Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
1.
J Cyst Fibros ; 21(1): 164-171, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34049825

RESUMEN

BACKGROUND: Mechanisms governing the diversity of CFTR gene expression throughout the body are complex. Multiple intronic and distal regulatory elements are responsible for regulating differential CFTR expression across tissues. METHODS: Drawing on published data, 18 high-priority genomic regions were identified and interrogated for CFTR-enhancer function using CRISPR/dCas9-based epigenome editing tools. Each region was evaluated by dCas9p300 and dCas9KRAB for its ability to enhance or repress CFTR expression, respectively. RESULTS: Multiple genomic regions were tested for enhancer activity using CRISPR/dCas9 epigenome editing. dCas9p300 mediates a significant increase in CFTR mRNA levels when targeted to the promoter and a region 44 kb upstream of the transcriptional start site in a CFTR-low expressing cell line. Multiple gRNAs targeting the promoter induced a robust increase in CFTR protein levels. In contrast, dCas9KRAB-mediated repression is much more robust with 10 of the 18 evaluated genomic regions inducing CFTR protein knockdown. To evaluate the therapeutic efficacy of modulating CFTR gene regulation, dCas9p300 was used to induce elevated levels of CFTR from the endogenous locus in ΔF508/ΔF508 human bronchial epithelial cells. Ussing chamber studies demonstrated a synergistic increase in ion transport in response to CRISPR-induced expression of ΔF508 CFTR mRNA along with VX809 treatment. CONCLUSIONS: CRISPR/dCas9-based epigenome-editing provides a previously unexplored tool for interrogating CFTR enhancer function. Here, we demonstrate that therapeutic interventions that increase the expression of CFTR may improve the efficacy of CFTR modulators. A better understanding CFTR regulatory mechanisms could uncover novel therapeutic interventions for the development of cystic fibrosis therapies.


Asunto(s)
Proteína 9 Asociada a CRISPR/genética , Sistemas CRISPR-Cas/genética , Regulador de Conductancia de Transmembrana de Fibrosis Quística/genética , Fibrosis Quística/genética , Edición Génica/métodos , Fibrosis Quística/tratamiento farmacológico , Regulador de Conductancia de Transmembrana de Fibrosis Quística/uso terapéutico , Epigenoma , Regulación de la Expresión Génica , Células HEK293 , Humanos , ARN Guía de Kinetoplastida/genética
2.
SLAS Discov ; 25(8): 823-842, 2020 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-32026742

RESUMEN

Many diseases, such as diabetes, autoimmune diseases, cancer, and neurological disorders, are caused by a dysregulation of a complex interplay of genes. Genome-wide association studies have identified thousands of disease-linked polymorphisms in the human population. However, detailing the causative gene expression or functional changes underlying those associations has been elusive in many cases. Functional genomics is an emerging field of research that aims to deconvolute the link between genotype and phenotype by making use of large -omic data sets and next-generation gene and epigenome editing tools to perturb genes of interest. Here we review how functional genomic tools can be used to better understand the biological interplay between genes, improve disease modeling, and identify novel drug targets. Incorporation of functional genomic capabilities into conventional drug development pipelines is predicted to expedite the development of first-in-class therapeutics.


Asunto(s)
Descubrimiento de Drogas/tendencias , Predisposición Genética a la Enfermedad , Genoma Humano/genética , Genómica/tendencias , Sistemas CRISPR-Cas/genética , Epigenoma/genética , Edición Génica , Estudio de Asociación del Genoma Completo , Genotipo , Humanos , Fenotipo , ARN Interferente Pequeño/genética
3.
Nucleic Acids Res ; 45(20): 11684-11699, 2017 Nov 16.
Artículo en Inglés | MEDLINE | ID: mdl-28977539

RESUMEN

Our current understanding of cellular transdifferentiation systems is limited. It is oftentimes unknown, at a genome-wide scale, how much transdifferentiated cells differ quantitatively from both the starting cells and the target cells. Focusing on transdifferentiation of primary human skin fibroblasts by forced expression of myogenic transcription factor MyoD, we performed quantitative analyses of gene expression and chromatin accessibility profiles of transdifferentiated cells compared to fibroblasts and myoblasts. In this system, we find that while many of the early muscle marker genes are reprogrammed, global gene expression and accessibility changes are still incomplete when compared to myoblasts. In addition, we find evidence of epigenetic memory in the transdifferentiated cells, with reminiscent features of fibroblasts being visible both in chromatin accessibility and gene expression. Quantitative analyses revealed a continuum of changes in chromatin accessibility induced by MyoD, and a strong correlation between chromatin-remodeling deficiencies and incomplete gene expression reprogramming. Classification analyses identified genetic and epigenetic features that distinguish reprogrammed from non-reprogrammed sites, and suggested ways to potentially improve transdifferentiation efficiency. Our approach for combining gene expression, DNA accessibility, and protein-DNA binding data to quantify and characterize the efficiency of cellular transdifferentiation on a genome-wide scale can be applied to any transdifferentiation system.


Asunto(s)
Transdiferenciación Celular/genética , Reprogramación Celular/genética , Ensamble y Desensamble de Cromatina/genética , Proteína MioD/genética , Western Blotting , Células Cultivadas , Cromatina/genética , Cromatina/metabolismo , Fibroblastos/citología , Fibroblastos/metabolismo , Perfilación de la Expresión Génica/métodos , Ontología de Genes , Células HEK293 , Humanos , Microscopía Confocal , Proteína MioD/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Piel/citología
4.
Nat Methods ; 12(12): 1143-9, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26501517

RESUMEN

Epigenome editing with the CRISPR (clustered, regularly interspaced, short palindromic repeats)-Cas9 platform is a promising technology for modulating gene expression to direct cell phenotype and to dissect the causal epigenetic mechanisms of gene regulation. Fusions of nuclease-inactive dCas9 to the Krüppel-associated box (KRAB) repressor (dCas9-KRAB) can silence target gene expression, but the genome-wide specificity and the extent of heterochromatin formation catalyzed by dCas9-KRAB are not known. We targeted dCas9-KRAB to the HS2 enhancer, a distal regulatory element that orchestrates the expression of multiple globin genes, and observed highly specific induction of H3K9 trimethylation (H3K9me3) at the enhancer and decreased chromatin accessibility of both the enhancer and its promoter targets. Targeted epigenetic modification of HS2 silenced the expression of multiple globin genes, with minimal off-target changes in global gene expression. These results demonstrate that repression mediated by dCas9-KRAB is sufficiently specific to disrupt the activity of individual enhancers via local modification of the epigenome.


Asunto(s)
Proteínas Asociadas a CRISPR/genética , Sistemas CRISPR-Cas/genética , Repeticiones Palindrómicas Cortas Agrupadas y Regularmente Espaciadas/genética , Epigénesis Genética , Epigenómica/métodos , Elementos Reguladores de la Transcripción/genética , Elementos de Facilitación Genéticos , Regulación Viral de la Expresión Génica , Globinas/genética , Células HEK293 , Humanos , Células K562 , Lentivirus/genética , ARN Guía de Kinetoplastida/genética , Proteínas Represoras/genética , Proteínas Virales/genética
5.
Nat Commun ; 6: 6244, 2015 Feb 18.
Artículo en Inglés | MEDLINE | ID: mdl-25692716

RESUMEN

The CRISPR/Cas9 genome-editing platform is a promising technology to correct the genetic basis of hereditary diseases. The versatility, efficiency and multiplexing capabilities of the CRISPR/Cas9 system enable a variety of otherwise challenging gene correction strategies. Here, we use the CRISPR/Cas9 system to restore the expression of the dystrophin gene in cells carrying dystrophin mutations that cause Duchenne muscular dystrophy (DMD). We design single or multiplexed sgRNAs to restore the dystrophin reading frame by targeting the mutational hotspot at exons 45-55 and introducing shifts within exons or deleting one or more exons. Following gene editing in DMD patient myoblasts, dystrophin expression is restored in vitro. Human dystrophin is also detected in vivo after transplantation of genetically corrected patient cells into immunodeficient mice. Importantly, the unique multiplex gene-editing capabilities of the CRISPR/Cas9 system facilitate the generation of a single large deletion that can correct up to 62% of DMD mutations.


Asunto(s)
Sistemas CRISPR-Cas/genética , Distrofina/genética , Genoma , Distrofia Muscular de Duchenne/genética , Mutación , Animales , Separación Celular , Modelos Animales de Enfermedad , Exones , Citometría de Flujo , Eliminación de Gen , Terapia Genética/métodos , Células HEK293 , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Masculino , Ratones , Ratones SCID , Plásmidos/metabolismo , Reacción en Cadena de la Polimerasa
6.
Mol Ther ; 23(3): 523-32, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25492562

RESUMEN

Duchenne muscular dystrophy (DMD) is caused by genetic mutations that result in the absence of dystrophin protein expression. Oligonucleotide-induced exon skipping can restore the dystrophin reading frame and protein production. However, this requires continuous drug administration and may not generate complete skipping of the targeted exon. In this study, we apply genome editing with zinc finger nucleases (ZFNs) to permanently remove essential splicing sequences in exon 51 of the dystrophin gene and thereby exclude exon 51 from the resulting dystrophin transcript. This approach can restore the dystrophin reading frame in ~13% of DMD patient mutations. Transfection of two ZFNs targeted to sites flanking the exon 51 splice acceptor into DMD patient myoblasts led to deletion of this genomic sequence. A clonal population was isolated with this deletion and following differentiation we confirmed loss of exon 51 from the dystrophin mRNA transcript and restoration of dystrophin protein expression. Furthermore, transplantation of corrected cells into immunodeficient mice resulted in human dystrophin expression localized to the sarcolemmal membrane. Finally, we quantified ZFN toxicity in human cells and mutagenesis at predicted off-target sites. This study demonstrates a powerful method to restore the dystrophin reading frame and protein expression by permanently deleting exons.


Asunto(s)
Distrofina/genética , Exones , Terapia Genética/métodos , Edición de ARN , ARN Mensajero/genética , Dedos de Zinc/genética , Animales , Secuencia de Bases , Distrofina/biosíntesis , Distrofina/química , Electroporación , Endonucleasas/genética , Endonucleasas/metabolismo , Humanos , Ratones , Ratones Endogámicos NOD , Ratones SCID , Datos de Secuencia Molecular , Distrofia Muscular de Duchenne/genética , Distrofia Muscular de Duchenne/metabolismo , Distrofia Muscular de Duchenne/patología , Distrofia Muscular de Duchenne/terapia , Mioblastos/metabolismo , Mioblastos/patología , Sistemas de Lectura Abierta , Plásmidos/química , Plásmidos/genética , Empalme del ARN , ARN Mensajero/química , ARN Mensajero/metabolismo , Eliminación de Secuencia
7.
ACS Synth Biol ; 4(6): 689-99, 2015 Jun 19.
Artículo en Inglés | MEDLINE | ID: mdl-25494287

RESUMEN

Genetic reprogramming holds great potential for disease modeling, drug screening, and regenerative medicine. Genetic reprogramming of mammalian cells is typically achieved by forced expression of natural transcription factors that control master gene networks and cell lineage specification. However, in many instances, the natural transcription factors do not induce a sufficiently robust response to completely reprogram cell phenotype. In this study, we demonstrate that protein engineering of the master transcription factor MyoD can enhance the conversion of human dermal fibroblasts and adult stem cells to a skeletal myocyte phenotype. Fusion of potent transcriptional activation domains to MyoD led to increased myogenic gene expression, myofiber formation, cell fusion, and global reprogramming of the myogenic gene network. This work supports a general strategy for synthetically enhancing the direct conversion between cell types that can be applied in both synthetic biology and regenerative medicine.


Asunto(s)
Proteína MioD/metabolismo , Células Madre Adultas/citología , Células Madre Adultas/metabolismo , Secuencia de Aminoácidos , Linaje de la Célula , Transdiferenciación Celular , Reprogramación Celular , Dermis/citología , Fibroblastos/citología , Fibroblastos/metabolismo , Regulación de la Expresión Génica , Redes Reguladoras de Genes , Vectores Genéticos/metabolismo , Células HEK293 , Humanos , Lentivirus/genética , Datos de Secuencia Molecular , Desarrollo de Músculos , Fibras Musculares Esqueléticas/citología , Fibras Musculares Esqueléticas/metabolismo , Proteína MioD/química , Proteína MioD/genética , Ingeniería de Proteínas , Estructura Terciaria de Proteína
8.
Stem Cell Reports ; 3(6): 940-7, 2014 Dec 09.
Artículo en Inglés | MEDLINE | ID: mdl-25448066

RESUMEN

Gene activation by the CRISPR/Cas9 system has the potential to enable new approaches to science and medicine, but the technology must be enhanced to robustly control cell behavior. We show that the fusion of two transactivation domains to Cas9 dramatically enhances gene activation to a level that is necessary to reprogram cell phenotype. Targeted activation of the endogenous Myod1 gene locus with this system led to stable and sustained reprogramming of mouse embryonic fibroblasts into skeletal myocytes. The levels of myogenic marker expression obtained by the activation of endogenous Myod1 gene were comparable to that achieved by overexpression of lentivirally delivered MYOD1 transcription factor.


Asunto(s)
Sistemas CRISPR-Cas , Linaje de la Célula/genética , Reprogramación Celular , Animales , Línea Celular , Expresión Génica , Regulación de la Expresión Génica , Ratones , Proteína MioD/genética , Proteína MioD/metabolismo , Especificidad de Órganos/genética , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo
10.
Nucleic Acids Res ; 42(19): e147, 2014 Oct 29.
Artículo en Inglés | MEDLINE | ID: mdl-25122746

RESUMEN

Engineered DNA-binding proteins that manipulate the human genome and transcriptome have enabled rapid advances in biomedical research. In particular, the RNA-guided CRISPR/Cas9 system has recently been engineered to create site-specific double-strand breaks for genome editing or to direct targeted transcriptional regulation. A unique capability of the CRISPR/Cas9 system is multiplex genome engineering by delivering a single Cas9 enzyme and two or more single guide RNAs (sgRNAs) targeted to distinct genomic sites. This approach can be used to simultaneously create multiple DNA breaks or to target multiple transcriptional activators to a single promoter for synergistic enhancement of gene induction. To address the need for uniform and sustained delivery of multiplex CRISPR/Cas9-based genome engineering tools, we developed a single lentiviral system to express a Cas9 variant, a reporter gene and up to four sgRNAs from independent RNA polymerase III promoters that are incorporated into the vector by a convenient Golden Gate cloning method. Each sgRNA is efficiently expressed and can mediate multiplex gene editing and sustained transcriptional activation in immortalized and primary human cells. This delivery system will be significant to enabling the potential of CRISPR/Cas9-based multiplex genome engineering in diverse cell types.


Asunto(s)
Proteínas Asociadas a CRISPR/genética , Sistemas CRISPR-Cas , Ingeniería Celular , Proteínas de Unión al ADN/genética , Vectores Genéticos , Lentivirus/genética , Proteínas Asociadas a CRISPR/metabolismo , Células Cultivadas , Proteínas de Unión al ADN/metabolismo , Fibroblastos/metabolismo , Genoma , Células HEK293 , Humanos , ARN Guía de Kinetoplastida/metabolismo , Transactivadores/genética , Activación Transcripcional
11.
Methods ; 69(2): 188-97, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-25010559

RESUMEN

Engineered DNA-binding proteins that can be targeted to specific sites in the genome to manipulate gene expression have enabled many advances in biomedical research. This includes generating tools to study fundamental aspects of gene regulation and the development of a new class of gene therapies that alter the expression of endogenous genes. Designed transcription factors have entered clinical trials for the treatment of human diseases and others are in preclinical development. High-throughput and user-friendly platforms for designing synthetic DNA-binding proteins present innovative methods for deciphering cell biology and designing custom synthetic gene circuits. We review two platforms for designing synthetic transcription factors for manipulating gene expression: Transcription activator-like effectors (TALEs) and the RNA-guided clustered regularly interspaced short palindromic repeats (CRISPR)/Cas9 system. We present an overview of each technology and a guide for designing and assembling custom TALE- and CRISPR/Cas9-based transcription factors. We also discuss characteristics of each platform that are best suited for different applications.


Asunto(s)
Sistemas CRISPR-Cas/genética , Repeticiones Palindrómicas Cortas Agrupadas y Regularmente Espaciadas/genética , Proteínas de Unión al ADN/genética , Regulación de la Expresión Génica , Ingeniería Genética/métodos , Factores de Transcripción/genética , Secuencia de Bases , Proteínas de Unión al ADN/química , Humanos , Datos de Secuencia Molecular , Estructura Secundaria de Proteína , Factores de Transcripción/química
12.
Nat Methods ; 10(10): 973-6, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23892895

RESUMEN

Technologies for engineering synthetic transcription factors have enabled many advances in medical and scientific research. In contrast to existing methods based on engineering of DNA-binding proteins, we created a Cas9-based transactivator that is targeted to DNA sequences by guide RNA molecules. Coexpression of this transactivator and combinations of guide RNAs in human cells induced specific expression of endogenous target genes, demonstrating a simple and versatile approach for RNA-guided gene activation.


Asunto(s)
Repeticiones Palindrómicas Cortas Agrupadas y Regularmente Espaciadas , Ingeniería de Proteínas/métodos , Edición de ARN , Factores de Transcripción/genética , Activación Transcripcional , Células HEK293 , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Proteína Antagonista del Receptor de Interleucina 1/genética , Ribonucleasas/genética , ARN Pequeño no Traducido
13.
Mol Ther ; 21(9): 1718-26, 2013 09.
Artículo en Inglés | MEDLINE | ID: mdl-23732986

RESUMEN

Genome editing with engineered nucleases has recently emerged as an approach to correct genetic mutations by enhancing homologous recombination with a DNA repair template. However, many genetic diseases, such as Duchenne muscular dystrophy (DMD), can be treated simply by correcting a disrupted reading frame. We show that genome editing with transcription activator-like effector nucleases (TALENs), without a repair template, can efficiently correct the reading frame and restore the expression of a functional dystrophin protein that is mutated in DMD. TALENs were engineered to mediate highly efficient gene editing at exon 51 of the dystrophin gene. This led to restoration of dystrophin protein expression in cells from Duchenne patients, including skeletal myoblasts and dermal fibroblasts that were reprogrammed to the myogenic lineage by MyoD. Finally, exome sequencing of cells with targeted modifications of the dystrophin locus showed no TALEN-mediated off-target changes to the protein-coding regions of the genome, as predicted by in silico target site analysis. This strategy integrates the rapid and robust assembly of active TALENs with an efficient gene-editing method for the correction of genetic diseases caused by mutations in non-essential coding regions that cause frameshifts or premature stop codons.


Asunto(s)
Distrofina/biosíntesis , Distrofina/genética , Endonucleasas/metabolismo , Marcación de Gen , Terapia Genética/métodos , Distrofia Muscular de Duchenne/genética , Distrofia Muscular de Duchenne/terapia , Distrofina/metabolismo , Endonucleasas/genética , Exoma , Genoma Humano , Células HEK293 , Humanos , Distrofia Muscular de Duchenne/metabolismo , Sistemas de Lectura
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA