Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
1.
Cell ; 140(5): 678-91, 2010 Mar 05.
Artículo en Inglés | MEDLINE | ID: mdl-20211137

RESUMEN

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.


Asunto(s)
Histonas/análisis , Telómero/química , Animales , Sitios de Unión , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Células Madre Embrionarias/metabolismo , Genoma , Chaperonas de Histonas/genética , Chaperonas de Histonas/metabolismo , Histonas/genética , Histonas/metabolismo , Ratones , Ratones Endogámicos C57BL , Telómero/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Sitio de Iniciación de la Transcripción
2.
Mol Ther ; 27(4): 866-877, 2019 04 10.
Artículo en Inglés | MEDLINE | ID: mdl-30902585

RESUMEN

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


Asunto(s)
Sistemas de Liberación de Medicamentos/métodos , Edición Génica/métodos , Nanopartículas/administración & dosificación , ARN Mensajero/administración & dosificación , Nucleasas con Dedos de Zinc/administración & dosificación , Animales , Células Cultivadas , Dependovirus/genética , Femenino , Técnicas de Inactivación de Genes , Vectores Genéticos , Hepatocitos/metabolismo , Intrones/genética , Lípidos/química , Masculino , Ratones , Ratones Endogámicos C57BL , Prealbúmina/genética , Proproteína Convertasa 9/genética , ARN Mensajero/genética , Transgenes/genética , Nucleasas con Dedos de Zinc/farmacología
3.
Mol Ther ; 27(1): 178-187, 2019 01 02.
Artículo en Inglés | MEDLINE | ID: mdl-30528089

RESUMEN

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


Asunto(s)
Edición Génica/métodos , Terapia Genética/métodos , Mucopolisacaridosis I/terapia , Nucleasas con Dedos de Zinc/metabolismo , Animales , Modelos Animales de Enfermedad , Terapia de Reemplazo Enzimático , Femenino , Glicosaminoglicanos/metabolismo , Iduronidasa/metabolismo , Enfermedades por Almacenamiento Lisosomal/tratamiento farmacológico , Enfermedades por Almacenamiento Lisosomal/metabolismo , Enfermedades por Almacenamiento Lisosomal/terapia , Masculino , Ratones , Mucopolisacaridosis I/tratamiento farmacológico , Mucopolisacaridosis I/metabolismo , Nucleasas con Dedos de Zinc/genética
4.
Mol Ther ; 26(4): 1127-1136, 2018 04 04.
Artículo en Inglés | MEDLINE | ID: mdl-29580682

RESUMEN

Mucopolysaccharidosis type II (MPS II) is an X-linked recessive lysosomal disorder caused by deficiency of iduronate 2-sulfatase (IDS), leading to accumulation of glycosaminoglycans (GAGs) in tissues of affected individuals, progressive disease, and shortened lifespan. Currently available enzyme replacement therapy (ERT) requires lifelong infusions and does not provide neurologic benefit. We utilized a zinc finger nuclease (ZFN)-targeting system to mediate genome editing for insertion of the human IDS (hIDS) coding sequence into a "safe harbor" site, intron 1 of the albumin locus in hepatocytes of an MPS II mouse model. Three dose levels of recombinant AAV2/8 vectors encoding a pair of ZFNs and a hIDS cDNA donor were administered systemically in MPS II mice. Supraphysiological, vector dose-dependent levels of IDS enzyme were observed in the circulation and peripheral organs of ZFN+donor-treated mice. GAG contents were markedly reduced in tissues from all ZFN+donor-treated groups. Surprisingly, we also demonstrate that ZFN-mediated genome editing prevented the development of neurocognitive deficit in young MPS II mice (6-9 weeks old) treated at high vector dose levels. We conclude that this ZFN-based platform for expression of therapeutic proteins from the albumin locus is a promising approach for treatment of MPS II and other lysosomal diseases.


Asunto(s)
Metabolismo Energético , Dosificación de Gen , Edición Génica , Iduronato Sulfatasa/genética , Mucopolisacaridosis II/genética , Mucopolisacaridosis II/metabolismo , Fenotipo , Animales , Biomarcadores , Modelos Animales de Enfermedad , Endonucleasas/genética , Endonucleasas/metabolismo , Activación Enzimática , Técnicas de Transferencia de Gen , Hepatocitos/metabolismo , Intrones , Ratones , Mucopolisacaridosis II/patología , Mucopolisacaridosis II/fisiopatología , Dedos de Zinc/genética
5.
Mol Cell ; 42(2): 185-98, 2011 Apr 22.
Artículo en Inglés | MEDLINE | ID: mdl-21504830

RESUMEN

It has been suspected that cell-cycle progression might be functionally coupled with RNA processing. However, little is known about the role of the precise splicing control in cell-cycle progression. Here, we report that SON, a large Ser/Arg (SR)-related protein, is a splicing cofactor contributing to efficient splicing of cell-cycle regulators. Downregulation of SON leads to severe impairment of spindle pole separation, microtubule dynamics, and genome integrity. These molecular defects result from inadequate RNA splicing of a specific set of cell-cycle-related genes that possess weak splice sites. Furthermore, we show that SON facilitates the interaction of SR proteins with RNA polymerase II and other key spliceosome components, suggesting its function in efficient cotranscriptional RNA processing. These results reveal a mechanism for controlling cell-cycle progression through SON-dependent constitutive splicing at suboptimal splice sites, with strong implications for its role in cancer and other human diseases.


Asunto(s)
Ciclo Celular/genética , Proteínas de Unión al ADN/metabolismo , Genes cdc , Empalme del ARN , Empalmosomas/metabolismo , Segregación Cromosómica , Citocinesis , Proteínas de Unión al ADN/genética , Inestabilidad Genómica , Células HEK293 , Células HeLa , Humanos , Células K562 , Microtúbulos/metabolismo , Antígenos de Histocompatibilidad Menor , Interferencia de ARN , ARN Polimerasa II/metabolismo , Huso Acromático/metabolismo , Factores de Tiempo , Transfección
6.
Blood ; 126(15): 1777-84, 2015 Oct 08.
Artículo en Inglés | MEDLINE | ID: mdl-26297739

RESUMEN

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.


Asunto(s)
Albúminas/genética , Terapia de Reemplazo Enzimático , Terapia Genética , Genoma , Hígado/metabolismo , Transgenes/fisiología , Albúminas/metabolismo , Animales , Dependovirus/genética , Endonucleasas , Enfermedad de Fabry/genética , Enfermedad de Fabry/terapia , Factor IX/genética , Factor VIII/genética , Enfermedad de Gaucher/genética , Enfermedad de Gaucher/terapia , Vectores Genéticos/administración & dosificación , Hemofilia A/genética , Hemofilia A/terapia , Hemofilia B/genética , Hemofilia B/terapia , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Lisosomas/enzimología , Ratones , Ratones Endogámicos C57BL , Mucopolisacaridosis I/genética , Mucopolisacaridosis I/terapia , Mucopolisacaridosis II/genética , Mucopolisacaridosis II/terapia , Regiones Promotoras Genéticas/genética , Edición de ARN , ARN Mensajero/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Dedos de Zinc
7.
PLoS Genet ; 9(10): e1003765, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24130502

RESUMEN

Fusion protein RUNX1-ETO (AML1-ETO, RUNX1-RUNX1T1) is expressed as the result of the 8q22;21q22 translocation [t(8;21)], which is one of the most common chromosomal abnormalities found in acute myeloid leukemia. RUNX1-ETO is thought to promote leukemia development through the aberrant regulation of RUNX1 (AML1) target genes. Repression of these genes occurs via the recruitment of the corepressors N-COR and SMRT due to their interaction with ETO. Mechanisms of RUNX1-ETO target gene upregulation remain less well understood. Here we show that RUNX1-ETO9a, the leukemogenic alternatively spliced transcript expressed from t(8;21), upregulates target gene Alox5, which is a gene critically required for the promotion of chronic myeloid leukemia development by BCR-ABL. Loss of Alox5 expression reduces activity of RUNX1-ETO9a, MLL-AF9 and PML-RARα in vitro. However, Alox5 is not essential for the induction of leukemia by RUNX1-ETO9a in vivo. Finally, we demonstrate that the upregulation of Alox5 by RUNX1-ETO9a occurs via the C2H2 zinc finger transcription factor KLF6, a protein required for early hematopoiesis and yolk sac development. Furthermore, KLF6 is specifically upregulated by RUNX1-ETO in human leukemia cells. This identifies KLF6 as a novel mediator of t(8;21) target gene regulation, providing a new mechanism for RUNX1-ETO transcriptional control.


Asunto(s)
Araquidonato 5-Lipooxigenasa/genética , Subunidad alfa 2 del Factor de Unión al Sitio Principal/genética , Factores de Transcripción de Tipo Kruppel/genética , Leucemia Mieloide Aguda/genética , Proteínas Proto-Oncogénicas/genética , Empalme Alternativo , Animales , Línea Celular Tumoral , Aberraciones Cromosómicas , Regulación Leucémica de la Expresión Génica , Humanos , Factor 6 Similar a Kruppel , Leucemia Mieloide Aguda/patología , Ratones , Proteínas de Fusión Oncogénica/genética , Proteína 1 Compañera de Translocación de RUNX1 , Factores de Transcripción/genética
8.
Blood ; 121(18): 3714-7, 2013 May 02.
Artículo en Inglés | MEDLINE | ID: mdl-23426948

RESUMEN

AML1-ETO (RUNX1-ETO) fusion proteins are generated by the 8;21 translocation, commonly found in acute myeloid leukemia, which fuses the AML1 (RUNX1) and ETO (MTG8, RUNX1T1) genes. Previous studies have shown that AML1-ETO interferes with AML1 function but requires additional cooperating mutations to induce leukemia development. In mouse models, AML1-ETO forms lacking the C-terminus have been shown to have greatly enhanced leukemogenic potential. Here, we investigate the role of 2 AML1-ETO C-terminal-interacting proteins, N-CoR, a transcriptional corepressor, and SON, a splicing/transcription factor required for cell cycle progression, in AML1-ETO-induced leukemia development. AML1-ETO-W692A loses N-CoR binding at NHR4, displays attenuated transcriptional repression ability and decreased cellular dysregulation, and promotes leukemia in vivo. These results support the importance of the degree of dysregulation by AML1-ETO in cellular transformation and demonstrate that AML1-ETO-W692A can be used as an effective experimental model for determining which factors compromise the leukemogenic potential of AML1-ETO.


Asunto(s)
Transformación Celular Neoplásica/genética , Subunidad alfa 2 del Factor de Unión al Sitio Principal/genética , Leucemia/genética , Proteínas de Fusión Oncogénica/genética , Animales , Células Cultivadas , Subunidad alfa 2 del Factor de Unión al Sitio Principal/metabolismo , Regulación hacia Abajo/genética , Regulación Leucémica de la Expresión Génica , Células HEK293 , Humanos , Células K562 , Leucemia/patología , Ratones , Ratones Endogámicos C57BL , Co-Represor 1 de Receptor Nuclear/metabolismo , Proteínas de Fusión Oncogénica/metabolismo , Unión Proteica/genética , Proteína 1 Compañera de Translocación de RUNX1
9.
Nature ; 459(7245): 437-41, 2009 May 21.
Artículo en Inglés | MEDLINE | ID: mdl-19404259

RESUMEN

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.


Asunto(s)
Biotecnología/métodos , Desoxirribonucleasas/química , Desoxirribonucleasas/metabolismo , Marcación de Gen/métodos , Genoma de Planta/genética , Zea mays/genética , Dedos de Zinc , Desoxirribonucleasas/genética , Alimentos Modificados Genéticamente , Genes de Plantas/genética , Resistencia a los Herbicidas/genética , Herbicidas/farmacología , Herencia , Fosfatos de Inositol/metabolismo , Mutagénesis Sitio-Dirigida/métodos , Plantas Modificadas Genéticamente , Recombinación Genética/genética , Reproducibilidad de los Resultados
10.
J Biol Chem ; 288(8): 5381-8, 2013 Feb 22.
Artículo en Inglés | MEDLINE | ID: mdl-23322776

RESUMEN

SON is a DNA- and RNA-binding protein localized in nuclear speckles. Although its function in RNA splicing for effective cell cycle progression and genome stability was recently unveiled, other mechanisms of SON functions remain unexplored. Here, we report that SON regulates GATA-2, a key transcription factor involved in hematopoietic stem cell maintenance and differentiation. SON is highly expressed in undifferentiated hematopoietic stem/progenitor cells and leukemic blasts. SON knockdown leads to significant depletion of GATA-2 protein with marginal down-regulation of GATA-2 mRNA. We show that miR-27a is up-regulated upon SON knockdown and targets the 3'-UTR of GATA-2 mRNA in hematopoietic cells. Up-regulation of miR-27a was due to activation of the promoter of the miR-23a∼27a∼24-2 cluster, suggesting that SON suppresses this promoter to lower the microRNAs from this cluster. Our data revealed a previously unidentified role of SON in microRNA production via regulating the transcription process, thereby modulating GATA-2 at the protein level during hematopoietic differentiation.


Asunto(s)
Proteínas de Unión al ADN/fisiología , Factor de Transcripción GATA2/metabolismo , Regulación de la Expresión Génica , MicroARNs/metabolismo , Regiones no Traducidas 3' , Animales , Diferenciación Celular , Proteínas de Unión al ADN/genética , Hematopoyesis , Humanos , Células K562 , Leucemia Mieloide Aguda/metabolismo , Ratones , Ratones Endogámicos C57BL , Antígenos de Histocompatibilidad Menor , Modelos Biológicos , Regiones Promotoras Genéticas , Empalme del ARN , ARN Mensajero/metabolismo , Células U937 , Regulación hacia Arriba
11.
Genome Res ; 20(8): 1133-42, 2010 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-20508142

RESUMEN

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.


Asunto(s)
Técnicas de Transferencia de Gen , Genoma Humano , Genómica/métodos , Secuencias Reguladoras de Ácidos Nucleicos/genética , Dedos de Zinc/genética , Secuencia de Bases , Línea Celular , Endonucleasas/genética , Sitios Genéticos , Humanos , Datos de Secuencia Molecular , Proteómica/métodos
12.
Plant Biotechnol J ; 10(7): 783-91, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22520333

RESUMEN

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


Asunto(s)
3-Oxoacil-(Proteína Transportadora de Acil) Sintasa/genética , Brassica napus/enzimología , Brassica napus/genética , Ingeniería Genética/métodos , Factores de Transcripción/metabolismo , Activación Transcripcional/genética , Dedos de Zinc/genética , 3-Oxoacil-(Proteína Transportadora de Acil) Sintasa/metabolismo , Secuencia de Bases , Cruzamientos Genéticos , ADN Complementario/genética , Activación Enzimática , Ácidos Grasos/metabolismo , Regulación de la Expresión Génica de las Plantas , Genes de Plantas/genética , Datos de Secuencia Molecular , Hojas de la Planta/enzimología , Hojas de la Planta/genética , Plantas Modificadas Genéticamente , ARN Mensajero/genética , ARN Mensajero/metabolismo , Semillas/metabolismo , Factores de Transcripción/genética
13.
Nucleic Acids Res ; 38(15): e152, 2010 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-20530528

RESUMEN

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


Asunto(s)
Desoxirribonucleasas de Localización Especificada Tipo II/metabolismo , Marcación de Gen/métodos , Dedos de Zinc , Animales , Células CHO , Cromosomas de los Mamíferos/química , Cricetinae , Cricetulus , ADN/química , Roturas del ADN de Doble Cadena , Desoxirribonucleasas de Localización Especificada Tipo II/química , Genoma , Humanos , Células K562 , Homología de Secuencia de Ácido Nucleico
14.
Blood Cancer J ; 10(2): 16, 2020 02 06.
Artículo en Inglés | MEDLINE | ID: mdl-32029705

RESUMEN

Large-scale chromosomal translocations are frequent oncogenic drivers in acute myeloid leukemia (AML). These translocations often occur in critical transcriptional/epigenetic regulators and contribute to malignant cell growth through alteration of normal gene expression. Despite this knowledge, the specific gene expression alterations that contribute to the development of leukemia remain incompletely understood. Here, through characterization of transcriptional regulation by the RUNX1-ETO fusion protein, we have identified Ras-association domain family member 2 (RASSF2) as a critical gene that is aberrantly transcriptionally repressed in t(8;21)-associated AML. Re-expression of RASSF2 specifically inhibits t(8;21) AML development in multiple models. Through biochemical and functional studies, we demonstrate RASSF2-mediated functions to be dependent on interaction with Hippo kinases, MST1 and MST2, but independent of canonical Hippo pathway signaling. Using proximity-based biotin labeling we define the RASSF2-proximal proteome in leukemia cells and reveal association with Rac GTPase-related proteins, including an interaction with the guanine nucleotide exchange factor, DOCK2. Importantly, RASSF2 knockdown impairs Rac GTPase activation, and RASSF2 expression is broadly correlated with Rac-mediated signal transduction in AML patients. Together, these data reveal a previously unappreciated mechanistic link between RASSF2, Hippo kinases, and Rac activity with potentially broad functional consequences in leukemia.


Asunto(s)
Cromosomas Humanos Par 21/genética , Cromosomas Humanos Par 8/genética , Regulación Neoplásica de la Expresión Génica , Leucemia Mieloide Aguda/prevención & control , Proteínas de Fusión Oncogénica/metabolismo , Translocación Genética , Proteínas Supresoras de Tumor/metabolismo , Proteínas de Unión al GTP rac/metabolismo , Animales , Biomarcadores de Tumor/genética , Humanos , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/metabolismo , Leucemia Mieloide Aguda/patología , Ratones , Ratones Endogámicos C57BL , Proteínas de Fusión Oncogénica/genética , ARN Largo no Codificante , Células Tumorales Cultivadas , Proteínas Supresoras de Tumor/genética , Ensayos Antitumor por Modelo de Xenoinjerto , Proteínas de Unión al GTP rac/genética
15.
Mol Cell Biol ; 35(17): 3071-82, 2015 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-26124281

RESUMEN

Myelodysplastic syndromes (MDS) are a group of neoplasms characterized by ineffective myeloid hematopoiesis and various risks for leukemia. SRSF2, a member of the serine/arginine-rich (SR) family of splicing factors, is one of the mutation targets associated with poor survival in patients suffering from myelodysplastic syndromes. Here we report the biological function of SRSF2 in hematopoiesis by using conditional knockout mouse models. Ablation of SRSF2 in the hematopoietic lineage caused embryonic lethality, and Srsf2-deficient fetal liver cells showed significantly enhanced apoptosis and decreased levels of hematopoietic stem/progenitor cells. Induced ablation of SRSF2 in adult Mx1-Cre Srsf2(flox/flox) mice upon poly(I):poly(C) injection demonstrated a significant decrease in lineage(-) Sca(+) c-Kit(+) cells in bone marrow. To reveal the functional impact of myelodysplastic syndromes-associated mutations in SRSF2, we analyzed splicing responses on the MSD-L cell line and found that the missense mutation of proline 95 to histidine (P95H) and a P95-to-R102 in-frame 8-amino-acid deletion caused significant changes in alternative splicing. The affected genes were enriched in cancer development and apoptosis. These findings suggest that intact SRSF2 is essential for the functional integrity of the hematopoietic system and that its mutations likely contribute to development of myelodysplastic syndromes.


Asunto(s)
Células Sanguíneas/citología , Hematopoyesis/genética , Células Madre Hematopoyéticas/citología , Síndromes Mielodisplásicos/genética , Proteínas Nucleares/genética , Ribonucleoproteínas/genética , Secuencia de Aminoácidos , Animales , Apoptosis/genética , Células de la Médula Ósea/citología , Trasplante de Médula Ósea , Línea Celular , Supervivencia Celular/genética , Células HEK293 , Humanos , Hígado/patología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Mutación/genética , Poli I-C/farmacología , Interferencia de ARN , Precursores del ARN/metabolismo , Empalme del ARN/genética , ARN Interferente Pequeño , Factores de Empalme Serina-Arginina
16.
Leuk Lymphoma ; 55(4): 884-91, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23772668

RESUMEN

The 8;21 translocation is the most common chromosomal aberration occurring in acute myeloid leukemia (AML). This translocation causes expression of the RUNX1-ETO (AML1-ETO) fusion protein, which cooperates with additional mutations in leukemia development. We report here that interferons (IFNs) and IFN-stimulated genes are a group of genes consistently up-regulated by RUNX1-ETO in both human and murine models. RUNX1-ETO-induced up-regulation of IFN-stimulated genes occurs primarily via type I IFN signaling with a requirement for the IFNAR complex. Addition of exogenous IFN in vitro significantly reduces the increase in self-renewal potential induced by both RUNX1-ETO and its leukemogenic splicing isoform RUNX1-ETO9a. Finally, loss of type I IFN signaling via knockout of Ifnar1 significantly accelerates leukemogenesis in a t(8;21) murine model. This demonstrates the role of increased IFN signaling as an important factor inhibiting t(8;21) fusion protein function and leukemia development and supports the use of type I IFNs in the treatment of AML.


Asunto(s)
Subunidad alfa 2 del Factor de Unión al Sitio Principal/genética , Regulación Leucémica de la Expresión Génica/efectos de los fármacos , Interferón Tipo I/farmacología , Leucemia/genética , Proteínas de Fusión Oncogénica/genética , Proteínas Proto-Oncogénicas/genética , Factores de Transcripción/genética , Translocación Genética , Animales , Línea Celular Tumoral , Transformación Celular Neoplásica/genética , Cromosomas Humanos Par 21 , Cromosomas Humanos Par 8 , Modelos Animales de Enfermedad , Humanos , Leucemia/metabolismo , Ratones , Ratones Noqueados , Proteínas de Fusión Oncogénica/metabolismo , Proteína 1 Compañera de Translocación de RUNX1 , Receptor de Interferón alfa y beta/deficiencia , Receptor de Interferón alfa y beta/genética , Receptor de Interferón alfa y beta/metabolismo , Células U937
17.
Nat Biotechnol ; 27(9): 851-7, 2009 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-19680244

RESUMEN

Realizing the full potential of human embryonic stem cells (hESCs) and induced pluripotent stem cells (hiPSCs) requires efficient methods for genetic modification. However, techniques to generate cell type-specific lineage reporters, as well as reliable tools to disrupt, repair or overexpress genes by gene targeting, are inefficient at best and thus are not routinely used. Here we report the highly efficient targeting of three genes in human pluripotent cells using zinc-finger nuclease (ZFN)-mediated genome editing. First, using ZFNs specific for the OCT4 (POU5F1) locus, we generated OCT4-eGFP reporter cells to monitor the pluripotent state of hESCs. Second, we inserted a transgene into the AAVS1 locus to generate a robust drug-inducible overexpression system in hESCs. Finally, we targeted the PITX3 gene, demonstrating that ZFNs can be used to generate reporter cells by targeting non-expressed genes in hESCs and hiPSCs.


Asunto(s)
Desoxirribonucleasas/metabolismo , Células Madre Embrionarias/fisiología , Marcación de Gen/métodos , Células Madre Pluripotentes/fisiología , Dedos de Zinc/fisiología , Línea Celular , Desoxirribonucleasas/genética , Expresión Génica , Silenciador del Gen , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Humanos , Inmunohistoquímica , Factor 3 de Transcripción de Unión a Octámeros/genética , Factor 3 de Transcripción de Unión a Octámeros/metabolismo , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
18.
Plant Mol Biol ; 69(6): 699-709, 2009 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-19112554

RESUMEN

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.


Asunto(s)
Endonucleasas/metabolismo , Transgenes/genética , Dedos de Zinc/genética , Secuencia de Aminoácidos , Secuencia de Bases , Sitios de Unión , Células Cultivadas , Quitinasas/genética , Endonucleasas/genética , Glucuronidasa/genética , Glucuronidasa/metabolismo , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Datos de Secuencia Molecular , Reacción en Cadena de la Polimerasa , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Recombinación Genética , Nicotiana/citología , Nicotiana/genética , Nicotiana/metabolismo , Transfección/métodos
19.
Proc Natl Acad Sci U S A ; 104(9): 3055-60, 2007 Feb 27.
Artículo en Inglés | MEDLINE | ID: mdl-17360608

RESUMEN

Efficient incorporation of novel DNA sequences into a specific site in the genome of living human cells remains a challenge despite its potential utility to genetic medicine, biotechnology, and basic research. We find that a precisely placed double-strand break induced by engineered zinc finger nucleases (ZFNs) can stimulate integration of long DNA stretches into a predetermined genomic location, resulting in high-efficiency site-specific gene addition. Using an extrachromosomal DNA donor carrying a 12-bp tag, a 900-bp ORF, or a 1.5-kb promoter-transcription unit flanked by locus-specific homology arms, we find targeted integration frequencies of 15%, 6%, and 5%, respectively, within 72 h of treatment, and with no selection for the desired event. Importantly, we find that the integration event occurs in a homology-directed manner and leads to the accurate reconstruction of the donor-specified genotype at the endogenous chromosomal locus, and hence presumably results from synthesis-dependent strand annealing repair of the break using the donor DNA as a template. This site-specific gene addition occurs with no measurable increase in the rate of random integration. Remarkably, we also find that ZFNs can drive the addition of an 8-kb sequence carrying three distinct promoter-transcription units into an endogenous locus at a frequency of 6%, also in the absence of any selection. These data reveal the surprising versatility of the specialized polymerase machinery involved in double-strand break repair, illuminate a powerful approach to mammalian cell engineering, and open the possibility of ZFN-driven gene addition therapy for human genetic disease.


Asunto(s)
Desoxirribonucleasas/genética , Marcación de Gen/métodos , Técnicas de Transferencia de Gen , Ingeniería Genética/métodos , Genoma Humano/genética , Dedos de Zinc/genética , Secuencia de Bases , Estudios de Evaluación como Asunto , Humanos , Datos de Secuencia Molecular
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA