Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
Nucleic Acids Res ; 45(1): 353-366, 2017 Jan 09.
Article in English | MEDLINE | ID: mdl-27899587

ABSTRACT

Transposons are highly abundant in eukaryotic genomes, but their mobilization must be finely tuned to maintain host organism fitness and allow for transposon propagation. Forty percent of the human genome is comprised of transposable element sequences, and the most abundant cut-and-paste transposons are from the hAT superfamily. We found that the hAT transposase TcBuster from Tribolium castaneum formed filamentous structures, or rodlets, in human tissue culture cells, after gene transfer to adult mice, and ex vivo in cell-free conditions, indicating that host co-factors or cellular structures were not required for rodlet formation. Time-lapsed imaging of GFP-laced rodlets in human cells revealed that they formed quickly in a dynamic process involving fusion and fission. We delayed the availability of the transposon DNA and found that transposition declined after transposase concentrations became high enough for visible transposase rodlets to appear. In combination with earlier findings for maize Ac elements, these results give insight into transposase overproduction inhibition by demonstrating that the appearance of transposase protein structures and the end of active transposition are simultaneous, an effect with implications for genetic engineering and horizontal gene transfer.


Subject(s)
DNA Transposable Elements , Gene Expression Regulation , Insect Proteins/genetics , Transposases/genetics , Animals , Female , Genes, Reporter , HEK293 Cells , HeLa Cells , Humans , Insect Proteins/metabolism , Luciferases/genetics , Luciferases/metabolism , Male , Mice , Optical Imaging , Time-Lapse Imaging , Transposases/metabolism , Tribolium
2.
PLoS One ; 7(11): e42666, 2012.
Article in English | MEDLINE | ID: mdl-23166581

ABSTRACT

BACKGROUND: Transposons are useful tools for creating transgenic organisms, insertional mutagenesis, and genome engineering. TcBuster, a novel hAT-family transposon system derived from the red flour beetle Tribolium castaneum, was shown to be highly active in previous studies in insect embryoes. METHODOLOGY/PRINCIPAL FINDINGS: We tested TcBuster for its activity in human embryonic kidney 293 (HEK-293) cells. Excision footprints obtained from HEK-293 cells contained small insertions and deletions consistent with a hAT-type repair mechanism of hairpin formation and non-homologous end-joining. Genome-wide analysis of 23,417 piggyBac, 30,303 Sleeping Beauty, and 27,985 TcBuster integrations in HEK-293 cells revealed a uniquely different integration pattern when compared to other transposon systems with regards to genomic elements. TcBuster experimental conditions were optimized to assay TcBuster activity in HEK-293 cells by colony assay selection for a neomycin-containing transposon. Increasing transposon plasmid increased the number of colonies, whereas gene transfer activity dependent on codon-optimized transposase plasmid peaked at 100 ng with decreased colonies at the highest doses of transposase DNA. Expression of the related human proteins Buster1, Buster3, and SCAND3 in HEK-293 cells did not result in genomic integration of the TcBuster transposon. TcBuster, Tol2, and piggyBac were compared directly at different ratios of transposon to transposase and found to be approximately comparable while having their own ratio preferences. CONCLUSIONS/SIGNIFICANCE: TcBuster was found to be highly active in mammalian HEK-293 cells and represents a promising tool for mammalian genome engineering.


Subject(s)
DNA Transposable Elements/genetics , Gene Transfer Techniques , Genetic Engineering/methods , Mutagenesis, Insertional/methods , Tribolium/genetics , Animals , Base Sequence , DNA Primers/genetics , HEK293 Cells , Humans , INDEL Mutation/genetics , Molecular Sequence Data , Plasmids/genetics , Transposases/metabolism
3.
Mol Ther ; 19(9): 1636-44, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21730970

ABSTRACT

The ability to direct gene delivery to a user-defined chromosomal location would greatly improve gene transfer applications. The piggyBac transposon system is a nonviral gene transfer system proven effective in a variety of cells and tissues, including human cells. We fused a highly site-specific synthetic zinc-finger DNA-binding domain (ZFP) to the N-terminus of the piggyBac transposase and evaluated site-directed genomic integration in human cells. Chimeric ZFP-piggyBac transposase exhibited robust gene transfer activity, targeted binding to a cognate endogenous chromosomal ZFP site in the human genome, and site-directed transposon integration into an episomal plasmid target containing a single ZFP site in human cells. We evaluated the ability of ZFP-piggyBac to direct gene integration into an engineered chromosomal ZFP target site in the human genome and consistently observed a higher degree of ZFP-piggyBac site-directed genomic integration when compared to native piggyBac. Chromatin immunoprecipitation (ChIP) experiments revealed binding of native piggyBac to our engineered TTAA-containing chromosomal target which supported integration, but not a TTAA-deficient chromosomal target which lacked integration. Our results offer insight into the requirements for using a chimeric zinc finger-piggyBac transposase to direct integration into a user-defined chromosomal location.


Subject(s)
Chromosomes, Human/genetics , DNA Transposable Elements , DNA-Binding Proteins/genetics , Genetic Engineering/methods , Blotting, Western , Chromatin Immunoprecipitation/methods , DNA-Binding Proteins/metabolism , Genetic Vectors , Genome, Human , HEK293 Cells , Humans , Mutagenesis, Site-Directed , Plasmids , Real-Time Polymerase Chain Reaction , Transfection , Transposases/genetics , Transposases/metabolism , Zinc Fingers
4.
Proc Natl Acad Sci U S A ; 107(4): 1343-8, 2010 Jan 26.
Article in English | MEDLINE | ID: mdl-20080581

ABSTRACT

Generation of cultured human cells stably expressing one or more recombinant gene sequences is a widely used approach in biomedical research, biotechnology, and drug development. Conventional methods are not efficient and have severe limitations especially when engineering cells to coexpress multiple transgenes or multiprotein complexes. In this report, we harnessed the highly efficient, nonviral, and plasmid-based piggyBac transposon system to enable concurrent genomic integration of multiple independent transposons harboring distinct protein-coding DNA sequences. Flow cytometry of cell clones derived from a single multiplexed transfection demonstrated approximately 60% (three transposons) or approximately 30% (four transposons) stable coexpression of all delivered transgenes with selection for a single marker transposon. We validated multiplexed piggyBac transposon delivery by coexpressing large transgenes encoding a multisubunit neuronal voltage-gated sodium channel (SCN1A) containing a pore-forming subunit and two accessory subunits while using two additional genes for selection. Previously unobtainable robust sodium current was demonstrated through 38 passages, suitable for use on an automated high-throughput electrophysiology platform. Cotransfection of three large (up to 10.8 kb) piggyBac transposons generated a heterozygous SCN1A stable cell line expressing two separate alleles of the pore-forming subunit and two accessory subunits (total of four sodium channel subunits) with robust functional expression. We conclude that the piggyBac transposon system can be used to perform multiplexed stable gene transfer in cultured human cells, and this technology may be valuable for applications requiring concurrent expression of multiprotein complexes.


Subject(s)
DNA Transposable Elements , Transgenes , Cell Line , Gene Expression , Genetic Markers , Humans , NAV1.1 Voltage-Gated Sodium Channel , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Open Reading Frames , Protein Binding , Sodium Channels/genetics , Sodium Channels/metabolism
5.
Mol Ther ; 17(12): 2115-20, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19809403

ABSTRACT

Somatic cell gene transfer has permitted inducible gene expression in vivo through coinfection of multiple viruses. We hypothesized that the highly efficient plasmid-based piggyBac transposon system would enable long-term inducible gene expression in mice in vivo. We used a multiple-transposon delivery strategy to create a tetracycline-inducible expression system in vitro in human cells by delivering the two genes on separate transposons for inducible reporter gene expression along with a separate selectable transposon marker. Evaluation of stable cell lines revealed 100% of selected clones exhibited inducible expression via stable expression from three separate transposons simultaneously. We next tested and found that piggyBac-mediated gene transfer to liver or lung could achieve stable reporter gene expression in mice in vivo in either immunocompetent or immune deficient animals. A single injection of piggyBac transposons could achieve long-term inducible gene expression in the livers of mice in vivo, confirming our multiple-transposon strategy used in cultured cells. The plasmid-based piggyBac transposon system enables constitutive or inducible gene expression in vivo for potential therapeutic and biological applications without using viral vectors.


Subject(s)
DNA Transposable Elements/genetics , Gene Transfer Techniques , Genetic Vectors/administration & dosage , Green Fluorescent Proteins/metabolism , Transgenes/genetics , Animals , Blotting, Southern , Cells, Cultured , Female , Green Fluorescent Proteins/genetics , Humans , Kidney/cytology , Kidney/metabolism , Liver/cytology , Liver/metabolism , Liver/virology , Lung/cytology , Lung/metabolism , Lung/virology , Mice , Mice, Inbred C57BL , Mice, SCID , Virus Integration
6.
J Immunother ; 32(8): 837-44, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19752750

ABSTRACT

The piggyBac transposon system represents a promising nonviral tool for gene delivery and discovery, and may also be of value for clinical gene therapy. PiggyBac is a highly efficient integrating vector that stably transfects (approximately 40%) of primary human T cells for potential adoptive immunotherapy applications. To evaluate the potential genotoxicity of piggyBac, we compared 228 integration sites in primary human T cells to integrations in 2 other human-derived cell lines (HEK293 and HeLa) and randomly simulated integrations into the human genome. Our results revealed distinct differences between cell types. PiggyBac had a nonrandom integration profile and a preference for transcriptional units (approximately 50% into RefSeq genes in all cell types), CpG islands (18% in T cells and 8% in other human cells), and transcriptional start sites (<5 kb, 16% to 20% in all cell types). PiggyBac also preferred TTAA but not AT-rich regions of the human genome. We evaluated the expression of mapped genes into which piggyBac integrated, and found selection of more active genes in primary human T cells compared with other human cell types, possibly due to concomitant T-cell activation during transposition. Importantly, we found that in comparison to what has been reported for gammaretroviral and human lenitviral vectors, piggyBac had decreased integration frequency into or within 50 kb of the transcriptional start sites of known proto-oncogenes. Hence the piggyBac nonviral gene delivery system seems to represent a promising gene transfer system for clinical applications using human T lymphocytes.


Subject(s)
DNA Transposable Elements , Genetic Vectors , Immunotherapy, Adoptive , T-Lymphocytes/metabolism , CpG Islands/genetics , Gammaretrovirus , Genome-Wide Association Study , HeLa Cells , Humans , Lentivirus , Mutagenicity Tests , T-Lymphocytes/immunology , Transcription Initiation Site
SELECTION OF CITATIONS
SEARCH DETAIL
...