Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
1.
Sci Adv ; 7(12)2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-33741591

RESUMO

Neuronal tau reduction confers resilience against ß-amyloid and tau-related neurotoxicity in vitro and in vivo. Here, we introduce a novel translational approach to lower expression of the tau gene MAPT at the transcriptional level using gene-silencing zinc finger protein transcription factors (ZFP-TFs). Following a single administration of adeno-associated virus (AAV), either locally into the hippocampus or intravenously to enable whole-brain transduction, we selectively reduced tau messenger RNA and protein by 50 to 80% out to 11 months, the longest time point studied. Sustained tau lowering was achieved without detectable off-target effects, overt histopathological changes, or molecular alterations. Tau reduction with AAV ZFP-TFs was able to rescue neuronal damage around amyloid plaques in a mouse model of Alzheimer's disease (APP/PS1 line). The highly specific, durable, and controlled knockdown of endogenous tau makes AAV-delivered ZFP-TFs a promising approach for the treatment of tau-related human brain diseases.


Assuntos
Doença de Alzheimer , Fatores de Transcrição , Doença de Alzheimer/genética , Doença de Alzheimer/patologia , Doença de Alzheimer/terapia , Peptídeos beta-Amiloides/metabolismo , Animais , Encéfalo/metabolismo , Dependovirus/genética , Dependovirus/metabolismo , Modelos Animais de Doenças , Camundongos , Placa Amiloide/patologia , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Dedos de Zinco/genética , Proteínas tau/genética , Proteínas tau/metabolismo
2.
Nat Biotechnol ; 37(8): 945-952, 2019 08.
Artigo em Inglês | MEDLINE | ID: mdl-31359006

RESUMO

Engineered nucleases have gained broad appeal for their ability to mediate highly efficient genome editing. However the specificity of these reagents remains a concern, especially for therapeutic applications, given the potential mutagenic consequences of off-target cleavage. Here we have developed an approach for improving the specificity of zinc finger nucleases (ZFNs) that engineers the FokI catalytic domain with the aim of slowing cleavage, which should selectively reduce activity at low-affinity off-target sites. For three ZFN pairs, we engineered single-residue substitutions in the FokI domain that preserved full on-target activity but showed a reduction in off-target indels of up to 3,000-fold. By combining this approach with substitutions that reduced the affinity of zinc fingers, we developed ZFNs specific for the TRAC locus that mediated 98% knockout in T cells with no detectable off-target activity at an assay background of ~0.01%. We anticipate that this approach, and the FokI variants we report, will enable routine generation of nucleases for gene editing with no detectable off-target activity.


Assuntos
Clivagem do DNA , Edição de Genes/métodos , Linfócitos T , Sequência de Bases , DNA/genética , DNA/metabolismo , Citometria de Fluxo , Células-Tronco Hematopoéticas , Humanos , Células K562 , Domínios Proteicos , RNA Mensageiro
3.
Nat Med ; 24(11): 1691-1695, 2018 11.
Artigo em Inglês | MEDLINE | ID: mdl-30250142

RESUMO

Mutations of the mitochondrial genome (mtDNA) underlie a substantial portion of mitochondrial disease burden. These disorders are currently incurable and effectively untreatable, with heterogeneous penetrance, presentation and prognosis. To address the lack of effective treatment for these disorders, we exploited a recently developed mouse model that recapitulates common molecular features of heteroplasmic mtDNA disease in cardiac tissue: the m.5024C>T tRNAAla mouse. Through application of a programmable nuclease therapy approach, using systemically administered, mitochondrially targeted zinc-finger nucleases (mtZFN) delivered by adeno-associated virus, we induced specific elimination of mutant mtDNA across the heart, coupled to a reversion of molecular and biochemical phenotypes. These findings constitute proof of principle that mtDNA heteroplasmy correction using programmable nucleases could provide a therapeutic route for heteroplasmic mitochondrial diseases of diverse genetic origin.


Assuntos
Edição de Genes , Mitocôndrias Cardíacas/genética , Doenças Mitocondriais/genética , Nucleases de Dedos de Zinco/genética , Animais , DNA Mitocondrial/genética , Dependovirus/genética , Modelos Animais de Doenças , Humanos , Camundongos , Mitocôndrias Cardíacas/patologia , Doenças Mitocondriais/patologia , Doenças Mitocondriais/terapia , Mutação/genética , Prognóstico , RNA de Transferência/genética , Nucleases de Dedos de Zinco/uso terapêutico
4.
Mol Ther Methods Clin Dev ; 4: 137-148, 2017 Mar 17.
Artigo em Inglês | MEDLINE | ID: mdl-28344999

RESUMO

To develop an effective and sustainable cell therapy for sickle cell disease (SCD), we investigated the feasibility of targeted disruption of the BCL11A gene, either within exon 2 or at the GATAA motif in the intronic erythroid-specific enhancer, using zinc finger nucleases in human bone marrow (BM) CD34+ hematopoietic stem and progenitor cells (HSPCs). Both targeting strategies upregulated fetal globin expression in erythroid cells to levels predicted to inhibit hemoglobin S polymerization. However, complete inactivation of BCL11A resulting from bi-allelic frameshift mutations in BCL11A exon 2 adversely affected erythroid enucleation. In contrast, bi-allelic disruption of the GATAA motif in the erythroid enhancer of BCL11A did not negatively impact enucleation. Furthermore, BCL11A exon 2-edited BM-CD34+ cells demonstrated a significantly reduced engraftment potential in immunodeficient mice. Such an adverse effect on HSPC function was not observed upon BCL11A erythroid-enhancer GATAA motif editing, because enhancer-edited CD34+ cells achieved robust long-term engraftment and gave rise to erythroid cells with elevated levels of fetal globin expression when chimeric BM was cultured ex vivo. Altogether, our results support further clinical development of the BCL11A erythroid-specific enhancer editing in BM-CD34+ HSPCs as an autologous stem cell therapy in SCD patients.

5.
Stem Cell Reports ; 7(2): 139-48, 2016 08 09.
Artigo em Inglês | MEDLINE | ID: mdl-27396937

RESUMO

Wiskott-Aldrich syndrome (WAS) is an X-linked primary immunodeficiency disease caused by mutations in the gene encoding the WAS protein (WASp). Here, induced pluripotent stem cells (iPSCs) were derived from a WAS patient (WAS-iPSC) and the endogenous chromosomal WAS locus was targeted with a wtWAS-2A-eGFP transgene using zinc finger nucleases (ZFNs) to generate corrected WAS-iPSC (cWAS-iPSC). WASp and GFP were first expressed in the earliest CD34(+)CD43(+)CD45(-) hematopoietic precursor cells and later in all hematopoietic lineages examined. Whereas differentiation to non-lymphoid lineages was readily obtained from WAS-iPSCs, in vitro T lymphopoiesis from WAS-iPSC was deficient with few CD4(+)CD8(+) double-positive and mature CD3(+) T cells obtained. T cell differentiation was restored for cWAS-iPSCs. Similarly, defects in natural killer cell differentiation and function were restored on targeted correction of the WAS locus. These results demonstrate that the defects exhibited by WAS-iPSC-derived lymphoid cells were fully corrected and suggests the potential therapeutic use of gene-corrected WAS-iPSCs.


Assuntos
Terapia Genética , Células-Tronco Pluripotentes Induzidas/patologia , Linfopoese , Síndrome de Wiskott-Aldrich/patologia , Síndrome de Wiskott-Aldrich/terapia , Células-Tronco Hematopoéticas/metabolismo , Células-Tronco Hematopoéticas/patologia , Humanos , Células Matadoras Naturais/metabolismo , Linfócitos T/imunologia , Proteína da Síndrome de Wiskott-Aldrich/genética
6.
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
7.
Sci Rep ; 6: 21757, 2016 Feb 23.
Artigo em Inglês | MEDLINE | ID: mdl-26902653

RESUMO

Mismatch of human leukocyte antigens (HLA) adversely impacts the outcome of patients after allogeneic hematopoietic stem-cell transplantation (alloHSCT). This translates into the clinical requirement to timely identify suitable HLA-matched donors which in turn curtails the chances of recipients, especially those from a racial minority, to successfully undergo alloHSCT. We thus sought to broaden the existing pool of registered unrelated donors based on analysis that eliminating the expression of the HLA-A increases the chance for finding a donor matched at HLA-B, -C, and -DRB1 regardless of a patient's race. Elimination of HLA-A expression in HSC was achieved using artificial zinc finger nucleases designed to target HLA-A alleles. Significantly, these engineered HSCs maintain their ability to engraft and reconstitute hematopoiesis in immunocompromised mice. This introduced loss of HLA-A expression decreases the need to recruit large number of donors to match with potential recipients and has particular importance for patients whose HLA repertoire is under-represented in the current donor pool. Furthermore, the genetic engineering of stem cells provides a translational approach to HLA-match a limited number of third-party donors with a wide number of recipients.


Assuntos
Desoxirribonucleases/genética , Deleção de Genes , Antígenos HLA-A/genética , Transplante de Células-Tronco Hematopoéticas/etnologia , Células-Tronco Hematopoéticas/imunologia , Alelos , Animais , Desoxirribonucleases/metabolismo , Seleção do Doador/ética , Expressão Gênica , Engenharia Genética/métodos , Antígenos HLA-A/imunologia , Antígenos HLA-B/genética , Antígenos HLA-B/imunologia , Antígenos HLA-C/genética , Antígenos HLA-C/imunologia , Cadeias HLA-DRB1/genética , Cadeias HLA-DRB1/imunologia , Acessibilidade aos Serviços de Saúde/ética , Transplante de Células-Tronco Hematopoéticas/ética , Células-Tronco Hematopoéticas/citologia , Teste de Histocompatibilidade , Humanos , Camundongos , Grupos Raciais , Transplante Heterólogo , Transplante Homólogo , Doadores não Relacionados , Dedos de Zinco
8.
Sci Rep ; 6: 21645, 2016 Feb 22.
Artigo em Inglês | MEDLINE | ID: mdl-26898342

RESUMO

We describe a fundamentally novel feat of animal genetic engineering: the precise and efficient substitution of an agronomic haplotype into a domesticated species. Zinc finger nuclease in-embryo editing of the RELA locus generated live born domestic pigs with the warthog RELA orthologue, associated with resilience to African Swine Fever. The ability to efficiently achieve interspecies allele introgression in one generation opens unprecedented opportunities for agriculture and basic research.


Assuntos
Resistência à Doença/genética , Edição de Genes/métodos , Engenharia Genética , Ligases/genética , Febre Suína Africana/genética , Febre Suína Africana/virologia , Vírus da Febre Suína Africana/patogenicidade , Alelos , Animais , Genoma , Haplótipos , Suínos
9.
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
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.
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
12.
Stem Cell Reports ; 2(6): 838-52, 2014 Jun 03.
Artigo em Inglês | MEDLINE | ID: mdl-24936470

RESUMO

Genetically engineered human pluripotent stem cells (hPSCs) have been proposed as a source for transplantation therapies and are rapidly becoming valuable tools for human disease modeling. However, many applications are limited due to the lack of robust differentiation paradigms that allow for the isolation of defined functional tissues. Here, using an endogenous LGR5-GFP reporter, we derived adult stem cells from hPSCs that gave rise to functional human intestinal tissue comprising all major cell types of the intestine. Histological and functional analyses revealed that such human organoid cultures could be derived with high purity and with a composition and morphology similar to those of cultures obtained from human biopsies. Importantly, hPSC-derived organoids responded to the canonical signaling pathways that control self-renewal and differentiation in the adult human intestinal stem cell compartment. This adult stem cell system provides a platform for studying human intestinal disease in vitro using genetically engineered hPSCs.


Assuntos
Células-Tronco Adultas/citologia , Células-Tronco Adultas/metabolismo , Perfilação da Expressão Gênica/métodos , Intestinos/citologia , Células-Tronco Pluripotentes/citologia , Células-Tronco Pluripotentes/metabolismo , Células Cultivadas , Humanos , Receptores Acoplados a Proteínas G/metabolismo
13.
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
14.
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
15.
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
16.
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
17.
Nat Biotechnol ; 29(2): 143-8, 2011 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-21179091

RESUMO

Nucleases that cleave unique genomic sequences in living cells can be used for targeted gene editing and mutagenesis. Here we develop a strategy for generating such reagents based on transcription activator-like effector (TALE) proteins from Xanthomonas. We identify TALE truncation variants that efficiently cleave DNA when linked to the catalytic domain of FokI and use these nucleases to generate discrete edits or small deletions within endogenous human NTF3 and CCR5 genes at efficiencies of up to 25%. We further show that designed TALEs can regulate endogenous mammalian genes. These studies demonstrate the effective application of designed TALE transcription factors and nucleases for the targeted regulation and modification of endogenous genes.


Assuntos
Técnicas de Química Combinatória/métodos , Engenharia Genética , Mutagênese Sítio-Dirigida/métodos , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Sequência de Bases , Sítios de Ligação , DNA/genética , DNA/metabolismo , Desoxirribonucleases de Sítio Específico do Tipo II/genética , Desoxirribonucleases de Sítio Específico do Tipo II/metabolismo , Genoma , Humanos , Células K562 , Dados de Sequência Molecular , Receptores CCR5/genética , Fator A de Crescimento do Endotélio Vascular/genética , Xanthomonas
18.
J Neurosci ; 30(49): 16469-74, 2010 Dec 08.
Artigo em Inglês | MEDLINE | ID: mdl-21147986

RESUMO

Loss of dopaminergic neurons is primarily responsible for the onset and progression of Parkinson's disease (PD); thus, neuroprotective and/or neuroregenerative strategies remain critical to the treatment of this increasingly prevalent disease. Here we explore a novel approach to neurotrophic factor-based therapy by engineering zinc finger protein transcription factors (ZFP TFs) that activate the expression of the endogenous glial cell line-derived neurotrophic factor (GDNF) gene. We show that GDNF activation can be achieved with exquisite genome-wide specificity. Furthermore, in a rat model of PD, striatal delivery of an adeno-associated viral vector serotype 2 encoding the GDNF activator resulted in improvements in forelimb akinesia, sensorimotor neglect, and amphetamine-induced rotations caused by 6-hydroxydopamine (6-OHDA) lesion. Our results suggest that an engineered ZFP TF can drive sufficient GDNF expression in the brain to provide functional neuroprotection against 6-OHDA; therefore, targeted activation of the endogenous gene may provide a method for delivering appropriate levels of GDNF to PD patients.


Assuntos
Terapia Genética/métodos , Fatores Neurotróficos Derivados de Linhagem de Célula Glial/uso terapêutico , Fármacos Neuroprotetores/uso terapêutico , Doença de Parkinson/terapia , Engenharia de Proteínas/métodos , Anfetamina/administração & dosagem , Animais , Linhagem Celular , Modelos Animais de Doenças , Dopaminérgicos/administração & dosagem , Ensaio de Imunoadsorção Enzimática/métodos , Regulação da Expressão Gênica/efeitos dos fármacos , Vetores Genéticos/fisiologia , Fatores Neurotróficos Derivados de Linhagem de Célula Glial/biossíntese , Fatores Neurotróficos Derivados de Linhagem de Célula Glial/genética , Proteínas de Fluorescência Verde/genética , Haplorrinos , Humanos , Lentivirus/fisiologia , Camundongos , Análise em Microsséries/métodos , Atividade Motora/efeitos dos fármacos , Oxidopamina/toxicidade , Doença de Parkinson/complicações , Doença de Parkinson/etiologia , RNA Mensageiro/metabolismo , Ratos , Fatores de Tempo , Transfecção , Tirosina 3-Mono-Oxigenase/metabolismo , Dedos de Zinco/genética
19.
Plant Mol Biol ; 69(6): 699-709, 2009 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-19112554

RESUMO

Targeted transgene integration in plants remains a significant technical challenge for both basic and applied research. Here it is reported that designed zinc finger nucleases (ZFNs) can drive site-directed DNA integration into transgenic and native gene loci. A dimer of designed 4-finger ZFNs enabled intra-chromosomal reconstitution of a disabled gfp reporter gene and site-specific transgene integration into chromosomal reporter loci following co-transformation of tobacco cell cultures with a donor construct comprised of sequences necessary to complement a non-functional pat herbicide resistance gene. In addition, a yeast-based assay was used to identify ZFNs capable of cleaving a native endochitinase gene. Agrobacterium delivery of a Ti plasmid harboring both the ZFNs and a donor DNA construct comprising a pat herbicide resistance gene cassette flanked by short stretches of homology to the endochitinase locus yielded up to 10% targeted, homology-directed transgene integration precisely into the ZFN cleavage site. Given that ZFNs can be designed to recognize a wide range of target sequences, these data point toward a novel approach for targeted gene addition, replacement and trait stacking in plants.


Assuntos
Endonucleases/metabolismo , Transgenes/genética , Dedos de Zinco/genética , Sequência de Aminoácidos , Sequência de Bases , Sítios de Ligação , Células Cultivadas , Quitinases/genética , Endonucleases/genética , Glucuronidase/genética , Glucuronidase/metabolismo , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Dados de Sequência Molecular , Reação em Cadeia da Polimerase , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Recombinação Genética , Nicotiana/citologia , Nicotiana/genética , Nicotiana/metabolismo , Transfecção/métodos
20.
Diabetes ; 55(6): 1847-54, 2006 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-16731852

RESUMO

Peripheral neuropathy is a common, irreversible complication of diabetes. We investigated whether gene transfer of an engineered zinc finger protein transcription factor (ZFP-TF) designed to upregulate expression of the endogenous vascular endothelial growth factor (VEGF)-A gene could protect against experimental diabetic neuropathy. ZFP-TF-driven activation of the endogenous gene results in expression of all of the VEGF-A isoforms, a fact that may be of significance for recapitulation of the proper biological responses stimulated by this potent neuroprotective growth factor. We show here that this engineered ZFP-TF activates VEGF-A in appropriate cells in culture and that the secreted VEGF-A protein induced by the ZFP protects neuroblastoma cell lines from a serum starvation insult in vitro. Importantly, single and repeat intramuscular injections of formulated plasmid DNA encoding the VEGF-A-activating ZFP-TF resulted in protection of both sensory and motor nerve conduction velocities in a streptozotocin-induced rat model of diabetes. These data suggest that VEGF-A-activating ZFP-TFs may ultimately be of clinical utility in the treatment of this disease.


Assuntos
Neuropatias Diabéticas/terapia , Terapia Genética/métodos , Fatores de Transcrição/fisiologia , Fator A de Crescimento do Endotélio Vascular/fisiologia , Dedos de Zinco/genética , Animais , Linhagem Celular , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/genética , Sobrevivência Celular/fisiologia , Células Cultivadas , Meios de Cultura Livres de Soro/farmacologia , Diabetes Mellitus Experimental/induzido quimicamente , Diabetes Mellitus Experimental/complicações , Neuropatias Diabéticas/etiologia , Neuropatias Diabéticas/fisiopatologia , Expressão Gênica , Vetores Genéticos/genética , Humanos , Ratos , Retroviridae/genética , Estreptozocina/toxicidade , Fatores de Transcrição/genética , Transfecção , Fator A de Crescimento do Endotélio Vascular/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA