1
|
Huang Y, Mei H, Deng C, Wang W, Yuan C, Nie Y, Li JD, Liu J. EXTL3 and NPC1 are mammalian host factors for Autographa californica multiple nucleopolyhedrovirus infection. Nat Commun 2024; 15:7711. [PMID: 39231976 PMCID: PMC11374996 DOI: 10.1038/s41467-024-52193-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Accepted: 08/28/2024] [Indexed: 09/06/2024] Open
Abstract
Baculovirus is an obligate parasitic virus of the phylum Arthropoda. Baculovirus including Autographa californica multiple nucleopolyhedrovirus (AcMNPV) has been widely used in the laboratory and industrial preparation of proteins or protein complexes. Due to its large packaging capacity and non-replicative and non-integrative natures in mammals, baculovirus has been proposed as a gene therapy vector for transgene delivery. However, the mechanism of baculovirus transduction in mammalian cells has not been fully illustrated. Here, we employed a cell surface protein-focused CRISPR screen to identify host dependency factors for baculovirus transduction in mammalian cells. The screening experiment uncovered a series of baculovirus host factors in human cells, including exostosin-like glycosyltransferase 3 (EXTL3) and NPC intracellular cholesterol transporter 1 (NPC1). Further investigation illustrated that EXTL3 affected baculovirus attachment and entry by participating in heparan sulfate biosynthesis. In addition, NPC1 promoted baculovirus transduction by mediating membrane fusion and endosomal escape. Moreover, in vivo, baculovirus transduction in Npc1-/+ mice showed that disruption of Npc1 gene significantly reduced baculovirus transduction in mouse liver. In summary, our study revealed the functions of EXTL3 and NPC1 in baculovirus attachment, entry, and endosomal escape in mammalian cells, which is useful for understanding baculovirus transduction in human cells.
Collapse
Affiliation(s)
- Yuege Huang
- Furong Laboratory, Center for Medical Genetics, School of Life Sciences, Central South University, Changsha, Hunan, China
| | - Hong Mei
- Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech University, Shanghai, China.
| | - Chunchen Deng
- Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech University, Shanghai, China
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| | - Wei Wang
- Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech University, Shanghai, China
| | - Chao Yuan
- Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech University, Shanghai, China
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| | - Yan Nie
- Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech University, Shanghai, China
| | - Jia-Da Li
- Furong Laboratory, Center for Medical Genetics, School of Life Sciences, Central South University, Changsha, Hunan, China.
- Hunan Key Laboratory of Animal Models for Human Diseases, Changsha, Hunan, China.
| | - Jia Liu
- Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech University, Shanghai, China.
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China.
- Shanghai Clinical Research and Trial Center, Shanghai, China.
- Guangzhou Laboratory, Guangzhou International Bio Island, Guangzhou, Guangdong, China.
- Shanghai Asiflyerbio Biotechnology, Shanghai, China.
| |
Collapse
|
2
|
Xue Q, Swevers L, Taning CNT. Plant and insect virus-like particles: emerging nanoparticles for agricultural pest management. PEST MANAGEMENT SCIENCE 2023; 79:2975-2991. [PMID: 37103223 DOI: 10.1002/ps.7514] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Revised: 04/20/2023] [Accepted: 04/27/2023] [Indexed: 06/05/2023]
Abstract
Virus-like particles (VLPs) represent a biodegradable, biocompatible nanomaterial made from viral coat proteins that can improve the delivery of antigens, drugs, nucleic acids, and other substances, with most applications in human and veterinary medicine. Regarding agricultural viruses, many insect and plant virus coat proteins have been shown to assemble into VLPs accurately. In addition, some plant virus-based VLPs have been used in medical studies. However, to our knowledge, the potential application of plant/insect virus-based VLPs in agriculture remains largely underexplored. This review focuses on why and how to engineer coat proteins of plant/insect viruses as functionalized VLPs, and how to exploit VLPs in agricultural pest control. The first part of the review describes four different engineering strategies for loading cargo at the inner or the outer surface of VLPs depending on the type of cargo and purpose. Second, the literature on plant and insect viruses the coat proteins of which have been confirmed to self-assemble into VLPs is reviewed. These VLPs are good candidates for developing VLP-based agricultural pest control strategies. Lastly, the concepts of plant/insect virus-based VLPs for delivering insecticidal and antiviral components (e.g., double-stranded RNA, peptides, and chemicals) are discussed, which provides future prospects of VLP application in agricultural pest control. In addition, some concerns are raised about VLP production on a large scale and the short-term resistance of hosts to VLP uptake. Overall, this review is expected to stimulate interest and research exploring plant/insect virus-based VLP applications in agricultural pest management. © 2023 Society of Chemical Industry.
Collapse
Affiliation(s)
- Qi Xue
- Department of Plants and Crops, Faculty of Bioscience Engineering, Ghent University, Ghent, Belgium
| | - Luc Swevers
- Insect Molecular Genetics and Biotechnology, Institute of Biosciences and Applications, National Centre for Scientific Research "Demokritos", Athens, Greece
| | - Clauvis Nji Tizi Taning
- Department of Plants and Crops, Faculty of Bioscience Engineering, Ghent University, Ghent, Belgium
| |
Collapse
|
3
|
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
|
4
|
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
|
5
|
B S A, Puthumana J, Sukumaran V, Vazhappilly CG, Kombiyil S, Philip R, Singh ISB. A Novel Approach of Transducing Recombinant Baculovirus into Primary Lymphoid Cells of Penaeus monodon for Developing Continuous Cell Line. MARINE BIOTECHNOLOGY (NEW YORK, N.Y.) 2021; 23:517-528. [PMID: 34241714 DOI: 10.1007/s10126-021-10043-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Accepted: 06/03/2021] [Indexed: 06/13/2023]
Abstract
Cell line development from shrimp is not a novel venture as researchers across the globe have been trying to have crustacean cell lines over 30 years. The reason for not attaining a crustacean or precisely a shrimp cell line is believed to be the replicative senescence and the inability to maintain telomere length in vitro. Moreover, spontaneous in vitro transformations do not happen in shrimp cells. Oncogenic induction in primary cell culture is one of the ways to attain in vitro transformation by way of disrupting the mechanisms which involve cellular senescence. In this context, a recombinant baculovirus with shrimp viral promoter IHHNV-P2 was used for the transduction aimed at immortalization. An oncogene, H-ras, was successfully amplified and cloned in to the baculoviral vector, downstream to shrimp viral promoter IHHNV-P2 and upstream to GFP. Recombinant baculovirus with H-ras was generated and used for transduction into shrimp lymphoid cells during early dividing stage. Accordingly, fibroblast-like primary cell culture got developed, and H-ras and GFP expression could be confirmed. The study suggests that the simple method of incubating recombinant baculovirus with minced tissue enables in vitro transduction during early dividing stage of the cells, and the transduction efficiency gets enhanced by adding 5 mM sodium butyrate to the culture medium.
Collapse
Affiliation(s)
- Anoop B S
- National Centre for Aquatic Animal Health, Cochin University of Science and Technology, Fine Arts Avenue, Kochi, Kerala, 682016, India
| | - Jayesh Puthumana
- National Centre for Aquatic Animal Health, Cochin University of Science and Technology, Fine Arts Avenue, Kochi, Kerala, 682016, India
| | - Vrinda Sukumaran
- National Centre for Aquatic Animal Health, Cochin University of Science and Technology, Fine Arts Avenue, Kochi, Kerala, 682016, India
- CSIR-National Institute of Oceanography, Regional Centre, Cochin, 682019, India
| | - Cijo George Vazhappilly
- National Centre for Aquatic Animal Health, Cochin University of Science and Technology, Fine Arts Avenue, Kochi, Kerala, 682016, India
- Department of Biotechnology, American University of Ras Al Khaimah, Ras Al Khaimah, UAE
| | - Salini Kombiyil
- National Centre for Aquatic Animal Health, Cochin University of Science and Technology, Fine Arts Avenue, Kochi, Kerala, 682016, India
| | - Rosamma Philip
- Department of Marine Biology, Microbiology and Biochemistry, School of Marine Sciences, Cochin University of Science and Technology, Fine Arts Avenue, Kochi, Kerala, 682016, India
| | - Isaac Sarojini Bright Singh
- National Centre for Aquatic Animal Health, Cochin University of Science and Technology, Fine Arts Avenue, Kochi, Kerala, 682016, India.
| |
Collapse
|
6
|
Schaly S, Ghebretatios M, Prakash S. Baculoviruses in Gene Therapy and Personalized Medicine. Biologics 2021; 15:115-132. [PMID: 33953541 PMCID: PMC8088983 DOI: 10.2147/btt.s292692] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2020] [Accepted: 02/22/2021] [Indexed: 12/18/2022]
Abstract
This review will outline the role of baculoviruses in gene therapy and future potential in personalized medicine. Baculoviruses are a safe, non-toxic, non-integrative vector with a large cloning capacity. Baculoviruses are also a highly adaptable, low-cost vector with a broad tissue and host tropism due to their ability to infect both quiescent and proliferating cells. Moreover, they only replicate in insect cells, not mammalian cells, improving their biosafety. The beneficial properties of baculoviruses make it an attractive option for gene delivery. The use of baculoviruses in gene therapy has advanced significantly, contributing to vaccine production, anti-cancer therapies and regenerative medicine. Currently, baculoviruses are primarily used for recombinant protein production and vaccines. This review will also discuss methods to optimize baculoviruses protein production and mammalian cell entry, limitations and potential for gene therapy and personalized medicine. Limitations such as transient gene expression, complement activation and virus fragility are discussed in details as they can be overcome through further genetic modifications and other methods. This review concludes that baculoviruses are an excllent candidate for gene therapy, personalized medicine and other biotherapeutic applications.
Collapse
Affiliation(s)
- Sabrina Schaly
- Biomedical Technology and Cell Therapy Research Laboratory, Department of Biomedical Engineering, Faculty of Medicine, McGill University, Montreal, Quebec, H3A 2B4, Canada
| | - Merry Ghebretatios
- Biomedical Technology and Cell Therapy Research Laboratory, Department of Biomedical Engineering, Faculty of Medicine, McGill University, Montreal, Quebec, H3A 2B4, Canada
| | - Satya Prakash
- Biomedical Technology and Cell Therapy Research Laboratory, Department of Biomedical Engineering, Faculty of Medicine, McGill University, Montreal, Quebec, H3A 2B4, Canada
| |
Collapse
|
7
|
Binder JL, Chander P, Deretic V, Weick JP, Bhaskar K. Optical induction of autophagy via Transcription factor EB (TFEB) reduces pathological tau in neurons. PLoS One 2020; 15:e0230026. [PMID: 32208437 PMCID: PMC7092971 DOI: 10.1371/journal.pone.0230026] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2019] [Accepted: 02/19/2020] [Indexed: 01/25/2023] Open
Abstract
Pathological accumulation of microtubule associated protein tau in neurons is a major neuropathological hallmark of Alzheimer's disease (AD) and related tauopathies. Several attempts have been made to promote clearance of pathological tau (p-Tau) from neurons. Transcription factor EB (TFEB) has shown to clear p-Tau from neurons via autophagy. However, sustained TFEB activation and autophagy can create burden on cellular bioenergetics and can be deleterious. Here, we modified previously described two-plasmid systems of Light Activated Protein (LAP) from bacterial transcription factor-EL222 and Light Responsive Element (LRE) to encode TFEB. Upon blue-light (465 nm) illumination, the conformation changes in LAP induced LRE-driven expression of TFEB, its nuclear entry, TFEB-mediated expression of autophagy-lysosomal genes and clearance of p-Tau from neuronal cells and AD patient-derived human iPSC-neurons. Turning the blue-light off reversed the expression of TFEB-target genes and attenuated p-Tau clearance. Together, these results suggest that optically regulated TFEB expression unlocks the potential of opto-therapeutics to treat AD and other dementias.
Collapse
Affiliation(s)
- Jessica L. Binder
- Department of Molecular Genetics and Microbiology, University of New Mexico Health Sciences Center, Albuquerque, New Mexico, United States of America
| | - Praveen Chander
- Department of Neurosciences, University of New Mexico Health Sciences Center, Albuquerque, New Mexico, United States of America
| | - Vojo Deretic
- Department of Molecular Genetics and Microbiology, University of New Mexico Health Sciences Center, Albuquerque, New Mexico, United States of America
- Autophagy Inflammation and Metabolism Center of Biomedical Research Excellence (CoBRE), University of New Mexico Health Sciences Center, Albuquerque, New Mexico, United States of America
| | - Jason P. Weick
- Department of Neurosciences, University of New Mexico Health Sciences Center, Albuquerque, New Mexico, United States of America
| | - Kiran Bhaskar
- Department of Molecular Genetics and Microbiology, University of New Mexico Health Sciences Center, Albuquerque, New Mexico, United States of America
| |
Collapse
|
8
|
Wang Z, Li M, Ji Y, Yang M, Yang W, Wang J, Li W. Development of a novel bivalent baculovirus vectors for complement resistance and sustained transgene expression and its application in anti-angiogenesis gene therapy. Biomed Pharmacother 2019; 123:109765. [PMID: 31846843 DOI: 10.1016/j.biopha.2019.109765] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2019] [Revised: 12/05/2019] [Accepted: 12/05/2019] [Indexed: 11/28/2022] Open
Abstract
Baculovirus (BV) is a potential gene delivery vector but only mediates transient transgene expression and easily inactivated by human complement. To this end, we intend to develop a novel bivalent BV vector for complement resistance and sustained transgene expression, and evaluate its effect in anti-angiogenesis gene therapy. The results showed that the hybrid bivalent BV significantly prolonged the expression of enhanced green fluorescent protein (eGFP) in vitro for at least 90 days at over 109 a.u. total fluorescence intensity, and exhibited significantly higher complement resistance. The control BV-mediated eGFP expression gradually declined within 15 days and showed lower transduction efficiency. In vivo studies confirmed that the hybrid bivalent BV exhibited longer duration of eGFP expression and higher transduction efficacy than the control BVs. Based on these findings, we further constructed a hybrid BV expressing the antiangiogenic fusion protein containing human endostatin and angiostatin (hEA). The hybrid BV-expressed hEA significantly prolonged the expression level of hEA with enhanced anti-angiogenic activities compared to the control groups, as evidenced by ELISA, cell proliferation, migration and tubular formation assays. With the stable expression of hEA, the hybrid BV conferred hEA more significant inhibitory effect on hepatocellular carcinoma tumor growth and significantly extended the life span of mice. These data implicate that the SB-based BV surface display system may have broad prospects as a novel platform for gene therapy of tumors.
Collapse
Affiliation(s)
- Zhisheng Wang
- School of Pharmacy, Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region, 750004, PR China; Laboratory Animal Center, Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region, 750004, PR China.
| | - Mengting Li
- School of Pharmacy, Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region, 750004, PR China
| | - Yonggan Ji
- School of Pharmacy, Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region, 750004, PR China
| | - Mengmeng Yang
- Laboratory Animal Center, Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region, 750004, PR China
| | - Wen Yang
- Laboratory Animal Center, Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region, 750004, PR China
| | - Jinbao Wang
- Laboratory Animal Center, Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region, 750004, PR China
| | - Wei Li
- Department of Urology, Shenzhen Longhua District Central Hospital, Shenzhen, 518110, PR China.
| |
Collapse
|
9
|
Mansouri M, Berger P. Baculovirus for gene delivery to mammalian cells: Past, present and future. Plasmid 2018; 98:1-7. [PMID: 29842913 DOI: 10.1016/j.plasmid.2018.05.002] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2018] [Revised: 05/25/2018] [Accepted: 05/25/2018] [Indexed: 12/11/2022]
Abstract
Baculovirus is an insect virus which has been used for more than thirty years for production of recombinant proteins in insect cells. However, baculovirus can also be harnessed for efficient gene delivery to mammalian cells if it is equipped with mammalian promoters. This technology is known as BacMam and has been used for gene delivery to immortalized cell lines, stem cells, and primary cells, as well as for gene delivery in animals. Baculovirus has unique features when compared to mammalian viruses. Besides the fact that it is replication-incompetent and does not integrate into the host genome, it has large capacity for foreign DNA. This capacity can for example be used to deliver multiple genes for reprogramming of stem cells, or for delivery of large homology constructs for genome editing. In this review, we provide a brief overview of baculovirus-based gene delivery and its recent applications in therapy and basic research. We also describe how baculovirus is manipulated for efficient transduction in mammalian cells and we highlight possible future improvements.
Collapse
Affiliation(s)
- Maysam Mansouri
- Paul Scherrer Institute, Biomolecular Research, Applied Molecular Biology, CH-5232 Villigen, Switzerland; ETH Zürich, Department of Biosystems Science and Engineering, Mattenstrasse 26, CH-4058 Basel, Switzerland
| | - Philipp Berger
- Paul Scherrer Institute, Biomolecular Research, Applied Molecular Biology, CH-5232 Villigen, Switzerland.
| |
Collapse
|
10
|
Liu X, Li Y, Hu X, Yi Y, Zhang Z. Gene delivery and gene expression in vertebrate using baculovirus Bombyx mori nucleopolyhedrovirus vector. Oncotarget 2017; 8:106017-106025. [PMID: 29285311 PMCID: PMC5739698 DOI: 10.18632/oncotarget.22522] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2017] [Accepted: 10/28/2017] [Indexed: 01/03/2023] Open
Abstract
The baculovirus Autographa californica multicapsid nucleopolyhedrovirus (AcMNPV) has been investigated as a possible tool for gene therapy, but its inhibition by complement proteins in human serum limits its applicability. Here, we used the baculovirus Bombyx mori nucleopolyhedrovirus (BmNPV) to construct a gene delivery vector in which a reporter gene is driven by a cytomegalovirus IE promoter. Enhanced green fluorescent protein (EGFP) and luciferase reporter genes were used to test the efficiency of gene delivery. In vitro complement inactivation data showed that the recombinant BmNPV vector was more stable in human serum than the recombinant AcMNPV vector. The recombinant BmNPV vector successfully delivered the reporter genes into different tissues and organs in mice and chicks. These results demonstrate that the BmNPV vector is more stability against complement inactivation in human serum than the AcMNPV vector, and indicate that it may be useful as an effective gene delivery vector for gene therapy in vertebrates.
Collapse
Affiliation(s)
- Xingjian Liu
- Biotechnology Research Institute, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Yinü Li
- Biotechnology Research Institute, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Xiaoyuan Hu
- Biotechnology Research Institute, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Yongzhu Yi
- The Sericultural Research Institute, Chinese Academy of Agricultural Sciences, Zhenjiang, China
| | - Zhifang Zhang
- Biotechnology Research Institute, Chinese Academy of Agricultural Sciences, Beijing, China
| |
Collapse
|
11
|
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
|
12
|
Levin E, Diekmann H, Fischer D. Highly efficient transduction of primary adult CNS and PNS neurons. Sci Rep 2016; 6:38928. [PMID: 27958330 PMCID: PMC5153636 DOI: 10.1038/srep38928] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2016] [Accepted: 11/15/2016] [Indexed: 12/19/2022] Open
Abstract
Delivery and expression of recombinant genes, a key methodology for many applications in biological research, remains a challenge especially for mature neurons. Here, we report easy, highly efficient and well tolerated transduction of adult peripheral and central neuronal populations of diverse species in culture using VSV-G pseudo-typed, recombinant baculovirus (BacMam). Transduction rates of up to 80% were reliably achieved at high multiplicity of infection without apparent neuro-cytopathic effects. Neurons could be transduced either shortly after plating or after several days in culture. Co-incubation with two different baculoviruses attained near complete co-localization of fluorescent protein expression, indicating multigene delivery. Finally, evidence for functional protein expression is provided by means of cre-mediated genetic recombination and neurite outgrowth assays. Recombinant protein was already detected within hours after transduction, thereby enabling functional readouts even in relatively short-lived neuronal cultures. Altogether, these results substantiate the usefulness of baculovirus-mediated transduction of mature neurons for future research in neuroscience.
Collapse
Affiliation(s)
- Evgeny Levin
- Division of Experimental Neurology, Medical Faculty, Heinrich-Heine-University, Merowingerplatz 1a, 40225 Düsseldorf, Germany
| | - Heike Diekmann
- Division of Experimental Neurology, Medical Faculty, Heinrich-Heine-University, Merowingerplatz 1a, 40225 Düsseldorf, Germany
| | - Dietmar Fischer
- Division of Experimental Neurology, Medical Faculty, Heinrich-Heine-University, Merowingerplatz 1a, 40225 Düsseldorf, Germany
| |
Collapse
|
13
|
Puthumana J, Philip R, Bright Singh IS. Transgene expression in Penaeus monodon cells: evaluation of recombinant baculoviral vectors with shrimp specific hybrid promoters. Cytotechnology 2016; 68:1147-59. [PMID: 25982944 PMCID: PMC4960163 DOI: 10.1007/s10616-015-9872-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2014] [Accepted: 03/27/2015] [Indexed: 12/30/2022] Open
Abstract
It has been realized that shrimp cell immortalization may not be accomplished without in vitro transformation by expressing immortalizing gene in cells. In this process, efficiency of transgene expression is confined to the ability of vectors to transmit gene of interests to the genome. Over the years, unavailability of such vectors has been hampering application of such a strategy in shrimp cells. We report the use of recombinant baculovirus mediated transduction using hybrid promoter system for transgene expression in lymphoid cells of Penaeus monodon. Two recombinant baculovirus vectors with shrimp viral promoters (WSSV-Ie1 and IHHNV-P2) were constructed (BacIe1-GFP and BacP2-GFP) and green fluorescent protein (GFP) used as the transgene. The GFP expression in cells under the control of hybrid promoters, PH-Ie1 or PH-P2, were analyzed and confirmed in shrimp cells. The results indicate that the recombinant baculovirus with shrimp specific viral promoters (hybrid) can be employed for delivery of foreign genes to shrimp cells for in vitro transformation.
Collapse
Affiliation(s)
- Jayesh Puthumana
- National Centre for Aquatic Animal Health, Cochin University of Science and Technology, Fine Arts Avenue, Kochi, 682016, Kerala, India
| | - Rosamma Philip
- Department of Marine Biology, Microbiology and Biochemistry, Cochin University of Science and Technology, Fine Arts Avenue, Kochi, 682016, Kerala, India
| | - I S Bright Singh
- National Centre for Aquatic Animal Health, Cochin University of Science and Technology, Fine Arts Avenue, Kochi, 682016, Kerala, India.
| |
Collapse
|
14
|
Liao JC. Bone Marrow Mesenchymal Stem Cells Expressing Baculovirus-Engineered Bone Morphogenetic Protein-7 Enhance Rabbit Posterolateral Fusion. Int J Mol Sci 2016; 17:ijms17071073. [PMID: 27399674 PMCID: PMC4964449 DOI: 10.3390/ijms17071073] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2016] [Revised: 06/20/2016] [Accepted: 06/27/2016] [Indexed: 02/06/2023] Open
Abstract
Previous studies have suggested that bone marrow-derived mesenchymal stem cells (BMDMSCs) genetically modified with baculoviral bone morphogenetic protein-2 (Bac-BMP-2) vectors could achieve successful fusion in a femur defect model or in a spinal fusion model. In this study, BMDMSCs expressing BMP-7 (Bac-BMP-7-BMDMSCs) were generated. We hypothesized that Bac-BMP-7-BMDMSCs could secrete more BMP-7 than untransduced BMDMSCs in vitro and achieve spinal posterolateral fusion in a rabbit model. Eighteen rabbits underwent posterolateral fusion at L4-5. Group I (n = 6) was implanted with collagen-β-tricalcium phosphate (TCP)-hydroxyapatite (HA), Group II (n = 6) was implanted with collagen-β-TCP-HA plus BMDMSCs, and Group III (n = 6) was implanted with collagen-β-TCP-HA plus Bac-BMP-7-BMDMSCs. In vitro production of BMP-7 was quantified with an enzyme-linked immunosorbent assay (ELISA). Spinal fusion was examined using computed tomography (CT), manual palpation, and histological analysis. ELISA demonstrated that Bac-BMP-7-BMDMSCs produced four-fold to five-fold more BMP-7 than did BMDMSCs. In the CT results, 6 fused segments were observed in Group I (50%, 6/12), 8 in Group II (67%, 8/12), and 12 in Group III (100%, 12/12). The fusion rate, determined by manual palpation, was 0% (0/6) in Group I, 0% (0/6) in Group II, and 83% (5/6) in Group III. Histology showed that Group III had more new bone and matured marrow formation. In conclusion, BMDMSCs genetically transduced with the Bac-BMP-7 vector could express more BMP-7 than untransduced BMDMSCs. These Bac-BMP-7-BMDMSCs on collagen-β-TCP-HA scaffolds were able to induce successful spinal fusion in rabbits.
Collapse
Affiliation(s)
- Jen-Chung Liao
- Department of Orthopedic Surgery, Bone and Joint Research Center, Chang Gung Memorial Hospital, Chang Gung University, Taoyuan 333, Taiwan.
| |
Collapse
|
15
|
Riikonen R, Matilainen H, Rajala N, Pentikainen O, Johnson M, Heino J, Oker-Blom C. Functional Display of an α2 Integrin-Specific Motif (RKK) on the Surface of Baculovirus Particles. Technol Cancer Res Treat 2016; 4:437-45. [PMID: 16029062 DOI: 10.1177/153303460500400411] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
The use of baculovirus vectors shows promise as a tool for gene delivery into mammalian cells. These insect viruses have been shown to transduce a variety of mammalian cell lines, and gene transfer has also been demonstrated in vivo. In this study, we generated two recombinant baculovirus vectors displaying an integrin-specific motif, RKK, as a part of two different loops of the green fluorescent protein (GFP) fused with the major envelope protein gp64 of Autographa californica M nucleopolyhedrovirus. By enzyme linked immunosorbent assays, these viruses were shown to bind a peptide representing the receptor binding site of an α2 integrin, the α2I-domain. However, the interaction was not strong enough to overcome binding of wild type gp64 to the unknown cellular receptor(s) on the surface of α2 integrin-expressing cells (CHO-α2β1) or enhance the viral uptake. After treatment of these cells with phospholipase C, internalization of all viruses was blocked or decreased significantly. However, one of the RKK displaying viruses, AcGFP(K)gp64, was still able to internalize into CHO-α2β1 cells, although at a lower level as compared to non-treated cells. This may indicate the possible utilization of a PLC independent alternative route via, in this case, the α2β1 integrin.
Collapse
Affiliation(s)
- Reetta Riikonen
- University of Jyvaskyla, Dept. of Biological and Environmental Science, PO Box 35, FIN-40351 Jyvaskyla, Finland
| | | | | | | | | | | | | |
Collapse
|
16
|
Liao JC. Cell Therapy Using Bone Marrow-Derived Stem Cell Overexpressing BMP-7 for Degenerative Discs in a Rat Tail Disc Model. Int J Mol Sci 2016; 17:ijms17020147. [PMID: 26805824 PMCID: PMC4783881 DOI: 10.3390/ijms17020147] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2015] [Revised: 01/13/2016] [Accepted: 01/14/2016] [Indexed: 01/08/2023] Open
Abstract
Degenerative discs can cause low back pain. Cell-based transplantation or growth factors therapy have been suggested as a strategy to stimulate disc regeneration. Bone marrow-derived mesenchymal stem cells (BMDMSC) containing bone morphogenetic protein-7 (BMP-7) gene were constructed. We evaluated the effectiveness of these BMP-7 overexpressing cells on degenerative discs in rat tails. In vitro and in vivo studies were designed. In the first stage, the rats were divided into two group according to discs punctured by different needle gauges (18 gauge and 22 gauge). In the second stage, the ideal size of needle was used to induce rat tail disc degeneration. These animals are divided into three groups according to timing of treatment (zero-week, two-week, four-week). Each group was divided into three treating subgroups: control group, BMDMSC group, and Baculo-BMP-7-BMDMSC group. Each rat undergoes radiography examination every two weeks. After eight weeks, the discs were histologically examined with hematoxylin and eosin stain and Alcian blue stain. The 18-gauge group exhibited significant decrease in disc height index (%) than 22-gauge group at eight weeks at both Co6-7 (58.1% ± 2.8% vs. 63.7% ± 1.0%, p = 0.020) and Co8-9 discs (62.7% ± 2.8% vs. 62.8% ± 1.5%, p = 0.010). Baculo-BMP-7-BMDMSCs group showed significant difference in disc height index compared to the BMDMSCs group at both Co6-7 (93.7% ± 1.5% vs. 84.8% ± 1.0%, p = 0.011) and Co8-9 (86.0% ± 2.1% vs. 81.8% ± 1.7%, p = 0.012). In Baculo-BMP-7-BMDMSCs group, the zero-week treatment subgroup showed significant better in disc height index compared to two-week treatment group (p = 0.044), and four-week treatment group (p = 0.011). The zero-week treatment subgroup in Baculo-BMP-7-BMDMSCs group also had significant lower histology score than two-week treatment (4.3 vs. 5.7, p = 0.045) and four-week treatment (4.3 vs. 6.0, p = 0.031). In conclusion, Baculo-BMP-7-BMDMSC can slow down the progression of disc degeneration, but could not provide evidence of regeneration. Early treatment might obtain more distinct results.
Collapse
Affiliation(s)
- Jen-Chung Liao
- Department of Orthopedics Surgery, Bone and Joint Research Center, Chang Gung Memorial Hospital, Chang Gung University, No._5, Fu-Shin Street; Kweishian, Taoyuan 333, Taiwan.
| |
Collapse
|
17
|
Chang MO, Suzuki T, Kitajima M, Takaku H. Baculovirus Infection of Human Monocyte-Derived Dendritic Cells Restricts HIV-1 Replication. AIDS Res Hum Retroviruses 2015; 31:1023-31. [PMID: 26178669 DOI: 10.1089/aid.2015.0060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Acquired immune deficiency syndrome (AIDS) is mainly caused by infection with human immunodeficiency virus-1 (HIV-1) and still poses a global threat for which we lack a protective or therapeutic vaccine. Dendritic cells (DCs) play a major role in the onset of HIV infection, providing one of the primary sites of HIV replication, and also act as viral reservoirs in vivo. Previous studies have shown that baculovirus (BV) induces strong host immune responses against infections and malignancies. In this study, we infected human monocyte-derived DCs with recombinant BV (AcCAG-gag) and showed that AcCAG-gag-infected human DCs underwent maturation and produced interferon alpha and other proinflammatory cytokines accompanied by increases in the mRNA and protein expression levels of APOBEC3 (A3A, A3F, and A3G), proteins associated with the inhibition of HIV-1 replication. Surprisingly, HIV-1 inhibition is also observed in human DCs infected with a wild-type BV, as determined by the production of inflammatory cytokines, the expression of A3, and a reduction in the p24 level. Our findings outline the mechanism underlying the inhibition of HIV-1 in BV-infected human DCs and pave the way for the use of BV as an effective tool for immunotherapy against HIV-1.
Collapse
Affiliation(s)
- Myint Oo Chang
- 1 High Technology Research Centre, Chiba Institute of Technology , Chiba, Japan
| | - Tomoyuki Suzuki
- 2 Department of Life and Environmental Sciences, Chiba Institute of Technology , Chiba, Japan
| | - Masayuki Kitajima
- 2 Department of Life and Environmental Sciences, Chiba Institute of Technology , Chiba, Japan
- 3 Department of Immunology and Pathology, Research Institute National Center for Global Health and Medicine , Chiba, Japan
| | - Hiroshi Takaku
- 1 High Technology Research Centre, Chiba Institute of Technology , Chiba, Japan
- 2 Department of Life and Environmental Sciences, Chiba Institute of Technology , Chiba, Japan
- 4 Research Institute, Chiba Institute of Technology , Chiba, Japan
| |
Collapse
|
18
|
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
|
19
|
Kolangath SM, Basagoudanavar SH, Hosamani M, Saravanan P, Tamil Selvan RP. Baculovirus mediated transduction: analysis of vesicular stomatitis virus glycoprotein pseudotyping. Virusdisease 2014; 25:441-6. [PMID: 25674620 DOI: 10.1007/s13337-014-0229-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2014] [Accepted: 09/17/2014] [Indexed: 11/29/2022] Open
Abstract
The recombinant baculoviruses were constructed to investigate the necessity of VSV-G pseudotyping for mammalian cell transduction. The viruses were designed to express green fluorescent protein (GFP) gene under the control of cytomegalovirus promoter, with or without pseudotyping with VSV-G. VSV-G was placed under the control of polyhedrin promoter that is recognized by insect cells, allowing the formation of pseudotyped baculovirus. The study findings demonstrate that the pseudotyping of baculovirus significantly enhanced transduction efficiency compared to non-pseudotyped baculovirus, resulting in consequent distinction in the expression of GFP in mammalian cells. The results confirmed that pseudotyping is important for baculovirus mediated gene delivery. Further, when full-length VSV-G pseudotyping was compared with truncated VSV-G containing GED domain (G-stem of ectodomain in conjunction with the TM and CT domains of the glycoprotein), latter was relatively less efficient in transducing mammalian cells. This study demonstrated that pseudotyping with full-length VSV-G had better transduction efficiency in mammalian cells. However, at higher multiplicity of infection, both full-length and truncated VSV-G showed equivalent transduction. This study established the significance of pseudotyping of baculovirus with full-length VSV-G for efficient transduction of mammalian cells, utilizing the highly sensitive GFP marker system. These findings have significant implications in designing of baculovirus vector based antigen delivery for developing new generation vaccines.
Collapse
Affiliation(s)
- Sujit M Kolangath
- ICAR-Indian Veterinary Research Institute, Hebbal, Bangalore, 560 024 India
| | - S H Basagoudanavar
- ICAR-Indian Veterinary Research Institute, Hebbal, Bangalore, 560 024 India
| | - M Hosamani
- ICAR-Indian Veterinary Research Institute, Hebbal, Bangalore, 560 024 India
| | - P Saravanan
- ICAR-Indian Veterinary Research Institute, Hebbal, Bangalore, 560 024 India
| | - R P Tamil Selvan
- ICAR-Indian Veterinary Research Institute, Hebbal, Bangalore, 560 024 India
| |
Collapse
|
20
|
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
|
21
|
Ye Y, Cheng X, Zhang J, Tong T, Lin W, Liao M, Fan H. Induction of robust immunity response in mice by dual-expression-system-based recombinant baculovirus expressing the capsid protein of porcine circovirus type 2. Virol J 2013; 10:316. [PMID: 24161107 PMCID: PMC4231451 DOI: 10.1186/1743-422x-10-316] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2013] [Accepted: 10/22/2013] [Indexed: 01/14/2023] Open
Abstract
Background Porcine circovirus type 2 (PCV2) is associated with post-weaning multisystemic wasting syndrome (PMWS), an emerging swine disease that causes progressive weight loss, dyspnea, tachypnea, anemia, jaundice, and diarrhea in piglets. Although baculovirus is an enveloped virus that infects insects in nature, it has emerged as a vaccine vector, and we used it to develop a novel candidate vaccine for a preventive or therapeutic strategy to control PCV2 infections. Methods Immunoblotting analysis of recombinant baculovirus and immunofluorescent staining of baculovirus-infected cells were followed using anti-ORF2 monoclonal antibodies. The BALB/c mice were immunized intramuscularly with this baculovirus. The titers of antibodies were mensurated with a Cap-protein-specific enzyme-linked immunosorbent assay (ELISA) and a serum neutralization assay. The IFN-γ response in splenocytes harvested from immunized mice was measured by ELISA. Student's t-test was used to compare immune responses of different groups. Results In this study, we successfully constructed a dual-expression-system-based recombinant baculovirus BV-GD-ORF2, which can display the PCV2 capsid (Cap) protein and VSV-G protein on the viral envelope and also expressing Cap protein on transduced mammalian cells, thereby functioning as both a subunit and a DNA vaccine. After infection, the Cap protein was expressed and displayed on the viral surface, as demonstrated with an indirect fluorescence assay and immunoblotting. The vaccination of mice with recombinant baculovirus BV-GD-ORF2 successfully induced robust Cap-protein-specific humoral and cellular immune responses. Conclusions Our findings collectively demonstrate that the recombinant baculovirus BV-GD-ORF2 is a potential vaccine against PCV2 infections.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Huiying Fan
- Key Laboratory of Animal Vaccine Development, Ministry of Agriculture, Guangzhou 510642, China.
| |
Collapse
|
22
|
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
|
23
|
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
|
24
|
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
|
25
|
Drugmand JC, Schneider YJ, Agathos SN. Insect cells as factories for biomanufacturing. Biotechnol Adv 2012; 30:1140-57. [DOI: 10.1016/j.biotechadv.2011.09.014] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2011] [Revised: 09/13/2011] [Accepted: 09/16/2011] [Indexed: 10/17/2022]
|
26
|
Zhu B, Ye J, Lu P, Jiang R, Yang X, Fu ZF, Chen H, Cao S. Induction of antigen-specific immune responses in mice by recombinant baculovirus expressing premembrane and envelope proteins of West Nile virus. Virol J 2012; 9:132. [PMID: 22799608 PMCID: PMC3424137 DOI: 10.1186/1743-422x-9-132] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2011] [Accepted: 07/16/2012] [Indexed: 02/10/2023] Open
Abstract
Background West Nile Virus (WNV) is an emerging arthropod-born flavivirus with increasing distribution worldwide that is responsible for a large proportion of viral encephalitis in humans and horses. Given that there are no effective antiviral drugs available for treatment of the disease, efforts have been directed to develop vaccines to prevent WNV infection. Recently baculovirus has emerged as a novel and attractive gene delivery vehicle for mammalian cells. Results In the present study, recombinant baculoviruses expressing WNV premembrane (prM) and envelope (E) proteins under the cytomegalovirus (CMV) promoter with or without vesicular stomatitis virus glycoprotein (VSV/G) were constructed. The recombinant baculoviruses designated Bac-G-prM/E and Bac-prM/E, efficiently express E protein in mammalian cells. Intramuscular injection of the two recombinant baculoviruses (at doses of 108 or 109 PFU/mouse) induced the production of WNV-specific antibodies, neutralizing antibodies as well as gamma interferon (IFN-γ) in a dose-dependent pattern. Interestingly, the recombinant baculovirus Bac-G-prM/E was found to be a more efficient immunogen than Bac-prM/E to elicit a robust immune response upon intramuscular injection. In addition, inoculation of baculovirus resulted in the secretion of inflammatory cytokines, such as TNF-α, IL-2 and IL-6. Conclusions These recombinant baculoviruses are capable of eliciting robust humoral and cellular immune responses in mice, and may be considered as novel vaccine candidates for West Nile Virus.
Collapse
Affiliation(s)
- Bibo Zhu
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, Hubei, People's Republic of China
| | | | | | | | | | | | | | | |
Collapse
|
27
|
Application of baculovirus as a delivery vehicle for study of transcription and translation mechanism of parvovirus in non-permissive mammalian cells. J Virol Methods 2012; 183:1-7. [DOI: 10.1016/j.jviromet.2012.02.009] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2011] [Revised: 02/19/2012] [Accepted: 02/28/2012] [Indexed: 01/14/2023]
|
28
|
Westenberg M, Soedling HM, Hirani N, Nicholson LJ, Mann DA, Dolphin CT. Seamless replacement of Autographa californica multiple nucleopolyhedrovirus gp64 with each of five novel type II alphabaculovirus fusion sequences generates pseudotyped virus that fails to transduce mammalian cells. J Gen Virol 2012; 93:1583-1590. [PMID: 22492915 PMCID: PMC3542734 DOI: 10.1099/vir.0.041921-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
Autographa californica multiple nucleopolyhedrovirus (AcMNPV), a member of the type I alphabaculoviruses, is able to transduce and deliver a functional gene to a range of non-host cells, including many mammalian lines and primary cells, a property mediated by the envelope fusion protein GP64. AcMNPV is non-cytopathic and inherently replication deficient in non-host cells. As such, AcMNPV represents a possible new class of gene therapy vector with potential future clinical utility. Whilst not a problem for in vitro gene delivery, the broad tropism displayed for non-host cells is less desirable in a gene therapy vector. The fusion protein F of type II alphabaculoviruses can substitute functionally for GP64, and such pseudotyped viruses display a severely impaired capacity for non-host-cell transduction. Thus, surface decoration of such an F-pseudotyped AcMNPV with cell-binding ligands may restore transduction competence and generate vectors with desirable cell-targeting characteristics. By seamlessly swapping the native gp64 coding sequence with each of five sequences encoding different F proteins, a set of F-pseudotyped AcMNPV was generated. This report details their relative abilities both to functionally replace GP64 in viral growth and to transduce human Saos-2 and HeLa cells. All five supported viable infections in insect cell cultures and one, the Mamestra configurata NPV (MacoNPV) F pseudotype, could be amplified to titres close to those of native AcMNPV. In contrast, none was able to transduce the Saos-2 and HeLa cell lines. The robust support provided by MacoNPV F in virus production makes the corresponding pseudotype a viable scaffold to display surface ligands to direct selective mammalian cell targeting.
Collapse
Affiliation(s)
- Marcel Westenberg
- Pharmaceutical Science Research Division, King's College London, Franklin-Wilkins Building, 150 Stamford Street, London SE1 9NH, UK
| | - Helen M Soedling
- Pharmaceutical Science Research Division, King's College London, Franklin-Wilkins Building, 150 Stamford Street, London SE1 9NH, UK
| | - Nisha Hirani
- Pharmaceutical Science Research Division, King's College London, Franklin-Wilkins Building, 150 Stamford Street, London SE1 9NH, UK
| | - Linda J Nicholson
- Division of Cancer Studies, King's College London, Guy's Campus, London SE1 1UL, UK
| | - Derek A Mann
- Liver Research Group, Institute of Cellular Medicine, 4th Floor, Catherine Cookson Building Medical School, Newcastle University, Newcastle NE2 4HH, UK
| | - Colin T Dolphin
- Pharmaceutical Science Research Division, King's College London, Franklin-Wilkins Building, 150 Stamford Street, London SE1 9NH, UK
| |
Collapse
|
29
|
Li SF, Wang HL, Hu ZH, Deng F. Genetic modification of baculovirus expression vectors. Virol Sin 2012; 27:71-82. [PMID: 22491998 DOI: 10.1007/s12250-012-3236-y] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2011] [Accepted: 02/27/2012] [Indexed: 10/28/2022] Open
Abstract
As a protein expression vector, the baculovirus demonstrates many advantages over other vectors. With the development of biotechnology, baculoviral vectors have been genetically modified to facilitate high level expression of heterologous proteins in both insect and mammalian cells. These modifications include utilization of different promoters and signal peptides, deletion or replacement of viral genes for increasing protein secretion, integration of polycistronic expression cassette for producing protein complexes, and baculovirus pseudotyping, promoter accommodation or surface display for enhancing mammalian cell targeting gene delivery. This review summarizes the development and the current state of art of the baculovirus expression system. Further development of baculovirus expression systems will make them even more feasible and accessible for advanced applications.
Collapse
Affiliation(s)
- Shu-fen Li
- State Key Laboratory of Virology and Joint-Lab of Invertebrate Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan 430071, China
| | | | | | | |
Collapse
|
30
|
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
|
31
|
Ames RS, Fornwald JA, Nuthulaganti P, Trill JJ, Foley JJ, Buckley PT, Kost TA, Wu Z, Romanos MA. BacMam Recombinant Baculoviruses in G Protein–Coupled Receptor Drug Discovery. ACTA ACUST UNITED AC 2011. [DOI: 10.3109/10606820490514969] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
32
|
Tang XC, Lu HR, Ross TM. Baculovirus-produced influenza virus-like particles in mammalian cells protect mice from lethal influenza challenge. Viral Immunol 2011; 24:311-9. [PMID: 21830902 DOI: 10.1089/vim.2011.0016] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Influenza virus-like particles (VLPs) are effective vaccines against influenza infection, which can be produced either in insect cells by recombinant baculovirus (BV) infection or in mammalian cells by DNA plasmid transfection. However, VLPs produced from baculovirus/insect cells are difficult to purify due to baculovirus contamination; VLPs produced by plasmid transfection are limited by scale-up capability. In this study, a BacMam BV, in which three CMV-promoters drive the hemagglutinin, neuraminidase, and matrix of influenza virus was constructed. This baculovirus can deliver these genes into mammalian cells/hosts and subsequently influenza VLPs can be produced and secreted from transduced cells. Transduction conditions were optimized and influenza VLPs were purified from transduced 293T cells. Mice were vaccinated with BV transduction-produced VLPs, plasmid transfection-produced VLPs, and BacMam BV. Two vaccinations of each vaccine induced high hemagglutination-inhibition (HAI) titers and prevented influenza virus infection. In contrast, following a single vaccination, all mice vaccinated with each vaccine had significantly lower lung viral titers compared to unvaccinated mice. Remarkably, mice vaccinated with a single dose of BV transduction-produced VLPs survived challenge, whereas mice vaccinated with one dose of BacMam BV- or plasmid transfection-produced VLPs had 60-80% survival. This finding is particularly significant for producing easily purified VLPs. The BacMam system is an alternative strategy for VLP production, which is easy to scale up and purify. Besides, BacMam BV can be used as a gene delivery vector to produce VLPs in vivo, to stimulate immune responses.
Collapse
Affiliation(s)
- Xian-Chun Tang
- Center for Vaccine Research, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | | | | |
Collapse
|
33
|
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
|
34
|
Functional integration of grafted neural stem cell-derived dopaminergic neurons monitored by optogenetics in an in vitro Parkinson model. PLoS One 2011; 6:e17560. [PMID: 21394212 PMCID: PMC3048875 DOI: 10.1371/journal.pone.0017560] [Citation(s) in RCA: 82] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2010] [Accepted: 01/19/2011] [Indexed: 12/11/2022] Open
Abstract
Intrastriatal grafts of stem cell-derived dopamine (DA) neurons induce behavioral recovery in animal models of Parkinson's disease (PD), but how they functionally integrate in host neural circuitries is poorly understood. Here, Wnt5a-overexpressing neural stem cells derived from embryonic ventral mesencephalon of tyrosine hydroxylase-GFP transgenic mice were expanded as neurospheres and transplanted into organotypic cultures of wild type mouse striatum. Differentiated GFP-labeled DA neurons in the grafts exhibited mature neuronal properties, including spontaneous firing of action potentials, presence of post-synaptic currents, and functional expression of DA D2 autoreceptors. These properties resembled those recorded from identical cells in acute slices of intrastriatal grafts in the 6-hydroxy-DA-induced mouse PD model and from DA neurons in intact substantia nigra. Optogenetic activation or inhibition of grafted cells and host neurons using channelrhodopsin-2 (ChR2) and halorhodopsin (NpHR), respectively, revealed complex, bi-directional synaptic interactions between grafted cells and host neurons and extensive synaptic connectivity within the graft. Our data demonstrate for the first time using optogenetics that ectopically grafted stem cell-derived DA neurons become functionally integrated in the DA-denervated striatum. Further optogenetic dissection of the synaptic wiring between grafted and host neurons will be crucial to clarify the cellular and synaptic mechanisms underlying behavioral recovery as well as adverse effects following stem cell-based DA cell replacement strategies in PD.
Collapse
|
35
|
Nizamani ZA, Keil GM, Albina E, Holz C, Minet C, Kwiatek O, Libeau G, Servan de Almeida R. Potential of adenovirus and baculovirus vectors for the delivery of shRNA against morbilliviruses. Antiviral Res 2011; 90:98-101. [PMID: 21356246 DOI: 10.1016/j.antiviral.2011.02.007] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2010] [Revised: 01/17/2011] [Accepted: 02/18/2011] [Indexed: 11/30/2022]
Abstract
Morbilliviruses are important pathogens of humans, ruminants, carnivores and marine mammals. Although good vaccines inducing long-term immunity are available, recurrent outbreaks of measles, canine distemper and peste des petits ruminants (PPR) are observed. In control strategies, antivirals thus could be useful to confine virus spread and application of interfering RNAs is a promising approach, provided they can be delivered efficiently into the host cells. We have constructed recombinant adenovirus and baculovirus vectors expressing short hairpin RNAs (shRNAs) against the PPR virus (PPRV) and compared them in vitro. It was found that both recombinant viruses inhibited PPRV replication with the baculovirus vector, which inhibited generation of infectious progeny by more than 2 log10 and the nucleoprotein expression of PPRV by 73%, being the more efficient. The results show that baculoviral shRNA-expressing vectors have the potential for therapeutic use against morbillivirus infections.
Collapse
|
36
|
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
|
37
|
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
|
38
|
Suzuki H, Saitoh H, Suzuki T, Takaku H. Baculovirus-mediated bispecific short-hairpin small-interfering RNAs have remarkable ability to cope with both influenza viruses A and B. Oligonucleotides 2010; 19:307-16. [PMID: 19899951 DOI: 10.1089/oli.2009.0189] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Influenza viruses A and B cause widespread infections of the human respiratory tract; however, existing vaccines and drug therapy are of limited value for their treatment. Here, we show that bispecific short-hairpin small-interfering RNA constructs containing an 8-nucleotide intervening spacer, targeted against influenza virus A or influenza virus B, can inhibit the production of both types of virus in infected cell lines. This multiple vector showed remarkable ability to cope with both influenza viruses A and B. Furthermore, the Autographa californica multiple nuclear polyhedrosis virus can infect a range of mammalian cells, facilitating its use as a baculovirus vector for gene delivery into cells. In this study, baculovirus-mediated bispecific short-hairpin RNA expression markedly inhibited both influenza viruses A and B production.
Collapse
Affiliation(s)
- Hitoshi Suzuki
- Department of Life and Environmental Sciences, Faculty of Engineering, Chiba Institute of Technology, Chiba, Japan.
| | | | | | | |
Collapse
|
39
|
Fujita R, Ohtsuka D, Sahara K, Asano S, Bando H. An HDAC inhibitor increases AcMNPV gene expression in mammalian cells. Arch Virol 2010; 155:577-81. [PMID: 20186445 DOI: 10.1007/s00705-010-0614-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2009] [Accepted: 01/08/2010] [Indexed: 11/28/2022]
Abstract
The baculovirus Autographa californica multiple nucleopolyhedrovirus (AcMNPV) is used as a safer viral vector in mammalian cells with potential applications in gene therapy. However, the mechanism for the insusceptibility of mammalian cells to proliferative infection by entomopathogenic viruses is not well understood. Here, we studied the significance of epigenetic modifications such as histone acetylation, histone methylation and HP1 accumulation for AcMNPV gene expression in mammalian BHK cells. Real-time PCR and chromatin immunoprecipitation with sodium butyrate revealed an important relationship between viral gene expression and histone acetylation, with implications for a mechanism of suppression of AcMNPV gene expression in BHK cells.
Collapse
Affiliation(s)
- Ryosuke Fujita
- Laboratory of Applied Molecular Entomology, Division of Applied Bioscience, Graduate School of Agriculture, Hokkaido University, Sapporo 001-8589, Japan
| | | | | | | | | |
Collapse
|
40
|
Zhou X, Li B, Wang J, Yin H, Zhang Y. The feasibility of using a baculovirus vector to deliver the sodium-iodide symporter gene as a reporter. Nucl Med Biol 2010; 37:299-308. [PMID: 20346869 DOI: 10.1016/j.nucmedbio.2009.12.007] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2009] [Revised: 11/17/2009] [Accepted: 12/08/2009] [Indexed: 10/19/2022]
Abstract
PURPOSE To evaluate the efficiency of baculovirus vectors in transducing FTC-133 cells and to examine the feasibility of using baculovirus vectors for the delivery of the sodium-iodide symporter (NIS) gene as a reporter through co-transduction to monitor the expression of the target gene. METHOD Two recombinant baculoviruses were constructed to express NIS and green fluorescent protein (GFP) respectively. FTC-133, 8050C, SW1116, A549 cells, were infected with Bac-GFP. The infection efficiency of Bac-GFP and the intensity of fluorescence, in either the presence or absence of sodium butyrate, were monitored by flow cytometry. The iodine uptake by FTC-133 cells infected with Bac-NIS was measured using a gamma counter. FTC-133 cells were infected with a mixture of equal amounts of Bac-NIS and Bac-GFP at different setting of multiplicity of infection (MOI). The changes of GFP fluorescence intensity and iodine uptake were monitored 24 h after infection in the coinfected cells. RESULTS We have successfully constructed recombinant baculoviruses carrying NIS and GFP under the control of the cytomegalovirus IE-1 promoter. We found that transduced efficiency of baculovirus in 8505C, SW1116, A549 cells are low in absence of sodium butyrate. Yet Bac-GFP infects FTC-133 cells at a high efficiency, 77.67%, 85.57% and 93.23% with MOI of 100, 200 and 400, respectively. The fluorescence intensity of the Bac-GFP infected tumor cells correlated positively with the MOI of the virus. Sodium butyrate induction increased both the infection efficiency and the fluorescence intensity, but increase of infection efficiency was insignificant in FTC-133 cells. Reporter gene (GFP) expression in FTC-133 is stable within 7 days after infection. The radioactivity incorporated by the tumor cells infected with Bac-NIS correlated positively with the MOI of Bac-NIS as well. In tumor cells co-infected with Bac-NIS and Bac-GFP, the amount of radioactivity incorporated significantly correlated with the GFP fluorescence intensity (r=0.922). CONCLUSION Baculovirus vectors are powerful vehicles for studying FTC-133 tumor cells in gene delivery. It is feasible to use a baculovirus vector to deliver NIS as a reporter gene to monitor the expression of target genes. This is therefore an effective approach for the detection of target gene expression in gene therapy.
Collapse
Affiliation(s)
- Xiang Zhou
- Department of Nuclear Medicine, Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | | | | | | | | |
Collapse
|
41
|
Georgopoulos LJ, Elgue G, Sanchez J, Dussupt V, Magotti P, Lambris JD, Tötterman TH, Maitland NJ, Nilsson B. Preclinical evaluation of innate immunity to baculovirus gene therapy vectors in whole human blood. Mol Immunol 2009; 46:2911-7. [PMID: 19665799 DOI: 10.1016/j.molimm.2009.07.008] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2009] [Revised: 06/26/2009] [Accepted: 07/13/2009] [Indexed: 11/18/2022]
Abstract
Interactions of gene therapy vectors with human blood components upon intravenous administration have a significant effect on vector efficacy and patient safety. Here we describe methods to evaluate these interactions and their effects in whole human blood, using baculovirus vectors as a model. Opsonisation of baculovirus particles by binding of IgM and C3b was demonstrated, which is likely to be the cause of the significant blood cell-associated virus that was detected. Preventing formation of the complement C5b-9 (membrane attack) complex maintained infectivity of baculovirus particles as shown by studying the effects of two specific complement inhibitors, Compstatin and a C5a receptor antagonist. Formation of macroscopic blood clots after 4h was prevented by both complement inhibitors. Pro- and anti-inflammatory cytokines Il-1beta, IL-6, IL-8 and TNF-alpha were produced at variable levels between volunteers and complement inhibitors showed patient-specific effects on cytokine levels. Whilst both complement inhibitors could play a role in protecting patients from aggressive inflammatory reactions, only Compstatin maintained virus infectivity. We conclude that this ex vivo model, used here for the first time with infectious agents, is a valuable tool in evaluating human innate immune responses to gene therapy vectors or to predict the response of individual patients as part of a clinical trial or treatment. The use of complement inhibitors for therapeutic viruses should be considered on a patient-specific basis.
Collapse
Affiliation(s)
- Lindsay J Georgopoulos
- YCR Cancer Research Unit, Department of Biology (Area 13), University of York, Heslington, York YO10 5DD, UK.
| | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Baculovirus transduction of mesenchymal stem cells triggers the toll-like receptor 3 pathway. J Virol 2009; 83:10548-56. [PMID: 19656899 DOI: 10.1128/jvi.01250-09] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Human mesenchymal stem cells (hMSCs) can be genetically modified with viral vectors and hold promise as a cell source for regenerative medicine, yet how hMSCs respond to viral vector transduction remains poorly understood, leaving the safety concerns unaddressed. Here, we explored the responses of hMSCs against an emerging DNA viral vector, baculovirus (BV), and discovered that BV transduction perturbed the transcription of 816 genes associated with five signaling pathways. Surprisingly, Toll-like receptor-3 (TLR3), a receptor that generally recognizes double-stranded RNA, was apparently upregulated by BV transduction, as confirmed by microarray, PCR array, flow cytometry, and confocal microscopy. Cytokine array data showed that BV transduction triggered robust secretion of interleukin-6 (IL-6) and IL-8 but not of other inflammatory cytokines and beta interferon (IFN-beta). BV transduction activated the signaling molecules (e.g., Toll/interleukin-1 receptor domain-containing adaptor-inducing IFN-beta, NF-kappaB, and IFN regulatory factor 3) downstream of TLR3, while silencing the TLR3 gene with small interfering RNA considerably abolished cytokine expression and promoted cell migration. These data demonstrate, for the first time, that a DNA viral vector can activate the TLR3 pathway in hMSCs and lead to a cytokine expression profile distinct from that in immune cells. These findings underscore the importance of evaluating whether the TLR3 signaling cascade plays roles in the immune response provoked by other DNA vectors (e.g., adenovirus). Nonetheless, BV transduction barely disturbed surface marker expression and induced only transient and mild cytokine responses, thereby easing the safety concerns of using BV for hMSCs engineering.
Collapse
|
43
|
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
|
44
|
Wang X, Li L, Ding S, Huang X, Zhang J, Yin J, Zhong J. Chicken HS4 insulator significantly improves baculovirus-mediated foreign gene expression in insect cells by modifying the structure of neighbouring chromatin in virus minichromosome. J Biotechnol 2009; 142:193-9. [DOI: 10.1016/j.jbiotec.2009.06.012] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2008] [Revised: 06/07/2009] [Accepted: 06/09/2009] [Indexed: 01/05/2023]
|
45
|
High-efficiency transient transduction of human embryonic stem cell-derived neurons with baculoviral vectors. Mol Ther 2009; 17:1585-93. [PMID: 19532141 DOI: 10.1038/mt.2009.124] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
Transient genetic manipulation of human neurons without chromosomal integration of the transgene would be valuable but has been challenging due to the quiescent nature of these postmitotic cells. In this study, we developed a set of baculoviral vectors for transient transduction in nondividing neurons derived from human embryonic stem cells (hESCs). Using a baculoviral vector equipped with the woodchuck hepatitis virus posttranscriptional regulatory element (WPRE), we observed a quick onset of transgene expression as early as day 1 after baculoviral transduction and a high efficiency of up to 80%. Strong transgene expression in the cultured human neurons was observed for more than 1 month and the signal was easily detectable even after 3 months. Using two baculoviral vectors carrying different transgenes, we found that co-transduction at a single neuron level was possible. After transplantation into the brain of nude mice, the baculovirus-transduced human neurons were integrated into the mouse brain and maintained transgene expression for at least 4 weeks, portending the usefulness of this technique in assisting neural transplantation. Therefore, by mediating efficient transient gene expression, baculoviral vectors can provide useful tools for both basic gene function studies in human neurons and therapeutic applications of these cells.
Collapse
|
46
|
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]
|
47
|
(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
|
48
|
He F, Madhan S, Kwang J. Baculovirus vector as a delivery vehicle for influenza vaccines. Expert Rev Vaccines 2009; 8:455-67. [PMID: 19348561 DOI: 10.1586/erv.09.2] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
The baculovirus vector has emerged as an efficient delivery vehicle for influenza vaccines. In addition to the ease and safety in expeditious production, recent improvements in baculovirus engineering to display foreign proteins on the surface and to express transgenes with suitable promoters in various cell lines have become milestones in the development of the baculovirus expression system. Surface-displayed and shuttle promoter-mediated baculovirus vaccines for influenza present advantages in immunogenicity and safety, as studied in several animal models. A variety of strategies, including the modification of envelope proteins for surface display, the selection of novel promoters for in vivo transductions and advancements in downstream processing, aid the improvement of baculovirus-based influenza vaccines and represent progress toward next-generation vaccines for influenza.
Collapse
Affiliation(s)
- Fang He
- Animal Health Biotechnology, Temasek Life Sciences Laboratory, National University of Singapore, 117604 Singapore.
| | | | | |
Collapse
|
49
|
Grabowska AK, Lipińska AD, Rohde J, Szewczyk B, Bienkowska-Szewczyk K, Rziha HJ. New baculovirus recombinants expressing Pseudorabies virus (PRV) glycoproteins protect mice against lethal challenge infection. Vaccine 2009; 27:3584-91. [PMID: 19464538 DOI: 10.1016/j.vaccine.2009.03.067] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2008] [Revised: 03/14/2009] [Accepted: 03/18/2009] [Indexed: 11/24/2022]
Abstract
The present study demonstrates the protective potential of novel baculovirus recombinants, which express the glycoproteins gB, gC, or gD of Pseudorabies virus (PRV; Alphaherpesvirus of swine) and additionally contain the glycoprotein G of Vesicular Stomatitis Virus (VSV-G) in the virion (Bac-G-PRV). To evaluate the protective capacity, mixtures of equal amounts of the PRV gB-, gC-, and gD-expressing baculoviruses were used for immunization. Three intramuscular immunizations with that Bac-G-PRV mixture could protect mice against a lethal PRV challenge infection. To achieve complete protection high titers of Bac-G-PRV and three immunizations were necessary. This immunization with Bac-G-PRV resulted in the induction of high titers of PRV-specific serum antibodies of the IgG2a subclass and of interferon (IFN)-gamma, indicating a Th1-type immune response. Moreover, splenocytes of immunized mice exhibited natural killer cell activity accompanied by the production of IFN-alpha and IFN-gamma. Collectively, the presented data demonstrate for the first time that co-expression of VSV-G in baculovirus recombinant vaccines can improve the induction of a protective immune response against foreign antigens.
Collapse
Affiliation(s)
- Agnieszka K Grabowska
- Dept. of Mol. Virology, Faculty of Biotechnology, University of Gdansk, Gdansk, Poland
| | | | | | | | | | | |
Collapse
|
50
|
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
|