Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 47
Filter
1.
Cell ; 140(5): 678-91, 2010 Mar 05.
Article in English | MEDLINE | ID: mdl-20211137

ABSTRACT

The incorporation of histone H3 variants has been implicated in the epigenetic memory of cellular state. Using genome editing with zinc-finger nucleases to tag endogenous H3.3, we report genome-wide profiles of H3 variants in mammalian embryonic stem cells and neuronal precursor cells. Genome-wide patterns of H3.3 are dependent on amino acid sequence and change with cellular differentiation at developmentally regulated loci. The H3.3 chaperone Hira is required for H3.3 enrichment at active and repressed genes. Strikingly, Hira is not essential for localization of H3.3 at telomeres and many transcription factor binding sites. Immunoaffinity purification and mass spectrometry reveal that the proteins Atrx and Daxx associate with H3.3 in a Hira-independent manner. Atrx is required for Hira-independent localization of H3.3 at telomeres and for the repression of telomeric RNA. Our data demonstrate that multiple and distinct factors are responsible for H3.3 localization at specific genomic locations in mammalian cells.


Subject(s)
Histones/analysis , Telomere/chemistry , Animals , Binding Sites , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Embryonic Stem Cells/metabolism , Genome , Histone Chaperones/genetics , Histone Chaperones/metabolism , Histones/genetics , Histones/metabolism , Mice , Mice, Inbred C57BL , Telomere/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , Transcription Initiation Site
2.
Proc Biol Sci ; 287(1934): 20201894, 2020 09 09.
Article in English | MEDLINE | ID: mdl-32900312

ABSTRACT

Some camouflaged animals hide colour signals and display them only transiently. These hidden colour signals are often conspicuous and are used as a secondary defence to warn or startle predators (deimatic displays) and/or to confuse them (flash displays). The hidden signals used in these displays frequently resemble typical aposematic signals, so it is possible that prey with hidden signals have evolved to employ colour patterns of a form that predators have previously learned to associate with unprofitability. Here, we tested this hypothesis by conducting two experiments that examined the effect of predator avoidance learning on the efficacy of deimatic and flash displays. We found that the survival benefits of both deimatic and flash displays were substantially higher against predators that had previously learned to associate the hidden colours with unprofitability than against naive predators. These findings help explain the phenological patterns we found in 1568 macro-lepidopteran species on three continents: species with hidden signals tend to occur later in the season than species without hidden signals.


Subject(s)
Avoidance Learning , Pigmentation , Predatory Behavior , Animals , Color , Cues , Seasons
3.
Mol Ther ; 27(4): 866-877, 2019 04 10.
Article in English | MEDLINE | ID: mdl-30902585

ABSTRACT

It has previously been shown that engineered zinc finger nucleases (ZFNs) can be packaged into adeno-associated viruses (AAVs) and delivered intravenously into mice, non-human primates, and most recently, humans to induce highly efficient therapeutic genome editing in the liver. Lipid nanoparticles (LNPs) are synthetic delivery vehicles that enable repeat administration and are not limited by the presence of preexisting neutralizing antibodies in patients. Here, we show that mRNA encoding ZFNs formulated into LNP can enable >90% knockout of gene expression in mice by targeting the TTR or PCSK9 gene, at mRNA doses 10-fold lower than has ever been reported. Additionally, co-delivering mRNA-LNP containing ZFNs targeted to intron 1 of the ALB locus with AAV packaged with a promoterless human IDS or FIX therapeutic transgene can result in high levels of targeted integration and subsequent therapeutically relevant levels of protein expression in mice. Finally, we show repeat administration of ZFN mRNA-LNP after a single AAV donor dose results in significantly increased levels of genome editing and transgene expression compared to a single dose. These results demonstrate LNP-mediated ZFN mRNA delivery can drive highly efficient levels of in vivo genome editing and can potentially offer a new treatment modality for a variety of diseases.


Subject(s)
Drug Delivery Systems/methods , Gene Editing/methods , Nanoparticles/administration & dosage , RNA, Messenger/administration & dosage , Zinc Finger Nucleases/administration & dosage , Animals , Cells, Cultured , Dependovirus/genetics , Female , Gene Knockout Techniques , Genetic Vectors , Hepatocytes/metabolism , Introns/genetics , Lipids/chemistry , Male , Mice , Mice, Inbred C57BL , Prealbumin/genetics , Proprotein Convertase 9/genetics , RNA, Messenger/genetics , Transgenes/genetics , Zinc Finger Nucleases/pharmacology
4.
Nucleic Acids Res ; 46(10): 4845-4871, 2018 06 01.
Article in English | MEDLINE | ID: mdl-29718463

ABSTRACT

Protein engineering is used to generate novel protein folds and assemblages, to impart new properties and functions onto existing proteins, and to enhance our understanding of principles that govern protein structure. While such approaches can be employed to reprogram protein-protein interactions, modifying protein-DNA interactions is more difficult. This may be related to the structural features of protein-DNA interfaces, which display more charged groups, directional hydrogen bonds, ordered solvent molecules and counterions than comparable protein interfaces. Nevertheless, progress has been made in the redesign of protein-DNA specificity, much of it driven by the development of engineered enzymes for genome modification. Here, we summarize the creation of novel DNA specificities for zinc finger proteins, meganucleases, TAL effectors, recombinases and restriction endonucleases. The ease of re-engineering each system is related both to the modularity of the protein and the extent to which the proteins have evolved to be capable of readily modifying their recognition specificities in response to natural selection. The development of engineered DNA binding proteins that display an ideal combination of activity, specificity, deliverability, and outcomes is not a fully solved problem, however each of the current platforms offers unique advantages, offset by behaviors and properties requiring further study and development.


Subject(s)
DNA-Binding Proteins/metabolism , DNA/metabolism , Protein Engineering/methods , Recombinant Proteins/metabolism , Base Pairing , DNA/chemistry , DNA Cleavage , DNA Restriction Enzymes/chemistry , DNA Restriction Enzymes/genetics , DNA Restriction Enzymes/metabolism , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/genetics , Deoxyribonucleases/chemistry , Deoxyribonucleases/genetics , Deoxyribonucleases/metabolism , Gene Editing , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinases/chemistry , Recombinases/genetics , Recombinases/metabolism , Transcription Activator-Like Effectors/chemistry , Transcription Activator-Like Effectors/genetics , Transcription Activator-Like Effectors/metabolism , Zinc Fingers
6.
Nat Methods ; 12(5): 465-71, 2015 May.
Article in English | MEDLINE | ID: mdl-25799440

ABSTRACT

Transcription activator-like effector (TALE) proteins have gained broad appeal as a platform for targeted DNA recognition, largely owing to their simple rules for design. These rules relate the base specified by a single TALE repeat to the identity of two key residues (the repeat variable diresidue, or RVD) and enable design for new sequence targets via modular shuffling of these units. A key limitation of these rules is that their simplicity precludes options for improving designs that are insufficiently active or specific. Here we address this limitation by developing an expanded set of RVDs and applying them to improve the performance of previously described TALEs. As an extreme example, total conversion of a TALE nuclease to new RVDs substantially reduced off-target cleavage in cellular studies. By providing new RVDs and design strategies, these studies establish options for developing improved TALEs for broader application across medicine and biotechnology.


Subject(s)
Gene Expression Regulation/physiology , Genome , RNA Editing/physiology , Transcription Factors/metabolism , Animals , Base Sequence , DNA/genetics , Enzyme-Linked Immunosorbent Assay , Genetic Markers , Transcription Factors/genetics
7.
Blood ; 126(15): 1777-84, 2015 Oct 08.
Article in English | MEDLINE | ID: mdl-26297739

ABSTRACT

Site-specific genome editing provides a promising approach for achieving long-term, stable therapeutic gene expression. Genome editing has been successfully applied in a variety of preclinical models, generally focused on targeting the diseased locus itself; however, limited targeting efficiency or insufficient expression from the endogenous promoter may impede the translation of these approaches, particularly if the desired editing event does not confer a selective growth advantage. Here we report a general strategy for liver-directed protein replacement therapies that addresses these issues: zinc finger nuclease (ZFN) -mediated site-specific integration of therapeutic transgenes within the albumin gene. By using adeno-associated viral (AAV) vector delivery in vivo, we achieved long-term expression of human factors VIII and IX (hFVIII and hFIX) in mouse models of hemophilia A and B at therapeutic levels. By using the same targeting reagents in wild-type mice, lysosomal enzymes were expressed that are deficient in Fabry and Gaucher diseases and in Hurler and Hunter syndromes. The establishment of a universal nuclease-based platform for secreted protein production would represent a critical advance in the development of safe, permanent, and functional cures for diverse genetic and nongenetic diseases.


Subject(s)
Albumins/genetics , Enzyme Replacement Therapy , Genetic Therapy , Genome , Liver/metabolism , Transgenes/physiology , Albumins/metabolism , Animals , Dependovirus/genetics , Endonucleases , Fabry Disease/genetics , Fabry Disease/therapy , Factor IX/genetics , Factor VIII/genetics , Gaucher Disease/genetics , Gaucher Disease/therapy , Genetic Vectors/administration & dosage , Hemophilia A/genetics , Hemophilia A/therapy , Hemophilia B/genetics , Hemophilia B/therapy , High-Throughput Nucleotide Sequencing , Humans , Lysosomes/enzymology , Mice , Mice, Inbred C57BL , Mucopolysaccharidosis I/genetics , Mucopolysaccharidosis I/therapy , Mucopolysaccharidosis II/genetics , Mucopolysaccharidosis II/therapy , Promoter Regions, Genetic/genetics , RNA Editing , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Zinc Fingers
8.
Genome Res ; 22(7): 1316-26, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22434427

ABSTRACT

Zinc-finger nucleases (ZFNs) drive highly efficient genome editing by generating a site-specific DNA double-strand break (DSB) at a predetermined site in the genome. Subsequent repair of this break via the nonhomologous end-joining (NHEJ) or homology-directed repair (HDR) pathways results in targeted gene disruption or gene addition, respectively. Here, we report that ZFNs can be engineered to induce a site-specific DNA single-strand break (SSB) or nick. Using the CCR5-specific ZFNs as a model system, we show that introduction of a nick at this target site stimulates gene addition using a homologous donor template but fails to induce significant levels of the small insertions and deletions (indels) characteristic of repair via NHEJ. Gene addition by these CCR5-targeted zinc finger nickases (ZFNickases) occurs in both transformed and primary human cells at efficiencies of up to ∼1%-8%. Interestingly, ZFNickases targeting the AAVS1 "safe harbor" locus revealed similar in vitro nicking activity, a marked reduction of indels characteristic of NHEJ, but stimulated far lower levels of gene addition-suggesting that other, yet to be identified mediators of nick-induced gene targeting exist. Introduction of site-specific nicks at distinct endogenous loci provide an important tool for the study of DNA repair. Moreover, the potential for a SSB to direct repair pathway choice (i.e., HDR but not NHEJ) may prove advantageous for certain therapeutic applications such as the targeted correction of human disease-causing mutations.


Subject(s)
Deoxyribonucleases, Type II Site-Specific/metabolism , Gene Targeting/methods , Genome, Human , Recombinant Fusion Proteins/metabolism , Zinc Fingers , Amino Acid Sequence , Catalytic Domain , Cell Line, Transformed , Cell Line, Tumor , Cloning, Molecular , DNA Breaks, Double-Stranded , DNA Breaks, Single-Stranded , DNA End-Joining Repair , Deoxyribonucleases, Type II Site-Specific/genetics , Fibroblasts/cytology , Fibroblasts/metabolism , Genetic Vectors , Histones/metabolism , Humans , INDEL Mutation , Molecular Sequence Data , Protein Engineering/methods , Receptors, CCR5/genetics , Recombinant Fusion Proteins/genetics , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Transformation, Genetic
9.
Blood ; 122(19): 3283-7, 2013 Nov 07.
Article in English | MEDLINE | ID: mdl-24085764

ABSTRACT

Monogenic diseases, including hemophilia, represent ideal targets for genome-editing approaches aimed at correcting a defective gene. Here we report that systemic adeno-associated virus (AAV) vector delivery of zinc finger nucleases (ZFNs) and corrective donor template to the predominantly quiescent livers of adult mice enables production of high levels of human factor IX in a murine model of hemophilia B. Further, we show that off-target cleavage can be substantially reduced while maintaining robust editing by using obligate heterodimeric ZFNs engineered to minimize unwanted cleavage attributable to homodimerization of the ZFNs. These results broaden the therapeutic potential of AAV/ZFN-mediated genome editing in the liver and could expand this strategy to other nonreplicating cell types.


Subject(s)
Endonucleases/genetics , Factor IX/biosynthesis , Genetic Therapy/methods , Genetic Vectors , Genome , Hemophilia B/therapy , Zinc Fingers/genetics , Animals , Dependovirus/genetics , Disease Models, Animal , Endonucleases/metabolism , Factor IX/genetics , Factor IX/metabolism , Hemophilia B/genetics , Hemophilia B/pathology , Liver/metabolism , Male , Mice , Mice, Transgenic , Protein Multimerization
10.
Blood ; 122(8): 1341-9, 2013 Aug 22.
Article in English | MEDLINE | ID: mdl-23741009

ABSTRACT

Long-term engraftment of allogeneic cells necessitates eluding immune-mediated rejection, which is currently achieved by matching for human leukocyte antigen (HLA) expression, immunosuppression, and/or delivery of donor-derived cells to sanctuary sites. Genetic engineering provides an alternative approach to avoid clearance of cells that are recognized as "non-self" by the recipient. To this end, we developed designer zinc finger nucleases and employed a "hit-and-run" approach to genetic editing for selective elimination of HLA expression. Electro-transfer of mRNA species coding for these engineered nucleases completely disrupted expression of HLA-A on human T cells, including CD19-specific T cells. The HLA-A(neg) T-cell pools can be enriched and evade lysis by HLA-restricted cytotoxic T-cell clones. Recognition by natural killer cells of cells that had lost HLA expression was circumvented by enforced expression of nonclassical HLA molecules. Furthermore, we demonstrate that zinc finger nucleases can eliminate HLA-A expression from embryonic stem cells, which broadens the applicability of this strategy beyond infusing HLA-disparate immune cells. These findings establish that clinically appealing cell types derived from donors with disparate HLA expression can be genetically edited to evade an immune response and provide a foundation whereby cells from a single donor can be administered to multiple recipients.


Subject(s)
Deoxyribonucleases/genetics , Histocompatibility Antigens Class I/metabolism , Stem Cell Transplantation/methods , Transplantation, Homologous , Antigens, CD19/metabolism , Base Sequence , Cell Differentiation , Cytotoxicity, Immunologic/immunology , Electroporation , Embryonic Stem Cells/cytology , Gene Transfer Techniques , HEK293 Cells , Humans , Leukocytes, Mononuclear/cytology , Molecular Sequence Data , Protein Engineering , T-Lymphocytes/immunology , Zinc Fingers
11.
Nature ; 459(7245): 437-41, 2009 May 21.
Article in English | MEDLINE | ID: mdl-19404259

ABSTRACT

Agricultural biotechnology is limited by the inefficiencies of conventional random mutagenesis and transgenesis. Because targeted genome modification in plants has been intractable, plant trait engineering remains a laborious, time-consuming and unpredictable undertaking. Here we report a broadly applicable, versatile solution to this problem: the use of designed zinc-finger nucleases (ZFNs) that induce a double-stranded break at their target locus. We describe the use of ZFNs to modify endogenous loci in plants of the crop species Zea mays. We show that simultaneous expression of ZFNs and delivery of a simple heterologous donor molecule leads to precise targeted addition of an herbicide-tolerance gene at the intended locus in a significant number of isolated events. ZFN-modified maize plants faithfully transmit these genetic changes to the next generation. Insertional disruption of one target locus, IPK1, results in both herbicide tolerance and the expected alteration of the inositol phosphate profile in developing seeds. ZFNs can be used in any plant species amenable to DNA delivery; our results therefore establish a new strategy for plant genetic manipulation in basic science and agricultural applications.


Subject(s)
Biotechnology/methods , Deoxyribonucleases/chemistry , Deoxyribonucleases/metabolism , Gene Targeting/methods , Genome, Plant/genetics , Zea mays/genetics , Zinc Fingers , Deoxyribonucleases/genetics , Food, Genetically Modified , Genes, Plant/genetics , Herbicide Resistance/genetics , Herbicides/pharmacology , Heredity , Inositol Phosphates/metabolism , Mutagenesis, Site-Directed/methods , Plants, Genetically Modified , Recombination, Genetic/genetics , Reproducibility of Results
12.
Nat Methods ; 8(1): 74-9, 2011 Jan.
Article in English | MEDLINE | ID: mdl-21131970

ABSTRACT

Zinc-finger nucleases (ZFNs) drive efficient genome editing by introducing a double-strand break into the targeted gene. Cleavage is induced when two custom-designed ZFNs heterodimerize upon binding DNA to form a catalytically active nuclease complex. The importance of this dimerization event for subsequent cleavage activity has stimulated efforts to engineer the nuclease interface to prevent undesired homodimerization. Here we report the development and application of a yeast-based selection system designed to functionally interrogate the ZFN dimer interface. We identified critical residues involved in dimerization through the isolation of cold-sensitive nuclease domains. We used these residues to engineer ZFNs that have superior cleavage activity while suppressing homodimerization. The improvements were portable to orthogonal domains, allowing the concomitant and independent cleavage of two loci using two different ZFN pairs. These ZFN architectures provide a general means for obtaining highly efficient and specific genome modification.


Subject(s)
Endonucleases/metabolism , Zinc Fingers/physiology , DNA/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Dimerization , Endonucleases/genetics , Genome , Saccharomyces cerevisiae/enzymology , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Zinc Fingers/genetics
13.
Blood ; 119(24): 5697-705, 2012 Jun 14.
Article in English | MEDLINE | ID: mdl-22535661

ABSTRACT

Clinical-grade T cells are genetically modified ex vivo to express a chimeric antigen receptor (CAR) to redirect specificity to a tumor associated antigen (TAA) thereby conferring antitumor activity in vivo. T cells expressing a CD19-specific CAR recognize B-cell malignancies in multiple recipients independent of major histocompatibility complex (MHC) because the specificity domains are cloned from the variable chains of a CD19 monoclonal antibody. We now report a major step toward eliminating the need to generate patient-specific T cells by generating universal allogeneic TAA-specific T cells from one donor that might be administered to multiple recipients. This was achieved by genetically editing CD19-specific CAR(+) T cells to eliminate expression of the endogenous αß T-cell receptor (TCR) to prevent a graft-versus-host response without compromising CAR-dependent effector functions. Genetically modified T cells were generated using the Sleeping Beauty system to stably introduce the CD19-specific CAR with subsequent permanent deletion of α or ß TCR chains with designer zinc finger nucleases. We show that these engineered T cells display the expected property of having redirected specificity for CD19 without responding to TCR stimulation. CAR(+)TCR(neg) T cells of this type may potentially have efficacy as an off-the-shelf therapy for investigational treatment of B-lineage malignancies.


Subject(s)
Antigens, CD19/immunology , Epitopes/immunology , Genetic Engineering , Immunotherapy/methods , Receptors, Antigen, T-Cell/immunology , Recombinant Proteins/immunology , T-Lymphocytes/immunology , Adult , Antigen-Presenting Cells/immunology , Antigens, Neoplasm/immunology , CD28 Antigens/metabolism , CD3 Complex/metabolism , Cells, Cultured , Endonucleases/metabolism , Gene Knockout Techniques , Humans , K562 Cells , Lymphocyte Activation/immunology , Zinc Fingers
14.
Proc Natl Acad Sci U S A ; 108(17): 7052-7, 2011 Apr 26.
Article in English | MEDLINE | ID: mdl-21471457

ABSTRACT

The frog Xenopus, an important research organism in cell and developmental biology, currently lacks tools for targeted mutagenesis. Here, we address this problem by genome editing with zinc-finger nucleases (ZFNs). ZFNs directed against an eGFP transgene in Xenopus tropicalis induced mutations consistent with nonhomologous end joining at the target site, resulting in mosaic loss of the fluorescence phenotype at high frequencies. ZFNs directed against the noggin gene produced tadpoles and adult animals carrying up to 47% disrupted alleles, and founder animals yielded progeny carrying insertions and deletions in the noggin gene with no indication of off-target effects. Furthermore, functional tests demonstrated an allelic series of activity between three germ-line mutant alleles. Because ZFNs can be designed against any locus, our data provide a generally applicable protocol for gene disruption in Xenopus.


Subject(s)
Alleles , Carrier Proteins/genetics , Deoxyribonucleases/genetics , Gene Targeting/methods , Xenopus Proteins/genetics , Animals , Animals, Genetically Modified , Carrier Proteins/metabolism , Deoxyribonucleases/metabolism , Xenopus , Xenopus Proteins/metabolism , Zinc Fingers
15.
Nat Commun ; 15(1): 1181, 2024 Feb 15.
Article in English | MEDLINE | ID: mdl-38360922

ABSTRACT

Nucleobase editors represent an emerging technology that enables precise single-base edits to the genomes of eukaryotic cells. Most nucleobase editors use deaminase domains that act upon single-stranded DNA and require RNA-guided proteins such as Cas9 to unwind the DNA prior to editing. However, the most recent class of base editors utilizes a deaminase domain, DddAtox, that can act upon double-stranded DNA. Here, we target DddAtox fragments and a FokI-based nickase to the human CIITA gene by fusing these domains to arrays of engineered zinc fingers (ZFs). We also identify a broad variety of Toxin-Derived Deaminases (TDDs) orthologous to DddAtox that allow us to fine-tune properties such as targeting density and specificity. TDD-derived ZF base editors enable up to 73% base editing in T cells with good cell viability and favorable specificity.


Subject(s)
Cytidine Deaminase , Gene Editing , Humans , Cytidine Deaminase/genetics , Cytidine Deaminase/metabolism , DNA/metabolism , Zinc Fingers , Cytidine/genetics , CRISPR-Cas Systems
16.
Genome Res ; 20(8): 1133-42, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20508142

ABSTRACT

Isogenic settings are routine in model organisms, yet remain elusive for genetic experiments on human cells. We describe the use of designed zinc finger nucleases (ZFNs) for efficient transgenesis without drug selection into the PPP1R12C gene, a "safe harbor" locus known as AAVS1. ZFNs enable targeted transgenesis at a frequency of up to 15% following transient transfection of both transformed and primary human cells, including fibroblasts and hES cells. When added to this locus, transgenes such as expression cassettes for shRNAs, small-molecule-responsive cDNA expression cassettes, and reporter constructs, exhibit consistent expression and sustained function over 50 cell generations. By avoiding random integration and drug selection, this method allows bona fide isogenic settings for high-throughput functional genomics, proteomics, and regulatory DNA analysis in essentially any transformed human cell type and in primary cells.


Subject(s)
Gene Transfer Techniques , Genome, Human , Genomics/methods , Regulatory Sequences, Nucleic Acid/genetics , Zinc Fingers/genetics , Base Sequence , Cell Line , Endonucleases/genetics , Genetic Loci , Humans , Molecular Sequence Data , Proteomics/methods
17.
PLoS Pathog ; 7(4): e1002020, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21533216

ABSTRACT

HIV-1 entry requires the cell surface expression of CD4 and either the CCR5 or CXCR4 coreceptors on host cells. Individuals homozygous for the ccr5Δ32 polymorphism do not express CCR5 and are protected from infection by CCR5-tropic (R5) virus strains. As an approach to inactivating CCR5, we introduced CCR5-specific zinc-finger nucleases into human CD4+ T cells prior to adoptive transfer, but the need to protect cells from virus strains that use CXCR4 (X4) in place of or in addition to CCR5 (R5X4) remains. Here we describe engineering a pair of zinc finger nucleases that, when introduced into human T cells, efficiently disrupt cxcr4 by cleavage and error-prone non-homologous DNA end-joining. The resulting cells proliferated normally and were resistant to infection by X4-tropic HIV-1 strains. CXCR4 could also be inactivated in ccr5Δ32 CD4+ T cells, and we show that such cells were resistant to all strains of HIV-1 tested. Loss of CXCR4 also provided protection from X4 HIV-1 in a humanized mouse model, though this protection was lost over time due to the emergence of R5-tropic viral mutants. These data suggest that CXCR4-specific ZFNs may prove useful in establishing resistance to CXCR4-tropic HIV for autologous transplant in HIV-infected individuals.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Deoxyribonucleases/immunology , HIV Infections/immunology , HIV-1/immunology , Receptors, CXCR4/immunology , Animals , CD4-Positive T-Lymphocytes/metabolism , CD4-Positive T-Lymphocytes/transplantation , Cell Proliferation , Deoxyribonucleases/biosynthesis , Deoxyribonucleases/genetics , Disease Models, Animal , Genetic Engineering , HIV Infections/genetics , HIV Infections/metabolism , HIV Infections/therapy , HIV-1/genetics , HIV-1/metabolism , Humans , Macaca mulatta , Mice , Receptors, CCR5/genetics , Receptors, CCR5/immunology , Receptors, CCR5/metabolism , Receptors, CXCR4/genetics , Receptors, CXCR4/metabolism , Transplantation, Autologous , Transplantation, Heterologous , Virus Internalization
18.
Plant Biotechnol J ; 10(7): 783-91, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22520333

ABSTRACT

Targeted gene regulation via designed transcription factors has great potential for precise phenotypic modification and acceleration of novel crop trait development. Canola seed oil composition is dictated largely by the expression of genes encoding enzymes in the fatty acid biosynthetic pathway. In the present study, zinc finger proteins (ZFPs) were designed to bind DNA sequences common to two canola ß-ketoacyl-ACP Synthase II (KASII) genes downstream of their transcription start site. Transcriptional activators (ZFP-TFs) were constructed by fusing these ZFP DNA-binding domains to the VP16 transcriptional activation domain. Following transformation using Agrobacterium, transgenic events expressing ZFP-TFs were generated and shown to have elevated KASII transcript levels in the leaves of transgenic T(0) plants when compared to 'selectable marker only' controls as well as of T(1) progeny plants when compared to null segregants. In addition, leaves of ZFP-TF-expressing T(1) plants contained statistically significant decreases in palmitic acid (consistent with increased KASII activity) and increased total C18. Similarly, T(2) seed displayed statistically significant decreases in palmitic acid, increased total C18 and reduced total saturated fatty acid contents. These results demonstrate that designed ZFP-TFs can be used to regulate the expression of endogenous genes to elicit specific phenotypic modifications of agronomically relevant traits in a crop species.


Subject(s)
3-Oxoacyl-(Acyl-Carrier-Protein) Synthase/genetics , Brassica napus/enzymology , Brassica napus/genetics , Genetic Engineering/methods , Transcription Factors/metabolism , Transcriptional Activation/genetics , Zinc Fingers/genetics , 3-Oxoacyl-(Acyl-Carrier-Protein) Synthase/metabolism , Base Sequence , Crosses, Genetic , DNA, Complementary/genetics , Enzyme Activation , Fatty Acids/metabolism , Gene Expression Regulation, Plant , Genes, Plant/genetics , Molecular Sequence Data , Plant Leaves/enzymology , Plant Leaves/genetics , Plants, Genetically Modified , RNA, Messenger/genetics , RNA, Messenger/metabolism , Seeds/metabolism , Transcription Factors/genetics
19.
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
20.
Nature ; 435(7042): 646-51, 2005 Jun 02.
Article in English | MEDLINE | ID: mdl-15806097

ABSTRACT

Permanent modification of the human genome in vivo is impractical owing to the low frequency of homologous recombination in human cells, a fact that hampers biomedical research and progress towards safe and effective gene therapy. Here we report a general solution using two fundamental biological processes: DNA recognition by C2H2 zinc-finger proteins and homology-directed repair of DNA double-strand breaks. Zinc-finger proteins engineered to recognize a unique chromosomal site can be fused to a nuclease domain, and a double-strand break induced by the resulting zinc-finger nuclease can create specific sequence alterations by stimulating homologous recombination between the chromosome and an extrachromosomal DNA donor. We show that zinc-finger nucleases designed against an X-linked severe combined immune deficiency (SCID) mutation in the IL2Rgamma gene yielded more than 18% gene-modified human cells without selection. Remarkably, about 7% of the cells acquired the desired genetic modification on both X chromosomes, with cell genotype accurately reflected at the messenger RNA and protein levels. We observe comparably high frequencies in human T cells, raising the possibility of strategies based on zinc-finger nucleases for the treatment of disease.


Subject(s)
DNA/metabolism , Endodeoxyribonucleases/chemistry , Endodeoxyribonucleases/metabolism , Gene Targeting/methods , Receptors, Interleukin-2/genetics , Severe Combined Immunodeficiency/genetics , Zinc Fingers , Alleles , CD4-Positive T-Lymphocytes/metabolism , Cell Line , Cells, Cultured , Chromosomes, Human, X/genetics , DNA/genetics , DNA Damage/genetics , DNA Repair/genetics , Genes, Reporter/genetics , Genetic Linkage/genetics , Genetic Therapy/methods , Humans , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, Interleukin-2/metabolism , Recombination, Genetic/genetics , Sequence Homology, Nucleic Acid , Severe Combined Immunodeficiency/therapy , Substrate Specificity
SELECTION OF CITATIONS
SEARCH DETAIL