1
|
Garcia Fallit M, Pidre ML, Asad AS, Peña Agudelo JA, Vera MB, Nicola Candia AJ, Sagripanti SB, Pérez Kuper M, Amorós Morales LC, Marchesini A, Gonzalez N, Caruso CM, Romanowski V, Seilicovich A, Videla-Richardson GA, Zanetti FA, Candolfi M. Evaluation of Baculoviruses as Gene Therapy Vectors for Brain Cancer. Viruses 2023; 15:608. [PMID: 36992317 PMCID: PMC10051617 DOI: 10.3390/v15030608] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2022] [Revised: 02/19/2023] [Accepted: 02/19/2023] [Indexed: 02/25/2023] Open
Abstract
We aimed to assess the potential of baculoviral vectors (BV) for brain cancer gene therapy. We compared them with adenoviral vectors (AdV), which are used in neuro-oncology, but for which there is pre-existing immunity. We constructed BVs and AdVs encoding fluorescent reporter proteins and evaluated their transduction efficiency in glioma cells and astrocytes. Naïve and glioma-bearing mice were intracranially injected with BVs to assess transduction and neuropathology. Transgene expression was also assessed in the brain of BV-preimmunized mice. While the expression of BVs was weaker than AdVs in murine and human glioma cell lines, BV-mediated transgene expression in patient-derived glioma cells was similar to AdV-mediated transduction and showed strong correlation with clathrin expression, a protein that interacts with the baculovirus glycoprotein GP64, mediating BV endocytosis. BVs efficiently transduced normal and neoplastic astrocytes in vivo, without apparent neurotoxicity. BV-mediated transgene expression was stable for at least 21 days in the brain of naïve mice, but it was significantly reduced after 7 days in mice systemically preimmunized with BVs. Our findings indicate that BVs efficiently transduce glioma cells and astrocytes without apparent neurotoxicity. Since humans do not present pre-existing immunity against BVs, these vectors may constitute a valuable tool for the delivery of therapeutic genes into the brain.
Collapse
Affiliation(s)
- Matías Garcia Fallit
- Instituto de Investigaciones Biomédicas (INBIOMED, UBA-CONICET), Facultad de Medicina, Universidad de Buenos Aires, Consejo Nacional de Investigaciones Científicas y Técnicas, Ciudad Autónoma de Buenos Aires C1121A6B, Argentina
- Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Ciudad Autónoma de Buenos Aires C1428BFA, Argentina
| | - Matías L. Pidre
- Instituto de Biotecnología y Biología Molecular (IBBM, UNLP-CONICET), Facultad de Ciencias Exactas, Universidad Nacional de La Plata, La Plata B1900, Argentina
| | - Antonela S. Asad
- Instituto de Investigaciones Biomédicas (INBIOMED, UBA-CONICET), Facultad de Medicina, Universidad de Buenos Aires, Consejo Nacional de Investigaciones Científicas y Técnicas, Ciudad Autónoma de Buenos Aires C1121A6B, Argentina
| | - Jorge A. Peña Agudelo
- Instituto de Investigaciones Biomédicas (INBIOMED, UBA-CONICET), Facultad de Medicina, Universidad de Buenos Aires, Consejo Nacional de Investigaciones Científicas y Técnicas, Ciudad Autónoma de Buenos Aires C1121A6B, Argentina
| | - Mariana B. Vera
- Departamento de Biología Celular e Histología, Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Ciudad Autónoma de Buenos Aires C1121A6B, Argentina
| | - Alejandro J. Nicola Candia
- Instituto de Investigaciones Biomédicas (INBIOMED, UBA-CONICET), Facultad de Medicina, Universidad de Buenos Aires, Consejo Nacional de Investigaciones Científicas y Técnicas, Ciudad Autónoma de Buenos Aires C1121A6B, Argentina
| | - Sofia B. Sagripanti
- Instituto de Investigaciones Biomédicas (INBIOMED, UBA-CONICET), Facultad de Medicina, Universidad de Buenos Aires, Consejo Nacional de Investigaciones Científicas y Técnicas, Ciudad Autónoma de Buenos Aires C1121A6B, Argentina
| | - Melanie Pérez Kuper
- Instituto de Investigaciones Biomédicas (INBIOMED, UBA-CONICET), Facultad de Medicina, Universidad de Buenos Aires, Consejo Nacional de Investigaciones Científicas y Técnicas, Ciudad Autónoma de Buenos Aires C1121A6B, Argentina
| | - Leslie C. Amorós Morales
- Instituto de Biotecnología y Biología Molecular (IBBM, UNLP-CONICET), Facultad de Ciencias Exactas, Universidad Nacional de La Plata, La Plata B1900, Argentina
| | - Abril Marchesini
- Instituto de Biotecnología y Biología Molecular (IBBM, UNLP-CONICET), Facultad de Ciencias Exactas, Universidad Nacional de La Plata, La Plata B1900, Argentina
| | - Nazareno Gonzalez
- Instituto de Investigaciones Biomédicas (INBIOMED, UBA-CONICET), Facultad de Medicina, Universidad de Buenos Aires, Consejo Nacional de Investigaciones Científicas y Técnicas, Ciudad Autónoma de Buenos Aires C1121A6B, Argentina
| | - Carla M. Caruso
- Instituto de Investigaciones Biomédicas (INBIOMED, UBA-CONICET), Facultad de Medicina, Universidad de Buenos Aires, Consejo Nacional de Investigaciones Científicas y Técnicas, Ciudad Autónoma de Buenos Aires C1121A6B, Argentina
| | - Víctor Romanowski
- Instituto de Biotecnología y Biología Molecular (IBBM, UNLP-CONICET), Facultad de Ciencias Exactas, Universidad Nacional de La Plata, La Plata B1900, Argentina
| | - Adriana Seilicovich
- Instituto de Investigaciones Biomédicas (INBIOMED, UBA-CONICET), Facultad de Medicina, Universidad de Buenos Aires, Consejo Nacional de Investigaciones Científicas y Técnicas, Ciudad Autónoma de Buenos Aires C1121A6B, Argentina
- Departamento de Biología Celular e Histología, Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Ciudad Autónoma de Buenos Aires C1121A6B, Argentina
| | - Guillermo A. Videla-Richardson
- Fundación Para la Lucha Contra las Enfermedades Neurológicas de la Infancia (FLENI), Ciudad Autónoma de Buenos Aires C1121A6B, Argentina
| | - Flavia A. Zanetti
- Instituto de Ciencia y Tecnología ‘‘Dr. Cesar Milstein”, CONICET, Saladillo 2468 (C1440FFX), Ciudad Autónoma de Buenos Aires C1428, Argentina
| | - Marianela Candolfi
- Instituto de Investigaciones Biomédicas (INBIOMED, UBA-CONICET), Facultad de Medicina, Universidad de Buenos Aires, Consejo Nacional de Investigaciones Científicas y Técnicas, Ciudad Autónoma de Buenos Aires C1121A6B, Argentina
| |
Collapse
|
2
|
Lu B, Tang Q, Wang Q, Liu X, Peng H, Zhu B, Xie L, Li Z, Wang H, Zheng Z, Wang L, Li B. Recovery Infectious Enterovirus 71 by Bac-to-Bac Expression System in vitro and in vivo. Front Microbiol 2022; 13:825111. [PMID: 35356523 PMCID: PMC8959925 DOI: 10.3389/fmicb.2022.825111] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Accepted: 01/19/2022] [Indexed: 11/13/2022] Open
Abstract
Enterovirus 71 (EV71) is one of the most important etiological agents for hand-foot-mouth disease. Compared with coxsackievirus A16 infection, EV71 infection is often associated with severe central nervous system complications, such as encephalitis, encephalomyelitis, and acute flaccid paralysis in infants and young children. In this study, we constructed a recombinant baculovirus with T7 ribonucleic acid polymerase under the control of a cytomegalovirus promoter and simultaneously engineered the T7 promoter upstream of a full-length EV71 complementary deoxyribonucleic acid. After transduction into mammalian cells, typical cytopathic effects (CPEs) and VP1 signals were detected in cells transfected with recombinant baculovirus. Additionally, viral particles located in the cytoplasm of human rhabdomyosarcoma cells (Rd) and Vero cells were observed by electron microscope, indicating that EV71 was recovered using a Bac-to-Bac expression system in vitro. After four passages, the rescued virus had a growth curve and plaque morphology similar to those of the parental virus. Furthermore, the Vp1 gene and the protein from the mouse brain were detected by reverse transcription polymerase chain reaction and immunohistochemistry after intracerebral injection of purified recombinant baculovirus. Typical CPEs were observed after inoculation of the supernatant from mouse brain to Rd cells, revealing a reconstruction of EV71 in vivo. Thus, we established a new approach to rescue EV71 based on a baculovirus expression system in vitro and in vivo, which may provide a safe and convenient platform for fundamental research and a strategy to rescue viruses that currently lack suitable cell culture and animal models.
Collapse
Affiliation(s)
- Baojing Lu
- The Key Laboratory of Microbiology and Parasitology of Anhui Province, The Key Laboratory of Zoonoses of High Institutions in Anhui, Department of Microbiology and Parasitology, School of Basic Medical Sciences, Anhui Medical University, Hefei, China
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Hefei, China
| | - Qi Tang
- The Key Laboratory of Microbiology and Parasitology of Anhui Province, The Key Laboratory of Zoonoses of High Institutions in Anhui, Department of Microbiology and Parasitology, School of Basic Medical Sciences, Anhui Medical University, Hefei, China
| | - Qianyun Wang
- The Key Laboratory of Microbiology and Parasitology of Anhui Province, The Key Laboratory of Zoonoses of High Institutions in Anhui, Department of Microbiology and Parasitology, School of Basic Medical Sciences, Anhui Medical University, Hefei, China
| | - Xuejuan Liu
- The Key Laboratory of Microbiology and Parasitology of Anhui Province, The Key Laboratory of Zoonoses of High Institutions in Anhui, Department of Microbiology and Parasitology, School of Basic Medical Sciences, Anhui Medical University, Hefei, China
| | - Hui Peng
- The Key Laboratory of Microbiology and Parasitology of Anhui Province, The Key Laboratory of Zoonoses of High Institutions in Anhui, Department of Microbiology and Parasitology, School of Basic Medical Sciences, Anhui Medical University, Hefei, China
| | - Binbin Zhu
- The Key Laboratory of Microbiology and Parasitology of Anhui Province, The Key Laboratory of Zoonoses of High Institutions in Anhui, Department of Microbiology and Parasitology, School of Basic Medical Sciences, Anhui Medical University, Hefei, China
| | - Li Xie
- Department of Tuberculosis Prevention, Shenzhen Center for Chronic Disease Control, Shenzhen, China
| | - Zeng Li
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Hefei, China
| | - Hanzhong Wang
- CAS Key Laboratory of Special Pathogens and Biosafety, Center for Emerging Infectious Diseases, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China
| | - Zhenhua Zheng
- CAS Key Laboratory of Special Pathogens and Biosafety, Center for Emerging Infectious Diseases, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China
| | - Linding Wang
- The Key Laboratory of Microbiology and Parasitology of Anhui Province, The Key Laboratory of Zoonoses of High Institutions in Anhui, Department of Microbiology and Parasitology, School of Basic Medical Sciences, Anhui Medical University, Hefei, China
| | - Bao Li
- The Comprehensive Lab, School of Basic Medical Science, Anhui Medical University, Hefei, China
| |
Collapse
|
3
|
Meysami P, Rezaei F, Marashi SM, Amiri MM, Bakker E, Mokhtari-Azad T. Antitumor effects of a recombinant baculovirus displaying anti-HER2 scFv expressing Apoptin in HER2 positive SK-BR-3 breast cancer cells. Future Virol 2019. [DOI: 10.2217/fvl-2018-0187] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Aim: Since HER2 targeted therapies have shown clinical benefit in breast cancer, in the present study recombinant baculovirus (BV) displaying anti-HER2 single-chain variable domain fragment (scFv) expressing Apoptin was generated. Methods: The binding specificity and surface display of anti-HER2 scFv were evaluated using enzyme-linked immunosorbent assay (ELISA) and electron microscopy, respectively. The targeting properties and cytotoxic effect on breast cancer cells determined by fluorescence microscopy and MTT assays. Results: The results demonstrated that recombinant BV could specifically bind to HER2-overexpressing SK-BR-3 cells but not to the HER2 negative MCF-7 cells and reduced the viability of SK-BR-3 cells by expressing Apoptin. Conclusion: These results suggest that the antitumor effect of Apoptin in combination with HER2 targeting of this recombinant BV makes it a promising vector in targeted cancer therapy.
Collapse
Affiliation(s)
- Parisa Meysami
- Department of Virology, School of Public Health, Tehran University of Medical Sciences, Tehran 1471613151, Iran
| | - Farhad Rezaei
- Department of Virology, School of Public Health, Tehran University of Medical Sciences, Tehran 1471613151, Iran
| | - Sayed Mahdi Marashi
- Department of Virology, School of Public Health, Tehran University of Medical Sciences, Tehran 1471613151, Iran
| | - Mohammad Mehdi Amiri
- Department of Immunology, School of Public Health, Tehran University of Medical Sciences, Tehran 1471613151, Iran
| | - Emyr Bakker
- School of Medicine, University of Central Lancashire, Preston, UK
| | - Talat Mokhtari-Azad
- Department of Virology, School of Public Health, Tehran University of Medical Sciences, Tehran 1471613151, Iran
| |
Collapse
|
4
|
Pseudotyped baculovirus is an effective gene expression tool for studying molecular function during axolotl limb regeneration. Dev Biol 2017; 433:262-275. [PMID: 29198566 DOI: 10.1016/j.ydbio.2017.10.008] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2017] [Revised: 10/08/2017] [Accepted: 10/11/2017] [Indexed: 12/31/2022]
Abstract
Axolotls can regenerate complex structures through recruitment and remodeling of cells within mature tissues. Accessing the underlying mechanisms at a molecular resolution is crucial to understand how injury triggers regeneration and how it proceeds. However, gene transformation in adult tissues can be challenging. Here we characterize the use of pseudotyped baculovirus (BV) as an effective gene transfer method both for cells within mature limb tissue and within the blastema. These cells remain competent to participate in regeneration after transduction. We further characterize the effectiveness of BV for gene overexpression studies by overexpressing Shh in the blastema, which yields a high penetrance of classic polydactyly phenotypes. Overall, our work establishes BV as a powerful tool to access gene function in axolotl limb regeneration.
Collapse
|
5
|
Karste K, Bleckmann M, van den Heuvel J. Not Limited to E. coli: Versatile Expression Vectors for Mammalian Protein Expression. Methods Mol Biol 2017; 1586:313-324. [PMID: 28470614 DOI: 10.1007/978-1-4939-6887-9_20] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
Recombinant protein expression is not limited to E. coli or other prokaryotic systems. It is inevitable to use eukaryotic systems in order to express challenging mammalian proteins. Eukaryotic systems are able to perform complex posttranslational modifications like protein processing, phosphorylation, glycosylation, which are essential for stability and functionality of many proteins. Different eukaryotic protein expression systems employing yeast, insect, or mammalian cell lines are established with each having its own advantages and disadvantages. Often it is quite difficult to decide which will be the most optimal expression system as this depends highly on the protein itself. Expression in stable cell lines requires substantial screening of expressible constructs prior to developing a stable expression cell line. To achieve fast screening by transient expression in multiple hosts, versatile vectors can be applied. In this chapter, we present an overview of the most common multi-host vectors, which allow for fast expression analysis without tedious (re)cloning of the gene of interest in several different protein production systems. The protocols in this chapter describe the latest methods for fast transient expression in insect and mammalian cell lines.
Collapse
Affiliation(s)
- Katharina Karste
- Helmholtz Zentrum für Infektionsforschung GmbH, Inhoffenstrasse 7, 38124, Braunschweig, Germany
| | - Maren Bleckmann
- Helmholtz Zentrum für Infektionsforschung GmbH, Inhoffenstrasse 7, 38124, Braunschweig, Germany
| | - Joop van den Heuvel
- Helmholtz Zentrum für Infektionsforschung GmbH, Inhoffenstrasse 7, 38124, Braunschweig, Germany.
| |
Collapse
|
6
|
Balasundaram G, Kwang TW, Wang S. cDNA microarray assays to evaluate immune responses following intracranial injection of baculoviral vectors in non-human primates. J Neurochem 2016; 140:320-333. [DOI: 10.1111/jnc.13884] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2016] [Revised: 10/27/2016] [Accepted: 10/27/2016] [Indexed: 12/19/2022]
Affiliation(s)
- Ghayathri Balasundaram
- Institute of Bioengineering and Nanotechnology; Singapore
- Department of Biological Sciences; National University of Singapore; Singapore
| | | | - Shu Wang
- Institute of Bioengineering and Nanotechnology; Singapore
- Department of Biological Sciences; National University of Singapore; Singapore
| |
Collapse
|
7
|
Abstract
Viral vectors have frequently been applied in gene therapy with the final goal of treating various diseases in the areas of neurology, neurodegeneration, metabolic disease, and cancer. Vectors have been engineered based on AAV, adenoviruses, alphaviruses, herpes simplex viruses, lentiviruses, and retroviruses. Some vectors are suitable for short-term episomal transgene expression, whereas others are integrated into the host cell genome to provide long-term expression. Additionally, hybrid vectors with favorable features from different viruses have been developed. Therapeutic genes of choice have typically been toxic genes such as thymidine kinase, pro-apoptotic genes like Bax, and immunostimulatory genes (for instance, interleukin-12). A large number of animal studies have demonstrated proof of concept of viral gene therapy. Many types of viral vectors have been employed in more than 700 clinical trials that have been carried out or are currently in progress.
Collapse
Affiliation(s)
- Kenneth Lundstrom
- Regulon Inc., Chemin des Croisettes 22, CH-1066 Epalinges, Switzerland.
| |
Collapse
|
8
|
Makkonen KE, Airenne K, Ylä-Herttulala S. Baculovirus-mediated gene delivery and RNAi applications. Viruses 2015; 7:2099-125. [PMID: 25912715 PMCID: PMC4411692 DOI: 10.3390/v7042099] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2015] [Revised: 04/02/2015] [Accepted: 04/16/2015] [Indexed: 12/11/2022] Open
Abstract
Baculoviruses are widely encountered in nature and a great deal of data is available about their safety and biology. Recently, these versatile, insect-specific viruses have demonstrated their usefulness in various biotechnological applications including protein production and gene transfer. Multiple in vitro and in vivo studies exist and support their use as gene delivery vehicles in vertebrate cells. Recently, baculoviruses have also demonstrated high potential in RNAi applications in which several advantages of the virus make it a promising tool for RNA gene transfer with high safety and wide tropism.
Collapse
Affiliation(s)
- Kaisa-Emilia Makkonen
- Virtanen Institute, Department of Biotechnology and Molecular Medicine, University of Eastern Finland, Kuopio 70211 Finland.
| | - Kari Airenne
- Virtanen Institute, Department of Biotechnology and Molecular Medicine, University of Eastern Finland, Kuopio 70211 Finland.
| | - Seppo Ylä-Herttulala
- Virtanen Institute, Department of Biotechnology and Molecular Medicine, University of Eastern Finland, Kuopio 70211 Finland.
- Gene Therapy Unit, Kuopio University Hospital, Kuopio 70211, Finland.
- Science Service Center, Kuopio University Hospital, Kuopio 70211, Finland.
| |
Collapse
|
9
|
Turunen TAK, Laakkonen JP, Alasaarela L, Airenne KJ, Ylä-Herttuala S. Sleeping Beauty-baculovirus hybrid vectors for long-term gene expression in the eye. J Gene Med 2014; 16:40-53. [PMID: 24464652 DOI: 10.1002/jgm.2756] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2013] [Revised: 11/18/2013] [Accepted: 01/22/2014] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND A baculovirus vector is capable of efficiently transducing many nondiving and diving cell types. However, the potential of baculovirus is restricted for many gene delivery applications as a result of the transient gene expression that it mediates. The plasmid-based Sleeping Beauty (SB) transposon system integrates transgenes into target cell genome efficiently with a genomic integration pattern that is generally considered safer than the integration of many other integrating vectors; yet efficient delivery of therapeutic genes into cells of target tissues in vivo is a major challenge for nonviral gene therapy. In the present study, SB was introduced into baculovirus to obtain novel hybrid vectors that would combine the best features of the two vector systems (i.e. effective gene delivery and efficient integration into the genome), thus circumventing the major limitations of these vectors. METHODS We constructed and optimized SB-baculovirus hybrid vectors that bear either SB100x transposase or SB transposon in the forward or reverse orientations with respect to the viral backbone The functionality of the novel hybrid vectors was investigated in cell cultures and in a proof-of-concept study in the mouse eye. RESULTS The hybrid vectors showed high and sustained transgene expression that remained stable and demonstrated no signs of decline during the 2 months follow-up in vitro. These results were verified in the mouse eye where persistent transgene expression was detected two months after intravitreal injection. CONCLUSIONS Our results confirm that (i) SB-baculovirus hybrid vectors mediate long-term gene expression in vitro and in vivo, and (ii) the hybrid vectors are potential new tools for the treatment of ocular diseases.
Collapse
Affiliation(s)
- Tytteli Anni Kaarina Turunen
- A. I. Virtanen Institute for Molecular Sciences, Department of Biotechnology and Molecular Medicine, University of Eastern Finland, Kuopio, Finland
| | | | | | | | | |
Collapse
|
10
|
Paul A, Hasan A, Rodes L, Sangaralingam M, Prakash S. Bioengineered baculoviruses as new class of therapeutics using micro and nanotechnologies: principles, prospects and challenges. Adv Drug Deliv Rev 2014; 71:115-30. [PMID: 24503281 DOI: 10.1016/j.addr.2014.01.004] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2013] [Revised: 01/08/2014] [Accepted: 01/13/2014] [Indexed: 12/15/2022]
Abstract
Designing a safe and efficient gene delivery system is required for success of gene therapy trials. Although a wide variety of viral, non-viral and polymeric nanoparticle based careers have been widely studied, the current gene delivery vehicles are limited by their suboptimal, non-specific therapeutic efficacy and acute immunological reactions, leading to unwanted side effects. Recently, there has been a growing interest in insect-cell-originated baculoviruses as gene delivery vehicles for diverse biomedical applications. Specifically, the emergence of diverse types of surface functionalized and bioengineered baculoviruses is posed to edge over currently available gene delivery vehicles. This is primarily because baculoviruses are comparatively non-pathogenic and non-toxic as they cannot replicate in mammalian cells and do not invoke any cytopathic effect. Moreover, emerging advanced studies in this direction have demonstrated that hybridizing the baculovirus surface with different kinds of bioactive therapeutic molecules, cell-specific targeting moieties, protective polymeric grafts and nanomaterials can significantly improve the preclinical efficacy of baculoviruses. This review presents a comprehensive overview of the recent advancements in the field of bioengineering and biotherapeutics to engineer baculovirus hybrids for tailored gene therapy, and articulates in detail the potential and challenges of these strategies for clinical realization. In addition, the article illustrates the rapid evolvement of microfluidic devices as a high throughput platform for optimizing baculovirus production and treatment conditions.
Collapse
Affiliation(s)
- Arghya Paul
- Biomedical Technology and Cell Therapy Research Laboratory, Department of Biomedical Engineering and Artificial Cells and Organs Research Centre, Faculty of Medicine, McGill University, 3775 University Street, Montreal, Québec H3A 2B4, Canada; Harvard-MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Center for Biomedical Engineering, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge, MA 02139, USA
| | - Anwarul Hasan
- Harvard-MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Center for Biomedical Engineering, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge, MA 02139, USA
| | - Laetitia Rodes
- Biomedical Technology and Cell Therapy Research Laboratory, Department of Biomedical Engineering and Artificial Cells and Organs Research Centre, Faculty of Medicine, McGill University, 3775 University Street, Montreal, Québec H3A 2B4, Canada
| | - Mugundhine Sangaralingam
- Biomedical Technology and Cell Therapy Research Laboratory, Department of Biomedical Engineering and Artificial Cells and Organs Research Centre, Faculty of Medicine, McGill University, 3775 University Street, Montreal, Québec H3A 2B4, Canada
| | - Satya Prakash
- Biomedical Technology and Cell Therapy Research Laboratory, Department of Biomedical Engineering and Artificial Cells and Organs Research Centre, Faculty of Medicine, McGill University, 3775 University Street, Montreal, Québec H3A 2B4, Canada.
| |
Collapse
|
11
|
Innate immune response induced by baculovirus attenuates transgene expression in mammalian cells. J Virol 2013; 88:2157-67. [PMID: 24335288 DOI: 10.1128/jvi.03055-13] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
The baculovirus Autographa californica nucleopolyhedrovirus (AcNPV) has been widely used to achieve a high level of foreign gene expression in insect cells, as well as for efficient gene transduction into mammalian cells without any replication. In addition to permitting efficient gene delivery, baculovirus has been shown to induce host innate immune responses in various mammalian cells and in mice. In this study, we examined the effects of the innate immune responses on gene expression by recombinant baculoviruses in cultured cells. The reporter gene expression in IRF3-deficient mouse embryonic fibroblasts (MEFs) infected with the recombinant baculovirus was shown to be enhanced in accordance with the suppression of beta interferon (IFN-β) production. Furthermore, efficient gene transduction by the recombinant baculovirus was achieved in MEFs deficient for stimulator of interferon genes (STING), TANK binding kinase 1 (TBK1), IFN regulatory factor 3 (IRF3), or IFN-β promoter stimulator 1 (IPS-1), but not in those deficient for IRF7, MyD88, or Z-DNA binding protein 1 (ZBP1)/DAI. Enhancement of gene expression by the recombinant baculovirus was also observed in human hepatoma cell lines replicating hepatitis C virus (HCV), in which innate immunity was impaired by the cleavage of IPS-1 by the viral protease. In addition, infection with the recombinant baculovirus expressing the BH3-only protein, BIMS, a potent inducer of apoptosis, resulted in a selective cell death in the HCV replicon cells. These results indicate that innate immune responses induced by infection with baculovirus attenuate transgene expression, and this characteristic might be useful for a selective gene transduction into cells with impaired innate immunity arising from infection with various viruses.
Collapse
|
12
|
Swift SL, Rivera GC, Dussupt V, Leadley RM, Hudson LC, MA de Ridder C, Kraaij R, Burns JE, Maitland NJ, Georgopoulos LJ. Evaluating baculovirus as a vector for human prostate cancer gene therapy. PLoS One 2013; 8:e65557. [PMID: 23755250 PMCID: PMC3675042 DOI: 10.1371/journal.pone.0065557] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2013] [Accepted: 04/26/2013] [Indexed: 02/07/2023] Open
Abstract
Gene therapy represents an attractive strategy for the non-invasive treatment of prostate cancer, where current clinical interventions show limited efficacy. Here, we evaluate the use of the insect virus, baculovirus (BV), as a novel vector for human prostate cancer gene therapy. Since prostate tumours represent a heterogeneous environment, a therapeutic approach that achieves long-term regression must be capable of targeting multiple transformed cell populations. Furthermore, discrimination in the targeting of malignant compared to non-malignant cells would have value in minimising side effects. We employed a number of prostate cancer models to analyse the potential for BV to achieve these goals. In vitro, both traditional prostate cell lines as well as primary epithelial or stromal cells derived from patient prostate biopsies, in two- or three-dimensional cultures, were used. We also evaluated BV in vivo in murine prostate cancer xenograft models. BV was capable of preferentially transducing invasive malignant prostate cancer cell lines compared to early stage cancers and non-malignant samples, a restriction that was not a function of nuclear import. Of more clinical relevance, primary patient-derived prostate cancer cells were also efficiently transduced by BV, with robust rates observed in epithelial cells of basal phenotype, which expressed BV-encoded transgenes faster than epithelial cells of a more differentiated, luminal phenotype. Maximum transduction capacity was observed in stromal cells. BV was able to penetrate through three-dimensional structures, including in vitro spheroids and in vivo orthotopic xenografts. BV vectors containing a nitroreductase transgene in a gene-directed enzyme pro-drug therapy approach were capable of efficiently killing malignant prostate targets following administration of the pro-drug, CB1954. Thus, BV is capable of transducing a large proportion of prostate cell types within a heterogeneous 3-D prostate tumour, can facilitate cell death using a pro-drug approach, and shows promise as a vector for the treatment of prostate cancer.
Collapse
Affiliation(s)
- Stephanie L. Swift
- Yorkshire Cancer Research Unit, Department of Biology, University of York, Heslington, York, United Kingdom
| | - Guillermo C. Rivera
- Yorkshire Cancer Research Unit, Department of Biology, University of York, Heslington, York, United Kingdom
| | - Vincent Dussupt
- Yorkshire Cancer Research Unit, Department of Biology, University of York, Heslington, York, United Kingdom
| | - Regina M. Leadley
- Yorkshire Cancer Research Unit, Department of Biology, University of York, Heslington, York, United Kingdom
| | - Lucy C. Hudson
- Yorkshire Cancer Research Unit, Department of Biology, University of York, Heslington, York, United Kingdom
| | | | - Robert Kraaij
- Department of Urology, Erasmus MC, Rotterdam, The Netherlands
| | - Julie E. Burns
- Yorkshire Cancer Research Unit, Department of Biology, University of York, Heslington, York, United Kingdom
| | - Norman J. Maitland
- Yorkshire Cancer Research Unit, Department of Biology, University of York, Heslington, York, United Kingdom
- * E-mail:
| | - Lindsay J. Georgopoulos
- Yorkshire Cancer Research Unit, Department of Biology, University of York, Heslington, York, United Kingdom
| |
Collapse
|
13
|
Erbs E, Pradhan AA, Matifas A, Kieffer BL, Massotte D. Evaluation of cre recombinase delivery in mammalian cells using baculovirus infection. J Biotechnol 2013; 166:182-6. [PMID: 23732834 DOI: 10.1016/j.jbiotec.2013.05.011] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2013] [Revised: 05/16/2013] [Accepted: 05/23/2013] [Indexed: 10/26/2022]
Abstract
In vivo conditional knock-out of a protein is a method of choice to decipher its biological function. It can be achieved by encoding the cre-recombinase on a recombinant virus to exert spatio-temporal control of its expression and enzymatic activity and, subsequently, of the target gene deletion. Recombinant baculoviruses have been successfully used to express a wide range of proteins in insect cells. More recently, their potential to infect mammalian cells has been addressed but, so far, their ability to yield a conditional knock-out as a result of efficient in vivo cre-recombinase gene delivery has not been examined. Cre-recombinase fused to the green fluorescent protein was cloned under the control of the CAG promoter in a recombinant Autographa californica baculovirus expressing the vesicular stomatitis virus envelope G protein for increased mammalian cell infection. Gene delivery was evaluated in vitro in mammalian cells, neuroblastoma and mouse primary neuronal cultures as well as in vivo in the mouse brain. Infection with adeno-associated viruses encoding the cre-recombinase fused to the green fluorescent protein was performed as a positive control. Our results indicate that baculovirus infection leads to functional cre-recombinase expression in non-neuronal and neuroblastoma cell lines but not in mouse primary neuronal cultures or brain.
Collapse
Affiliation(s)
- Eric Erbs
- Department of Neurobiology and Translational Medicine, IGBMC, Illkirch, France.
| | | | | | | | | |
Collapse
|
14
|
Airenne KJ, Hu YC, Kost TA, Smith RH, Kotin RM, Ono C, Matsuura Y, Wang S, Ylä-Herttuala S. Baculovirus: an insect-derived vector for diverse gene transfer applications. Mol Ther 2013; 21:739-49. [PMID: 23439502 PMCID: PMC3616530 DOI: 10.1038/mt.2012.286] [Citation(s) in RCA: 134] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2012] [Accepted: 12/11/2012] [Indexed: 01/23/2023] Open
Abstract
Insect-derived baculoviruses have emerged as versatile and safe workhorses of biotechnology. Baculovirus expression vectors (BEVs) have been applied widely for crop and forest protection, as well as safe tools for recombinant protein production in insect cells. However, BEVs ability to efficiently transduce noninsect cells is still relatively poorly recognized despite the fact that efficient baculovirus-mediated in vitro and ex vivo gene delivery into dormant and dividing vertebrate cells of diverse origin has been described convincingly by many authors. Preliminary proof of therapeutic potential has also been established in preclinical studies. This review summarizes the advantages and current status of baculovirus-mediated gene delivery. Stem cell transduction, preclinical animal studies, tissue engineering, vaccination, cancer gene therapy, viral vector production, and drug discovery are covered.
Collapse
Affiliation(s)
- Kari J Airenne
- Department of Biotechnology and Molecular Medicine, A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Yu-Chen Hu
- Department of Chemical Engineering, National Tsing Hua University, Hsinchu, Taiwan
- Institute of Biomedical Engineering, National Tsing Hua University, Hsinchu, Taiwan
| | - Thomas A Kost
- Biological Reagents and Assay Development, GlaxoSmithKline R&D, Research Triangle Park, North Carolina, USA
| | - Richard H Smith
- Molecular Virology and Gene Therapy Laboratory, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Robert M Kotin
- Molecular Virology and Gene Therapy Laboratory, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Chikako Ono
- Department of Molecular Virology, Research Institute for Microbial Diseases, Osaka University, Suita, Japan
| | - Yoshiharu Matsuura
- Department of Molecular Virology, Research Institute for Microbial Diseases, Osaka University, Suita, Japan
| | - Shu Wang
- Institute of Bioengineering and Nanotechnology, Singapore, Singapore
- Department of Biological Sciences, National University of Singapore, Singapore, Singapore
| | - Seppo Ylä-Herttuala
- Department of Biotechnology and Molecular Medicine, A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
- Research Unit, Kuopio University Hospital, Kuopio, Finland
- Gene Therapy Unit, Kuopio University Hospital, Kuopio, Finland
| |
Collapse
|
15
|
A new finding concerning adenoviral-mediated gene transfer: A high-level, cell-specific transgene expression in the neural stem cells of adult mice. J Virol Methods 2012; 186:1-6. [DOI: 10.1016/j.jviromet.2012.07.012] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2012] [Revised: 07/05/2012] [Accepted: 07/09/2012] [Indexed: 01/28/2023]
|
16
|
Heikura T, Nieminen T, Roschier MM, Karvinen H, Kaikkonen MU, Mähönen AJ, Lesch HP, Rissanen TT, Laitinen OH, Airenne KJ, Ylä-Herttuala S. Baculovirus-mediated vascular endothelial growth factor-D(ΔNΔC) gene transfer induces angiogenesis in rabbit skeletal muscle. J Gene Med 2012; 14:35-43. [PMID: 22162149 DOI: 10.1002/jgm.1637] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
BACKGROUND Occluded arteries and ischemic tissues cannot always be treated by angioplasty, stenting or by-pass-surgery. Under such circumstances, viral gene therapy may be useful in inducing increased blood supply to ischemic area. There is evidence of improved blood flow in ischemic skeletal muscle and myocardium in both animal and human studies using adenoviral vascular endothelial growth factor (VEGF) gene therapy. However, the expression is transient and repeated gene transfers with the same vector are inefficient due to immune responses. METHODS Different baculoviral vectors pseudotyped with or without vesicular stomatitis virus glycoprotein (VSV-G) and/or carrying woodchuck hepatitis virus post-transcriptional regulatory element (Wpre) were tested both in vitro and in vivo. VEGF-D(ΔNΔC) was used as therapeutic transgene and lacZ as a control. In vivo efficacy was evaluated as capillary enlargement and transgene expression in New Zealand White (NZW) rabbit skeletal muscle. RESULTS A statistically significant capillary enlargement was detected 6 days after gene transfer in transduced areas compared to the control gene transfers with baculovirus and adenovirus encoding β-galactosidase (lacZ). Substantially improved gene transfer efficiency was achieved with a modified baculovirus pseudotyped with VSV-G and carrying Wpre. Dose escalation experiments revealed that either too large volume or too many virus particles caused inflammation and necrosis in the target tissue, whereas 10(9) plaque forming units injected in multiple aliquots resulted in transgene expression with only mild immune reactions. CONCLUSIONS We show the first evidence of biologically significant baculoviral gene transfer in skeletal muscle of NZW rabbits using VEGF-D(ΔNΔC) as a therapeutic transgene.
Collapse
Affiliation(s)
- Tommi Heikura
- Department of Biotechnology and Molecular Medicine, AI Virtanen Institute, University of Eastern Finland, Kuopio, Finland
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
17
|
Abstract
There are many methods presently available to produce recombinant proteins in mammalian systems. The BacMam system is a simple straightforward method which overlaps two well-established technologies, namely the BEVS insect cell system and the transduction of mammalian cells in vitro. This chapter describes a method for the study of gene expression in mammalian cells in a series of simple steps. Protocols outlined include the design and construction of the recombinant baculovirus, cell culture techniques required to maintain both insect and mammalian cells, generation of baculovirus stocks, and methods to obtain maximal and reproducible gene expression in mammalian cells. Currently available statistical techniques using factorial design of experiment to optimize conditions for recombinant protein in vitro are outlined. Then details with respect to process scale-up in disposable bioreactors are included.
Collapse
|
18
|
Kaikkonen MU, Ylä-Herttuala S, Airenne KJ. How to avoid complement attack in baculovirus-mediated gene delivery. J Invertebr Pathol 2011; 107 Suppl:S71-9. [PMID: 21784233 DOI: 10.1016/j.jip.2011.05.007] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2010] [Accepted: 01/03/2011] [Indexed: 11/30/2022]
Abstract
Serum inactivation of baculovirus vectors is a significant barrier to the development of these highly efficient vectors for therapeutic gene delivery. In this review we will describe the efforts taken to avoid complement attack by passive or active measures. Evidently good targets for baculovirus-mediated gene delivery include immunoprivileged tissues, such as eye, brain and testis. Similarly baculovirus vectors have also proven their efficacy in an ex vivo setting for tissue engineering. Active measures to inhibit complement include the use of pharmacological inhibitors of complement as well as surface engineering of the baculoviral vectors through the use of synthetic polymers, pseudotyping or display of complement inhibitors. Lessons learned from these studies will significantly increase the possibility of using baculovirus vectors for therapeutic applications.
Collapse
Affiliation(s)
- Minna U Kaikkonen
- AI Virtanen Institute, Department of Biotechnology and Molecular Medicine, University of Eastern Finland, PO Box 1627, FI-70211 Kuopio, Finland
| | | | | |
Collapse
|
19
|
Birks SM, Danquah JO, King L, Vlasak R, Gorecki DC, Pilkington GJ. Targeting the GD3 acetylation pathway selectively induces apoptosis in glioblastoma. Neuro Oncol 2011; 13:950-60. [PMID: 21807667 DOI: 10.1093/neuonc/nor108] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
The expression of ganglioside GD3, which plays crucial roles in normal brain development, decreases in adults but is upregulated in neoplastic cells, where it regulates tumor invasion and survival. Normally a buildup of GD3 induces apoptosis, but this does not occur in gliomas due to formation of 9-O-acetyl GD3 by the addition of an acetyl group to the terminal sialic acid of GD3; this renders GD3 unable to induce apoptosis. Using human biopsy-derived glioblastoma cell cultures, we have carried out a series of molecular manipulations targeting GD3 acetylation pathways. Using immunocytochemistry, flow cytometry, western blotting, and transwell assays, we have shown the existence of a critical ratio between GD3 and 9-O-acetyl GD3, which promotes tumor survival. Thus, we have demonstrated for the first time in primary glioblastoma that cleaving the acetyl group restores GD3, resulting in a reduction in tumor cell viability while normal astrocytes remain unaffected. Additionally, we have shown that glioblastoma viability is reduced due to the induction of mitochondrially mediated apoptosis and that this occurs after mitochondrial membrane depolarization. Three methods of cleaving the acetyl group using hemagglutinin esterase were investigated, and we have shown that the baculovirus vector transduces glioma cells as well as normal astroctyes with a relatively high efficacy. A recombinant baculovirus containing hemagglutinin esterase could be developed for the clinic as an adjuvant therapy for glioma.
Collapse
Affiliation(s)
- Suzanne M Birks
- Cellular and Molecular Neuro-oncology Research Group, Institute Biomedical and Biomolecular Sciences, University of Portsmouth, Portsmouth, UK.
| | | | | | | | | | | |
Collapse
|
20
|
Lesch HP, Makkonen KE, Laitinen A, Määttä AM, Närvänen O, Airenne KJ, Ylä-Herttuala S. Requirements for baculoviruses for clinical gene therapy applications. J Invertebr Pathol 2011; 107 Suppl:S106-12. [PMID: 21784225 DOI: 10.1016/j.jip.2011.05.010] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2010] [Accepted: 01/23/2011] [Indexed: 11/30/2022]
Affiliation(s)
- Hanna P Lesch
- AI Virtanen Institute Department of Biotechnology and Molecular Medicine, University of Eastern Finland/Kuopio, Finland
| | | | | | | | | | | | | |
Collapse
|
21
|
Guo R, Tian L, Han B, Xu H, Zhang M, Li B. Feasibility of a novel positive feedback effect of 131I-promoted Bac-Egr1-hNIS expression in malignant glioma via baculovirus. Nucl Med Biol 2011; 38:599-604. [DOI: 10.1016/j.nucmedbio.2010.11.005] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2010] [Revised: 11/14/2010] [Accepted: 11/16/2010] [Indexed: 11/29/2022]
|
22
|
Chen CY, Lin CY, Chen GY, Hu YC. Baculovirus as a gene delivery vector: recent understandings of molecular alterations in transduced cells and latest applications. Biotechnol Adv 2011; 29:618-31. [PMID: 21550393 PMCID: PMC7126054 DOI: 10.1016/j.biotechadv.2011.04.004] [Citation(s) in RCA: 107] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2011] [Revised: 04/15/2011] [Accepted: 04/15/2011] [Indexed: 12/13/2022]
Abstract
Baculovirus infects insects in nature and is non-pathogenic to humans, but can transduce a broad range of mammalian and avian cells. Thanks to the biosafety, large cloning capacity, low cytotoxicity and non-replication nature in the transduced cells as well as the ease of manipulation and production, baculovirus has gained explosive popularity as a gene delivery vector for a wide variety of applications. This article extensively reviews the recent understandings of the molecular mechanisms pertinent to baculovirus entry and cellular responses, and covers the latest advances in the vector improvements and applications, with special emphasis on antiviral therapy, cancer therapy, regenerative medicine and vaccine.
Collapse
Affiliation(s)
- Chi-Yuan Chen
- Department of Chemical Engineering, National Tsing Hua University, Hsinchu 30013, Taiwan
| | | | | | | |
Collapse
|
23
|
Abstract
Viral vector is the most effective means of gene transfer to modify specific cell type or tissue and can be manipulated to express therapeutic genes. Several virus types are currently being investigated for use to deliver genes to cells to provide either transient or permanent transgene expression. These include adenoviruses (Ads), retroviruses (γ-retroviruses and lentiviruses), poxviruses, adeno-associated viruses, baculoviruses, and herpes simplex viruses. The choice of virus for routine clinical use will depend on the efficiency of transgene expression, ease of production, safety, toxicity, and stability. This chapter provides an introductory overview of the general characteristics of viral vectors commonly used in gene transfer and their advantages and disadvantages for gene therapy use.
Collapse
Affiliation(s)
- James N Warnock
- School of Chemical & Bioprocess Engineering and Conway Institute for Biomolecular and Biomedical Research, University College Dublin, Belfield, Dublin, Ireland
| | | | | |
Collapse
|
24
|
Sodium butyrate enhances the expression of baculovirus-mediated sodium/iodide symporter gene in A549 lung adenocarcinoma cells. Nucl Med Commun 2010; 31:916-21. [PMID: 20683361 DOI: 10.1097/mnm.0b013e32833dedd7] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
OBJECTIVE Increased expression of sodium/iodide symporter (NIS) is required for reporter gene imaging and effective radioiodine treatment of tumor. We investigated whether increased accumulation of iodine can be induced by sodium butyrate through a newly developed baculoviral transfer of the human NIS (hNIS) gene in A549 human lung adenocarcinoma. METHODS A recombinant baculovirus [Bac-cytomegalovirus (CMV)-hNIS] encoding hNIS gene under the control of the CMV promoter was constructed. After A549 cells were transfected with Bac-CMV-hNIS in the presence of sodium butyrate, the expression of hNIS protein was detected by immunofluorescence and western blot analysis. The uptake and efflux of iodine were determined after the incubation of the transfected cells with I-iodide in the presence or absence of sodium butyrate. RESULTS Immunocytochemical staining and western blot analysis showed increased hNIS protein expression in A549 cells transfected with Bac-CMV-hNIS after sodium butyrate treatment. Bac-CMV-hNIS transfected A549 cells accumulated up to about nine times more I than nontransfected cells; the amount of I uptake increased in a sodium butyrate in dose-dependent manner (P<0.001). However, rapid efflux of radioactivity was observed, with 50% lost during the first 2 min after I-containing medium had been replaced by a nonradioactive medium. CONCLUSION Our results indicated that an improved efficiency of baculovirus-mediated hNIS reporter gene imaging in lung adenocarcinoma is possible with treatment with sodium butyrate. However, additional conditions need to be defined to reduce the rapid efflux of radioiodine for the purpose of radionuclide therapy.
Collapse
|
25
|
Guo H, Choudhury Y, Yang J, Chen C, Tay FC, Lim TM, Wang S. Antiglioma effects of combined use of a baculovirual vector expressing wild-type p53 and sodium butyrate. J Gene Med 2010; 13:26-36. [PMID: 21259406 DOI: 10.1002/jgm.1522] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2010] [Revised: 10/12/2010] [Accepted: 11/02/2010] [Indexed: 11/06/2022] Open
Abstract
BACKGROUND Combination therapy is usually desirable for successful cancer treatment, especially in cancers that are resistant to single forms of therapy. METHODS To achieve an optimal therapeutic effect against glioblastoma, we tested a strategy that combines baculovirus-mediated transfer of the p53 tumor suppressor gene with the use of sodium butyrate, a histone deacetylase inhibitor. This strategy was designed based on the findings that the transduction efficiency of baculovirus in mammalian cells can be markedly enhanced by the addition of histone deacetylase inhibitors and that these inhibitors are effective in inducing cell cycle arrest, differentiation, or apoptosis in tumor cells. RESULTS We observed a synergistic effect of the combination of the two treatments in provoking apoptosis in glioblastoma cells with mutant p53. In a mouse glioma xenograft model, the tumor inhibitory effect of baculovirus-expressed p53 was significantly enhanced by co-administration of sodium butyrate. CONCLUSIONS These findings suggest a new approach to treat glioblastoma using baculovirus-mediated gene transfer in combination with administration of histone deacetylase inhibitor.
Collapse
Affiliation(s)
- Haiyan Guo
- Institute of Bioengineering and Nanotechnology, Singapore
| | | | | | | | | | | | | |
Collapse
|
26
|
Kaikkonen MU, Maatta AI, Ylä-Herttuala S, Airenne KJ. Screening of complement inhibitors: shielded baculoviruses increase the safety and efficacy of gene delivery. Mol Ther 2010; 18:987-92. [PMID: 20179675 PMCID: PMC2890102 DOI: 10.1038/mt.2010.25] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
One of the major obstacles in the use of baculovirus vectors for in vivo gene transfer is the virus inactivation by serum complement. In this study, we investigated the effect of decay-accelerating factor (DAF), factor H (FH)-like protein-1 (FHL-1), C4b-binding protein (C4BP), and membrane cofactor protein (MCP) on protection of baculovirus vectors from the complement-mediated inactivation. Complement regulatory proteins were displayed on baculovirus surface as fusions to membrane anchor of the vesicular stomatitis virus-G (VSV-G) protein. This strategy resulted in abundant expression of recombinant proteins on the viral envelope while viral titers comparable to control virus were reached. The surface-modified vectors exhibited complement resistance in vitro, DAF showing the highest level of protection. Intraportal delivery of DAF-displaying baculovirus resulted in increased survival and enhanced gene expression in immunocompetent mice. Mice receiving DAF-displaying baculovirus also exhibited lower level of liver inflammation as evidenced by aspartate aminotransferase (AST). In line with this, macrophages treated with DAF baculovirus produced lower levels of inflammatory cytokines IL-1beta, IL-6, and IL-12p40 compared to control virus. These results suggest that DAF-display can protect the vector against complement inactivation but also reduce complement-mediated inflammation injury. In conclusion, complement shielded baculovirus vectors represent attractive tools for effective in vivo gene delivery.
Collapse
Affiliation(s)
- Minna U Kaikkonen
- Department of Biotechnology and Molecular Medicine, AI Virtanen Institute, University of Eastern Finland, Kuopio, Finland
| | | | | | | |
Collapse
|
27
|
Mäkelä AR, Ernst W, Grabherr R, Oker-Blom C. Baculovirus-based display and gene delivery systems. Cold Spring Harb Protoc 2010; 2010:pdb.top72. [PMID: 20194476 DOI: 10.1101/pdb.top72] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/28/2023]
Abstract
The baculovirus expression vector system has been used extensively to produce numerous proteins originating from both prokaryotic and eukaryotic sources. In addition to easy cloning techniques and abundant viral propagation, the system's insect cell environment provides eukaryotic post-translational modification machinery. The recently established eukaryotic molecular biology tool, the baculovirus display vector system (BDVS), allows the combination of genotype with phenotype, enabling presentation of foreign peptides or even complex proteins on the baculoviral envelope or capsid. This strategy is important because it can be used to enhance viral binding and entry to mammalian cells as well as to produce antibodies against the displayed antigen. In addition, the technology should enable modifications of intracellular behavior, that is, trafficking of recombinant "nanoparticles," a highly relevant feature for studies of targeted gene or protein delivery. This article discusses the design and potential uses of insect-derived baculoviral display vectors.
Collapse
|
28
|
Del Bigio MR. Ependymal cells: biology and pathology. Acta Neuropathol 2010; 119:55-73. [PMID: 20024659 DOI: 10.1007/s00401-009-0624-y] [Citation(s) in RCA: 234] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2009] [Revised: 12/03/2009] [Accepted: 12/04/2009] [Indexed: 11/28/2022]
Abstract
The literature was reviewed to summarize the current understanding of the role of ciliated ependymal cells in the mammalian brain. Previous reviews were summarized. Publications from the past 10 years highlight interactions between ependymal cells and the subventricular zone and the possible role of restricted ependymal populations in neurogenesis. Ependymal cells provide trophic support and possibly metabolic support for progenitor cells. Channel proteins such as aquaporins may be important for determining water fluxes at the ventricle wall. The junctional and anchoring proteins are now fairly well understood, as are proteins related to cilia function. Defects in ependymal adhesion and cilia function can cause hydrocephalus through several different mechanisms, one possibility being loss of patency of the cerebral aqueduct. Ependymal cells are susceptible to infection by a wide range of common viruses; while they may act as a line of first defense, they eventually succumb to repeated attacks in long-lived organisms. Ciliated ependymal cells are almost certainly important during brain development. However, the widespread absence of ependymal cells from the adult human lateral ventricles suggests that they may have only regionally restricted value in the mature brain of large size.
Collapse
Affiliation(s)
- Marc R Del Bigio
- Department of Pathology, University of Manitoba, Winnipeg, MB, Canada.
| |
Collapse
|
29
|
Mähönen AJ, Makkonen KE, Laakkonen JP, Ihalainen TO, Kukkonen SP, Kaikkonen MU, Vihinen-Ranta M, Ylä-Herttuala S, Airenne KJ. Culture medium induced vimentin reorganization associates with enhanced baculovirus-mediated gene delivery. J Biotechnol 2009; 145:111-9. [PMID: 19903502 DOI: 10.1016/j.jbiotec.2009.11.003] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2009] [Revised: 09/16/2009] [Accepted: 11/03/2009] [Indexed: 01/04/2023]
Abstract
Baculoviruses can express transgenes under mammalian promoters in a wide range of vertebrate cells. However, the success of transgene expression is dependent on both the appropriate cell type and culture conditions. We studied the mechanism behind the substantial effect of the cell culture medium on efficiency of the baculovirus transduction in different cell lines. We tested six cell culture mediums; the highest transduction efficiency was detected in the presence of RPMI 1640 medium. Vimentin, a major component of type III intermediate filaments, was reorganized in the optimized medium, which associated with enhanced nuclear entry of baculoviruses. Accordingly, the phosphorylation pattern of vimentin was changed in the studied cell lines. These results suggest that vimentin has an important role in baculovirus entry into vertebrate cells. Enhanced gene delivery in the optimized medium was observed also with adenoviruses and lentiviruses. The results highlight the general importance of the culture medium in the assembly of the cytoskeleton network and in viral gene delivery.
Collapse
Affiliation(s)
- Anssi J Mähönen
- A.I. Virtanen Institute, Department of Biotechnology and Molecular Medicine, University of Kuopio Kuopio, Finland.
| | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Polyethylenimine coating to produce serum-resistant baculoviral vectors for in vivo gene delivery. Biomaterials 2009; 30:5767-74. [PMID: 19577293 DOI: 10.1016/j.biomaterials.2009.06.020] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2009] [Accepted: 06/09/2009] [Indexed: 12/11/2022]
Abstract
Recombinant baculoviral vectors efficiently transduce many types of mammalian cells. However, their in vivo applications are hampered by the sensitivity of the virus to complement-mediated inactivation. Based on our observation that the surface charge of baculovirus is negative at neutral pH, we developed a procedure to coat baculoviral vectors with positively charged polyethylenimine 25 kDa, a commonly tested non-viral gene delivery vector, through electrostatic interaction. This coating was effective in protecting baculoviral vectors against human and rat serum-mediated inactivation in vitro, providing transduction efficiencies comparable with that generated by the control virus used under a serum-free condition. Enhanced in vivo gene expression in the liver and spleen was observed after tail vein injection of the coated viruses into mice. When injected directly into human tumor xenografts in nude mice, the coated viruses suppressed tumor development more effectively than uncoated viral vectors. These findings demonstrated the usefulness of using a simple coating method to circumvent a major obstacle to in vivo application of baculoviral vectors. The method may also serve as a flexible platform technology for improved use of the vectors, for example introducing a targeting ligand and minimizing immune responses.
Collapse
|
31
|
Boulaire J, Zhao Y, Wang S. Gene expression profiling to define host response to baculoviral transduction in the brain. J Neurochem 2009; 109:1203-14. [DOI: 10.1111/j.1471-4159.2009.06015.x] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
|
32
|
(Strept)avidin-displaying lentiviruses as versatile tools for targeting and dual imaging of gene delivery. Gene Ther 2009; 16:894-904. [PMID: 19440224 DOI: 10.1038/gt.2009.47] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Lentiviruses have shown great promise for human gene therapy. However, no optimal strategies are yet available for noninvasive imaging of virus biodistribution and subsequent transduction in vivo. We have developed a dual-imaging strategy based on avidin-biotin system allowing easy exchange of the surface ligand on HIV-derived lentivirus envelope. This was achieved by displaying avidin or streptavidin fused to the transmembrane anchor of vesicular stomatitis virus G protein on gp64-pseudotyped envelopes. Avidin and streptavidin were efficiently incorporated on virus particles, which consequently showed binding to biotin in ELISA. These vectors, conjugated to biotinylated radionuclides and engineered to express a ferritin transgene, enabled for the first-time dual imaging of virus biodistribution and transduction pattern by single-photon emission computed tomography and magnetic resonance imaging after stereotactic injection into rat brain. In addition, vector retargeting to cancer cells overexpressing CD46, epidermal growth factor and transferrin receptors using biotinylated ligands and antibodies was demonstrated in vitro. In conclusion, we have generated novel lentivirus vectors for noninvasive imaging and targeting of lentivirus-mediated gene delivery. This study suggests that these novel vectors could be applicable for the treatment of central nervous system disorders and cancer.
Collapse
|
33
|
Improved adenovirus type 5 vector-mediated transduction of resistant cells by piggybacking on coxsackie B-adenovirus receptor-pseudotyped baculovirus. J Virol 2009; 83:6048-66. [PMID: 19357170 DOI: 10.1128/jvi.00012-09] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Taking advantage of the wide tropism of baculoviruses (BVs), we constructed a recombinant BV (BV(CAR)) pseudotyped with human coxsackie B-adenovirus receptor (CAR), the high-affinity attachment receptor for adenovirus type 5 (Ad5), and used the strategy of piggybacking Ad5-green fluorescent protein (Ad5GFP) vector on BV(CAR) to transduce various cells refractory to Ad5 infection. We found that transduction of all cells tested, including human primary cells and cancer cell lines, was significantly improved using the BV(CAR)-Ad5GFP biviral complex compared to that obtained with Ad5GFP or BV(CAR)GFP alone. We determined the optimal conditions for the formation of the complex and found that a high level of BV(CAR)-Ad5GFP-mediated transduction occurred at relatively low adenovirus vector doses, compared with transduction by Ad5GFP alone. The increase in transduction was dependent on the direct coupling of BV(CAR) to Ad5GFP via CAR-fiber knob interaction, and the cell attachment of the BV(CAR)-Ad5GFP complex was mediated by the baculoviral envelope glycoprotein gp64. Analysis of the virus-cell binding reaction indicated that the presence of BV(CAR) in the complex provided kinetic benefits to Ad5GFP compared to the effects with Ad5GFP alone. The endocytic pathway of BV(CAR)-Ad5GFP did not require Ad5 penton base RGD-integrin interaction. Biodistribution of BV(CAR)-Ad5Luc complex in vivo was studied by intravenous administration to nude BALB/c mice and compared to Ad5Luc injected alone. No significant difference in viscerotropism was found between the two inocula, and the liver remained the preferred localization. In vitro, coagulation factor X drastically increased the Ad5GFP-mediated transduction of CAR-negative cells but had no effect on the efficiency of transduction by the BV(CAR)-Ad5GFP complex. Various situations in vitro or ex vivo in which our BV(CAR)-Ad5 duo could be advantageously used as gene transfer biviral vector are discussed.
Collapse
|
34
|
Clathrin-independent entry of baculovirus triggers uptake of E. coli in non-phagocytic human cells. PLoS One 2009; 4:e5093. [PMID: 19352496 PMCID: PMC2662421 DOI: 10.1371/journal.pone.0005093] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2008] [Accepted: 03/05/2009] [Indexed: 12/29/2022] Open
Abstract
The prototype baculovirus, Autographa californica multiple nucleopolyhedrovirus, an insect pathogen, holds great potential as a gene therapy vector. To develop transductional targeting and gene delivery by baculovirus, we focused on characterizing the nature and regulation of its uptake in human cancer cells. Baculovirus entered the cells along fluid-phase markers from the raft areas into smooth-surfaced vesicles devoid of clathrin. Notably, regulators associated with macropinocytosis, namely EIPA, Pak1, Rab34, and Rac1, had no significant effect on viral transduction, and the virus did not induce fluid-phase uptake. The internalization and nuclear uptake was, however, affected by mutants of RhoA, and of Arf6, a regulator of clathrin-independent entry. Furthermore, the entry of baculovirus induced ruffle formation and triggered the uptake of fluorescent E. coli bioparticles. To conclude, baculovirus enters human cells via a clathrin-independent pathway, which is able to trigger bacterial uptake. This study increases our understanding of virus entry strategies and gives new insight into baculovirus-mediated gene delivery in human cells.
Collapse
|
35
|
Balani P, Boulaire J, Zhao Y, Zeng J, Lin J, Wang S. High mobility group box2 promoter-controlled suicide gene expression enables targeted glioblastoma treatment. Mol Ther 2009; 17:1003-11. [PMID: 19240692 DOI: 10.1038/mt.2009.22] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Achievement of specific tumor cell targeting remains a challenge for glioma gene therapy. We observed that the human high mobility group box2 (HMGB2) gene had a low level of expression in normal human brain tissues, but was significantly upregulated in glioblastoma tissues. With progressive truncation of a 5'-upstream sequence of the HMGB2 gene, we identified a 0.5-kb fragment displaying a high transcriptional activity in glioblastoma cells, but a low activity in normal brain cells. To test the feasibility of using the HMGB2 promoter sequence in targeted cancer therapy, we constructed a baculoviral vector expressing the herpes simplex virus thymidine kinase (HSVtk) gene driven by the HMGB2 promoter. Transduction with the viral vector induced cell death in glioblastoma cell lines in the presence of ganciclovir (GCV), but did not affect the survival of human astrocytes and neurons. In a mouse xenograft model, intratumor injection of the baculoviral vector suppressed the growth of human glioblastoma cells and prolonged the survival of tumor-bearing mice. Our results suggest that the novel 5' sequence of HMGB2 gene has a potential to be used as an efficient, tumor-selective promoter in targeted vectors for glioblastoma gene therapy.
Collapse
Affiliation(s)
- Poonam Balani
- Institute of Bioengineering and Nanotechnology, Singapore
| | | | | | | | | | | |
Collapse
|
36
|
Interstitial tissue-specific gene expression in mouse testis by intra-tunica albuguineal injection of recombinant baculovirus. Asian J Androl 2009; 11:342-50. [PMID: 19169267 DOI: 10.1038/aja.2008.54] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
The purpose of this study is to establish a gene delivery system for interstitial tissue-specific protein expression in mice testes using modified recombinant baculovirus. Green fluorescent protein (GFP)-expressing recombinant baculovirus (GFP-baculovirus), in which the insect cell-specific polyhedron promoter was replaced by the cytomegalovirus (CMV)-IE promoter, was used to transfect testicular cells in vitro, and for intra-tunica albuguineal injection of the interstitial tissue of the testis. GFP expression was monitored in frozen testes sections by fluorescence microscopy. Expression of GFP in testicular tissues was also assessed by reverse transcription polymerase chain reaction (RT-PCR), and protein expression was assessed by Western blot. Testicular cells in vitro were infected efficiently by modified recombinant GFP-baculovirus. Intra-tunica albuguineal injection of GFP-baculovirus into the mouse testis resulted in a high level of GFP expression in the interstitial tissues. RT-PCR analysis clearly showed GFP gene expression in the testis, particularly interstitial tissues. Intra-tunica albuguineal injection of a modified baculovirus that encoded recombinant rat insulin-like growth factor binding protein (IGFBP)-5 resulted in an increase in IGFBP-5 in testis and semen. In conclusion, we have developed an efficient delivery system for gene expression in vivo in testicular cells, particularly cells of the interstitial tissue using intra-tunica albuguineal injection of a modified recombinant baculovirus. This method will be particularly relevant for application that requires gene delivery and protein expression in the testicular cells of the outer seminiferous tubule of the testis.
Collapse
|
37
|
Mäkelä AR, Enbäck J, Laakkonen JP, Vihinen-Ranta M, Laakkonen P, Oker-Blom C. Tumor targeting of baculovirus displaying a lymphatic homing peptide. J Gene Med 2009; 10:1019-31. [PMID: 18655234 DOI: 10.1002/jgm.1222] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
BACKGROUND Tumor-associated cells and vasculature express attractive molecular markers for site-specific vector targeting. To attain tumor-selective tropism, we recently developed a baculovirus vector displaying the lymphatic homing peptide LyP-1, originally identified by ex vivo/in vivo screening of phage display libraries, on the viral envelope by fusion to the transmembrane anchor of vesicular stomatitis virus G-protein. METHODS In the present study, we explored the specificity and kinetics of viral binding and internalization as well as in vivo tumor homing of the LyP-1 displaying virus to elucidate the applicability of baculovirus for targeted therapies. RESULTS We demonstrated that the LyP-1 peptide contributes to saturable binding of baculovirus in human MDA-MB-435 and HepG2 carcinoma cells and escalates the kinetics of viral internalization leading to earlier nuclear accumulation and enhanced transgene expression. The LyP-1 displaying virus also showed stronger competitiveness against transduction with wild-type baculovirus, suggesting involvement of a specific receptor in cellular attachment and entry. Following intravenous injections, the modified virus accumulated within the human MDA-MB-435 and MDA-MB-231 carcinoma xenografts in mice with higher specificity and efficiency than the control virus. Targeting of the modified virus was more specific in the MDA-MB-435 than in the MDA-MB-231 xenografts as demonstrated by higher tumor accumulation and lower distribution in nontarget organs. No apparent cytotoxicity was associated with the surface modification. CONCLUSIONS This first demonstration of in vivo tumor targeting of a systemically administered, tropism-modified baculoviral vector highlights the potential of baculovirus-mediated targeted therapies.
Collapse
Affiliation(s)
- Anna R Mäkelä
- NanoScience Center, Department of Biological and Environmental Science, University of Jyväskylä, Finland.
| | | | | | | | | | | |
Collapse
|
38
|
Mäkelä AR, Närvänen A, Oker-Blom C. Peptide-mediated interference with baculovirus transduction. J Biotechnol 2008; 134:20-32. [PMID: 18294718 DOI: 10.1016/j.jbiotec.2007.12.010] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2007] [Revised: 12/14/2007] [Accepted: 12/17/2007] [Indexed: 10/22/2022]
Abstract
Baculovirus represents a multifunctional platform with potential for biomedical applications including disease therapies. The importance of F3, a tumor-homing peptide, in baculovirus transduction was previously recognized by the ability of F3 to augment viral binding and gene delivery to human cancer cells following display on the viral envelope. Here, F3 was utilized as a molecular tool to expand understanding of the poorly characterized baculovirus-mammalian cell interactions. Baculovirus-mediated transduction of HepG2 hepatocarcinoma cells was strongly inhibited by coincubating the virus with synthetic F3 or following incorporation of F3 into viral nucleocapsid by genetic engineering, the former suggesting direct interaction of the soluble peptide with the virus particles. Since internalization and nuclear accumulation of the virus were significantly inhibited or delayed, but the kinetics of viral binding, initial uptake, and endosomal release were unaffected, F3 likely interferes with cytoplasmic trafficking and subsequent nuclear transport of the virus. A polyclonal antibody raised against nucleolin, the internalizing receptor of F3, failed to inhibit cellular binding, but considerably reduced viral transduction efficiency, proposing the involvement of nucleolin in baculovirus entry. Together, these results render the F3 peptide a tool for elucidating the mechanism and molecular details conferring to baculovirus-mediated gene transduction in mammalian cells.
Collapse
Affiliation(s)
- Anna R Mäkelä
- NanoScience Center, Department of Biological and Environmental Science, PO Box 35, FIN-40014 University of Jyväskylä, Finland.
| | | | | |
Collapse
|
39
|
|
40
|
Tani H, Abe T, Matsunaga TM, Moriishi K, Matsuura Y. Baculovirus vector for gene delivery and vaccine development. Future Virol 2008. [DOI: 10.2217/17460794.3.1.35] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The baculovirus Autographa californica multiple nucleopolyhedrovirus has been widely used not only to acheive a high level of foreign gene expression in insect cells, but also for efficient gene transduction into mammalian cells. Recombinant and pseudotyped baculoviruses possessing chimeric or foreign ligands have been constructed to improve the efficiency of gene transduction and to confer specificity for gene delivery into mammalian cells, respectively. Baculoviral DNA CpG motifs induce proinflammatory cytokines through a Toll-like receptor (TLR9)/MyD88-dependent signaling pathway. Other baculovirus components produce type I interferons via a TLR-independent pathway. Baculovirus exhibits a strong adjuvant property and recombinant baculoviruses encoding microbial antigens elicit antibodies to the antigens and provide protective immunity in mice. This review deals with recent progress in the application of baculovirus vectors to gene delivery and vaccine development, and discusses the future prospects of baculovirus vectors.
Collapse
Affiliation(s)
- Hideki Tani
- Osaka University, Department of Molecular Virology, Research Institute for Microbial Diseases, Osaka, Japan
| | - Takayuki Abe
- Osaka University, Department of Molecular Virology, Research Institute for Microbial Diseases, Osaka, Japan
| | - Tomoko M Matsunaga
- Osaka University, Department of Molecular Virology, Research Institute for Microbial Diseases, Osaka, Japan
| | - Kohji Moriishi
- Osaka University, Department of Molecular Virology, Research Institute for Microbial Diseases, Osaka, Japan
| | - Yoshiharu Matsuura
- Osaka University, Department of Molecular Virology, Research Institute for Microbial Diseases, 3-1 Yamada-oka, Suita, Osaka 565-0871, Japan
| |
Collapse
|
41
|
Laakkonen JP, Kaikkonen MU, Ronkainen PHA, Ihalainen TO, Niskanen EA, Häkkinen M, Salminen M, Kulomaa MS, Ylä-Herttuala S, Airenne KJ, Vihinen-Ranta M. Baculovirus-mediated immediate-early gene expression and nuclear reorganization in human cells. Cell Microbiol 2007; 10:667-81. [PMID: 18042259 DOI: 10.1111/j.1462-5822.2007.01074.x] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Baculovirus, Autographa californica multiple nucleopolyhedrovirus (AcMNPV), has the ability to transduce mammalian cell lines without replication. The general objective of this study was to detect the transcription and expression of viral immediate-early genes in human cells and to examine the interactions between viral components and subnuclear structures. Viral capsids were seen in large, discrete foci in nuclei of both dividing and non-dividing human cells. Concurrently, the transcription of viral immediate-early transregulator genes (ie-1, ie-2) and translation of IE-2 protein were detected. Quantitative microscopy imaging and analysis showed that virus transduction altered the size of promyelocytic leukaemia nuclear bodies, which are suggested to be involved in replication and transcription of various viruses. Furthermore, altered distribution of the chromatin marker Draq5 and histone core protein (H2B) in transduced cells indicated that the virus was able to induce remodelling of the host cell chromatin. To conclude, this study shows that the non-replicative insect virus, baculovirus and its proteins can induce multiple changes in the cellular machinery of human cells.
Collapse
Affiliation(s)
- Johanna P Laakkonen
- NanoScience Center, Department of Biological and Environmental Science, University of Jyväskylä, Jyväskylä, Finland.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Wang J, Li B, Cai C, Zhang Y, Wang S, Hu S, Tian X, Zhang M. Efficient transduction of spiral ganglion neurons in vitro by baculovirus vectors. Neuroreport 2007; 18:1329-33. [PMID: 17762707 DOI: 10.1097/wnr.0b013e3282010b16] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Degeneration of peripheral auditory neurons constitutes one of the main causes of sensorineural hearing loss. Gene delivery to the inner ear is central to the development of gene therapy for hearing impairment. Thus we investigated the effectiveness of baculovirus-derived vectors to transduce spiral ganglion neurons. We found that baculovirus could efficiently transduce spiral ganglion neurons in vitro and that the highest transduced cell rate could be over 75%. The level of transgene expression exhibited viral dose dependence and was enhanced by the addition of butyrate. Thus, baculovirus is a novel and promising tool for gene transfer into the cochlear nervous system, both in studies of the function of foreign genes and in the development of gene-therapy strategies.
Collapse
Affiliation(s)
- Jun Wang
- Department of Otolaryngology, Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | | | | | | | | | | | | | | |
Collapse
|
43
|
Mähönen AJ, Airenne KJ, Purola S, Peltomaa E, Kaikkonen MU, Riekkinen MS, Heikura T, Kinnunen K, Roschier MM, Wirth T, Ylä-Herttuala S. Post-transcriptional regulatory element boosts baculovirus-mediated gene expression in vertebrate cells. J Biotechnol 2007; 131:1-8. [PMID: 17617485 DOI: 10.1016/j.jbiotec.2007.05.022] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2006] [Revised: 05/09/2007] [Accepted: 05/21/2007] [Indexed: 10/23/2022]
Abstract
Baculoviruses can express transgenes in a wide range of vertebrate cells. However, in some cells transgene expression is weak. To enhance transgene expression, we studied the effect of the Woodchuck hepatitis virus post-transcriptional regulatory element (WPRE) on baculovirus (BV)-mediated gene expression of several transgenes. A significant increase in BV-mediated gene expression was detected in several cell lines. A 10-fold increase in transgene expression was observed with the WPRE as determined by the percentage of positive cells and mean fluorescence intensity (MFI). Furthermore, a combination of optimized cell culture medium and WPRE virus led to more than a 60-fold increase in gene expression. In accordance, elevated mRNA and protein levels were detected in WPRE-virus transduced cells. In HepG2 and RaaSMC, WPRE-mediated enhancement was comparable to the previously shown positive effect of sodium butyrate on BV-mediated gene expression. Thus, inclusion of the WPRE into a baculovirus vector provides a simple means to improve BV-mediated gene expression in vertebrate cells.
Collapse
Affiliation(s)
- Anssi J Mähönen
- A.I. Virtanen Institute, Department of Biotechnology and Molecular Medicine, Kuopio, Finland
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Räty JK, Liimatainen T, Huhtala T, Kaikkonen MU, Airenne KJ, Hakumäki JM, Närvänen A, Ylä-Herttuala S. SPECT/CT imaging of baculovirus biodistribution in rat. Gene Ther 2007; 14:930-8. [PMID: 17410181 DOI: 10.1038/sj.gt.3302934] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Non-invasive imaging provides essential information regarding the biodistribution of gene therapy vectors and it can also be used for the development of targeted vectors. In this study, we have utilized micro Single-photon emission computed tomography to visualize biodistribution of a (99m)Tc-polylys-ser-DTPA-biotin-labelled avidin-displaying baculovirus, Baavi, after intrafemoral (i.f.), intraperitoneal (i.p.), intramuscular (i.m.) and intracerebroventricular (i.c.v.) administration. The imaging results suggest that the virus can spread via the lymphatic network after different administration routes, also showing accumulation in the nasal area after systemic administration. Extensive expression in the kidneys and spleen was seen after i.p. administration, which was confirmed by reverse transcriptase-polymerase chain reaction and immunohistochemistry. Additionally, transduction of kidneys was seen with i.m. and i.f. administrations. We conclude that baculovirus may be beneficial for the treatment of kidney diseases after i.p. administration route.
Collapse
Affiliation(s)
- J K Räty
- Department of Biotechnology and Molecular Medicine, AI Virtanen Institute for Molecular Sciences, University of Kuopio, Kuopio, Finland
| | | | | | | | | | | | | | | |
Collapse
|
45
|
Kaneko H, Suzuki H, Abe T, Miyano-Kurosaki N, Takaku H. Inhibition of HIV-1 replication by vesicular stomatitis virus envelope glycoprotein pseudotyped baculovirus vector-transduced ribozyme in mammalian cells. Biochem Biophys Res Commun 2006; 349:1220-7. [PMID: 16979590 DOI: 10.1016/j.bbrc.2006.08.184] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2006] [Accepted: 08/25/2006] [Indexed: 11/18/2022]
Abstract
The baculovirus has recently emerged as a promising vector for in vivo gene therapy. To investigate its potential as a delivery vector for an anti-virus ribozyme targeting HIV-1, we constructed recombinant baculovirus vectors bearing a ribozyme-synthesizing cassette driven by the tRNA(i)(Met) promoter with enhanced transduction efficiency by displaying vesicular stomatitis virus glycoprotein (VSV-G) on the viral envelope. Transduction of HeLa CD4(+) cells with a recombinant baculovirus delivering the HIV-1 U5 gene-specific ribozyme dramatically suppressed HIV-1 expression in this cell line. The VSV-G pseudotyped baculovirus vector-transduced ribozyme potently inhibited HIV-1 replication compared to a recombinant baculovirus vector-transduced ribozyme lacking VSV-G. The use of a baculovirus vector might be beneficial for application in gene therapy.
Collapse
Affiliation(s)
- Hiroyasu Kaneko
- Department of Life and Environmental Science, Faculty of Engineering, Chiba Institute of Technology, 2-17-1 Tsudanuma, Narashino, Chiba 275-0016, Japan
| | | | | | | | | |
Collapse
|
46
|
Räty JK, Liimatainen T, Wirth T, Airenne KJ, Ihalainen TO, Huhtala T, Hamerlynck E, Vihinen-Ranta M, Närvänen A, Ylä-Herttuala S, Hakumäki JM. Magnetic resonance imaging of viral particle biodistribution in vivo. Gene Ther 2006; 13:1440-6. [PMID: 16855615 DOI: 10.1038/sj.gt.3302828] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
We describe here a technique for the visualization of viral vector delivery by magnetic resonance imaging (MRI) in vivo. By conjugating avidin-coated baculoviral vectors (Baavi) with biotinylated ultra-small superparamagnetic iron oxide particles (USPIO), we are able to produce vector-related MRI contrast in the choroid plexus cells of rat brain in vivo over a period of 14 days. Ten microlitres of 2.5 x 10(10) PFU/ml nuclear-targeted LacZ-encoding Baavi with bUSPIO coating was injected into rat brain ventricles and visualized by MRI at 4.7 T. As baculoviruses exhibit restricted cell-type specificity in the rat brain, altered MRI contrast was detected in the choroid plexus of the injected ventricles. No specific signal loss was detected when wild-type baculoviruses or intact biotinylated USPIO particles were injected into the lateral ventricles. Cryosectioned brains were stained for nuclear-targeted beta-galactosidase gene expression, which was found to colocalize with MRI contrast. This study provides the first proof of principle for robust and non-invasive viral vector MRI by using avidin-displaying viruses in vivo. Considering the widespread use of MRI in current medical imaging, the approach is likely to provide numerous future applications in imaging of therapeutic gene transfer.
Collapse
Affiliation(s)
- J K Räty
- Department of Biotechnology and Molecular Medicine, University of Kuopio, Kuopio, Finland.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Mäkelä AR, Matilainen H, White DJ, Ruoslahti E, Oker-Blom C. Enhanced baculovirus-mediated transduction of human cancer cells by tumor-homing peptides. J Virol 2006; 80:6603-11. [PMID: 16775347 PMCID: PMC1488948 DOI: 10.1128/jvi.00528-06] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2006] [Accepted: 04/20/2006] [Indexed: 01/05/2023] Open
Abstract
Tumor cells and vasculature offer specific targets for the selective delivery of therapeutic genes. To achieve tumor-specific gene transfer, baculovirus tropism was manipulated by viral envelope modification using baculovirus display technology. LyP-1, F3, and CGKRK tumor-homing peptides, originally identified by in vivo screening of phage display libraries, were fused to the transmembrane anchor of vesicular stomatitis virus G protein and displayed on the baculoviral surface. The fusion proteins were successfully incorporated into budded virions, which showed two- to fivefold-improved binding to human breast carcinoma (MDA-MB-435) and hepatocarcinoma (HepG2) cells. The LyP-1 peptide inhibited viral binding to MDA-MB-435 cells with a greater magnitude and specificity than the CGKRK and F3 peptides. Maximal 7- and 24-fold increases in transduction, determined by transgene expression level, were achieved for the MDA-MB-435 and HepG2 cells, respectively. The internalization of each virus was inhibited by ammonium chloride treatment, suggesting the use of a similar endocytic entry route. The LyP-1 and F3 peptides showed an apparent inhibitory effect in transduction of HepG2 cells with the corresponding display viruses. Together, these results imply that the efficiency of baculovirus-mediated gene delivery can be significantly enhanced in vitro when tumor-targeting ligands are used and therefore highlight the potential of baculovirus vectors in cancer gene therapy.
Collapse
Affiliation(s)
- Anna R Mäkelä
- NanoScience Center, Department of Biological and Environmental Science, P.O. Box 35, FIN-40014 University of Jyväskylä, Finland
| | | | | | | | | |
Collapse
|
48
|
Wang CY, Wang S. Astrocytic expression of transgene in the rat brain mediated by baculovirus vectors containing an astrocyte-specific promoter. Gene Ther 2006; 13:1447-56. [PMID: 16724097 DOI: 10.1038/sj.gt.3302771] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Therapeutic gene expression in glial cells has been tested for the treatment of neurological diseases in animal models. Many of such studies used the promoter of the glial fibrillary acidic protein (GFAP) to restrict gene expression to astrocytes. We have investigated in the current study whether it is possible to improve the transcriptional activity of the cellular promoter, while maintaining its cell-type specificity. We constructed an expression cassette containing a hybrid cytomegalovirus (CMV) enhancer/GFAP promoter and placed it into baculovirus vectors, a type of viral vectors capable of transducing astrocytes. In another vector design, we used inverted terminal repeats (ITRs) from adeno-associated virus (AAV) to flank the expression cassette. The recombinant baculoviruses with the hybrid promoter improved gene expression levels over two orders of magnitude in glial cell lines and by 10-fold in the rat brain when compared to the baculoviruses with the GFAP promoter alone. The expression was further improved by ITR flanking, reaching levels higher than that mediated by the baculovirus vectors with the CMV immediate-early enhancer/promoter (CMV promoter). Using these recombinant baculoviruses, we observed extended in vivo transgene expression in the rat brain at 90 days postinjection, by which time the gene expression from baculovirus vectors with the GFAP or CMV promoter had already become undetectable. The astrocyte specificity of the GFAP promoter was preserved in the engineered expression cassette with the CMV enhancer and the AAV ITRs, as demonstrated by immunohistological analysis of brain samples and an axonal retrograde transport assay. Taken together, our findings suggest that these baculovirus vectors may serve as useful tools for astrocyte-specific gene expression in the brain.
Collapse
Affiliation(s)
- C Y Wang
- Institute of Bioengineering and Nanotechnology, National University of Singapore, Singapore, Singapore
| | | |
Collapse
|
49
|
Raschperger E, Thyberg J, Pettersson S, Philipson L, Fuxe J, Pettersson RF. The coxsackie- and adenovirus receptor (CAR) is an in vivo marker for epithelial tight junctions, with a potential role in regulating permeability and tissue homeostasis. Exp Cell Res 2006; 312:1566-80. [PMID: 16542650 DOI: 10.1016/j.yexcr.2006.01.025] [Citation(s) in RCA: 104] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2005] [Revised: 01/16/2006] [Accepted: 01/18/2006] [Indexed: 10/24/2022]
Abstract
The coxsackie- and adenovirus receptor (CAR) is a transmembrane protein belonging to the immunoglobulin superfamily. The function of CAR as a virus receptor has been extensively analyzed, while its physiological role and expression pattern in adult tissues have remained less clear. CAR associates with epithelial tight junctions in vitro and mediates cell-cell adhesion. Using a set of affinity-purified antibodies, we show that CAR is predominantly expressed in epithelial cells lining the body cavities in adult mice, where it specifically co-localizes with the tight junction components ZO-1 and occludin. Notably, CAR could not be detected in endothelial cells of the vasculature, including brain capillaries. CAR expression correlated positively with the maturity of tight junctions and inversely with permeability. With a few exceptions, the two known CAR isoforms were co-expressed in most epithelial cells analyzed. A CAR mutant lacking the intracellular tail over-expressed in transgenic mice was diffusely localized over the plasma membrane, showing the importance of this domain for correct subcellular localization in vivo. We conclude that CAR is localized to epithelial tight junctions in vivo where it may play a role in the regulation of epithelial permeability and tissue homeostasis.
Collapse
|
50
|
Kenoutis C, Efrose RC, Swevers L, Lavdas AA, Gaitanou M, Matsas R, Iatrou K. Baculovirus-mediated gene delivery into Mammalian cells does not alter their transcriptional and differentiating potential but is accompanied by early viral gene expression. J Virol 2006; 80:4135-46. [PMID: 16571829 PMCID: PMC1440473 DOI: 10.1128/jvi.80.8.4135-4146.2006] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2005] [Accepted: 02/01/2006] [Indexed: 12/22/2022] Open
Abstract
Gene delivery to neural cells is central to the development of transplantation therapies for neurological diseases. In this study, we used a baculovirus derived from the domesticated silk moth, Bombyx mori, as vector for transducing a human cell line (HEK293) and primary cultures of rat Schwann cells. Under optimal conditions of infection with a recombinant baculovirus containing the reporter green fluorescent protein gene under mammalian promoter control, the infected cells express the transgene with high efficiency. Toxicity assays and transcriptome analyses suggest that baculovirus infection is not cytotoxic and does not induce differential transcriptional responses in HEK293 cells. Infected Schwann cells retain their characteristic morphological and molecular phenotype as determined by immunocytochemistry for the marker proteins S-100, glial fibrillary acidic protein, and p75 nerve growth factor receptor. Moreover, baculovirus-infected Schwann cells are capable of differentiating in vitro and express the P0 myelination marker. However, transcripts for several immediate-early viral genes also accumulate in readily detectable levels in the transduced cells. This transcriptional activity raises concerns regarding the long-term safety of baculovirus vectors for gene therapy applications. Potential approaches for overcoming the identified problem are discussed.
Collapse
Affiliation(s)
- Christos Kenoutis
- Insect Molecular Genetics and Biotechnology Group, Institute of Biology, National Centre for Scientific Research Demokritos, P.O. Box 60228, 153 10 Aghia Paraskevi Attikis (Athens), Greece
| | | | | | | | | | | | | |
Collapse
|