Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
Mol Ther ; 27(1): 178-187, 2019 01 02.
Article in English | MEDLINE | ID: mdl-30528089

ABSTRACT

Mucopolysaccharidosis type I (MPS I) is a severe disease due to deficiency of the lysosomal hydrolase α-L-iduronidase (IDUA) and the subsequent accumulation of the glycosaminoglycans (GAG), leading to progressive, systemic disease and a shortened lifespan. Current treatment options consist of hematopoietic stem cell transplantation, which carries significant mortality and morbidity risk, and enzyme replacement therapy, which requires lifelong infusions of replacement enzyme; neither provides adequate therapy, even in combination. A novel in vivo genome-editing approach is described in the murine model of Hurler syndrome. A corrective copy of the IDUA gene is inserted at the albumin locus in hepatocytes, leading to sustained enzyme expression, secretion from the liver into circulation, and subsequent uptake systemically at levels sufficient for correction of metabolic disease (GAG substrate accumulation) and prevention of neurobehavioral deficits in MPS I mice. This study serves as a proof-of-concept for this platform-based approach that should be broadly applicable to the treatment of a wide array of monogenic diseases.


Subject(s)
Gene Editing/methods , Genetic Therapy/methods , Mucopolysaccharidosis I/therapy , Zinc Finger Nucleases/metabolism , Animals , Disease Models, Animal , Enzyme Replacement Therapy , Female , Glycosaminoglycans/metabolism , Iduronidase/metabolism , Lysosomal Storage Diseases/drug therapy , Lysosomal Storage Diseases/metabolism , Lysosomal Storage Diseases/therapy , Male , Mice , Mucopolysaccharidosis I/drug therapy , Mucopolysaccharidosis I/metabolism , Zinc Finger Nucleases/genetics
2.
ACS Chem Biol ; 9(11): 2594-602, 2014 Nov 21.
Article in English | MEDLINE | ID: mdl-25188731

ABSTRACT

Copy number variation resulting in excess PMP22 protein causes the peripheral neuropathy Charcot-Marie-Tooth disease, type 1A. To broadly interrogate chemically sensitive transcriptional pathways controlling PMP22 protein levels, we used the targeting precision of TALEN-mediated genome editing to embed reporters within the genetic locus harboring the Peripheral Myelin Protein 22 (Pmp22) gene. Using a Schwann cell line with constitutively high endogenous levels of Pmp22, we obtained allelic insertion of secreted bioluminescent reporters with sufficient signal to enable a 1536-well assay. Our findings from the quantitative high-throughput screening (qHTS) of several thousand drugs and clinically investigated compounds using this assay design both overlapped and expanded results from a previous assay using a randomly inserted reporter gene controlled by a single regulatory element of the Pmp22 gene. A key difference was the identification of a kinase-controlled inhibitory pathway of Pmp22 transcription revealed by the activity of the Protein kinase C (PKC)-modulator bryostatin.


Subject(s)
Charcot-Marie-Tooth Disease/genetics , Genome, Human , High-Throughput Nucleotide Sequencing , Humans
3.
Biotechnol Bioeng ; 110(3): 871-80, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23042119

ABSTRACT

Targeted DNA integration is commonly used to eliminate position effects on transgene expression. Integration can be targeted to specific sites in the genome via both homology-based and homology-independent processes. Both pathways start the integration process with a site-specific break in the chromosome, typically from a zinc-finger nuclease (ZFN). We previously described an efficient homology-independent targeted integration technique that captures short (<100 bp) pieces of DNA at chromosomal breaks created by ZFNs. We show here that inclusion of a nuclease target site on the donor plasmid followed by in vivo nuclease cleavage of both the donor and the chromosome results in efficient integration of large, transgene-sized DNA molecules into the chromosomal double-strand break. Successful targeted integration via in vivo donor linearization is demonstrated at five distinct loci in two mammalian cell types, highlighting the generality of the approach. Finally, we show that CHO cells, a cell type recalcitrant to homology-based integration, are proficient at capture of in vivo-linearized transgene donors. Moreover, we demonstrate knockout of the hamster FUT8 gene via the simultaneous ZFN- or TALE nuclease-mediated integration of an antibody cassette. Our results enable efficient targeted transgene addition to cells and organisms that fare poorly with traditional homology-driven approaches.


Subject(s)
Chromosomes/metabolism , DNA, Circular/metabolism , Deoxyribonucleases/metabolism , Gene Targeting , Plasmids/metabolism , Recombination, Genetic , Transgenes , Animals , Cell Line , DNA, Circular/genetics , Humans , Mutagenesis, Insertional
5.
Proc Natl Acad Sci U S A ; 108(29): 12013-7, 2011 Jul 19.
Article in English | MEDLINE | ID: mdl-21730124

ABSTRACT

Zinc-finger nucleases (ZFNs) are powerful tools for producing gene knockouts (KOs) with high efficiency. Whereas ZFN-mediated gene disruption has been demonstrated in laboratory animals such as mice, rats, and fruit flies, ZFNs have not been used to disrupt an endogenous gene in any large domestic species. Here we used ZFNs to induce a biallelic knockout of the porcine α1,3-galactosyltransferase (GGTA1) gene. Primary porcine fibroblasts were treated with ZFNs designed against the region coding for the catalytic core of GGTA1, resulting in biallelic knockout of ∼1% of ZFN-treated cells. A galactose (Gal) epitope counter-selected population of these cells was used in somatic cell nuclear transfer (SCNT). Of the resulting six fetuses, all completely lacked Gal epitopes and were phenotypically indistinguishable from the starting donor cell population, illustrating that ZFN-mediated genetic modification did not interfere with the cloning process. Neither off-target cleavage events nor integration of the ZFN-coding plasmid was detected. The GGTA1-KO phenotype was confirmed by a complement lysis assay that demonstrated protection of GGTA1-KO fibroblasts relative to wild-type cells. Cells from GGTA1-KO fetuses and pooled, transfected cells were used to produce live offspring via SCNT. This study reports the production of cloned pigs carrying a biallelic ZFN-induced knockout of an endogenous gene. These findings open a unique avenue toward the creation of gene KO pigs, which could benefit both agriculture and biomedicine.


Subject(s)
Cloning, Organism/methods , Deoxyribonucleases/metabolism , Galactosyltransferases/genetics , Gene Knockout Techniques/methods , Sus scrofa/genetics , Animals , Base Sequence , DNA Primers/genetics , Fibroblasts , Flow Cytometry , Molecular Sequence Data , Nuclear Transfer Techniques , Sequence Analysis, DNA , Transplantation, Heterologous/methods
6.
Nat Biotechnol ; 29(8): 731-4, 2011 Jul 07.
Article in English | MEDLINE | ID: mdl-21738127

ABSTRACT

Targeted genetic engineering of human pluripotent cells is a prerequisite for exploiting their full potential. Such genetic manipulations can be achieved using site-specific nucleases. Here we engineered transcription activator-like effector nucleases (TALENs) for five distinct genomic loci. At all loci tested we obtained human embryonic stem cell (ESC) and induced pluripotent stem cell (iPSC) clones carrying transgenic cassettes solely at the TALEN-specified location. Our data suggest that TALENs employing the specific architectures described here mediate site-specific genome modification in human pluripotent cells with similar efficiency and precision as do zinc-finger nucleases (ZFNs).


Subject(s)
Embryonic Stem Cells/physiology , Endonucleases/metabolism , Gene Targeting/methods , Genetic Engineering/methods , Induced Pluripotent Stem Cells/physiology , Transcription Factors/metabolism , Base Sequence , Endonucleases/genetics , Homeodomain Proteins/genetics , Humans , Molecular Sequence Data , Myosin-Light-Chain Phosphatase/genetics , Octamer Transcription Factor-3/genetics , Transcription Factors/genetics , Zinc Fingers
7.
Nat Cell Biol ; 13(3): 331-7, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21297641

ABSTRACT

Clathrin-mediated endocytosis (CME) is the best-studied pathway by which cells selectively internalize molecules from the plasma membrane and surrounding environment. Previous live-cell imaging studies using ectopically overexpressed fluorescent fusions of endocytic proteins indicated that mammalian CME is a highly dynamic but inefficient and heterogeneous process. In contrast, studies of endocytosis in budding yeast using fluorescent protein fusions expressed at physiological levels from native genomic loci have revealed a process that is very regular and efficient. To analyse endocytic dynamics in mammalian cells in which endogenous protein stoichiometry is preserved, we targeted zinc finger nucleases (ZFNs) to the clathrin light chain A and dynamin-2 genomic loci and generated cell lines expressing fluorescent protein fusions from each locus. The genome-edited cells exhibited enhanced endocytic function, dynamics and efficiency when compared with previously studied cells, indicating that CME is highly sensitive to the levels of its protein components. Our study establishes that ZFN-mediated genome editing is a robust tool for expressing protein fusions at endogenous levels to faithfully report subcellular localization and dynamics.


Subject(s)
Clathrin/metabolism , Endocytosis , Animals , Base Sequence , Cell Lineage , Cell Membrane/metabolism , Dynamin II/metabolism , Genome , Green Fluorescent Proteins/metabolism , Humans , Microscopy, Fluorescence/methods , Models, Genetic , Molecular Sequence Data , Polymerase Chain Reaction
8.
Nucleic Acids Res ; 38(15): e152, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20530528

ABSTRACT

We previously demonstrated high-frequency, targeted DNA addition mediated by the homology-directed DNA repair pathway. This method uses a zinc-finger nuclease (ZFN) to create a site-specific double-strand break (DSB) that facilitates copying of genetic information into the chromosome from an exogenous donor molecule. Such donors typically contain two approximately 750 bp regions of chromosomal sequence required for homology-directed DNA repair. Here, we demonstrate that easily-generated linear donors with extremely short (50 bp) homology regions drive transgene integration into 5-10% of chromosomes. Moreover, we measure the overhangs produced by ZFN cleavage and find that oligonucleotide donors with single-stranded 5' overhangs complementary to those made by ZFNs are efficiently ligated in vivo to the DSB. Greater than 10% of all chromosomes directly incorporate this exogenous DNA via a process that is dependent upon and guided by complementary 5' overhangs on the donor DNA. Finally, we extend this non-homologous end-joining (NHEJ)-based technique by directly inserting donor DNA comprising recombinase sites into large deletions created by the simultaneous action of two separate ZFN pairs. Up to 50% of deletions contained a donor insertion. Targeted DNA addition via NHEJ complements our homology-directed targeted integration approaches, adding versatility to the manipulation of mammalian genomes.


Subject(s)
Deoxyribonucleases, Type II Site-Specific/metabolism , Gene Targeting/methods , Zinc Fingers , Animals , CHO Cells , Chromosomes, Mammalian/chemistry , Cricetinae , Cricetulus , DNA/chemistry , DNA Breaks, Double-Stranded , Deoxyribonucleases, Type II Site-Specific/chemistry , Genome , Humans , K562 Cells , Sequence Homology, Nucleic Acid
9.
Biotechnol Bioeng ; 105(2): 330-40, 2010 Feb 01.
Article in English | MEDLINE | ID: mdl-19777580

ABSTRACT

Anoxic and metabolic stresses in large-scale cell culture during biopharmaceutical production can induce apoptosis. Strategies designed to ameliorate the problem of apoptosis in cell culture have focused on mRNA knockdown of pro-apoptotic proteins and over-expression of anti-apoptotic ones. Apoptosis in cell culture involves mitochondrial permeabilization by the pro-apoptotic Bak and Bax proteins; activity of either protein is sufficient to permit apoptosis. We demonstrate here the complete and permanent elimination of both the Bak and Bax proteins in combination in Chinese hamster ovary (CHO) cells using zinc-finger nuclease-mediated gene disruption. Zinc-finger nuclease cleavage of BAX and BAK followed by inaccurate DNA repair resulted in knockout of both genes. Cells lacking Bax and Bak grow normally but fail to activate caspases in response to apoptotic stimuli. When grown using scale-down systems under conditions that mimic growth in large-scale bioreactors they are significantly more resistant to apoptosis induced by starvation, staurosporine, and sodium butyrate. When grown under starvation conditions, BAX- and BAK-deleted cells produce two- to fivefold more IgG than wild-type CHO cells. Under normal growth conditions in suspension culture in shake flasks, double-knockout cultures achieve equal or higher cell densities than unmodified wild-type cultures and reach viable cell densities relevant for large-scale industrial protein production.


Subject(s)
Apoptosis , Biotechnology/methods , Cricetulus/genetics , bcl-2-Associated X Protein/genetics , Animals , Base Sequence , CHO Cells , Cloning, Molecular , Cricetinae , Female , Gene Deletion , Immunoglobulin G/biosynthesis , Molecular Sequence Data , Ovary/cytology , Zinc Fingers
10.
Proc Natl Acad Sci U S A ; 105(15): 5809-14, 2008 Apr 15.
Article in English | MEDLINE | ID: mdl-18359850

ABSTRACT

Gene knockout is the most powerful tool for determining gene function or permanently modifying the phenotypic characteristics of a cell. Existing methods for gene disruption are limited by their efficiency, time to completion, and/or the potential for confounding off-target effects. Here, we demonstrate a rapid single-step approach to targeted gene knockout in mammalian cells, using engineered zinc-finger nucleases (ZFNs). ZFNs can be designed to target a chosen locus with high specificity. Upon transient expression of these nucleases the target gene is first cleaved by the ZFNs and then repaired by a natural-but imperfect-DNA repair process, nonhomologous end joining. This often results in the generation of mutant (null) alleles. As proof of concept for this approach we designed ZFNs to target the dihydrofolate reductase (DHFR) gene in a Chinese hamster ovary (CHO) cell line. We observed biallelic gene disruption at frequencies >1%, thus obviating the need for selection markers. Three new genetically distinct DHFR(-/-) cell lines were generated. Each new line exhibited growth and functional properties consistent with the specific knockout of the DHFR gene. Importantly, target gene disruption is complete within 2-3 days of transient ZFN delivery, thus enabling the isolation of the resultant DHFR(-/-) cell lines within 1 month. These data demonstrate further the utility of ZFNs for rapid mammalian cell line engineering and establish a new method for gene knockout with application to reverse genetics, functional genomics, drug discovery, and therapeutic recombinant protein production.


Subject(s)
Deoxyribonucleases/metabolism , Gene Deletion , Genetic Techniques , Animals , Cell Line , Gene Silencing , Methods , Mutagenesis, Site-Directed , Protein Engineering , Tetrahydrofolate Dehydrogenase/deficiency , Tetrahydrofolate Dehydrogenase/genetics , Zinc Fingers
SELECTION OF CITATIONS
SEARCH DETAIL
...