Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 76
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Cell ; 146(2): 318-31, 2011 Jul 22.
Article in English | MEDLINE | ID: mdl-21757228

ABSTRACT

Patient-specific induced pluripotent stem cells (iPSCs) derived from somatic cells provide a unique tool for the study of human disease, as well as a promising source for cell replacement therapies. One crucial limitation has been the inability to perform experiments under genetically defined conditions. This is particularly relevant for late age onset disorders in which in vitro phenotypes are predicted to be subtle and susceptible to significant effects of genetic background variations. By combining zinc finger nuclease (ZFN)-mediated genome editing and iPSC technology, we provide a generally applicable solution to this problem, generating sets of isogenic disease and control human pluripotent stem cells that differ exclusively at either of two susceptibility variants for Parkinson's disease by modifying the underlying point mutations in the α-synuclein gene. The robust capability to genetically correct disease-causing point mutations in patient-derived hiPSCs represents significant progress for basic biomedical research and an advance toward hiPSC-based cell replacement therapies.


Subject(s)
Parkinson Disease/pathology , Pluripotent Stem Cells , Point Mutation , Cell Line , Embryonic Stem Cells , Genetic Engineering , Genome-Wide Association Study , Humans , Mutagenesis , Oligonucleotides/metabolism , alpha-Synuclein/genetics
2.
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
3.
Genes Dev ; 28(17): 1885-99, 2014 Sep 01.
Article in English | MEDLINE | ID: mdl-25128433

ABSTRACT

Telomere length homeostasis is essential for the long-term survival of stem cells, and its set point determines the proliferative capacity of differentiated cell lineages by restricting the reservoir of telomeric repeats. Knockdown and overexpression studies in human tumor cells showed that the shelterin subunit TPP1 recruits telomerase to telomeres through a region termed the TEL patch. However, these studies do not resolve whether the TPP1 TEL patch is the only mechanism for telomerase recruitment and whether telomerase regulation studied in tumor cells is representative of nontransformed cells such as stem cells. Using genome engineering of human embryonic stem cells, which have physiological telomere length homeostasis, we establish that the TPP1 TEL patch is genetically essential for telomere elongation and thus long-term cell viability. Furthermore, genetic bypass, protein fusion, and intragenic complementation assays define two distinct additional mechanisms of TPP1 involvement in telomerase action at telomeres. We demonstrate that TPP1 provides an essential step of telomerase activation as well as feedback regulation of telomerase by telomere length, which is necessary to determine the appropriate telomere length set point in human embryonic stem cells. These studies reveal and resolve multiple TPP1 roles in telomere elongation and stem cell telomere length homeostasis.


Subject(s)
Telomerase/metabolism , Telomere Homeostasis/genetics , Telomere/enzymology , Embryonic Stem Cells , Enzyme Activation/genetics , Gene Knockout Techniques , Genetic Complementation Test , Humans , Proto-Oncogene Proteins c-ets/genetics , Proto-Oncogene Proteins c-ets/metabolism , Repressor Proteins/genetics , Repressor Proteins/metabolism , Shelterin Complex , Telomerase/genetics , Telomere/genetics , Telomere-Binding Proteins/genetics , Telomere-Binding Proteins/metabolism , ETS Translocation Variant 6 Protein
5.
Nature ; 500(7462): 296-300, 2013 Aug 15.
Article in English | MEDLINE | ID: mdl-23863942

ABSTRACT

Down's syndrome is a common disorder with enormous medical and social costs, caused by trisomy for chromosome 21. We tested the concept that gene imbalance across an extra chromosome can be de facto corrected by manipulating a single gene, XIST (the X-inactivation gene). Using genome editing with zinc finger nucleases, we inserted a large, inducible XIST transgene into the DYRK1A locus on chromosome 21, in Down's syndrome pluripotent stem cells. The XIST non-coding RNA coats chromosome 21 and triggers stable heterochromatin modifications, chromosome-wide transcriptional silencing and DNA methylation to form a 'chromosome 21 Barr body'. This provides a model to study human chromosome inactivation and creates a system to investigate genomic expression changes and cellular pathologies of trisomy 21, free from genetic and epigenetic noise. Notably, deficits in proliferation and neural rosette formation are rapidly reversed upon silencing one chromosome 21. Successful trisomy silencing in vitro also surmounts the major first step towards potential development of 'chromosome therapy'.


Subject(s)
Chromosomes, Human, Pair 21/genetics , Dosage Compensation, Genetic , Down Syndrome/genetics , RNA, Long Noncoding/metabolism , Animals , Cell Line , Cell Proliferation , DNA Methylation , Down Syndrome/therapy , Gene Silencing , Humans , Induced Pluripotent Stem Cells , Male , Mice , Mutagenesis, Insertional , Neurogenesis , RNA, Long Noncoding/genetics , Sex Chromatin/genetics , X Chromosome Inactivation/genetics
7.
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
8.
Nat Methods ; 12(10): 927-30, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26322838

ABSTRACT

Regulatory regions harbor multiple transcription factor (TF) recognition sites; however, the contribution of individual sites to regulatory function remains challenging to define. We describe an approach that exploits the error-prone nature of genome editing-induced double-strand break repair to map functional elements within regulatory DNA at nucleotide resolution. We demonstrate the approach on a human erythroid enhancer, revealing single TF recognition sites that gate the majority of downstream regulatory function.


Subject(s)
Carrier Proteins/genetics , DNA Footprinting/methods , Genomics/methods , Nuclear Proteins/genetics , Regulatory Sequences, Nucleic Acid , Base Sequence , Binding Sites , DNA Breaks, Double-Stranded , DNA Repair , Enhancer Elements, Genetic , Erythrocytes/physiology , Erythropoiesis , Genome, Human , Humans , Mutation , Repressor Proteins , Transcription Factors/metabolism
10.
Nat Rev Genet ; 11(9): 636-46, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20717154

ABSTRACT

Reverse genetics in model organisms such as Drosophila melanogaster, Arabidopsis thaliana, zebrafish and rats, efficient genome engineering in human embryonic stem and induced pluripotent stem cells, targeted integration in crop plants, and HIV resistance in immune cells - this broad range of outcomes has resulted from the application of the same core technology: targeted genome cleavage by engineered, sequence-specific zinc finger nucleases followed by gene modification during subsequent repair. Such 'genome editing' is now established in human cells and a number of model organisms, thus opening the door to a range of new experimental and therapeutic possibilities.


Subject(s)
Endonucleases/genetics , Genetic Techniques , Genome , Zinc Fingers , Animals , Endonucleases/metabolism , Humans
13.
Nat Methods ; 9(10): 993-8, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22922501

ABSTRACT

Malaria afflicts over 200 million people worldwide, and its most lethal etiologic agent, Plasmodium falciparum, is evolving to resist even the latest-generation therapeutics. Efficient tools for genome-directed investigations of P. falciparum-induced pathogenesis, including drug-resistance mechanisms, are clearly required. Here we report rapid and targeted genetic engineering of this parasite using zinc-finger nucleases (ZFNs) that produce a double-strand break in a user-defined locus and trigger homology-directed repair. Targeting an integrated egfp locus, we obtained gene-deletion parasites with unprecedented speed (2 weeks), both with and without direct selection. ZFNs engineered against the parasite gene pfcrt, responsible for escape under chloroquine treatment, rapidly produced parasites that carried either an allelic replacement or a panel of specified point mutations. This method will enable a diverse array of genome-editing approaches to interrogate this human pathogen.


Subject(s)
Endonucleases/physiology , Genome, Protozoan , Plasmodium falciparum/genetics , Protein Engineering/methods , Zinc Fingers/physiology , Alleles , Base Sequence , Chloroquine/pharmacology , Drug Resistance/genetics , Endonucleases/genetics , Molecular Sequence Data , Plasmodium falciparum/drug effects , Zinc Fingers/genetics
15.
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
16.
Neurobiol Dis ; 62: 381-6, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24148854

ABSTRACT

Parkinson's disease associated mutations in leucine rich repeat kinase 2 (LRRK2) impair mitochondrial function and increase the vulnerability of induced pluripotent stem cell (iPSC)-derived neural cells from patients to oxidative stress. Since mitochondrial DNA (mtDNA) damage can compromise mitochondrial function, we examined whether LRRK2 mutations can induce damage to the mitochondrial genome. We found greater levels of mtDNA damage in iPSC-derived neural cells from patients carrying homozygous or heterozygous LRRK2 G2019S mutations, or at-risk individuals carrying the heterozygous LRRK2 R1441C mutation, than in cells from unrelated healthy subjects who do not carry LRRK2 mutations. After zinc finger nuclease-mediated repair of the LRRK2 G2019S mutation in iPSCs, mtDNA damage was no longer detected in differentiated neuroprogenitor and neural cells. Our results unambiguously link LRRK2 mutations to mtDNA damage and validate a new cellular phenotype that can be used for examining pathogenic mechanisms and screening therapeutic strategies.


Subject(s)
DNA Damage , DNA, Mitochondrial/metabolism , Neural Stem Cells/metabolism , Parkinson Disease/genetics , Parkinson Disease/metabolism , Protein Serine-Threonine Kinases/genetics , Targeted Gene Repair , Adult , Aged , DNA Repair , DNA, Mitochondrial/genetics , Female , Humans , Induced Pluripotent Stem Cells/metabolism , Leucine-Rich Repeat Serine-Threonine Protein Kinase-2 , Male , Middle Aged , Mutation , Zinc Fingers
17.
Chromosoma ; 122(1-2): 103-19, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23321980

ABSTRACT

Regulation of DNA replication is critical, and loss of control can lead to DNA amplification. Naturally occurring, developmentally regulated DNA amplification occurs in the DNA puffs of the late larval salivary gland giant polytene chromosomes in the fungus fly, Sciara coprophila. The steroid hormone ecdysone induces DNA amplification in Sciara, and the amplification origin of DNA puff II/9A contains a putative binding site for the ecdysone receptor (EcR). We report here the isolation, cloning, and characterizing of two ecdysone receptor isoforms in Sciara (ScEcR-A and ScEcR-B) and the heterodimeric partner, ultraspiracle (ScUSP). ScEcR-A is the predominant isoform in larval tissues and ScEcR-B in adult tissues, contrary to the pattern in Drosophila. Moreover, ScEcR-A is produced at amplification but is absent just prior. We discuss these results in relation to the model of ecdysone regulation of DNA amplification.


Subject(s)
DNA Replication/genetics , Diptera/genetics , Ecdysone/genetics , Receptors, Steroid/genetics , Amino Acid Sequence , Animals , Binding Sites , Diptera/cytology , Drosophila/cytology , Drosophila/genetics , Larva/genetics , Larva/metabolism , Protein Binding , Receptors, Steroid/isolation & purification , Receptors, Steroid/metabolism , Salivary Glands/cytology
18.
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
19.
Circ Res ; 111(12): 1494-503, 2012 Dec 07.
Article in English | MEDLINE | ID: mdl-22967807

ABSTRACT

RATIONALE: Molecular imaging has proven to be a vital tool in the characterization of stem cell behavior in vivo. However, the integration of reporter genes has typically relied on random integration, a method that is associated with unwanted insertional mutagenesis and positional effects on transgene expression. OBJECTIVE: To address this barrier, we used genome editing with zinc finger nuclease (ZFN) technology to integrate reporter genes into a safe harbor gene locus (PPP1R12C, also known as AAVS1) in the genome of human embryonic stem cells and human induced pluripotent stem cells for molecular imaging. METHODS AND RESULTS: We used ZFN technology to integrate a construct containing monomeric red fluorescent protein, firefly luciferase, and herpes simplex virus thymidine kinase reporter genes driven by a constitutive ubiquitin promoter into a safe harbor locus for fluorescence imaging, bioluminescence imaging, and positron emission tomography imaging, respectively. High efficiency of ZFN-mediated targeted integration was achieved in both human embryonic stem cells and induced pluripotent stem cells. ZFN-edited cells maintained both pluripotency and long-term reporter gene expression. Functionally, we successfully tracked the survival of ZFN-edited human embryonic stem cells and their differentiated cardiomyocytes and endothelial cells in murine models, demonstrating the use of ZFN-edited cells for preclinical studies in regenerative medicine. CONCLUSION: Our study demonstrates a novel application of ZFN technology to the targeted genetic engineering of human pluripotent stem cells and their progeny for molecular imaging in vitro and in vivo.


Subject(s)
Deoxyribonucleases/genetics , Embryonic Stem Cells/enzymology , Genetic Engineering/methods , Genome, Human/genetics , Induced Pluripotent Stem Cells/enzymology , RNA Editing/genetics , Zinc Fingers/genetics , Animals , Cell Differentiation/genetics , Cells, Cultured , Deoxyribonucleases/administration & dosage , Embryonic Stem Cells/cytology , Embryonic Stem Cells/transplantation , Gene Targeting/methods , Genes, Reporter/physiology , Humans , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/transplantation , Mice , Optical Imaging/methods
20.
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
SELECTION OF CITATIONS
SEARCH DETAIL