1
|
Terasawa K, Seike T, Sakamoto K, Ohtake K, Terada T, Iwata T, Watabe T, Yokoyama S, Hara‐Yokoyama M. Site-specific photo-crosslinking/cleavage for protein-protein interface identification reveals oligomeric assembly of lysosomal-associated membrane protein type 2A in mammalian cells. Protein Sci 2023; 32:e4823. [PMID: 37906694 PMCID: PMC10659947 DOI: 10.1002/pro.4823] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Revised: 10/16/2023] [Accepted: 10/21/2023] [Indexed: 11/02/2023]
Abstract
Genetic code expansion enables site-specific photo-crosslinking by introducing photo-reactive non-canonical amino acids into proteins at defined positions during translation. This technology is widely used for analyzing protein-protein interactions and is applicable in mammalian cells. However, the identification of the crosslinked region still remains challenging. Here, we developed a new method to identify the crosslinked region by pre-installing a site-specific cleavage site, an α-hydroxy acid (Nε -allyloxycarbonyl-α-hydroxyl-l-lysine acid, AllocLys-OH), into the target protein. Alkaline treatment cleaves the crosslinked complex at the position of the α-hydroxy acid residue and thus helps to identify which side of the cleavage site, either closer to the N-terminus or C-terminus, the crosslinked site is located within the target protein. A series of AllocLys-OH introductions narrows down the crosslinked region. By applying this method, we identified the crosslinked regions in lysosomal-associated membrane protein type 2A (LAMP2A), a receptor of chaperone-mediated autophagy, in mammalian cells. The results suggested that at least two interfaces are involved in the homophilic interaction, which requires a trimeric or higher oligomeric assembly of adjacent LAMP2A molecules. Thus, the combination of site-specific crosslinking and site-specific cleavage promises to be useful for revealing binding interfaces and protein complex geometries.
Collapse
Affiliation(s)
- Kazue Terasawa
- Department of Biochemistry, Graduate School of Medical and Dental SciencesTokyo Medical and Dental University (TMDU)TokyoJapan
- LiberoThera Co., Ltd.Chuo‐kuJapan
| | - Tatsuro Seike
- Department of Periodontology, Graduate School of Medical and Dental SciencesTokyo Medical and Dental University (TMDU)TokyoJapan
| | - Kensaku Sakamoto
- Laboratory for Nonnatural Amino Acid TechnologyRIKEN Center for Biosystems Dynamics ResearchYokohamaJapan
- Department of Drug Target Protein ResearchShinshu University School of MedicineNaganoJapan
| | - Kazumasa Ohtake
- Laboratory for Nonnatural Amino Acid TechnologyRIKEN Center for Biosystems Dynamics ResearchYokohamaJapan
- Department of Electrical Engineering and BioscienceWaseda UniversityTokyoJapan
| | - Tohru Terada
- Department of Biotechnology, Graduate School of Agricultural and Life SciencesThe University of TokyoTokyoJapan
| | - Takanori Iwata
- Department of Periodontology, Graduate School of Medical and Dental SciencesTokyo Medical and Dental University (TMDU)TokyoJapan
| | - Tetsuro Watabe
- Department of Biochemistry, Graduate School of Medical and Dental SciencesTokyo Medical and Dental University (TMDU)TokyoJapan
| | - Shigeyuki Yokoyama
- Department of Drug Target Protein ResearchShinshu University School of MedicineNaganoJapan
- Laboratory for Protein Function and Structural BiologyRIKEN Cluster for Science, Technology and Innovation HubYokohamaJapan
- Department of Structural Biology and Biochemistry, Graduate School of Medical and Dental SciencesTokyo Medical and Dental University (TMDU)TokyoJapan
| | - Miki Hara‐Yokoyama
- Department of Biochemistry, Graduate School of Medical and Dental SciencesTokyo Medical and Dental University (TMDU)TokyoJapan
| |
Collapse
|
2
|
Transfected plasmid DNA is incorporated into the nucleus via nuclear envelope reformation at telophase. Commun Biol 2022; 5:78. [PMID: 35058555 PMCID: PMC8776997 DOI: 10.1038/s42003-022-03021-8] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2021] [Accepted: 12/27/2021] [Indexed: 12/15/2022] Open
Abstract
AbstractDNA transfection is an important technology in life sciences, wherein nuclear entry of DNA is necessary to express exogenous DNA. Non-viral vectors and their transfection reagents are useful as safe transfection tools. However, they have no effect on the transfection of non-proliferating cells, the reason for which is not well understood. This study elucidates the mechanism through which transfected DNA enters the nucleus for gene expression. To monitor the behavior of transfected DNA, we introduce plasmid bearing lacO repeats and RFP-coding sequences into cells expressing GFP-LacI and observe plasmid behavior and RFP expression in living cells. RFP expression appears only after mitosis. Electron microscopy reveals that plasmids are wrapped with nuclear envelope (NE)‒like membranes or associated with chromosomes at telophase. The depletion of BAF, which is involved in NE reformation, delays plasmid RFP expression. These results suggest that transfected DNA is incorporated into the nucleus during NE reformation at telophase.
Collapse
|
3
|
Guo C, Chen F, Xiao Q, Catterall HB, Robinson JH, Wang Z, Mock M, Hubert R. Expression liabilities in a four-chain bispecific molecule. Biotechnol Bioeng 2021; 118:3744-3759. [PMID: 34110008 DOI: 10.1002/bit.27850] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Revised: 05/07/2021] [Accepted: 06/01/2021] [Indexed: 12/18/2022]
Abstract
Multispecific antibodies, often composed of three to five polypeptide chains, have become increasingly relevant in the development of biotherapeutics. These molecules have mechanisms of action that include redirecting T cells to tumors and blocking multiple pathogenic mediators simultaneously. One of the major challenges for asymmetric multispecific antibodies is generating a high proportion of the correctly paired antibody during production. To understand the causes and effects of chain mispairing impurities in a difficult to express multispecific hetero-IgG, we investigated consequences of individual and pairwise chain expression in mammalian transient expression hosts. We found that one of the two light chains (LC) was not secretion competent when transfected individually or cotransfected with the noncognate heavy chain (HC). Overexpression of this secretion impaired LC reduced cell growth while inducing endoplasmic reticulum stress and CCAAT/enhancer-binding protein homologous protein (CHOP) expression. The majority of this LC was observed as monomer with incomplete intrachain disulfide bonds when expressed individually. Russell bodies (RB) were induced when this LC was co-expressed with the cognate HC. Moreover, one HC paired promiscuously with noncognate LC. These results identify the causes for the low product quality observed from stable cell lines expressing this heteroIgG and suggest mitigation strategies to improve overall process productivity of the correctly paired multispecific antibody. The approach described here provides a general strategy for identifying the molecular and cellular liabilities associated with difficult to express multispecific antibodies.
Collapse
Affiliation(s)
- Cai Guo
- Department of Therapeutic Discovery, Amgen Research, Amgen Inc., Thousand Oaks, California, USA
| | - Fuyi Chen
- Department of Therapeutic Discovery, Amgen Research, Amgen Inc., Thousand Oaks, California, USA
| | - Qiang Xiao
- Department of Therapeutic Discovery, Amgen Research, Amgen Inc., Thousand Oaks, California, USA
| | - Hannah B Catterall
- Department of Therapeutic Discovery, Amgen Research, Amgen Inc., Thousand Oaks, California, USA
| | - John H Robinson
- Department of Therapeutic Discovery, Amgen Research, Amgen Inc., Thousand Oaks, California, USA
| | - Zhulun Wang
- Department of Therapeutic Discovery, Amgen Research, Amgen Inc., San Francisco, California, USA
| | - Marissa Mock
- Department of Therapeutic Discovery, Amgen Research, Amgen Inc., Thousand Oaks, California, USA
| | - René Hubert
- Department of Therapeutic Discovery, Amgen Research, Amgen Inc., Thousand Oaks, California, USA
| |
Collapse
|
4
|
Budge JD, Young RJ, Smales CM. Engineering of Chinese Hamster Ovary Cells With NDPK-A to Enhance DNA Nuclear Delivery Combined With EBNA1 Plasmid Maintenance Gives Improved Exogenous Transient Reporter, mAb and SARS-CoV-2 Spike Protein Expression. Front Bioeng Biotechnol 2021; 9:679448. [PMID: 34150735 PMCID: PMC8212061 DOI: 10.3389/fbioe.2021.679448] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Accepted: 05/10/2021] [Indexed: 12/15/2022] Open
Abstract
Transient gene expression (TGE) in mammalian cells is a method of rapidly generating recombinant protein material for initial characterisation studies that does not require time-consuming processes associated with stable cell line construction. High TGE yields are heavily dependent on efficient delivery of plasmid DNA across both the plasma and nuclear membranes. Here, we harness the protein nucleoside diphosphate kinase (NDPK-A) that contains a nuclear localisation signal (NLS) to enhance DNA delivery into the nucleus of CHO cells. We show that co-expression of NDPK-A during transient expression results in improved transfection efficiency in CHO cells, presumably due to enhanced transportation of plasmid DNA into the nucleus via the nuclear pore complex. Furthermore, introduction of the Epstein Barr Nuclear Antigen-1 (EBNA-1), a protein that is capable of inducing extrachromosomal maintenance, when coupled with complementary oriP elements on a transient plasmid, was utilised to reduce the effect of plasmid dilution. Whilst there was attenuated growth upon introduction of the EBNA-1 system into CHO cells, when both NDPK-A nuclear import and EBNA-1 mediated technologies were employed together this resulted in enhanced transient recombinant protein yields superior to those generated using either approach independently, including when expressing the complex SARS-CoV-2 spike (S) glycoprotein.
Collapse
Affiliation(s)
- James D Budge
- Industrial Biotechnology Centre, School of Biosciences, University of Kent, Canterbury, United Kingdom
| | - Robert J Young
- R&D Cell Engineering Group, Lonza Biologics, Chesterford Research Park, Saffron Walden, United Kingdom
| | - Christopher Mark Smales
- Industrial Biotechnology Centre, School of Biosciences, University of Kent, Canterbury, United Kingdom
| |
Collapse
|
5
|
Torres-Vanegas JD, Cruz JC, Reyes LH. Delivery Systems for Nucleic Acids and Proteins: Barriers, Cell Capture Pathways and Nanocarriers. Pharmaceutics 2021; 13:428. [PMID: 33809969 PMCID: PMC8004853 DOI: 10.3390/pharmaceutics13030428] [Citation(s) in RCA: 70] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Revised: 03/09/2021] [Accepted: 03/12/2021] [Indexed: 12/27/2022] Open
Abstract
Gene therapy has been used as a potential approach to address the diagnosis and treatment of genetic diseases and inherited disorders. In this line, non-viral systems have been exploited as promising alternatives for delivering therapeutic transgenes and proteins. In this review, we explored how biological barriers are effectively overcome by non-viral systems, usually nanoparticles, to reach an efficient delivery of cargoes. Furthermore, this review contributes to the understanding of several mechanisms of cellular internalization taken by nanoparticles. Because a critical factor for nanoparticles to do this relies on the ability to escape endosomes, researchers have dedicated much effort to address this issue using different nanocarriers. Here, we present an overview of the diversity of nanovehicles explored to reach an efficient and effective delivery of both nucleic acids and proteins. Finally, we introduced recent advances in the development of successful strategies to deliver cargoes.
Collapse
Affiliation(s)
- Julian D. Torres-Vanegas
- Grupo de Diseño de Productos y Procesos (GDPP), Department of Chemical and Food Engineering, Universidad de los Andes, Bogotá 111711, Colombia
| | - Juan C. Cruz
- Department of Biomedical Engineering, Universidad de los Andes, Bogotá 111711, Colombia
| | - Luis H. Reyes
- Grupo de Diseño de Productos y Procesos (GDPP), Department of Chemical and Food Engineering, Universidad de los Andes, Bogotá 111711, Colombia
| |
Collapse
|
6
|
Abstract
The genetic incorporation of noncanonical amino acids (ncAAs) into proteins has been realized in bacteria, yeast, and mammalian cells, and recently, in multicellular organisms including plants and animals. However, the addition of new building blocks to the genetic code of tissues from human origin has not yet been achieved. To this end, we report a self-replicating Epstein-Barr virus-based episomal vector for the long-term encoding of ncAAs in human hematopoietic stem cells and reconstitution of this genetically engineered hematopoietic system in mice.
Collapse
|
7
|
Yu F, Tan WJ, Lu Y, MacAry PA, Loh KS. The other side of the coin: Leveraging Epstein-Barr virus in research and therapy. Oral Oncol 2016; 60:112-7. [PMID: 27531881 PMCID: PMC7108324 DOI: 10.1016/j.oraloncology.2016.07.010] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2016] [Accepted: 07/09/2016] [Indexed: 12/22/2022]
Abstract
Epstein-Barr virus is (EBV) a ubiquitous virus prevalent in 90% of the human population. Transmitted through infected saliva, EBV is the causative agent of infectious mononucleosis (IM) and is further implicated in malignancies of lymphoid and epithelial origins. In the past few decades, research efforts primarily focused on dissecting the mechanism of EBV-induced oncogenesis. Here, we present an alternate facet of the oncovirus EBV, on its applications in research and therapy. Finally, discussions on the prospective utilization of EBV in nasopharyngeal carcinoma (NPC) diagnosis and therapy will also be presented.
Collapse
Affiliation(s)
- Fenggang Yu
- Department of Otolaryngology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore.
| | - Wei Jian Tan
- Department of Microbiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Yanan Lu
- Department of Otolaryngology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Paul A MacAry
- Department of Microbiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Kwok Seng Loh
- Department of Otolaryngology-Head and Neck Surgery, National University Health System, Singapore
| |
Collapse
|
8
|
Gottfried L, Lin X, Barravecchia M, Dean DA. Identification of an alveolar type I epithelial cell-specific DNA nuclear import sequence for gene delivery. Gene Ther 2016; 23:734-742. [PMID: 27367840 PMCID: PMC10141512 DOI: 10.1038/gt.2016.52] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2016] [Revised: 04/27/2016] [Accepted: 06/20/2016] [Indexed: 11/09/2022]
Abstract
The ability to restrict gene delivery and expression to particular cell types is of paramount importance for many types of gene therapy, especially in the lung. The alveolar epithelial type I (ATI) cell, in particular, is an attractive cell type to target, as it comprises 95% of the internal surface area of the lung. We demonstrate, through microinjection of fluorescently labeled plasmids, that a DNA sequence within the rat T1α promoter was able to mediate ATI cell-specific plasmid DNA nuclear import due to the binding of ATI-enriched transcription factors. Promoter deletion analysis and site-directed mutagenesis of specific transcription-factor-binding sites within the +101 to -200 bp region of the T1α promoter identified HNF3 and TTF-1 as critical transcription factors for import. To test for nuclear import in vivo, plasmids expressing GFP from the CMV promoter were delivered into the lungs of mice by electroporation and evaluated immunohistochemically 48 h later. Plasmids carrying the 1.3 kbp T1α sequence resulted in GFP expression almost exclusively in ATI cells. This represents a new and highly efficient way to target a specific lung epithelial cell type both in vitro and in vivo based on the restriction of DNA nuclear import.
Collapse
Affiliation(s)
- L Gottfried
- Department of Pediatrics, School of Medicine and Dentistry, University of Rochester, Rochester, NY, USA
| | - X Lin
- Department of Pediatrics, School of Medicine and Dentistry, University of Rochester, Rochester, NY, USA
| | - M Barravecchia
- Department of Pediatrics, School of Medicine and Dentistry, University of Rochester, Rochester, NY, USA
| | - D A Dean
- Department of Pediatrics, School of Medicine and Dentistry, University of Rochester, Rochester, NY, USA
| |
Collapse
|
9
|
Advanced Design of Dumbbell-shaped Genetic Minimal Vectors Improves Non-coding and Coding RNA Expression. Mol Ther 2016; 24:1581-91. [PMID: 27357627 DOI: 10.1038/mt.2016.138] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2015] [Accepted: 06/21/2016] [Indexed: 12/27/2022] Open
Abstract
Dumbbell-shaped DNA minimal vectors lacking nontherapeutic genes and bacterial sequences are considered a stable, safe alternative to viral, nonviral, and naked plasmid-based gene-transfer systems. We investigated novel molecular features of dumbbell vectors aiming to reduce vector size and to improve the expression of noncoding or coding RNA. We minimized small hairpin RNA (shRNA) or microRNA (miRNA) expressing dumbbell vectors in size down to 130 bp generating the smallest genetic expression vectors reported. This was achieved by using a minimal H1 promoter with integrated transcriptional terminator transcribing the RNA hairpin structure around the dumbbell loop. Such vectors were generated with high conversion yields using a novel protocol. Minimized shRNA-expressing dumbbells showed accelerated kinetics of delivery and transcription leading to enhanced gene silencing in human tissue culture cells. In primary human T cells, minimized miRNA-expressing dumbbells revealed higher stability and triggered stronger target gene suppression as compared with plasmids and miRNA mimics. Dumbbell-driven gene expression was enhanced up to 56- or 160-fold by implementation of an intron and the SV40 enhancer compared with control dumbbells or plasmids. Advanced dumbbell vectors may represent one option to close the gap between durable expression that is achievable with integrating viral vectors and short-term effects triggered by naked RNA.
Collapse
|
10
|
|
11
|
Abstract
Electroporation has been used extensively to transfer DNA to bacteria, yeast, and mammalian cells in culture for the past 30 years. Over this time, numerous advances have been made, from using fields to facilitate cell fusion, delivery of chemotherapeutic drugs to cells and tissues, and most importantly, gene and drug delivery in living tissues from rodents to man. Electroporation uses electrical fields to transiently destabilize the membrane allowing the entry of normally impermeable macromolecules into the cytoplasm. Surprisingly, at the appropriate field strengths, the application of these fields to tissues results in little, if any, damage or trauma. Indeed, electroporation has even been used successfully in human trials for gene delivery for the treatment of tumors and for vaccine development. Electroporation can lead to between 100 and 1000-fold increases in gene delivery and expression and can also increase both the distribution of cells taking up and expressing the DNA as well as the absolute amount of gene product per cell (likely due to increased delivery of plasmids into each cell). Effective electroporation depends on electric field parameters, electrode design, the tissues and cells being targeted, and the plasmids that are being transferred themselves. Most importantly, there is no single combination of these variables that leads to greatest efficacy in every situation; optimization is required in every new setting. Electroporation-mediated in vivo gene delivery has proven highly effective in vaccine production, transgene expression, enzyme replacement, and control of a variety of cancers. Almost any tissue can be targeted with electroporation, including muscle, skin, heart, liver, lung, and vasculature. This chapter will provide an overview of the theory of electroporation for the delivery of DNA both in individual cells and in tissues and its application for in vivo gene delivery in a number of animal models.
Collapse
Affiliation(s)
- Jennifer L Young
- Department of Pediatrics, University of Rochester, Rochester, NY, USA
| | - David A Dean
- Departments of Pediatrics and Biomedical Engineering, University of Rochester, Rochester, NY, USA
| |
Collapse
|
12
|
A novel nonviral gene delivery system: multifunctional envelope-type nano device. ADVANCES IN BIOCHEMICAL ENGINEERING/BIOTECHNOLOGY 2014; 119:197-230. [PMID: 19343308 DOI: 10.1007/10_2008_40] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/13/2023]
Abstract
In this review we introduce a new concept for developing a nonviral gene delivery system which we call "Programmed Packaging." Based on this concept, we succeeded in developing a multifunctional envelope-type nano device (MEND), which exerts high transfection activities equivalent to those of an adenovirus in a dividing cell. The use of MEND has been extended to in vivo applications. PEG/peptide/DOPE ternary conjugate (PPD)-MEND, a new in vivo gene delivery system for the targeting of tumor cells that dissociates surface-modified PEG in tumor tissue by matrix metalloproteinase (MMP) and exerts significant transfection activities, was developed. In parallel with the development of MEND, a quantitative gene delivery system, Confocal Image-assisted 3-dimensionally integrated quantification (CIDIQ), also was developed. This method identified the rate-limiting step of the nonviral gene delivery system by comparing it with adenoviral-mediated gene delivery. The results of this analysis provide a new direction for the development of rational nonviral gene delivery systems.
Collapse
|
13
|
Xu L, Jin X, Rainey GJ, Wu H, Gao C. A mammalian expression system for high throughput antibody screening. J Immunol Methods 2013; 395:45-53. [DOI: 10.1016/j.jim.2013.06.009] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2013] [Revised: 06/20/2013] [Accepted: 06/21/2013] [Indexed: 11/29/2022]
|
14
|
Jäger V, Büssow K, Wagner A, Weber S, Hust M, Frenzel A, Schirrmann T. High level transient production of recombinant antibodies and antibody fusion proteins in HEK293 cells. BMC Biotechnol 2013; 13:52. [PMID: 23802841 PMCID: PMC3699382 DOI: 10.1186/1472-6750-13-52] [Citation(s) in RCA: 158] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2012] [Accepted: 05/24/2013] [Indexed: 12/02/2022] Open
Abstract
Background The demand of monospecific high affinity binding reagents, particularly monoclonal antibodies, has been steadily increasing over the last years. Enhanced throughput of antibody generation has been addressed by optimizing in vitro selection using phage display which moved the major bottleneck to the production and purification of recombinant antibodies in an end-user friendly format. Single chain (sc)Fv antibody fragments require additional tags for detection and are not as suitable as immunoglobulins (Ig)G in many immunoassays. In contrast, the bivalent scFv-Fc antibody format shares many properties with IgG and has a very high application compatibility. Results In this study transient expression of scFv-Fc antibodies in human embryonic kidney (HEK) 293 cells was optimized. Production levels of 10-20 mg/L scFv-Fc antibody were achieved in adherent HEK293T cells. Employment of HEK293-6E suspension cells expressing a truncated variant of the Epstein Barr virus (EBV) nuclear antigen (EBNA) 1 in combination with production under serum free conditions increased the volumetric yield up to 10-fold to more than 140 mg/L scFv-Fc antibody. After vector optimization and process optimization the yield of an scFv-Fc antibody and a cytotoxic antibody-RNase fusion protein further increased 3-4-fold to more than 450 mg/L. Finally, an entirely new mammalian expression vector was constructed for single step in frame cloning of scFv genes from antibody phage display libraries. Transient expression of more than 20 different scFv-Fc antibodies resulted in volumetric yields of up to 600 mg/L and 400 mg/L in average. Conclusion Transient production of recombinant scFv-Fc antibodies in HEK293-6E in combination with optimized vectors and fed batch shake flasks cultivation is efficient and robust, and integrates well into a high-throughput recombinant antibody generation pipeline.
Collapse
|
15
|
Flynn RP, Zacharias J, Zhou X, Cannon ML, Philpott NJ. Non-integrating lentiviral vectors for specific killing of Epstein-Barr virus nuclear antigen 1-positive B cell lymphoma cells. J Gene Med 2012; 13:487-96. [PMID: 21850667 DOI: 10.1002/jgm.1601] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Epstein-Barr virus (EBV) causes a range of life-threatening B-lymphocyte malignancies but, despite the use of various strategies, treatment remains problematic. METHODS In the present study, we developed a non-integrating lentiviral vector (NILV) that mediates specific killing of EBV nuclear antigen 1 (EBNA1)-expressing cells with minimal toxicity to EBNA1-negative cells. The EBV family of repeats (FR) was cloned intok the NILV genome upstream of various transgenes. RESULTS The presence of the FR in the NILV genome induced transcriptional up-regulation and prolonged the expression of a transgene specifically in EBNA1-positive B cells. Transgene expression from an FR-containing NILV was also prolonged in EBV-transformed cells compared to an FR-negative NILV. We found that the delivery of an FR-containing NILV encoding herpes simplex virus 1 thymidine kinase (TK) lead to the killing of more than 99% of EBNA1-positive B cells with minimal toxicity to EBNA1-negative cells in the presence of gancyclovir. EBNA1-positive cells were not killed by an FR-negative vector containing the TK gene. An FR-TK-containing NILV also specifically killed EBNA1-containing cells in a mixed population of EBNA1-positive and EBNA1-negative cells, thus confirming that NILV-FR-TK-mediated killing is specific for EBNA1-expressing cells. CONCLUSIONS Transgene expression from our NILVs is both EBNA1-specific and dependent upon the presence of the FR. The results obtained in the present study indicate that NILVs have potential use in the treatment of EBV-associated B cell malignancies.
Collapse
Affiliation(s)
- Ryan P Flynn
- Division of Rheumatic and Autoimmune Diseases, Department of Medicine, Institute of Human Genetics, Masonic Cancer Center, University of Minnesota Medical School, Minneapolis, MN, USA
| | | | | | | | | |
Collapse
|
16
|
Abstract
The first protocols describing transient gene expression in mammalian cells for the rapid generation of recombinant proteins emerged more than 10 years ago as an alternative to the establishment of stable, often amplified clonal cell lines, and relieved somewhat the bias against mammalian cell systems as being too complicated, labor intensive, and tedious to serve as a source for tool proteins in industrial research and academia. Over the past decade, these attempts have been refined and optimized, giving rise to expression protocols applicable in every lab in dependence on available tools, equipment, and envisaged outcome. This chapter summarizes the development of transient expression technologies over the past decade up to its current status and provides an outlook into what may be the future of transient technology development.
Collapse
|
17
|
Matsushita T, Kawakubo Y, Sawano M, Funatsu K. Vitronectin enhances adhesion force and t-PA production of weakly adherent 293 cells exposed to a shear stress. Cytotechnology 2011; 32:181-90. [PMID: 19002979 DOI: 10.1023/a:1008140632747] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
The effects of shear stress on the adhesion andproductivity of 293 cells were studied quantitativelyand compared with those of Vero and human liver cells.These cells were cultured in polystyrene dishes byusing shear stress exposing equipment. 50% of 293cells cultured in 2% FBS supplemented medium detachedfrom the dish after 29 h of exposure to a shear stressof 0.10 Pa. On the other hand, 90% of Vero and humanliver cells remained on the dish under the samecondition. Observations with scanning electronmicroscopy about cell adhesion plaques on the surfaceof the dish showed that the area covered withlamellipodia and the number of microspikes for 293cells were found to be less than those of the othercell lines. Several attachment factors, especiallyvitronectin, were found to enhance the number ofmicrospikes and the adhesion force of 293 cells.Almost 100% of 293 cells remained on thevitronectin-coated dish after 40 h under 0.10 Pa ofshear stress. A higher shear stress (greater than 0.10Pa) caused a decrease in tissue plasminogen activator(t-PA) productivity of 293 cells. But 0.03 Pastimulated the t-PA secretion on the non-coated dish.Vitronectin also enhanced the t-PA secretion evenunder 0.10 Pa. These results indicate that theadhesion force of 293 cells is obviously weaker thanthat of the other cell lines, and vitronectin enhancesthe adhesion force and the productivity of 293 cellsexposed to a shear stress.
Collapse
Affiliation(s)
- T Matsushita
- Department of Chemical Systems and Engineering, Graduate School of Engineering, Kyushu University, 6-10-1 Hakozaki, Higashi-ku, Fukuoka, 812-8581, Japan
| | | | | | | |
Collapse
|
18
|
Abstract
Gene transfer within the cardiovascular system was first demonstrated in 1989 yet, despite extensive basic-science and clinical research, unequivocal benefit in the clinical setting remains to be demonstrated. Potential reasons for this include the fact that recombinant viral vectors, used in the majority of clinical studies, have inherent problems with immunogenicity that are difficult to circumvent. Attention has turned therefore to plasmid vectors, which possess many advantages over viruses in terms of safety and ease of use, and many clinical studies have now been performed using non-viral technology. This review will provide an overview of clinical trials for cardiovascular disease using plasmid vectors, recent developments in plasmid delivery and design, and potential directions for this modality of gene therapy.
Collapse
Affiliation(s)
- Paul D Williams
- Manchester Academic Health Science Centre, School of Biomedicine, Vascular Gene Therapy Unit, Core Technology Facility, The University of Manchester, 46 Grafton Street, Manchester M13 9NT, UK.
| | | |
Collapse
|
19
|
Zhou Y, Ren L, Zhu J, Yan S, Wang H, Song N, Li L, Ouyang H, Pang D. Construction of a recombinant human FGF1 expression vector for mammary gland-specific expression in human breast cancer cells. Mol Cell Biochem 2011; 354:39-46. [PMID: 21461910 DOI: 10.1007/s11010-011-0803-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2011] [Accepted: 03/24/2011] [Indexed: 11/24/2022]
Abstract
Human Fibroblast growth factor 1 (FGF1) has been recognized as a valuable protein drug for the treatment of many diseases because of its multiple functions in regulating a variety of biological processes involved in embryonic development, cell growth and differentiation, morphogenesis, tissue repair, and others. The aim of this study was to develop an FGF1 mammary gland-specific expression vector to produce FGF1 on a large scale from transgenic cows to meet the demand for FGF1 in medical use. In this study, we generated an FGF1 mammary gland-specific expression vector and validated its function in human MCF-7 cells. This vector was shown to successfully express functional FGF1, thus potentially enabling the generation of transgenic cows to be used as mammary gland bioreactors.
Collapse
Affiliation(s)
- Yang Zhou
- Department of Animal Biotechnology, College of Animal Science and Veterinary Medicine, Jilin University, Changchun, 130062 Jilin, China
| | | | | | | | | | | | | | | | | |
Collapse
|
20
|
BRG1 helps RNA polymerase II to overcome a nucleosomal barrier during elongation, in vivo. EMBO Rep 2010; 11:751-7. [PMID: 20829883 DOI: 10.1038/embor.2010.131] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2010] [Revised: 07/06/2010] [Accepted: 07/27/2010] [Indexed: 11/08/2022] Open
Abstract
RNA polymerase II (RNAPII) transcribes genes in a chromatin context. We have designed a system to investigate the role of chromatin remodelling during elongation in vivo, which involves inserting a strong nucleosome-positioning sequence between a promoter and a reporter gene. Our data indicate that a nucleosome positioned in the body of a transcription unit impairs RNAPII progression, provokes RNAPII accumulation upstream to the positioned nucleosome and reduces transcription. By using this system, we show that BRG1, the enzymatic motor of the SWI-SNF chromatin-remodelling complex, is recruited to the positioned nucleosome in a transcription elongation-dependent manner and facilitates traversal of the nucleosome by RNAPII.
Collapse
|
21
|
Development of cell cultures that express hepatitis B virus to high levels and accumulate cccDNA. J Virol Methods 2010; 169:52-60. [PMID: 20600328 DOI: 10.1016/j.jviromet.2010.06.015] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2010] [Revised: 06/10/2010] [Accepted: 06/21/2010] [Indexed: 01/20/2023]
Abstract
Establishment of an infection with hepatitis B virus (HBV) requires synthesis and maintenance of a covalently closed circular DNA (cccDNA) form of the viral genome in the nucleus of host cells. To facilitate the investigation of the synthesis of cccDNA, cell cultures were developed that express HBV to high levels. Cell lines derived from hepatoma cells Huh7 and HepG2 were created that express Epstein-Barr virus (EBV) nuclear antigen-1 and a fusion protein of the Tet repressor and Kox1 transcriptional repression domain stably. Transfection of these cell lines with an expression plasmid for HBV that contains the origin of plasmid replication of EBV (oriP) led to increases in the intracellular levels of HBV core protein ( approximately 8- to 51-fold) and encapsidated HBV DNA ( approximately 3- to 12-fold) in comparison to Huh7 and HepG2 cells. Virion production was also increased ( approximately 3- to 12-fold) in these cell cultures and an increase in the level of cccDNA ( approximately 3-fold) was observed in the Huh7-derived cell lines. In addition, these cell lines maintained the HBV expression plasmid upon selection and expressed HBV conditionally. Thus, these cell cultures exhibit several features that facilitate study of the synthesis of cccDNA and other aspects of replication of HBV.
Collapse
|
22
|
The SP-C promoter facilitates alveolar type II epithelial cell-specific plasmid nuclear import and gene expression. Gene Ther 2010; 17:541-9. [PMID: 20054353 PMCID: PMC3482943 DOI: 10.1038/gt.2009.166] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Although nonviral gene therapy has great potential for use in the lung, the relative lack of cell-specific targeting has limited its applications. We have developed a new approach for cell-specific targeting based on selective nuclear import of plasmids in non-dividing cells. Using a microinjection and in situ hybridization approach, we tested several potential DNA sequences for the ability to mediate plasmid nuclear import in alveolar type II epithelial (ATII) cells. Of these, only a sequence within the human surfactant protein C (SP-C) promoter was able to mediate nuclear localization of plasmid DNA specifically in ATII cells but not in other cell types. We have mapped the minimal import sequence to the proximal 318 nucleotides of the promoter, and demonstrate that binding sites for NFI, TTF-1, and GATA-6 and the proteins themselves are required for import activity. Using intratracheal delivery of DNA followed by electroporation, we demonstrate that the SP-C promoter sequence will enhance gene expression specifically in ATII cells in mouse lung. This represents a novel activity for the SP-C promoter and thus ATII cell-specific nuclear import of DNA may prove to be a safe and effective method for targeted and enhanced gene expression in ATII cells.
Collapse
|
23
|
Kim KS, Kim MS, Moon JH, Jeong MS, Kim J, Lee GM, Myung PK, Hong HJ. Enhancement of recombinant antibody production in HEK 293E cells by WPRE. BIOTECHNOL BIOPROC E 2009. [DOI: 10.1007/s12257-008-0221-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/03/2023]
|
24
|
Ward A, Anderson M, Craggs RI, Maltby J, Grahames C, Davies RA, Finch D, Pattison D, Oakes H, Mallinder PR. E. coli expression and purification of human and cynomolgus IL-15. Protein Expr Purif 2009; 68:42-8. [DOI: 10.1016/j.pep.2009.05.004] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2009] [Revised: 05/01/2009] [Accepted: 05/03/2009] [Indexed: 10/20/2022]
|
25
|
Munkonge FM, Amin V, Hyde SC, Green AM, Pringle IA, Gill DR, Smith JWS, Hooley RP, Xenariou S, Ward MA, Leeds N, Leung KY, Chan M, Hillery E, Geddes DM, Griesenbach U, Postel EH, Dean DA, Dunn MJ, Alton EWFW. Identification and functional characterization of cytoplasmic determinants of plasmid DNA nuclear import. J Biol Chem 2009; 284:26978-87. [PMID: 19638341 PMCID: PMC2785383 DOI: 10.1074/jbc.m109.034850] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2009] [Indexed: 01/07/2023] Open
Abstract
Import of exogenous plasmid DNA (pDNA) into mammalian cell nuclei represents a key intracellular obstacle to efficient non-viral gene delivery. This includes access of the pDNA to the nuclei of non-dividing cells where the presence of an intact nuclear membrane is limiting for gene transfer. Here we identify, isolate, and characterize, cytoplasmic determinants of pDNA nuclear import into digitonin-permeabilized HeLa cells. Depletion of putative DNA-binding proteins, on the basis of their ability to bind immobilized pDNA, abolished pDNA nuclear import supporting the critical role of cytoplasmic factors in this process. Elution of pDNA-bound proteins, followed by two-dimensional sodium dodecyl polyacrylamide gel electrophoresis identified several candidate DNA shuttle proteins. We show that two of these, NM23-H2, a ubiquitous c-Myc transcription-activating nucleoside diphosphate kinase, and the core histone H2B can both reconstitute pDNA nuclear import. Further, we demonstrate a significant increase in gene transfer in non-dividing HeLa cells transiently transfected with pDNA containing binding sequences from two of the DNA shuttle proteins, NM23-H2 and the homeobox transcription factor Chx10. These data support the hypothesis that exogenous pDNA binds to cytoplasmic shuttle proteins and is then translocated to the nucleus using the minimal import machinery. Importantly, increasing the binding of pDNA to shuttle proteins by re-engineering reporter plasmids with shuttle binding sequences enhances gene transfer. Increasing the potential for exogenously added pDNA to bind intracellular transport cofactors may enhance the potency of non-viral gene transfer.
Collapse
Affiliation(s)
- Felix M Munkonge
- Department of Gene Therapy, National Heart & Lung Institute, Faculty of Medicine, Imperial College London, London SW3 6LR, United Kingdom.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Miller AM, Dean DA. Tissue-specific and transcription factor-mediated nuclear entry of DNA. Adv Drug Deliv Rev 2009; 61:603-13. [PMID: 19393704 DOI: 10.1016/j.addr.2009.02.008] [Citation(s) in RCA: 73] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2008] [Accepted: 02/05/2009] [Indexed: 12/22/2022]
Abstract
Low levels of gene transfer and a lack of tissue-specific targeting of vectors have limited the therapeutic potential of non-viral gene therapy. This is due to the numerous cellular barriers that hinder nuclear delivery of vectors and the paucity of methods that restrict expression to specific cells types. In non-dividing cells, the nuclear envelope is an especially problematic hurdle to gene transfer. Given that the majority of target tissues are non-dividing in vivo, the nuclear membrane is a major obstacle to therapeutic gene transfer. In this review, the various barriers to gene transfer are discussed. In particular, the role of the nuclear pore complex (NPC) in regulating passage of plasmid vectors during interphase is reviewed. Several methods of modifying plasmid (pDNA) vectors to enhance nuclear import through the NPC are also discussed, including the use of tissue-specific transcription factors to mediate nuclear entry of pDNA in a cell-specific manner.
Collapse
Affiliation(s)
- Aaron M Miller
- Division of Pulmonary and Critical Care Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | | |
Collapse
|
27
|
|
28
|
Zinc coordination is required for and regulates transcription activation by Epstein-Barr nuclear antigen 1. PLoS Pathog 2009; 5:e1000469. [PMID: 19521517 PMCID: PMC2690687 DOI: 10.1371/journal.ppat.1000469] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2008] [Accepted: 05/12/2009] [Indexed: 12/17/2022] Open
Abstract
Epstein-Barr Nuclear Antigen 1 (EBNA1) is essential for Epstein-Barr virus to immortalize naïve B-cells. Upon binding a cluster of 20 cognate binding-sites termed the family of repeats, EBNA1 transactivates promoters for EBV genes that are required for immortalization. A small domain, termed UR1, that is 25 amino-acids in length, has been identified previously as essential for EBNA1 to activate transcription. In this study, we have elucidated how UR1 contributes to EBNA1's ability to transactivate. We show that zinc is necessary for EBNA1 to activate transcription, and that UR1 coordinates zinc through a pair of essential cysteines contained within it. UR1 dimerizes upon coordinating zinc, indicating that EBNA1 contains a second dimerization interface in its amino-terminus. There is a strong correlation between UR1-mediated dimerization and EBNA1's ability to transactivate cooperatively. Point mutants of EBNA1 that disrupt zinc coordination also prevent self-association, and do not activate transcription cooperatively. Further, we demonstrate that UR1 acts as a molecular sensor that regulates the ability of EBNA1 to activate transcription in response to changes in redox and oxygen partial pressure (pO2). Mild oxidative stress mimicking such environmental changes decreases EBNA1-dependent transcription in a lymphoblastoid cell-line. Coincident with a reduction in EBNA1-dependent transcription, reductions are observed in EBNA2 and LMP1 protein levels. Although these changes do not affect LCL survival, treated cells accumulate in G0/G1. These findings are discussed in the context of EBV latency in body compartments that differ strikingly in their pO2 and redox potential. Epstein-Barr virus (EBV) infects human B-cells and immortalizes them. Immortalization results in diseases that range from infectious mononucleosis to malignancies such as lymphomas. During immortalization, EBV expresses a small number of viral genes that modulate cellular proliferation and differentiation. One of the genes expressed by EBV, Epstein-Barr nuclear antigen 1 (EBNA1), activates the expression of the other viral genes required for immortalization. In this report, we have explored the mechanism by which EBNA1 activates gene expression. We have determined that EBNA1 uses the micronutrient zinc to self-associate, and that self-association is necessary for it to activate gene expression. Further, we have determined that environmental conditions such as oxygen tension and oxidative stress modulate EBNA1's capacity to self-associate, and therefore to activate gene expression. The gene expression profile and proliferative phenotype of EBV-infected cells is known to vary in differing environmental niches in the human body, such as lymph nodes and in peripheral circulation. We interpret our results to postulate that these differences arise as a consequence of varying oxygen tension in these microenvironments on EBNA1's capacity to activate viral gene expression. Our findings can be exploited to devise novel therapeutics against EBV-associated diseases that target EBNA1 through oxidative stress.
Collapse
|
29
|
Reflections on more than 10 years of TGE approaches. Protein Expr Purif 2009; 64:99-107. [DOI: 10.1016/j.pep.2008.10.017] [Citation(s) in RCA: 93] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2008] [Revised: 10/21/2008] [Accepted: 10/21/2008] [Indexed: 11/22/2022]
|
30
|
Feeney KM, Parish JL. Targeting mitotic chromosomes: a conserved mechanism to ensure viral genome persistence. Proc Biol Sci 2009; 276:1535-44. [PMID: 19203914 PMCID: PMC2660980 DOI: 10.1098/rspb.2008.1642] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Viruses that maintain their genomes as extrachromosomal circular DNA molecules and establish infection in actively dividing cells must ensure retention of their genomes within the nuclear envelope in order to prevent genome loss. The loss of nuclear membrane integrity during mitosis dictates that paired host cell chromosomes are captured and organized by the mitotic spindle apparatus before segregation to daughter cells. This prevents inaccurate chromosomal segregation and loss of genetic material. A similar mechanism may also exist for the nuclear retention of extrachromosomal viral genomes or episomes during mitosis, particularly for genomes maintained at a low copy number in latent infections. It has been heavily debated whether such a mechanism exists and to what extent this mechanism is conserved among diverse viruses. Research over the last two decades has provided a wealth of information regarding the mechanisms by which specific tumour viruses evade mitotic and DNA damage checkpoints. Here, we discuss the similarities and differences in how specific viruses tether episomal genomes to host cell chromosomes during mitosis to ensure long-term persistence.
Collapse
Affiliation(s)
- Katherine M Feeney
- Bute Medical School, University of St Andrews, St Andrews, Fife KY16 9TS, UK
| | | |
Collapse
|
31
|
Identifying sites bound by Epstein-Barr virus nuclear antigen 1 (EBNA1) in the human genome: defining a position-weighted matrix to predict sites bound by EBNA1 in viral genomes. J Virol 2009; 83:2930-40. [PMID: 19129441 DOI: 10.1128/jvi.01974-08] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
We identified binding sites for Epstein-Barr virus (EBV) nuclear antigen 1 (EBNA1) in the human genome using chromatin immunoprecipitation and microarrays. The sequences for these newly identified sites were used to generate a position-weighted matrix (PWM) for EBNA1's DNA-binding sites. This PWM helped identify additional DNA-binding sites for EBNA1 in the genomes of EBV, Kaposi's sarcoma-associated herpesvirus, and cercopithecine herpesvirus 15 (CeHV-15) (also called herpesvirus papio 15). In particular, a homologue of the Rep* locus in EBV was predicted in the genome of CeHV-15, which is notable because Rep* of EBV was not predicted by the previously developed consensus sequence for EBNA1's binding DNA. The Rep* of CeHV-15 functions as an origin of DNA synthesis in the EBV-positive cell line Raji; this finding thus builds on a set of DNA-binding sites for EBNA1 predicted in silico.
Collapse
|
32
|
Abstract
The timely availability of recombinant proteins in sufficient quantity and of validated quality is of utmost importance in driving drug discovery and the development of low molecular weight compounds, as well as for biotherapeutics. Transient gene expression (TGE) in mammalian cells has emerged as a promising technology for protein generation over the past decade as TGE meets all the prerequisites with respect to quantity and quality of the product as well as cost-effectiveness and speed of the process. Optimized protocols have been developed for both HEK293 and CHO cell lines which allow protein production at any desired scale up to >100 l and in milligram to gram quantities. Along with an overview on current scientific and technological knowledge, detailed protocols for expression of recombinant proteins on small, medium, and large scale are discussed in the following chapter.
Collapse
|
33
|
Wang L, Shan L, Lo KW, Yin J, Zhang Y, Sun R, Zhong J. Inhibition of nasopharyngeal carcinoma growth by RTA-expressing baculovirus vectors containing oriP. J Gene Med 2008; 10:1124-33. [PMID: 18642396 DOI: 10.1002/jgm.1237] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
BACKGROUND Nasopharyngeal carcinoma (NPC) is closely associated with latent Epstein-Barr virus (EBV) infection. Activation of latent EBV into lytic replication by introducing viral lytic gene BRLF1 (RTA) has been shown to be a potential approach to suppress the growth of EBV-associated NPC tumor. METHODS The baculovirus vectors with RTA expression cassette (BV-R), RTA and the EBV latent replication origin (OriP, BV-RO), or RTA, OriP and EBNA-1 gene (BV-ROE-CMV), were constructed and examined for their ability to mediate RTA expression, initiate lytic replication and induce cell death in EBV-positive cell lines Hone1-EBV, HK1-EBV and C666-1 in vitro. Their effect to inhibit the growth of EBV-positive NPC tumors was also evaluated in nude mice. RESULTS The baculovirus vectors BV-RO and BV-ROE-CMV mediated efficient expression of RTA in EBV-positive NPC cells. Lytic EBV replication and cell death were observed in these infected cells. Both vectors also significantly inhibited the growth of EBV-positive NPC tumor in nude mice. EBV early lytic gene expression and tumor cell death were observed in these treated tumors. CONCLUSIONS The presence of OriP improved the performance of the RTA-expressing baculovirus vectors to induce EBV lytic replication and cell death in vitro, and suppress the growth of EBV positive NPC tumors in vivo. By confining RTA and EBNA-1 expression to EBV-positive cells, BV-RO is expected to be a better candidate in application than BV-ROE-CMV in the long term.
Collapse
Affiliation(s)
- Leyao Wang
- Department of Microbiology and Microbial Engineering, School of Life Sciences, Fudan University, Shanghai, China
| | | | | | | | | | | | | |
Collapse
|
34
|
Adding l-lysine derivatives to the genetic code of mammalian cells with engineered pyrrolysyl-tRNA synthetases. Biochem Biophys Res Commun 2008; 371:818-22. [PMID: 18471995 DOI: 10.1016/j.bbrc.2008.04.164] [Citation(s) in RCA: 227] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2008] [Accepted: 04/28/2008] [Indexed: 11/21/2022]
Abstract
We report a method for site-specifically incorporating l-lysine derivatives into proteins in mammalian cells, based on the expression of the pyrrolysyl-tRNA synthetase (PylRS)-tRNA(Pyl) pair from Methanosarcina mazei. Different types of external promoters were tested for the expression of tRNA(Pyl) in Chinese hamster ovary cells. When tRNA(Pyl) was expressed from a gene cluster under the control of the U6 promoter, the wild-type PylRS-tRNA(Pyl) pair facilitated the most efficient incorporation of a pyrrolysine analog, N(epsilon)-tert-butyloxycarbonyl-l-lysine (Boc-lysine), into proteins at the amber position. This PylRS-tRNA(Pyl) system yielded the Boc-lysine-containing protein in an amount accounting for 1% of the total protein in human embryonic kidney (HEK) 293 cells. We also created a PylRS variant specific to N(epsilon)-benzyloxycarbonyl-l-lysine, to incorporate this long, bulky, non-natural lysine derivative into proteins in HEK293. The recently reported variant specific to N(epsilon)-acetyllysine was also expressed, resulting in the genetic encoding of this naturally-occurring lysine modification in mammalian cells.
Collapse
|
35
|
Young JL, Zimmer WE, Dean DA. Smooth muscle-specific gene delivery in the vasculature based on restriction of DNA nuclear import. Exp Biol Med (Maywood) 2008; 233:840-8. [PMID: 18445769 DOI: 10.3181/0712-rm-331] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
The two currently employed approaches restricting gene delivery and/or expression to desired cell types in vivo rely on cell surface targeting or cell-specific promoters. We have developed a third approach based on cell-specific nuclear transport of the delivered plasmid DNA. We have previously shown that plasmid nuclear import in non-dividing cells is sequence-specific and have identified a set of cell-specific DNA nuclear targeting sequences that can be used to limit DNA nuclear import to desired cell types. Specifically we have identified elements of the smooth muscle gamma actin (SMGA) promoter that direct plasmid nuclear import selectively in smooth muscle cells (SMCs) in vitro (Vacik et al, 1999, Gene Therapy 6:1006-1014). In the present study, we demonstrate that the SMC-specific DNA nuclear targeting sequence from the SMGA promoter drives nuclear accumulation of plasmids and subsequent gene expression exclusively in the smooth muscle cell layer of the vessel wall in the intact vasculature of rats using electroporation mediated delivery. These results demonstrate that certain DNA nuclear targeting sequences can be used to restrict DNA nuclear import to specific cell types providing a new, novel means of cell targeting for gene therapy.
Collapse
Affiliation(s)
- Jennifer L Young
- Division of Neonatology, University of Rochester, Box 850, 601 Elmwood Ave, Rochester, NY 14642, USA
| | | | | |
Collapse
|
36
|
Pashine A, Göpfert U, Chen J, Hoffmann E, Dietrich PS, Peng SL. Failed efficacy of soluble human CD83-Ig in allogeneic mixed lymphocyte reactions and experimental autoimmune encephalomyelitis: implications for a lack of therapeutic potential. Immunol Lett 2008; 115:9-15. [PMID: 18079004 DOI: 10.1016/j.imlet.2007.10.015] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2007] [Accepted: 10/24/2007] [Indexed: 01/09/2023]
Abstract
Soluble forms of CD83, a dendritic cell-specific surface glycoprotein, have been strongly proposed to be of therapeutic utility in inflammatory conditions such as multiple sclerosis and transplantation. We demonstrate here, however, that eukaryotically expressed, recombinant soluble human CD83-Ig molecules fail to achieve efficacy in model systems for those conditions: mouse experimental autoimmune encephalomyelitis models in vivo or in mixed lymphocyte reactions in vitro. These results raise concern as to the viability of a eukaryotically expressed soluble CD83 strategy for clinical therapeutic use.
Collapse
MESH Headings
- Animals
- Antigens, CD/blood
- Antigens, CD/immunology
- Antigens, CD/therapeutic use
- Dendritic Cells/immunology
- Encephalomyelitis, Autoimmune, Experimental/drug therapy
- Encephalomyelitis, Autoimmune, Experimental/immunology
- Encephalomyelitis, Autoimmune, Experimental/metabolism
- Humans
- Immunoglobulins/blood
- Immunoglobulins/immunology
- Immunoglobulins/therapeutic use
- Lymphocyte Culture Test, Mixed
- Membrane Glycoproteins/blood
- Membrane Glycoproteins/immunology
- Membrane Glycoproteins/therapeutic use
- Mice
- Mice, Inbred C57BL
- Recombinant Fusion Proteins/immunology
- Recombinant Fusion Proteins/therapeutic use
- CD83 Antigen
Collapse
|
37
|
Akamatsu Y, Pakabunto K, Xu Z, Zhang Y, Tsurushita N. Whole IgG surface display on mammalian cells: Application to isolation of neutralizing chicken monoclonal anti-IL-12 antibodies. J Immunol Methods 2007; 327:40-52. [PMID: 17719061 DOI: 10.1016/j.jim.2007.07.007] [Citation(s) in RCA: 51] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2007] [Revised: 06/14/2007] [Accepted: 07/11/2007] [Indexed: 10/23/2022]
Abstract
We have developed a mammalian cell surface display vector, suitable for directly isolating IgG molecules based on their antigen-binding affinity and biological activity. Using an Epstein-Barr virus-derived episomal vector, antibody libraries are displayed as whole IgG molecules on the cell surface and screened for specific antigen binding by a combination of magnetic beads and fluorescence-activated cell sorting. Plasmids encoding antibodies with desired binding characteristics are recovered from sorted cells and are converted to the form for production of soluble IgG. Transiently expressed soluble IgG antibodies are individually tested for binding to target antigens, as well as for biological activities, such as neutralization. This vector system was used to generate antibody display libraries derived from spleen cDNA of chickens immunized with human and mouse IL-12. Chicken-human chimeric IgG1 antibodies that neutralize human and mouse IL-12 were successfully isolated from the library. The mammalian surface display vector developed in this work facilitates the isolation of monoclonal antibodies from essentially any species.
Collapse
|
38
|
Lindner SE, Sugden B. The plasmid replicon of Epstein-Barr virus: mechanistic insights into efficient, licensed, extrachromosomal replication in human cells. Plasmid 2007; 58:1-12. [PMID: 17350094 PMCID: PMC2562867 DOI: 10.1016/j.plasmid.2007.01.003] [Citation(s) in RCA: 88] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2006] [Accepted: 01/05/2007] [Indexed: 12/24/2022]
Abstract
The genome of Epstein-Barr Virus (EBV) and plasmid derivatives of it are among the most efficient extrachromosomal replicons in mammalian cells. The latent origin of plasmid replication (oriP), when supplied with the viral Epstein-Barr Nuclear Antigen 1 (EBNA1) in trans, provides efficient duplication, partitioning and maintenance of plasmids bearing it. In this review, we detail what is known about the viral cis and trans elements required for plasmid replication. In addition, we describe how the cellular factors that EBV usurps are used to complement the functions of the viral constituents. Finally, we propose a model for the sequential assembly of an EBNA1-dependent origin of DNA synthesis into a pre-Replicative Complex (pre-RC), which functions by making use only of cellular enzymatic activities to carry out the replication of the viral plasmid.
Collapse
Affiliation(s)
- Scott E. Lindner
- McArdle Laboratory for Cancer Research, University of Wisconsin-Madison, Madison, WI 53706
| | - Bill Sugden
- McArdle Laboratory for Cancer Research, University of Wisconsin-Madison, Madison, WI 53706
| |
Collapse
|
39
|
Mocanu JD, Yip KW, Alajez NM, Shi W, Li JH, Lunt SJ, Moriyama EH, Wilson BC, Milosevic M, Lo KW, van Rooijen N, Busson P, Bastianutto C, Liu FF. Imaging the modulation of adenoviral kinetics and biodistribution for cancer gene therapy. Mol Ther 2007; 15:921-929. [PMID: 17356543 DOI: 10.1038/mt.sj.6300119] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2006] [Accepted: 01/04/2007] [Indexed: 11/08/2022] Open
Abstract
To explore systemic utilization of Epstein-Barr virus (EBV)-specific transcriptionally targeted adenoviruses, three vectors were constructed to examine kinetics, specificity, and biodistribution: adv.oriP.luc, expressing luciferase under EBV-specific control; adv.SV40luc, expressing luciferase constitutively; and adv.oriP.E1A.oriP.luc, a conditionally replicating adenovirus, expressing both luciferase and E1A. Bioluminescence imaging (BLI) was conducted on tumor-bearing severe combined immunodeficient (SCID) mice (C666-1, EBV-positive human nasopharyngeal cancer) treated intravenously (i.v.) with 3 x 10(8) infectious units (ifu) of the adenoviral vectors. At 72 hours, adv.oriPluc demonstrated an 8.4-fold higher tumor signal than adv.SV40luc; adv.oriP.E1A.oriP.luc was 26.7-fold higher; however, a significant liver signal was also observed, necessitating further action to improve biodistribution. Several compounds were examined to this end, including norepinephrine, serotonin, clodronate liposomes, and STI571, to determine whether any of these measures could improve adenoviral biodistribution. Each of these interventions was assessed using BLI in mice i.v. injected with adv.oriP.luc. STI571 achieved the highest increase in tumor-to-liver ratio (TLR; 6.6-fold), which was associated with a 59% reduction in tumor interstitial fluid pressure (IFP) along with a decrease in platelet-derived growth factor-beta receptor (PDGF beta R) activation. This study reports the favorable modulation by STI571 of the biodistribution of adenoviral vectors, providing a potential approach to improving therapeutic outcome.
Collapse
Affiliation(s)
- Joseph D Mocanu
- Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Shan L, Wang L, Yin J, Zhong P, Zhong J. An OriP/EBNA-1-based baculovirus vector with prolonged and enhanced transgene expression. J Gene Med 2007; 8:1400-6. [PMID: 17051599 DOI: 10.1002/jgm.978] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Abstract
BACKGROUND The baculovirus Autographa californica multiple nucleopolyhedrovirus (AcMNPV) has been explored as a gene delivery vehicle for a variety of mammalian cell lines. However, the transient expression nature due to its incapability to replicate in mammalian cells and insufficient transduction efficiency limit its application. METHODS Recombinant baculovirus vectors containing genetic elements from Epstein-Barr virus (EBV), OriP and EBNA-1, which are essential for the episomal maintenance of the EBV genome in latently infected cells, were constructed and tested for their ability to sustain and express transgene (enhanced green fluorescence protein (egfp)) in mammalian cells. RESULTS The recombinant baculovirus containing OriP and EBNA-1 genes driven by the cytomegalovirus (CMV) promoter was capable of persisting in a significant proportion of infected mammalian cells, HEK293, Vero, Cos-7, and Hone-1, without any selective pressure. In HEK293, the expression of EGFP lasted for 60 days with markedly enhanced expression level. The persistence of baculovirus genome correlated with the expression of EBNA-1. CONCLUSIONS The improved baculovirus vector could mediate prolonged and enhanced foreign gene expression in some mammalian cells. Furthermore, an adequate level of the EBNA-1 protein was essential for the maintenance of the OriP-containing baculovirus genome. The new vector has potential for use in gene therapy.
Collapse
Affiliation(s)
- Liang Shan
- Department of Microbiology and Microbial Engineering, School of Life Sciences, Fudan University, Shanghai 200433, China
| | | | | | | | | |
Collapse
|
41
|
Pham PL, Kamen A, Durocher Y. Large-scale transfection of mammalian cells for the fast production of recombinant protein. Mol Biotechnol 2007; 34:225-37. [PMID: 17172668 DOI: 10.1385/mb:34:2:225] [Citation(s) in RCA: 158] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/1999] [Revised: 11/30/1999] [Accepted: 11/30/1999] [Indexed: 11/11/2022]
Abstract
Recombinant proteins (r-proteins) are increasingly important in fundamental research and for clinical applications. As many of these r-proteins are of human or animal origin, cultivated mammalian cells are the host of choice to ensure their functional folding and proper posttranslational modifications. Large-scale transfection of human embryonic kidney 293 or Chinese hamster ovary cells is now an established technology that can be used in the production of hundreds of milligram to gram quantities of a r-protein in less than 1 mo from cloning of its cDNA. This chapter aims to provide an overview of large-scale transfection technology with a particular emphasis on calcium phosphate and polyethylenimine-mediated gene transfer.
Collapse
Affiliation(s)
- Phuong Lan Pham
- Laboratoire de Biotechnologie Vétérinaire et Alimentaire, Faculté de Médecine Vétérinaire, Université de Montréal, CP5000, Sainte-Hyacinthe (Québec) J2S 7C6, Canada
| | | | | |
Collapse
|
42
|
Vaughan EE, DeGiulio JV, Dean DA. Intracellular trafficking of plasmids for gene therapy: mechanisms of cytoplasmic movement and nuclear import. Curr Gene Ther 2006; 6:671-681. [PMID: 17168698 PMCID: PMC4400175 DOI: 10.2174/156652306779010688] [Citation(s) in RCA: 79] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Under physiologically relevant conditions, the levels of non-viral gene transfer are low at best. The reason for this is that many barriers exist for the efficient transfer of genes to cells, even before any gene expression can occur. While many transfection strategies focus on DNA condensation and overcoming the plasma membrane, events associated with the intracellular trafficking of the DNA complexes have not been as extensively studied. Once internalized, plasmids must travel potentially long distances through the cytoplasm to reach their next barrier, the nuclear envelope. This review summarizes the current progress on the cytoplasmic trafficking and nuclear transport of plasmids used for gene therapy applications. Both of these processes utilize specific and defined mechanisms to facilitate movement of DNA complexes through the cell. The continued elucidation and exploitation of these mechanisms will lead to improved strategies for transfection and successful gene therapy.
Collapse
Affiliation(s)
- Erin E. Vaughan
- Division of Pulmonary and Critical Care Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - James V. DeGiulio
- Division of Pulmonary and Critical Care Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - David A. Dean
- Division of Pulmonary and Critical Care Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| |
Collapse
|
43
|
Akita H, Hama S, Mizuguchi H, Harashima H. [Development of non-viral vector based on the quantitative comparison of intracellular trafficking with viral vector]. YAKUGAKU ZASSHI 2006; 126:1047-57. [PMID: 17077611 DOI: 10.1248/yakushi.126.1047] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
For the development of efficient gene vector, intracellular processes such as cellular uptake, endosomal release and nuclear delivery must be overcome. Viruses have also evolved and have developed sophisticated mechanisms for controlling intracellular trafficking for the efficient delivery of their genomes to nuclei in host cells for symbiosis. In the light of these mechanisms, various kinds of artificial devices have been developed to overcome the intracellular barriers. However, in the majority of studies, variation of the transfection activity before and after the modification of devices was evaluated, and intracellular trafficking remained unclear. Therefore, it is understand to recognize which of the intracellular barrier should be intensively improved to enhance the transfection activity. To clarify the rate-limited process in the current non-viral vector, we compared the intracellular trafficking between adenovirus and LipofectAMINE PLUS. As a result, we found that difference of the transfection efficiency between adenovirus and LipofectAMINE PLUS was dominantly derived from the differences on transcription activity. Therefore it is essential to consider the regulation of the intranuclear events to improve the transfection activity of artificial vector.
Collapse
Affiliation(s)
- Hidetaka Akita
- Faculty of Pharmaceutical Sciences, Hokkaido University, Kita-12 Nishi-6, Kita-ku, Sapporo City 060-0812, Japan.
| | | | | | | |
Collapse
|
44
|
Son JK, Oh ST, Cho SK, Yoon KH, Lee SK. Mechanism of prolonged gene expression by Epstein-Barr virus-based plasmid in porcine cells. Xenotransplantation 2006; 13:560-5. [PMID: 17059583 DOI: 10.1111/j.1399-3089.2006.00350.x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
BACKGROUND We previously showed that an Epstein-Barr virus (EBV)-based plasmid, pEBVGFP, exerts prolonged gene expression in porcine neonatal pancreatic cell clusters (NPCCs). In this study, the mechanism underlying this was investigated. METHODS GFP expression was analyzed in porcine cells transfected with pEBVGFP by FACS analysis and confocal microscopy. The possible integration of pEBVGFP into the chromosomal DNA was analyzed by Southern blot. Self-replication of the EBV-based plasmid in porcine cells was investigated by PCR. The NPCCs were immunostained to characterize cells transfected with pEBVGFP. RESULTS The EBV based plasmid provided prolonged GFP expression in porcine cells and duct cells were the main cells transfected among NPCCs. Southern blot showed that the transfected pEBVGFP stayed for a long time as an episome rather than integrating into the chromosomal DNA. pEBVGFP isolated from the transfected porcine cells had methylated CpG suggesting that they self-replicated in those cells. CONCLUSIONS The EBV-based plasmid may be useful for genetically manipulating porcine cells to enhance their value as xenotransplantation sources.
Collapse
Affiliation(s)
- Jung Kyu Son
- Research Institute of Immunobiology, Department of Biomedical Sciences, The Catholic University of Korea, Seoul, Korea
| | | | | | | | | |
Collapse
|
45
|
Howden SE, Wardan H, Voullaire L, McLenachan S, Williamson R, Ioannou P, Vadolas J. Chromatin-binding regions of EBNA1 protein facilitate the enhanced transfection of Epstein-Barr virus-based vectors. Hum Gene Ther 2006; 17:833-44. [PMID: 16942443 DOI: 10.1089/hum.2006.17.833] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Epstein-Barr virus (EBV)-based vectors can stably maintain large genomic fragments in mammalian cells, offering great potential for the treatment/correction of many acquired and inherited disorders. Numerous studies report marked increases in the transfection efficiency of EBV-based vectors after delivery into cell lines constitutively expressing Epstein-Barr nuclear antigen-1 (EBNA1), compared with cells not expressing EBNA1. We employ a novel strategy, involving the transfection of mRNA encoding EBNA1, to transiently express EBNA1 protein in human cells. Subsequently we show that the transfection efficiency of a 21-kb EBVbased vector is improved significantly when codelivered with mRNA encoding EBNA1. Similar increases in transfection efficiency were observed after delivery of the plasmid into cells constitutively expressing EBNA1. We also investigate the mechanism by which EBNA1 facilitates the transfection of EBV-based vectors, using mRNA encoding modified versions of the protein. Previous studies suggest that the EBNA1 DNA-binding domain (DBD), together with the nuclear localization signal (NLS), may enhance transfection of EBV plasmids by facilitating their nuclear transport. We demonstrate that an EBNA1 derivative comprising only the NLS and DBD does not facilitate transfection of EBV-based vectors. However, cells expressing an EBNA1 derivative devoid of a functional NLS but retaining the chromatin-binding regions, domains A and B, enhances plasmid transfection efficiency by up to 10-fold. Moreover, a variant of EBNA1 comprising two copies of domain A fused to the DBD enhances DNA transfection to an even greater extent than wild-type EBNA1. We therefore propose that EBNA1-mediated transfection of EBV-based vectors is dependent on the presence of chromatin- binding regions and the DBD, but not the NLS.
Collapse
Affiliation(s)
- Sara E Howden
- Cell and Gene Therapy Research Group, Department of Paediatrics, Murdoch Childrens Research Institute, University of Melbourne, Royal Children's Hospital, Parkville 3052, Australia.
| | | | | | | | | | | | | |
Collapse
|
46
|
Altmann M, Pich D, Ruiss R, Wang J, Sugden B, Hammerschmidt W. Transcriptional activation by EBV nuclear antigen 1 is essential for the expression of EBV's transforming genes. Proc Natl Acad Sci U S A 2006; 103:14188-93. [PMID: 16966603 PMCID: PMC1599932 DOI: 10.1073/pnas.0605985103] [Citation(s) in RCA: 105] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
EBV is a paradigm for human tumor viruses because, although it infects most people benignly, it also can cause a variety of cancers. Both in vivo and in vitro, EBV infects B lymphocytes in G0, induces them to become blasts, and can maintain their proliferation in cell culture or in vivo as tumors. How EBV succeeds in these contrasting cellular environments in expressing its genes that control the host has not been explained. We have genetically dissected the EBV nuclear antigen 1 (EBNA1) gene that is required for replication of the viral genome, to elucidate its possible role in the transcription of viral genes. Strikingly, EBNA1 is essential to drive transcription of EBV's transforming genes after infection of primary B lymphocytes.
Collapse
Affiliation(s)
- Markus Altmann
- Department of Gene Vectors, GSF-National Research Center for Environment and Health, Marchioninistrasse 25, 81377 München, Germany; and
| | - Dagmar Pich
- Department of Gene Vectors, GSF-National Research Center for Environment and Health, Marchioninistrasse 25, 81377 München, Germany; and
| | - Romana Ruiss
- Department of Gene Vectors, GSF-National Research Center for Environment and Health, Marchioninistrasse 25, 81377 München, Germany; and
| | - Jindong Wang
- McArdle Laboratory for Cancer Research, University of Wisconsin, Madison, WI 53706
| | - Bill Sugden
- McArdle Laboratory for Cancer Research, University of Wisconsin, Madison, WI 53706
| | - Wolfgang Hammerschmidt
- Department of Gene Vectors, GSF-National Research Center for Environment and Health, Marchioninistrasse 25, 81377 München, Germany; and
- To whom correspondence should be addressed. E-mail:
| |
Collapse
|
47
|
Kameda T, Smuga-Otto K, Thomson JA. A severe de novo methylation of episomal vectors by human ES cells. Biochem Biophys Res Commun 2006; 349:1269-77. [PMID: 16973130 DOI: 10.1016/j.bbrc.2006.08.175] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2006] [Accepted: 08/29/2006] [Indexed: 11/29/2022]
Abstract
Episomal vectors can allow efficient genetic modification of cells and have the potential advantage of avoiding chromosomal position of integration effects. Here we explore the use of an Epstein-Barr virus-based episomal vector with human embryonic stem (ES) cells, and find high initial transfection rates, but a rapid loss of reporter gene expression. Similar to mouse ES cells, human ES cells express high levels of the de novo DNA methyltransferases, and we detected dramatic CpG methylation and minor non-CpG methylation on the episomes recovered from the human ES cells 7 days after the transfection, which was not present on the same episome recovered from 293 cells. Interestingly, the oriP region of the episomes was relatively excluded from this methylation. These findings define some of the limitations of using episomal vectors with human ES cells and offer a unique platform for analyzing epigenetic gene silencing in human ES cells.
Collapse
Affiliation(s)
- Takashi Kameda
- The Genome Center of Wisconsin, University of Wisconsin-Madison, 53706, USA
| | | | | |
Collapse
|
48
|
Howden SE, Wardan H, Voullaire L, McLenachan S, Williamson R, Ioannou P, Vadolas J. Chromatin-Binding Regions of EBNA1 Protein Facilitate the Enhanced Transfection of Epstein-Barr Virus-Based Vectors. Hum Gene Ther 2006. [DOI: 10.1089/hum.2006.17.ft-229] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
|
49
|
Heidel J, Mishra S, Davis ME. Molecular conjugates. ADVANCES IN BIOCHEMICAL ENGINEERING/BIOTECHNOLOGY 2006; 99:7-39. [PMID: 16568887 DOI: 10.1007/10_002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/08/2023]
Abstract
Molecular conjugates are nanometer-sized entities consisting of synthetic materials (lipids, polycations, targeting agents, and so on) and nucleic acids. These composites are delivery vehicles that function to provide the transport of nucleic acids to sites of action. Recently, great progress has been made in the construction of these nonviral delivery vehicles and the understanding of how they function in cells and animals. Here, we review some of the important issues in assembling molecular conjugates and understanding their behavior in biological fluids, cells, and animals. One of the largest challenges in the field of molecular conjugates is how to integrate the components into a workable system that exploits the combined attributes of the components without suffering losses due to the assembly of the system. We discuss some of the difficulties involved in the assembly of a functioning delivery system for in vivo use.
Collapse
Affiliation(s)
- Jeremy Heidel
- Chemical Engineering, 210-41, California Institute of Technology, Pasadena 91125, USA
| | | | | |
Collapse
|
50
|
van Gaal EVB, Hennink WE, Crommelin DJA, Mastrobattista E. Plasmid engineering for controlled and sustained gene expression for nonviral gene therapy. Pharm Res 2006; 23:1053-74. [PMID: 16715361 DOI: 10.1007/s11095-006-0164-2] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2005] [Accepted: 01/03/2006] [Indexed: 01/18/2023]
Abstract
Gene therapy requires the introduction of genetic material in diseased cells with the aim of treating or ultimately curing a disease. Since the start of gene therapy clinical trials in 1990, gene therapy has proven to be possible, but studies to date have highlighted the difficulty of achieving efficient, specific, and long-term transgene expression. Efforts to improve gene therapy strategies over the past years were mainly aimed at solving the problem of delivery, without paying much attention to the optimization of the expression cassette. With the current understanding of the eukaryotic transcription machinery and advanced molecular biology techniques at our disposition, it has now become possible to create custom-made transgene expression cassettes optimized for gene therapy applications. In this review, we will discuss several strategies that have been explored to improve the level and duration of transgene expression, to increase control over expression, or to restrict transgene expression to specific cell types or tissues. Although still in its infancy, such strategies will eventually lead to improvement of nonviral gene therapy and expansion of the range of possible therapeutic applications.
Collapse
Affiliation(s)
- Ethlinn V B van Gaal
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, P.O Box 80082, 3508 TB, Utrecht, The Netherlands
| | | | | | | |
Collapse
|