a major antigen of classical swine fever virus, was dramatically increased by fusion expression with polyhedrin amino acids 19 to 110, and its preliminary immunogenicity was verified using experimental guinea pigs. This study suggests a new option for higher expression of useful foreign recombinant protein by using the partial polyhedrin in baculovirus.
Collapse
|
131
|
MacLeod MKL, David A, Jin N, Noges L, Wang J, Kappler JW, Marrack P. Influenza nucleoprotein delivered with aluminium salts protects mice from an influenza A virus that expresses an altered nucleoprotein sequence. PLoS One 2013; 8:e61775. [PMID: 23613928 PMCID: PMC3629017 DOI: 10.1371/journal.pone.0061775] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2012] [Accepted: 03/13/2013] [Indexed: 11/19/2022] Open
Abstract
Influenza virus poses a difficult challenge for protective immunity. This virus is adept at altering its surface proteins, the proteins that are the targets of neutralizing antibody. Consequently, each year a new vaccine must be developed to combat the current recirculating strains. A universal influenza vaccine that primes specific memory cells that recognise conserved parts of the virus could prove to be effective against both annual influenza variants and newly emergent potentially pandemic strains. Such a vaccine will have to contain a safe and effective adjuvant that can be used in individuals of all ages. We examine protection from viral challenge in mice vaccinated with the nucleoprotein from the PR8 strain of influenza A, a protein that is highly conserved across viral subtypes. Vaccination with nucleoprotein delivered with a universally used and safe adjuvant, composed of insoluble aluminium salts, provides protection against viruses that either express the same or an altered version of nucleoprotein. This protection correlated with the presence of nucleoprotein specific CD8 T cells in the lungs of infected animals at early time points after infection. In contrast, immunization with NP delivered with alum and the detoxified LPS adjuvant, monophosphoryl lipid A, provided some protection to the homologous viral strain but no protection against infection by influenza expressing a variant nucleoprotein. Together, these data point towards a vaccine solution for all influenza A subtypes.
Collapse
Affiliation(s)
- Megan K. L. MacLeod
- Howard Hughes Medical Institute and Integrated Department of Immunology, National Jewish Health, Denver, Colorado, United States of America
- * E-mail: (PM); (MKLM)
| | - Alexandria David
- Howard Hughes Medical Institute and Integrated Department of Immunology, National Jewish Health, Denver, Colorado, United States of America
| | - Niyun Jin
- Howard Hughes Medical Institute and Integrated Department of Immunology, National Jewish Health, Denver, Colorado, United States of America
| | - Laura Noges
- Howard Hughes Medical Institute and Integrated Department of Immunology, National Jewish Health, Denver, Colorado, United States of America
| | - Jieru Wang
- Department of Medicine, National Jewish Health, Denver, Colorado, United States of America
| | - John W. Kappler
- Howard Hughes Medical Institute and Integrated Department of Immunology, National Jewish Health, Denver, Colorado, United States of America
- Program in Biomolecular Structure, University of Colorado Denver, School of Medicine, Aurora, Colorado, United States of America
- Department of Medicine, University of Colorado Denver, School of Medicine, Aurora, Colorado, United States of America
| | - Philippa Marrack
- Howard Hughes Medical Institute and Integrated Department of Immunology, National Jewish Health, Denver, Colorado, United States of America
- Program in Biomolecular Structure, University of Colorado Denver, School of Medicine, Aurora, Colorado, United States of America
- Department of Biochemistry and Molecular Genetics, University of Colorado Denver, School of Medicine, Aurora, Colorado, United States of America
- * E-mail: (PM); (MKLM)
| |
Collapse
|
132
|
Gaunitz S, Jin C, Nilsson A, Liu J, Karlsson NG, Holgersson J. Mucin-type proteins produced in the Trichoplusia ni and Spodoptera frugiperda insect cell lines carry novel O-glycans with phosphocholine and sulfate substitutions. Glycobiology 2013; 23:778-96. [PMID: 23463814 DOI: 10.1093/glycob/cwt015] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
The O-glycans of a recombinant mucin-type protein expressed in insect cell lines derived from Trichoplusia ni (Hi-5) and Spodoptera frugiperda (Sf9) were characterized. The P-selectin glycoprotein ligand-1/mouse IgG2b (PSGL-1/mIgG2b) fusion protein carrying 106 potential O-glycosylation sites and 6 potential N-glycosylation sites was expressed and purified from the Hi-5 and Sf9 cell culture medium using affinity chromatography and gel filtration. Liquid chromatography mass spectrometry (LC-MS) of O-glycans released from PSGL-1/mIgG2b revealed a large repertoire of structurally diverse glycans, which is in contrast to previous reports of only simple glycans. O-Glycans containing hexuronic acid (HexA, here glucuronic acid and galacturonic acid) were found to be prevalent. Also sulfate (Hi-5 and Sf9) and phosphocholine (PC; Sf9) O-glycan substitutions were detected. Western blotting confirmed the presence of O-linked PC on PSGL-1/mIG2b produced in Sf9 cells. To our knowledge, this is the first structural characterization of PC-substituted O-glycans in any species. The MS analyses revealed that Sf9 oligosaccharides consisted of short oligosaccharides (<6 residues) low in hexose (Hex) and with terminating N-acetylhexosamine (HexNAc) units, whereas Hi-5 produced a family of large O-glycans with (HexNAc-HexA-Hex) repeats and sulfate substitution on terminal residues. In both cell lines, the core N-acetylgalactosamine was preferentially non-branched, but small amounts of O-glycan cores with single fucose or hexose branches were found.
Collapse
Affiliation(s)
- Stefan Gaunitz
- Division of Clinical Immunology and Transfusion Medicine, Department of Laboratory Medicine, Karolinska Institute, Karolinska University Hospital at Huddinge, SE-141 86 Huddinge, Sweden.
| | | | | | | | | | | |
Collapse
|
133
|
Hwang I, Choi D, See H, Kim W, Chung IS, Shon DH. Beneficial effects of the mixed adjuvant of CpG plus monophosphoryl lipid a in immunization with a recombinant protein vaccine for hepatitis A. ACTA ACUST UNITED AC 2013. [DOI: 10.1007/s13765-012-2398-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
|
134
|
Baculovirus replication induces the expression of heat shock proteins in vivo and in vitro. Arch Virol 2013; 158:1517-22. [PMID: 23443933 DOI: 10.1007/s00705-013-1640-8] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2012] [Accepted: 01/10/2013] [Indexed: 10/27/2022]
Abstract
A recent handful of studies have linked baculovirus infection with the induction of heat shock proteins, a highly conserved family of cytoprotective proteins. Here, we demonstrate baculovirus-stimulated upregulation of hsp70 transcription in the natural host, Helicoverpa zea. Larvae lethally infected with Helicoverpa zea single nucleopolyhedrovirus (HzSNPV) accumulated hsp70 transcripts throughout the 72-hour course of infection in the midgut, hemocytes, and fat body. While a maximal 17- or 15-fold induction of hsp70 was noted in the midgut and hemocytes, respectively, by 72 hours postinfection, the level of hsp70 transcription in the fat body of larvae was greater than two orders of magnitude higher than in mock-infected larvae. These results were largely mirrored in cultures of infected cells, and a potentiation effect was observed in cells that were both heat shocked and infected. In contrast, Spodoptera frugiperda multiple nucleopolyhedrovirus and ultraviolet-inactivated HzSNPV did not stimulate hsp70 transcription in these non-permissive larvae and in cell culture, respectively. Taken together, this report documents baculovirus-mediated upregulation of hsp70 in the host and demonstrates the requirement for productive infection for hsp70 induction in vitro and in vivo.
Collapse
|
135
|
Moyle PM, Hartas J, Henningham A, Batzloff MR, Good MF, Toth I. An efficient, chemically-defined semisynthetic lipid-adjuvanted nanoparticulate vaccine development system. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2013; 9:935-44. [PMID: 23428988 DOI: 10.1016/j.nano.2013.01.009] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/02/2012] [Revised: 11/23/2012] [Accepted: 01/23/2013] [Indexed: 01/22/2023]
Abstract
UNLABELLED A novel vaccine development platform that enables the site-specific conjugation of synthetic lipid adjuvants to recombinant proteins was produced. This technology facilitates the simple and efficient production of homogeneous, chemically-defined, semisynthetic lipoprotein vaccines. Using a polytope 'string-of-beads' approach, a synthetic gene incorporating seven Streptococcus pyogenes M protein strain-specific antigens, and a conserved M protein antigen (J14) was produced, expressed, and attached to a lipoamino acid based adjuvant (lipid core peptide; LCP). Nanoparticles (40 nm diameter) of an optimal size for stimulating antibody-mediated immunity were formed upon the addition of these lipoproteins to aqueous buffer (PBS). Systemic antigen-specific IgG antibodies were raised against all eight antigens in C57BL/6J mice, without the need to formulate with additional adjuvant. These antibodies bound cell surface M proteins of S. pyogenes strains represented within the polytope sequence, with higher antibody levels observed where a dendritic cell targeting peptide (DCpep) was incorporated within the LCP adjuvant. FROM THE CLINICAL EDITOR In this study, a novel vaccine development system is presented, combining adjuvants with recombinant protein antigens, and presenting the antigen in a nanoparticle system optimized for antibody production. They demonstrate efficient vaccination in a murine model system without the need for additional adjuvants.
Collapse
Affiliation(s)
- Peter M Moyle
- School of Chemistry and Molecular Biosciences, The University of Queensland, St Lucia 4072, QLD, Australia; Institute for Molecular Biosciences, The University of Queensland, St Lucia 4072, QLD, Australia.
| | | | | | | | | | | |
Collapse
|
136
|
Moyle PM, Toth I. Modern subunit vaccines: development, components, and research opportunities. ChemMedChem 2013; 8:360-76. [PMID: 23316023 DOI: 10.1002/cmdc.201200487] [Citation(s) in RCA: 337] [Impact Index Per Article: 28.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2012] [Revised: 12/08/2012] [Indexed: 12/11/2022]
Abstract
Traditional vaccines, based on the administration of killed or attenuated microorganisms, have proven to be among the most effective methods for disease prevention. Safety issues related to administering these complex mixtures, however, prevent their universal application. Through identification of the microbial components responsible for protective immunity, vaccine formulations can be simplified, enabling molecular-level vaccine characterization, improved safety profiles, prospects to develop new high-priority vaccines (e.g. for HIV, tuberculosis, and malaria), and the opportunity for extensive vaccine component optimization. This subunit approach, however, comes at the expense of decreased immunity, requiring the addition of immunostimulatory agents (adjuvants). As few adjuvants are currently used in licensed vaccines, adjuvant development represents an exciting area for medicinal chemists to play a role in the future of vaccine development. In addition, immune responses can be further customized though optimization of delivery systems, tuning the size of particulate vaccines, targeting specific cells of the immune system (e.g. dendritic cells), and adding components to aid vaccine efficacy in whole immunized populations (e.g. promiscuous T-helper epitopes). Herein we review the current state of the art and future direction in subunit vaccine development, with a focus on the described components and their potential to steer the immune response toward a desired response.
Collapse
Affiliation(s)
- Peter Michael Moyle
- School of Chemistry and Molecular Biosciences, The University of Queensland, St. Lucia, QLD 4072, Australia.
| | | |
Collapse
|
137
|
Chen CY, Lin SY, Cheng MC, Tsai CP, Hung CL, Lo KW, Hwang Y, Hu YC. Baculovirus vector as an avian influenza vaccine: hemagglutinin expression and presentation augment the vaccine immunogenicity. J Biotechnol 2013; 164:143-50. [PMID: 23313887 DOI: 10.1016/j.jbiotec.2012.12.013] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2012] [Revised: 12/17/2012] [Accepted: 12/21/2012] [Indexed: 12/11/2022]
Abstract
Baculovirus simultaneously displaying and expressing the avian influenza virus (AIV) hemagglutinin (HA) protein can induce potent anti-HA humoral and cellular immune responses. Based on the hypothesis that improving the antigen expression and presentation can further boost the AIV vaccine efficacies, we first constructed a baculoviral vector (Bac-HAW) with HA gene fused with the woodchuck hepatitis virus post-transcriptional regulatory element (WPRE) near its 3' end and expressed under the control of the hybrid CAG promoter. The WPRE fusion improved the HA expression and augmented the humoral and Th1 cellular immune responses after intramuscular administration into BALB/c mice. With Bac-HAW as the backbone, we next constructed Bac-HAMW which harbored the HA gene flanked with the signal sequence (MHCIss) and trafficking domain (MITD) of MHC class I molecule. In comparison with Bac-HAW, Bac-HAMW ameliorated the HA peptide presentation, significantly elevated the HA-specific humoral response (total IgG, IgG2a and hemagglutination inhibition titers) and favorably boosted the Th1 and IFN-γ(+)/CD8(+) T cell responses without extraneous adjuvants. These data collectively confirmed that enhancement of antigen expression and presentation by combining the WPRE and MHCIss/MITD fusion can potentiate the immunogenicity of the baculovirus-based vaccine, and implicates the potential of Bac-HAMW as an appealing AIV vaccine.
Collapse
Affiliation(s)
- Chi-Yuan Chen
- Department of Chemical Engineering, National Tsing Hua University, Hsinchu 30013, Taiwan
| | | | | | | | | | | | | | | |
Collapse
|
138
|
Abstract
Virus-like particles (VLPs) are formed by viral structural proteins that, when overexpressed, spontaneously self-assemble into particles that are antigenically indistinguishable from infectious virus or subviral particles. VLPs are appealing as vaccine candidates because their inherent properties (i.e., virus-sized, multimeric antigens, highly organised and repetitive structure, not infectious) are suitable for the induction of safe and efficient humoral and cellular immune responses. VLP-based vaccines have already been licensed for human and veterinary use, and many more vaccine candidates are currently in late stages of evaluation. Moreover, the development of VLPs as platforms for foreign antigen display has further broadened their potential applicability both as prophylactic and therapeutic vaccines. This chapter provides an overview on the design and use of VLPs for the development of new generation vaccines.
Collapse
Affiliation(s)
- Juan Bárcena
- Centro de Investigación en Sanidad Animal (INIA), Valdeolmos, 28130, Madrid, Spain,
| | | |
Collapse
|
139
|
Optimization of Insect Cell Based Protein Production Processes - Online Monitoring, Expression Systems, Scale Up. YELLOW BIOTECHNOLOGY II 2013; 136:65-100. [DOI: 10.1007/10_2013_205] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
|
140
|
Genzel Y, Frensing T, Reichl U. Herstellung moderner Grippeimpfstoffe. CHEM UNSERER ZEIT 2012. [DOI: 10.1002/ciuz.201200606] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
|
141
|
Yamaji H, Segawa M, Nakamura M, Katsuda T, Kuwahara M, Konishi E. Production of Japanese encephalitis virus-like particles using the baculovirus–insect cell system. J Biosci Bioeng 2012; 114:657-62. [DOI: 10.1016/j.jbiosc.2012.06.012] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2012] [Revised: 06/05/2012] [Accepted: 06/22/2012] [Indexed: 11/29/2022]
|
142
|
Mohana Subramanian B, Madhanmohan M, Sriraman R, Chandrasekhar Reddy RV, Yuvaraj S, Manikumar K, Rajalakshmi S, Nagendrakumar SB, Rana SK, Srinivasan VA. Development of foot-and-mouth disease virus (FMDV) serotype O virus-like-particles (VLPs) vaccine and evaluation of its potency. Antiviral Res 2012; 96:288-95. [PMID: 23043941 DOI: 10.1016/j.antiviral.2012.09.019] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2012] [Revised: 09/25/2012] [Accepted: 09/27/2012] [Indexed: 10/27/2022]
Abstract
Foot-and-mouth disease (FMD) is an economically significant viral disease that rampage dairy and other livestock industries in many countries. The disease is being controlled by the use of an inactivated vaccine. However, a recombinant marker vaccine, which avoids the use of live virus, may be an option for the unambiguous differentiation of infected animals from vaccinated animals. A recombinant baculovirus clone containing P1-2A-3C coding sequences of foot-and-mouth disease virus (FMDV) serotype O(1) Manisa was generated. The FMDV structural proteins along with the 3C protease were expressed in Sf9 cells and the generation of virus like particles (VLP) was studied. The recombinant protein was formulated as vaccine using an oil adjuvant, ISA 206 and potency of the vaccine was tested in cattle. The vaccine had a potency value (PD(50)) of 5.01 and most of the vaccinated animals exhibited neutralizing antibody titers after two immunizations.
Collapse
Affiliation(s)
- B Mohana Subramanian
- Research and Development Centre, Indian Immunologicals Limited, Gachibowli, Hyderabad 500032, India
| | | | | | | | | | | | | | | | | | | |
Collapse
|
143
|
Lai YK, Hsu JTA, Chu CC, Chang TY, Pan KL, Lin CC. Enhanced recombinant protein production and differential expression of molecular chaperones in sf-caspase-1-repressed stable cells after baculovirus infection. BMC Biotechnol 2012; 12:83. [PMID: 23134743 PMCID: PMC3505465 DOI: 10.1186/1472-6750-12-83] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2012] [Accepted: 11/04/2012] [Indexed: 12/27/2022] Open
Abstract
Background There are few studies that have examined the potential of RNA inference (RNAi) to increase protein production in the baculovirus expression vector system (BEVS). Spodoptera frugiperda (fall armyworm) (Sf)-caspase-1-repressed stable cells exhibit resistance to apoptosis and enhancement of recombinant protein production. However, the mechanism of recombinant protein augmentation in baculovirus-infected Caspase-repressed insect cells has not been elucidated. Results In the current study, we utilized RNAi-mediated Sf-caspase-1-repressed stable cells to clarify how the resistance to apoptosis can enhance both intracellular (firefly luciferase) and extracellular (secreted alkaline phosphatase [SEAP]) recombinant protein production in BEVS. Since the expression of molecular chaperones is strongly associated with the maximal production of exogenous proteins in BEVS, the differential expression of molecular chaperones in baculovirus-infected stable cells was also analyzed in this study. Conclusion The data indicated that the retention of expression of molecular chaperones in baculovirus-infected Sf-caspase-1-repressed stable cells give the higher recombinant protein accumulation.
Collapse
Affiliation(s)
- Yiu-Kay Lai
- Institute of Biotechnology, Department of Life Science, National Tsing Hua University, No. 101, Section 2, Kuang-Fu Road, Hsinchu 30013, Taiwan, R.O.C
| | | | | | | | | | | |
Collapse
|
144
|
Crisci E, Bárcena J, Montoya M. Virus-like particle-based vaccines for animal viral infections. ACTA ACUST UNITED AC 2012; 32:102-116. [PMID: 32287712 PMCID: PMC7115488 DOI: 10.1016/j.inmuno.2012.08.002] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2012] [Accepted: 08/20/2012] [Indexed: 12/20/2022]
Abstract
Vaccination is considered one of the most effective ways to control pathogens and prevent diseases in humans as well as in the veterinary field. Traditional vaccines against animal viral diseases are based on inactivated or attenuated viruses, but new subunit vaccines are gaining attention from researchers in animal vaccinology. Among these, virus-like particles (VLPs) represent one of the most appealing approaches opening up interesting frontiers in animal vaccines. VLPs are robust protein scaffolds exhibiting well-defined geometry and uniformity that mimic the overall structure of the native virions but lack the viral genome. They are often antigenically indistinguishable from the virus from which they were derived and present important advantages in terms of safety. VLPs can stimulate strong humoral and cellular immune responses and have been shown to exhibit self-adjuvanting abilities. In addition to their suitability as a vaccine for the homologous virus from which they are derived, VLPs can also be used as vectors for the multimeric presentation of foreign antigens. VLPs have therefore shown dramatic effectiveness as candidate vaccines; nevertheless, only one veterinary VLP-base vaccine is licensed. Here, we review and examine in detail the current status of VLPs as a vaccine strategy in the veterinary field, and discuss the potential advantages and challenges of this technology.
Collapse
Affiliation(s)
- Elisa Crisci
- Centre de Recerca en Sanitat Animal (CReSA), UAB-IRTA, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Spain
| | - Juan Bárcena
- Centro de Investigación en Sanidad Animal (CISA-INIA), Madrid, Spain
| | - María Montoya
- Centre de Recerca en Sanitat Animal (CReSA), UAB-IRTA, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Spain.,Institut de Recerca i Tecnologia Agroalimentàries (IRTA), Barcelona, Spain
| |
Collapse
|
145
|
Yamaji H, Nakamura M, Kuwahara M, Takahashi Y, Katsuda T, Konishi E. Efficient production of Japanese encephalitis virus-like particles by recombinant lepidopteran insect cells. Appl Microbiol Biotechnol 2012; 97:1071-9. [DOI: 10.1007/s00253-012-4371-y] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2012] [Revised: 08/09/2012] [Accepted: 08/12/2012] [Indexed: 12/24/2022]
|
146
|
Crisci E, Bárcena J, Montoya M. Virus-like particles: the new frontier of vaccines for animal viral infections. Vet Immunol Immunopathol 2012; 148:211-25. [PMID: 22705417 PMCID: PMC7112581 DOI: 10.1016/j.vetimm.2012.04.026] [Citation(s) in RCA: 102] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2011] [Revised: 04/25/2012] [Accepted: 04/26/2012] [Indexed: 12/12/2022]
Abstract
Vaccination continues to be the main approach to protect animals from infectious diseases. Until recently, all licensed vaccines were developed using conventional technologies. Subunit vaccines are, however, gaining attention from researchers in the field of veterinary vaccinology, and among these, virus-like particles (VLPs) represent one of the most appealing approaches. VLPs are robust protein cages in the nanometer range that mimic the overall structure of the native virions but lack the viral genome. They are often antigenically indistinguishable from the virus from which they were derived and present important advantages in terms of safety. VLPs can stimulate strong humoral and cellular immune responses and have been shown to exhibit self-adjuvanting abilities. In addition to their suitability as a vaccine for the homologous virus from which they are derived, VLPs can also be used as vectors for the multimeric presentation of foreign antigens. VLPs have therefore shown dramatic effectiveness as candidate vaccines. Here, we review the current status of VLPs as a vaccine technology in the veterinary field, and discuss the potential advantages and challenges of this technology.
Collapse
Affiliation(s)
- Elisa Crisci
- Centre de Recerca en Sanitat Animal (CReSA), UAB-IRTA, Campus de la Universitat Autònoma de Barcelona, 08193 Bellaterra, Barcelona, Spain
| | | | | |
Collapse
|
147
|
Martínez JL, Liu L, Petranovic D, Nielsen J. Pharmaceutical protein production by yeast: towards production of human blood proteins by microbial fermentation. Curr Opin Biotechnol 2012; 23:965-71. [PMID: 22503236 DOI: 10.1016/j.copbio.2012.03.011] [Citation(s) in RCA: 61] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2012] [Revised: 03/20/2012] [Accepted: 03/20/2012] [Indexed: 11/25/2022]
Abstract
Since the approval of recombinant insulin from Escherichia coli for its clinical use in the early 1980s, the amount of recombinant pharmaceutical proteins obtained by microbial fermentations has significantly increased. The recent advances in genomics together with high throughput analysis techniques (the so-called-omics approaches) and integrative approaches (systems biology) allow the development of novel microbial cell factories as valuable platforms for large scale production of therapeutic proteins. This review summarizes the main achievements and the current situation in the field of recombinant therapeutics using yeast Saccharomyces cerevisiae as a model platform, and discusses the future potential of this platform for production of blood proteins and substitutes.
Collapse
Affiliation(s)
- José L Martínez
- Novo Nordisk Center for Biosustainability, Department of Chemical & Biological Engineering, Chalmers University of Technology, SE412 96 Gothenburg, Sweden
| | | | | | | |
Collapse
|
148
|
Palomares LA, Mena JA, Ramírez OT. Simultaneous expression of recombinant proteins in the insect cell-baculovirus system: production of virus-like particles. Methods 2012; 56:389-95. [PMID: 22300754 DOI: 10.1016/j.ymeth.2012.01.004] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2011] [Revised: 01/16/2012] [Accepted: 01/17/2012] [Indexed: 12/11/2022] Open
Abstract
The insect cell-baculovirus system (IC-BEVS) is widely used for the production of recombinant viral proteins for vaccine applications. It is especially suitable for the production of virus-like particles, which often require the simultaneous production of several recombinant proteins. Here, the available tools and process requirements for the simultaneous production of several recombinant proteins using the IC-BEVS are discussed. The production of double-layered rotavirus like particles is used as a specific example for the simultaneous production of two recombinant proteins. Methods to quantify VLP in small samples are described. The multiplicity and time of infection are presented as tools to manipulate protein concentration, and the effect on protein concentration ratios on the assembly efficiency of double-layered rotavirus like particles is discussed. It was found that not only the ratio between the recombinant proteins is determinant of VLP assembly efficiency, but also that assembly efficiency is related to the characteristics of the assembled proteins. This is the first time that kinetics of VLP production are followed during cultures, and that the assembly efficiency is quantitatively determined.
Collapse
Affiliation(s)
- Laura A Palomares
- Departamento de Medicina Molecular y Bioprocesos, Instituto de Biotecnología, Universidad Nacional Autónoma de México, Apdo. Postal 510-3, Cuernavaca, Morelos, CP 62250, Mexico.
| | | | | |
Collapse
|