Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 59
Filtrar
1.
Cell ; 146(2): 318-31, 2011 Jul 22.
Artigo em Inglês | MEDLINE | ID: mdl-21757228

RESUMO

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.


Assuntos
Doença de Parkinson/patologia , Células-Tronco Pluripotentes , Mutação Puntual , Linhagem Celular , Células-Tronco Embrionárias , Engenharia Genética , Estudo de Associação Genômica Ampla , Humanos , Mutagênese , Oligonucleotídeos/metabolismo , alfa-Sinucleína/genética
2.
Nature ; 500(7462): 296-300, 2013 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-23863942

RESUMO

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'.


Assuntos
Cromossomos Humanos Par 21/genética , Mecanismo Genético de Compensação de Dose , Síndrome de Down/genética , RNA Longo não Codificante/metabolismo , Animais , Linhagem Celular , Proliferação de Células , Metilação de DNA , Síndrome de Down/terapia , Inativação Gênica , Humanos , Células-Tronco Pluripotentes Induzidas , Masculino , Camundongos , Mutagênese Insercional , Neurogênese , RNA Longo não Codificante/genética , Cromatina Sexual/genética , Inativação do Cromossomo X/genética
3.
Nat Methods ; 12(5): 465-71, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25799440

RESUMO

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.


Assuntos
Regulação da Expressão Gênica/fisiologia , Genoma , Edição de RNA/fisiologia , Fatores de Transcrição/metabolismo , Animais , Sequência de Bases , DNA/genética , Ensaio de Imunoadsorção Enzimática , Marcadores Genéticos , Fatores de Transcrição/genética
4.
Nat Methods ; 12(10): 927-30, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26322838

RESUMO

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.


Assuntos
Proteínas de Transporte/genética , Pegada de DNA/métodos , Genômica/métodos , Proteínas Nucleares/genética , Sequências Reguladoras de Ácido Nucleico , Sequência de Bases , Sítios de Ligação , Quebras de DNA de Cadeia Dupla , Reparo do DNA , Elementos Facilitadores Genéticos , Eritrócitos/fisiologia , Eritropoese , Genoma Humano , Humanos , Mutação , Proteínas Repressoras , Fatores de Transcrição/metabolismo
5.
Blood ; 127(20): 2416-26, 2016 05 19.
Artigo em Inglês | MEDLINE | ID: mdl-26980728

RESUMO

Genome editing in hematopoietic stem and progenitor cells (HSPCs) is a promising novel technology for the treatment of many human diseases. Here, we evaluated whether the disruption of the C-C chemokine receptor 5 (CCR5) locus in pigtailed macaque HSPCs by zinc finger nucleases (ZFNs) was feasible. We show that macaque-specific CCR5 ZFNs efficiently induce CCR5 disruption at levels of up to 64% ex vivo, 40% in vivo early posttransplant, and 3% to 5% in long-term repopulating cells over 6 months following HSPC transplant. These genome-edited HSPCs support multilineage engraftment and generate progeny capable of trafficking to secondary tissues including the gut. Using deep sequencing technology, we show that these ZFNs are highly specific for the CCR5 locus in primary cells. Further, we have adapted our clonal tracking methodology to follow individual CCR5 mutant cells over time in vivo, reinforcing that CCR5 gene-edited HSPCs are capable of long-term engraftment. Together, these data demonstrate that genome-edited HSPCs engraft, and contribute to multilineage repopulation after autologous transplantation in a clinically relevant large animal model, an important step toward the development of stem cell-based genome-editing therapies for HIV and potentially other diseases as well.


Assuntos
Transplante de Medula Óssea , Linhagem da Célula , Edição de Genes , Transplante de Células-Tronco Hematopoéticas , Macaca nemestrina/genética , Receptores CCR5/genética , Sequência de Aminoácidos , Animais , Linhagem Celular , Eletroporação , Estudos de Viabilidade , Técnicas de Silenciamento de Genes , Sobrevivência de Enxerto , Células-Tronco Hematopoéticas/citologia , Células-Tronco Hematopoéticas/metabolismo , Dados de Sequência Molecular , Mutação , Reação em Cadeia da Polimerase/métodos , RNA Mensageiro/genética , Receptores CCR5/deficiência , Análise de Sequência de DNA , Condicionamento Pré-Transplante , Transplante Autólogo , Irradiação Corporal Total , Dedos de Zinco
6.
Blood ; 126(15): 1777-84, 2015 Oct 08.
Artigo em Inglês | MEDLINE | ID: mdl-26297739

RESUMO

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.


Assuntos
Albuminas/genética , Terapia de Reposição de Enzimas , Terapia Genética , Genoma , Fígado/metabolismo , Transgenes/fisiologia , Albuminas/metabolismo , Animais , Dependovirus/genética , Endonucleases , Doença de Fabry/genética , Doença de Fabry/terapia , Fator IX/genética , Fator VIII/genética , Doença de Gaucher/genética , Doença de Gaucher/terapia , Vetores Genéticos/administração & dosagem , Hemofilia A/genética , Hemofilia A/terapia , Hemofilia B/genética , Hemofilia B/terapia , Sequenciamento de Nucleotídeos em Larga Escala , Humanos , Lisossomos/enzimologia , Camundongos , Camundongos Endogâmicos C57BL , Mucopolissacaridose I/genética , Mucopolissacaridose I/terapia , Mucopolissacaridose II/genética , Mucopolissacaridose II/terapia , Regiões Promotoras Genéticas/genética , Edição de RNA , RNA Mensageiro/genética , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Dedos de Zinco
7.
Blood ; 125(17): 2597-604, 2015 Apr 23.
Artigo em Inglês | MEDLINE | ID: mdl-25733580

RESUMO

Sickle cell disease (SCD) is characterized by a single point mutation in the seventh codon of the ß-globin gene. Site-specific correction of the sickle mutation in hematopoietic stem cells would allow for permanent production of normal red blood cells. Using zinc-finger nucleases (ZFNs) designed to flank the sickle mutation, we demonstrate efficient targeted cleavage at the ß-globin locus with minimal off-target modification. By co-delivering a homologous donor template (either an integrase-defective lentiviral vector or a DNA oligonucleotide), high levels of gene modification were achieved in CD34(+) hematopoietic stem and progenitor cells. Modified cells maintained their ability to engraft NOD/SCID/IL2rγ(null) mice and to produce cells from multiple lineages, although with a reduction in the modification levels relative to the in vitro samples. Importantly, ZFN-driven gene correction in CD34(+) cells from the bone marrow of patients with SCD resulted in the production of wild-type hemoglobin tetramers.


Assuntos
Anemia Falciforme/genética , Anemia Falciforme/terapia , Terapia Genética , Células-Tronco Hematopoéticas/metabolismo , Mutação , Globinas beta/genética , Anemia Falciforme/patologia , Animais , Antígenos CD34/análise , Sequência de Bases , Células da Medula Óssea/metabolismo , Células da Medula Óssea/patologia , Células Cultivadas , Endodesoxirribonucleases/metabolismo , Sangue Fetal/transplante , Loci Gênicos , Transplante de Células-Tronco Hematopoéticas , Células-Tronco Hematopoéticas/patologia , Humanos , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Dados de Sequência Molecular , Dedos de Zinco
8.
Nature ; 475(7355): 217-21, 2011 Jun 26.
Artigo em Inglês | MEDLINE | ID: mdl-21706032

RESUMO

Editing of the human genome to correct disease-causing mutations is a promising approach for the treatment of genetic disorders. Genome editing improves on simple gene-replacement strategies by effecting in situ correction of a mutant gene, thus restoring normal gene function under the control of endogenous regulatory elements and reducing risks associated with random insertion into the genome. Gene-specific targeting has historically been limited to mouse embryonic stem cells. The development of zinc finger nucleases (ZFNs) has permitted efficient genome editing in transformed and primary cells that were previously thought to be intractable to such genetic manipulation. In vitro, ZFNs have been shown to promote efficient genome editing via homology-directed repair by inducing a site-specific double-strand break (DSB) at a target locus, but it is unclear whether ZFNs can induce DSBs and stimulate genome editing at a clinically meaningful level in vivo. Here we show that ZFNs are able to induce DSBs efficiently when delivered directly to mouse liver and that, when co-delivered with an appropriately designed gene-targeting vector, they can stimulate gene replacement through both homology-directed and homology-independent targeted gene insertion at the ZFN-specified locus. The level of gene targeting achieved was sufficient to correct the prolonged clotting times in a mouse model of haemophilia B, and remained persistent after induced liver regeneration. Thus, ZFN-driven gene correction can be achieved in vivo, raising the possibility of genome editing as a viable strategy for the treatment of genetic disease.


Assuntos
Reparo do DNA/genética , Modelos Animais de Doenças , Marcação de Genes/métodos , Terapia Genética/métodos , Genoma/genética , Hemofilia B/genética , Hemostasia , Animais , Sequência de Bases , Linhagem Celular Tumoral , Quebras de DNA de Cadeia Dupla , Endonucleases/química , Endonucleases/genética , Endonucleases/metabolismo , Éxons/genética , Fator IX/análise , Fator IX/genética , Vetores Genéticos/genética , Células HEK293 , Hemofilia B/fisiopatologia , Humanos , Íntrons/genética , Fígado/metabolismo , Regeneração Hepática , Camundongos , Camundongos Endogâmicos C57BL , Mutação/genética , Fenótipo , Homologia de Sequência , Dedos de Zinco
9.
Nat Rev Genet ; 11(9): 636-46, 2010 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-20717154

RESUMO

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.


Assuntos
Endonucleases/genética , Técnicas Genéticas , Genoma , Dedos de Zinco , Animais , Endonucleases/metabolismo , Humanos
10.
Mol Ther ; 23(8): 1380-1390, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-25939491

RESUMO

Programmed cell death-1 (PD-1) is expressed on activated T cells and represents an attractive target for gene-editing of tumor targeted T cells prior to adoptive cell transfer (ACT). We used zinc finger nucleases (ZFNs) directed against the gene encoding human PD-1 (PDCD-1) to gene-edit melanoma tumor infiltrating lymphocytes (TIL). We show that our clinical scale TIL production process yielded efficient modification of the PD-1 gene locus, with an average modification frequency of 74.8% (n = 3, range 69.9-84.1%) of the alleles in a bulk TIL population, which resulted in a 76% reduction in PD-1 surface-expression. Forty to 48% of PD-1 gene-edited cells had biallelic PD-1 modification. Importantly, the PD-1 gene-edited TIL product showed improved in vitro effector function and a significantly increased polyfunctional cytokine profile (TNFα, GM-CSF, and IFNγ) compared to unmodified TIL in two of the three donors tested. In addition, all donor cells displayed an effector memory phenotype and expanded approximately 500-2,000-fold in vitro. Thus, further study to determine the efficiency and safety of adoptive cell transfer using PD-1 gene-edited TIL for the treatment of metastatic melanoma is warranted.


Assuntos
Endorribonucleases/genética , Regulação Neoplásica da Expressão Gênica , Linfócitos do Interstício Tumoral/imunologia , Melanoma/terapia , Receptor de Morte Celular Programada 1/genética , Dedos de Zinco , Alelos , Animais , Separação Celular , Citocinas/metabolismo , Feminino , Citometria de Fluxo , Fator Estimulador de Colônias de Granulócitos e Macrófagos/metabolismo , Proteínas de Fluorescência Verde/metabolismo , Humanos , Memória Imunológica , Imunoterapia Adotiva , Interferon gama/metabolismo , Ativação Linfocitária/imunologia , Camundongos , Metástase Neoplásica , Transplante de Neoplasias , Fenótipo , Receptor de Morte Celular Programada 1/metabolismo , Fator de Necrose Tumoral alfa/metabolismo
11.
Nat Methods ; 9(10): 993-8, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22922501

RESUMO

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.


Assuntos
Endonucleases/fisiologia , Genoma de Protozoário , Plasmodium falciparum/genética , Engenharia de Proteínas/métodos , Dedos de Zinco/fisiologia , Alelos , Sequência de Bases , Cloroquina/farmacologia , Resistência a Medicamentos/genética , Endonucleases/genética , Dados de Sequência Molecular , Plasmodium falciparum/efeitos dos fármacos , Dedos de Zinco/genética
12.
Blood ; 122(8): 1341-9, 2013 Aug 22.
Artigo em Inglês | MEDLINE | ID: mdl-23741009

RESUMO

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.


Assuntos
Desoxirribonucleases/genética , Antígenos de Histocompatibilidade Classe I/metabolismo , Transplante de Células-Tronco/métodos , Transplante Homólogo , Antígenos CD19/metabolismo , Sequência de Bases , Diferenciação Celular , Citotoxicidade Imunológica/imunologia , Eletroporação , Células-Tronco Embrionárias/citologia , Técnicas de Transferência de Genes , Células HEK293 , Humanos , Leucócitos Mononucleares/citologia , Dados de Sequência Molecular , Engenharia de Proteínas , Linfócitos T/imunologia , Dedos de Zinco
13.
Nature ; 459(7245): 437-41, 2009 May 21.
Artigo em Inglês | MEDLINE | ID: mdl-19404259

RESUMO

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.


Assuntos
Biotecnologia/métodos , Desoxirribonucleases/química , Desoxirribonucleases/metabolismo , Marcação de Genes/métodos , Genoma de Planta/genética , Zea mays/genética , Dedos de Zinco , Desoxirribonucleases/genética , Alimentos Geneticamente Modificados , Genes de Plantas/genética , Resistência a Herbicidas/genética , Herbicidas/farmacologia , Hereditariedade , Fosfatos de Inositol/metabolismo , Mutagênese Sítio-Dirigida/métodos , Plantas Geneticamente Modificadas , Recombinação Genética/genética , Reprodutibilidade dos Testes
14.
Blood ; 119(24): 5697-705, 2012 Jun 14.
Artigo em Inglês | MEDLINE | ID: mdl-22535661

RESUMO

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.


Assuntos
Antígenos CD19/imunologia , Epitopos/imunologia , Engenharia Genética , Imunoterapia/métodos , Receptores de Antígenos de Linfócitos T/imunologia , Proteínas Recombinantes/imunologia , Linfócitos T/imunologia , Adulto , Células Apresentadoras de Antígenos/imunologia , Antígenos de Neoplasias/imunologia , Antígenos CD28/metabolismo , Complexo CD3/metabolismo , Células Cultivadas , Endonucleases/metabolismo , Técnicas de Inativação de Genes , Humanos , Células K562 , Ativação Linfocitária/imunologia , Dedos de Zinco
15.
PLoS Genet ; 7(6): e1002080, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21655080

RESUMO

Nonhomologous end-joining (NHEJ) is the primary DNA repair pathway thought to underlie chromosomal translocations and other genomic rearrangements in somatic cells. The canonical NHEJ pathway, including DNA ligase IV (Lig4), suppresses genomic instability and chromosomal translocations, leading to the notion that a poorly defined, alternative NHEJ (alt-NHEJ) pathway generates these rearrangements. Here, we investigate the DNA ligase requirement of chromosomal translocation formation in mouse cells. Mammals have two other DNA ligases, Lig1 and Lig3, in addition to Lig4. As deletion of Lig3 results in cellular lethality due to its requirement in mitochondria, we used recently developed cell lines deficient in nuclear Lig3 but rescued for mitochondrial DNA ligase activity. Further, zinc finger endonucleases were used to generate DNA breaks at endogenous loci to induce translocations. Unlike with Lig4 deficiency, which causes an increase in translocation frequency, translocations are reduced in frequency in the absence of Lig3. Residual translocations in Lig3-deficient cells do not show a bias toward use of pre-existing microhomology at the breakpoint junctions, unlike either wild-type or Lig4-deficient cells, consistent with the notion that alt-NHEJ is impaired with Lig3 loss. By contrast, Lig1 depletion in otherwise wild-type cells does not reduce translocations or affect microhomology use. However, translocations are further reduced in Lig3-deficient cells upon Lig1 knockdown, suggesting the existence of two alt-NHEJ pathways, one that is biased toward microhomology use and requires Lig3 and a back-up pathway which does not depend on microhomology and utilizes Lig1.


Assuntos
DNA Ligases/metabolismo , Translocação Genética , Animais , Sequência de Bases , Núcleo Celular/metabolismo , DNA Ligase Dependente de ATP , DNA Ligases/genética , Reparo do DNA , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Camundongos , Modelos Genéticos , Dados de Sequência Molecular , Proteínas de Ligação a Poli-ADP-Ribose , Transfecção , Proteínas de Xenopus
16.
Proc Natl Acad Sci U S A ; 108(17): 7052-7, 2011 Apr 26.
Artigo em Inglês | MEDLINE | ID: mdl-21471457

RESUMO

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.


Assuntos
Alelos , Proteínas de Transporte/genética , Desoxirribonucleases/genética , Marcação de Genes/métodos , Proteínas de Xenopus/genética , Animais , Animais Geneticamente Modificados , Proteínas de Transporte/metabolismo , Desoxirribonucleases/metabolismo , Xenopus , Proteínas de Xenopus/metabolismo , Dedos de Zinco
17.
Genome Res ; 20(8): 1133-42, 2010 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-20508142

RESUMO

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.


Assuntos
Técnicas de Transferência de Genes , Genoma Humano , Genômica/métodos , Sequências Reguladoras de Ácido Nucleico/genética , Dedos de Zinco/genética , Sequência de Bases , Linhagem Celular , Endonucleases/genética , Loci Gênicos , Humanos , Dados de Sequência Molecular , Proteômica/métodos
18.
Plant Biotechnol J ; 11(9): 1126-34, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-23953646

RESUMO

Modern agriculture demands crops carrying multiple traits. The current paradigm of randomly integrating and sorting independently segregating transgenes creates severe downstream breeding challenges. A versatile, generally applicable solution is hereby provided: the combination of high-efficiency targeted genome editing driven by engineered zinc finger nucleases (ZFNs) with modular 'trait landing pads' (TLPs) that allow 'mix-and-match', on-demand transgene integration and trait stacking in crop plants. We illustrate the utility of nuclease-driven TLP technology by applying it to the stacking of herbicide resistance traits. We first integrated into the maize genome an herbicide resistance gene, pat, flanked with a TLP (ZFN target sites and sequences homologous to incoming DNA) using WHISKERS™-mediated transformation of embryogenic suspension cultures. We established a method for targeted transgene integration based on microparticle bombardment of immature embryos and used it to deliver a second trait precisely into the TLP via cotransformation with a donor DNA containing a second herbicide resistance gene, aad1, flanked by sequences homologous to the integrated TLP along with a corresponding ZFN expression construct. Remarkably, up to 5% of the embryo-derived transgenic events integrated the aad1 transgene precisely at the TLP, that is, directly adjacent to the pat transgene. Importantly and consistent with the juxtaposition achieved via nuclease-driven TLP technology, both herbicide resistance traits cosegregated in subsequent generations, thereby demonstrating linkage of the two independently transformed transgenes. Because ZFN-mediated targeted transgene integration is becoming applicable across an increasing number of crop species, this work exemplifies a simple, facile and rapid approach to trait stacking.


Assuntos
Endonucleases/genética , Marcação de Genes/métodos , Genoma de Planta/genética , Resistência a Herbicidas , Herbicidas/farmacologia , Zea mays/genética , Produtos Agrícolas , Endonucleases/metabolismo , Ligação Genética , Fenótipo , Folhas de Planta/genética , Folhas de Planta/metabolismo , Proteínas de Plantas/genética , Proteínas de Plantas/metabolismo , Plantas Geneticamente Modificadas , Transgenes , Dedos de Zinco
19.
PLoS Pathog ; 7(4): e1002020, 2011 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-21533216

RESUMO

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.


Assuntos
Linfócitos T CD4-Positivos/imunologia , Desoxirribonucleases/imunologia , Infecções por HIV/imunologia , HIV-1/imunologia , Receptores CXCR4/imunologia , Animais , Linfócitos T CD4-Positivos/metabolismo , Linfócitos T CD4-Positivos/transplante , Proliferação de Células , Desoxirribonucleases/biossíntese , Desoxirribonucleases/genética , Modelos Animais de Doenças , Engenharia Genética , Infecções por HIV/genética , Infecções por HIV/metabolismo , Infecções por HIV/terapia , HIV-1/genética , HIV-1/metabolismo , Humanos , Macaca mulatta , Camundongos , Receptores CCR5/genética , Receptores CCR5/imunologia , Receptores CCR5/metabolismo , Receptores CXCR4/genética , Receptores CXCR4/metabolismo , Transplante Autólogo , Transplante Heterólogo , Internalização do Vírus
20.
Mol Ther ; 20(8): 1508-15, 2012 08.
Artigo em Inglês | MEDLINE | ID: mdl-22828502

RESUMO

Selective inhibition of disease-related proteins underpins the majority of successful drug-target interactions. However, development of effective antagonists is often hampered by targets that are not druggable using conventional approaches. Here, we apply engineered zinc-finger protein transcription factors (ZFP TFs) to the endogenous phospholamban (PLN) gene, which encodes a well validated but recalcitrant drug target in heart failure. We show that potent repression of PLN expression can be achieved with specificity that approaches single-gene regulation. Moreover, ZFP-driven repression of PLN increases calcium reuptake kinetics and improves contractile function of cardiac muscle both in vitro and in an animal model of heart failure. These results support the development of the PLN repressor as therapy for heart failure, and provide evidence that delivery of engineered ZFP TFs to native organs can drive therapeutically relevant levels of gene repression in vivo. Given the adaptability of designed ZFPs for binding diverse DNA sequences and the ubiquity of potential targets (promoter proximal DNA), our findings suggest that engineered ZFP repressors represent a powerful tool for the therapeutic inhibition of disease-related genes, therefore, offering the potential for therapeutic intervention in heart failure and other poorly treated human diseases.


Assuntos
Proteínas de Ligação ao Cálcio/metabolismo , Insuficiência Cardíaca/metabolismo , Insuficiência Cardíaca/terapia , Fatores de Transcrição/metabolismo , Dedos de Zinco/fisiologia , Adenoviridae/genética , Animais , Western Blotting , Proteínas de Ligação ao Cálcio/genética , Linhagem Celular , Insuficiência Cardíaca/genética , Humanos , Cinética , Miócitos Cardíacos/citologia , Miócitos Cardíacos/metabolismo , Miócitos Cardíacos/fisiologia , Ratos , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fatores de Transcrição/genética , Dedos de Zinco/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA