Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Neurobiol Dis ; 147: 105148, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33184027

RESUMEN

Aging drives pathological accumulation of proteins such as tau, causing neurodegenerative dementia disorders like Alzheimer's disease. Previously we showed loss of function mutations in the gene encoding the poly(A) RNA binding protein SUT-2/MSUT2 suppress tau-mediated neurotoxicity in C. elegans neurons, cultured human cells, and mouse brain, while loss of PABPN1 had the opposite effect (Wheeler et al., 2019). Here we found that blocking poly(A) tail extension with cordycepin exacerbates tauopathy in cultured human cells, which is rescued by MSUT2 knockdown. To further investigate the molecular mechanisms of poly(A) RNA-mediated tauopathy suppression, we examined whether genes encoding poly(A) nucleases also modulated tauopathy in a C. elegans tauopathy model. We found that loss of function mutations in C. elegans ccr-4 and panl-2 genes enhanced tauopathy phenotypes in tau transgenic C. elegans while loss of parn-2 partially suppressed tauopathy. In addition, loss of parn-1 blocked tauopathy suppression by loss of parn-2. Epistasis analysis showed that sut-2 loss of function suppressed the tauopathy enhancement caused by loss of ccr-4 and SUT-2 overexpression exacerbated tauopathy even in the presence of parn-2 loss of function in tau transgenic C. elegans. Thus sut-2 modulation of tauopathy is epistatic to ccr-4 and parn-2. We found that human deadenylases do not colocalize with human MSUT2 in nuclear speckles; however, expression levels of TOE1, the homolog of parn-2, correlated with that of MSUT2 in post-mortem Alzheimer's disease patient brains. Alzheimer's disease patients with low TOE1 levels exhibited significantly increased pathological tau deposition and loss of NeuN staining. Taken together, this work suggests suppressing tauopathy cannot be accomplished by simply extending poly(A) tails, but rather a more complex relationship exists between tau, sut-2/MSUT2 function, and control of poly(A) RNA metabolism, and that parn-2/TOE1 may be altered in tauopathy in a similar way.


Asunto(s)
Enfermedad de Alzheimer/patología , Proteínas de Caenorhabditis elegans/metabolismo , Exorribonucleasas/metabolismo , Proteínas Nucleares/metabolismo , Proteínas de Unión a Poli(A)/metabolismo , Tauopatías/patología , Enfermedad de Alzheimer/metabolismo , Animales , Animales Modificados Genéticamente , Encéfalo/metabolismo , Encéfalo/patología , Caenorhabditis elegans , Humanos , Fenotipo , Tauopatías/metabolismo
2.
Methods ; 121-122: 16-28, 2017 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-28351759

RESUMEN

Genome editing using the CRISPR/Cas9 system requires the presence of guide RNAs bound to the Cas9 endonuclease as a ribonucleoprotein (RNP) complex in cells, which cleaves the host cell genome at sites specified by the guide RNAs. New genetic material may be introduced during repair of the double-stranded break via homology dependent repair (HDR) if suitable DNA templates are delivered with the CRISPR components. Early methods used plasmid or viral vectors to make these components in the host cell, however newer approaches using recombinant Cas9 protein with synthetic guide RNAs introduced directly as an RNP complex into cells shows faster onset of action with fewer off-target effects. This approach also enables use of chemically modified synthetic guide RNAs that have improved nuclease stability and reduces the risk of triggering an innate immune response in the host cell. This article provides detailed methods for genome editing using the RNP approach with synthetic guide RNAs using lipofection or electroporation in mammalian cells or using microinjection in murine zygotes, with or without addition of a single-stranded HDR template DNA.


Asunto(s)
Proteínas Bacterianas/genética , Sistemas CRISPR-Cas , Endonucleasas/genética , Edición Génica/métodos , Técnicas de Transferencia de Gen , ARN Guía de Kinetoplastida/genética , Ribonucleoproteínas/genética , Animales , Proteínas Bacterianas/metabolismo , Secuencia de Bases , Proteína 9 Asociada a CRISPR , Repeticiones Palindrómicas Cortas Agrupadas y Regularmente Espaciadas , ADN/genética , ADN/metabolismo , Electroporación , Endonucleasas/metabolismo , Marcación de Gen/métodos , Genoma , Células HEK293 , Humanos , Células Jurkat , Lípidos/química , Ratones , Microinyecciones , ARN Guía de Kinetoplastida/síntesis química , ARN Guía de Kinetoplastida/metabolismo , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Reparación del ADN por Recombinación , Ribonucleoproteínas/metabolismo , Cigoto/citología , Cigoto/metabolismo
3.
J Biol Chem ; 291(49): 25489-25504, 2016 Dec 02.
Artículo en Inglés | MEDLINE | ID: mdl-27756846

RESUMEN

We previously reported that delivery of a microRNA-138 mimic or siRNA against SIN3A to cultured cystic fibrosis (ΔF508/ΔF508) airway epithelia partially restored ΔF508-cystic fibrosis transmembrane conductance regulator (CFTR)-mediated cAMP-stimulated Cl- conductance. We hypothesized that dissecting this microRNA-138/SIN3A-regulated gene network would identify individual proteins contributing to the rescue of ΔF508-CFTR function. Among the genes in the network, we rigorously validated candidates using functional CFTR maturation and electrolyte transport assays in polarized airway epithelia. We found that depletion of the ubiquitin ligase SYVN1, the ubiquitin/proteasome system regulator NEDD8, or the F-box protein FBXO2 partially restored ΔF508-CFTR-mediated Cl- transport in primary cultures of human cystic fibrosis airway epithelia. Moreover, knockdown of SYVN1, NEDD8, or FBXO2 in combination with corrector compound 18 further potentiated rescue of ΔF508-CFTR-mediated Cl- conductance. This study provides new knowledge of the CFTR biosynthetic pathway. It suggests that SYVN1 and FBXO2 represent two distinct multiprotein complexes that may degrade ΔF508-CFTR in airway epithelia and identifies a new role for NEDD8 in regulating ΔF508-CFTR ubiquitination.


Asunto(s)
Secuencia de Aminoácidos , Proteínas de Ciclo Celular/metabolismo , Regulador de Conductancia de Transmembrana de Fibrosis Quística/metabolismo , Células Epiteliales/metabolismo , Proteínas F-Box/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Complejo de la Endopetidasa Proteasomal/metabolismo , Proteolisis , Mucosa Respiratoria/metabolismo , Eliminación de Secuencia , Ubiquitina-Proteína Ligasas/metabolismo , Ubiquitinación , Ubiquitinas/metabolismo , Proteínas de Ciclo Celular/genética , Regulador de Conductancia de Transmembrana de Fibrosis Quística/genética , Células Epiteliales/patología , Proteínas F-Box/genética , Técnicas de Silenciamiento del Gen , Células HeLa , Humanos , Transporte Iónico/genética , Proteína NEDD8 , Proteínas del Tejido Nervioso/genética , Complejo de la Endopetidasa Proteasomal/genética , Mucosa Respiratoria/fisiología , Ubiquitina/genética , Ubiquitina/metabolismo , Ubiquitina-Proteína Ligasas/genética
4.
Mol Pain ; 122016.
Artículo en Inglés | MEDLINE | ID: mdl-27306408

RESUMEN

BACKGROUND: Accumulating evidence suggests that the C-C chemokine ligand 2 (CCL2, or monocyte chemoattractant protein 1) acts as a neuromodulator in the central nervous system through its binding to the C-C chemokine receptor 2 (CCR2). Notably, it is well established that the CCL2/CCR2 axis plays a key role in neuron-glia communication as well as in spinal nociceptive transmission. Gene silencing through RNA interference has recently emerged as a promising avenue in research and drug development, including therapeutic management of chronic pain. In the present study, we used 27-mer Dicer-substrate small interfering RNA (DsiRNA) targeting CCR2 and assessed their ability to reverse the nociceptive behaviors induced by spinal CCL2 injection or following intraplantar injection of complete Freund's adjuvant. RESULTS: To this end, we first developed high-potency DsiRNAs designed to target different sequences distributed across the rat CCR2 (rCCR2) messenger RNA. For optimization, methyl groups were added to the two most potent DsiRNA candidates (Evader and M7 2'-O-methyl modified duplexes) in order to improve in vivo duplex stability and to reduce potential immunostimulatory activity. Our results demonstrated that all modified candidates formulated with the cell-penetrating peptide reagent Transductin showed strong RNAi activity following intrathecal delivery, exhibiting >50% rCCR2 knockdown in lumbar dorsal root ganglia. Accordingly, we found that these DsiRNA duplexes were able to reduce spinal microglia activation and were effective at blocking CCL2-induced mechanical hypersensitivity. Along with similar reductions of rCCR2 messenger RNA, both sequences and methylation patterns were similarly effective in inhibiting the CCL2 nociceptive action for the whole seven days testing period, compared to mismatch DsiRNA. DsiRNAs against CCR2 also reversed the hypernociceptive responses observed in the complete Freund's adjuvant-induced inflammatory chronic pain model. CONCLUSION: Altogether, these results validate CCR2 as a an appropriate molecular target for pain control and demonstrate that RNAi-based gene therapy represent an highly specific alternative to classical pharmacological approaches to treat central pathologies such as chronic pain.


Asunto(s)
Dolor/metabolismo , Dolor/prevención & control , ARN Interferente Pequeño/metabolismo , Receptores CCR2/antagonistas & inhibidores , Ribonucleasa III/metabolismo , Animales , Forma de la Célula , Fluorescencia , Ganglios Espinales/metabolismo , Regulación de la Expresión Génica , Células HEK293 , Humanos , Hiperalgesia/complicaciones , Hiperalgesia/metabolismo , Inflamación/complicaciones , Inflamación/patología , Masculino , Neuroglía/metabolismo , Dolor/complicaciones , ARN Mensajero/genética , ARN Mensajero/metabolismo , Ratas Sprague-Dawley , Receptores CCR2/genética , Reproducibilidad de los Resultados , Médula Espinal/metabolismo , Especificidad por Sustrato
5.
J Virol ; 88(2): 799-810, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24227841

RESUMEN

RNA-specific adenosine deaminase (ADAR)-mediated adenosine-to-inosine (A-to-I) editing is a critical arm of the antiviral response. However, mechanistic insights into how A-to-I RNA editing affects viral infection are lacking. We posited that inosine incorporation into RNA facilitates sensing of nonself RNA by innate immune sensors and accordingly investigated the impact of inosine-modified RNA on Toll-like receptor 7 and 8 (TLR7/8) sensing. Inosine incorporation into synthetic single-stranded RNA (ssRNA) potentiated tumor necrosis factor alpha (TNF-α) or alpha interferon (IFN-α) production in human peripheral blood mononuclear cells (PBMCs) in a sequence-dependent manner, indicative of TLR7/8 recruitment. The effect of inosine incorporation on TLR7/8 sensing was restricted to immunostimulatory ssRNAs and was not seen with inosine-containing short double-stranded RNAs or with a deoxy-inosine-modified ssRNA. Inosine-mediated increase of self-secondary structure of an ssRNA resulted in potentiated IFN-α production in human PBMCs through TLR7 recruitment, as established through the use of a TLR7 antagonist and Tlr7-deficient cells. There was a correlation between hyperediting of influenza A viral ssRNA and its ability to stimulate TNF-α, independent of 5'-triphosphate residues, and involving Adar-1. Furthermore, A-to-I editing of viral ssRNA directly enhanced mouse Tlr7 sensing, when present in proportions reproducing biologically relevant levels of RNA editing. Thus, we demonstrate for the first time that inosine incorporation into immunostimulatory ssRNA can potentiate TLR7/8 activation. Our results suggest a novel function of A-to-I RNA editing, which is to facilitate TLR7/8 sensing of phagocytosed viral RNA.


Asunto(s)
Subtipo H1N1 del Virus de la Influenza A/genética , Gripe Humana/inmunología , Gripe Humana/virología , Inosina/genética , Edición de ARN , ARN Viral/genética , Receptor Toll-Like 7/inmunología , Receptor Toll-Like 8/inmunología , Adenosina/genética , Adenosina/inmunología , Adenosina Desaminasa/genética , Adenosina Desaminasa/inmunología , Animales , Secuencia de Bases , Femenino , Humanos , Subtipo H1N1 del Virus de la Influenza A/inmunología , Gripe Humana/genética , Inosina/inmunología , Interferón-alfa/genética , Interferón-alfa/inmunología , Leucocitos Mononucleares/inmunología , Leucocitos Mononucleares/virología , Masculino , Ratones , Ratones Noqueados , Datos de Secuencia Molecular , Conformación de Ácido Nucleico , ARN Viral/química , ARN Viral/inmunología , Receptor Toll-Like 7/genética , Receptor Toll-Like 8/genética
6.
Proc Natl Acad Sci U S A ; 109(33): 13362-7, 2012 Aug 14.
Artículo en Inglés | MEDLINE | ID: mdl-22853952

RESUMEN

Production of functional proteins requires multiple steps, including gene transcription and posttranslational processing. MicroRNAs (miRNAs) can regulate individual stages of these processes. Despite the importance of the cystic fibrosis transmembrane conductance regulator (CFTR) channel for epithelial anion transport, how its expression is regulated remains uncertain. We discovered that miRNA-138 regulates CFTR expression through its interactions with the transcriptional regulatory protein SIN3A. Treating airway epithelia with an miR-138 mimic increased CFTR mRNA and also enhanced CFTR abundance and transepithelial Cl(-) permeability independent of elevated mRNA levels. An miR-138 anti-miR had the opposite effects. Importantly, miR-138 altered the expression of many genes encoding proteins that associate with CFTR and may influence its biosynthesis. The most common CFTR mutation, ΔF508, causes protein misfolding, protein degradation, and cystic fibrosis. Remarkably, manipulating the miR-138 regulatory network also improved biosynthesis of CFTR-ΔF508 and restored Cl(-) transport to cystic fibrosis airway epithelia. This miRNA-regulated network directs gene expression from the chromosome to the cell membrane, indicating that an individual miRNA can control a cellular process more broadly than recognized previously. This discovery also provides therapeutic avenues for restoring CFTR function to cells affected by the most common cystic fibrosis mutation.


Asunto(s)
Regulador de Conductancia de Transmembrana de Fibrosis Quística/biosíntesis , Regulador de Conductancia de Transmembrana de Fibrosis Quística/genética , Regulación de la Expresión Génica , Redes Reguladoras de Genes/genética , MicroARNs/metabolismo , Transporte Biológico , Cloruros/metabolismo , Epitelio/metabolismo , Epitelio/patología , Perfilación de la Expresión Génica , Células HeLa , Humanos , Pulmón/metabolismo , Pulmón/patología , MicroARNs/genética , Procesamiento Proteico-Postraduccional , Proteínas Represoras/antagonistas & inhibidores , Proteínas Represoras/metabolismo , Complejo Correpresor Histona Desacetilasa y Sin3
7.
J Virol ; 87(7): 3966-78, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23365428

RESUMEN

Latently infected cell reservoirs represent the main barrier to HIV eradication. Combination antiretroviral therapy (cART) effectively blocks viral replication but cannot purge latent provirus. One approach to HIV eradication could include cART to block new infections plus an agent to activate latent provirus. NF-κB activation induces HIV expression, ending latency. Before activation, IκB proteins sequester NF-κB dimers in the cytoplasm. Three canonical IκBs, IκBα, IκBß, and IκBε, exist, but the IκB proteins' role in HIV activation regulation is not fully understood. We studied the effects on HIV activation of targeting IκBs by single and pairwise small interfering RNA (siRNA) knockdown. After determining the relative abundance of the IκBs, the relative abundance of NF-κB subunits held by the IκBs, and the kinetics of IκB degradation and resynthesis following knockdown, we studied HIV activation by IκB knockdown, in comparison with those of known HIV activators, tumor necrosis factor alpha (TNF-α), tetradecanoyl phorbol acetate (TPA), and trichostatin A (TSA), in U1 monocytic and J-Lat 10.6 lymphocytic latently infected cells. We found that IκBα knockdown activated HIV in both U1 and J-Lat 10.6 cells, IκBß knockdown did not activate HIV, and, surprisingly, IκBε knockdown produced the most HIV activation, comparable to TSA activation. Our data show that HIV reactivation can be triggered by targeting two different IκB proteins and that IκBε may be an effective target for HIV latency reactivation in T-cell and macrophage lineages. IκBε knockdown may offer attractive therapeutic advantages for HIV activation because it is not essential for mammalian growth and development and because new siRNA delivery strategies may target siRNAs to HIV latently infected cells.


Asunto(s)
VIH-1/fisiología , Proteínas I-kappa B/metabolismo , FN-kappa B/metabolismo , Latencia del Virus/efectos de los fármacos , Latencia del Virus/fisiología , Línea Celular Tumoral , Dimerización , Técnicas de Silenciamiento del Gen , Humanos , Ácidos Hidroxámicos/farmacología , Proteínas I-kappa B/genética , Immunoblotting , Inmunoprecipitación , Modelos Lineales , ARN Interferente Pequeño/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Acetato de Tetradecanoilforbol/farmacología , Factor de Necrosis Tumoral alfa/farmacología , Latencia del Virus/genética
8.
FASEB J ; 27(9): 3741-52, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23756650

RESUMEN

Management of painful peripheral neuropathies remains challenging, since patients with chronic pain respond poorly to the available pharmacopeia. In recent years, the G-protein-coupled receptor neurotensin (NT) type 2 (NTS2) emerged as an attractive target for treating transitory pain states. To date, however, there is no evidence for its role in the regulation of chronic peripheral neuropathies. Here, we found that NTS2 receptors were largely localized to primary afferent fibers and superficial dorsal horns. Changes in the time course of the gene expression profile of NT, NTS1, and NTS2 were observed over a 28-d period following the sciatic nerve constriction [chronic constriction injury (CCI) model]. We next determined the effects of central delivery of selective-NTS2 agonists to CCI-treated rats on both mechanical allodynia (evoked withdrawal responses) and weight-bearing deficits (discomfort and quality-of-life proxies). The NTS2 analogs JMV431, levocabastine, and ß-lactotensin were all effective in reducing ongoing tactile allodynia in CCI-treated rats. Likewise, amitriptyline, pregabalin, and morphine significantly attenuated CCI-induced mechanical hypersensitivity. NTS2 agonists were also efficient in reversing weight-bearing and postural deficits caused by nerve damage, unlike reference analgesics currently used in the clinic. Thus, NTS2 agonists may offer new treatment avenues for limiting pain associated with peripheral neuropathies and improve functional rehabilitation and well-being.


Asunto(s)
Neuralgia/tratamiento farmacológico , Receptores de Neurotensina/metabolismo , Analgésicos/uso terapéutico , Animales , Western Blotting , Ganglios Espinales/efectos de los fármacos , Ganglios Espinales/metabolismo , Masculino , Microscopía Electrónica de Transmisión , Neuralgia/inducido químicamente , Oligopéptidos/uso terapéutico , Piperidinas/uso terapéutico , Ratas , Ratas Sprague-Dawley , Receptores de Neurotensina/agonistas , Médula Espinal/efectos de los fármacos , Médula Espinal/metabolismo
9.
Am J Respir Cell Mol Biol ; 49(4): 544-51, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23646886

RESUMEN

MicroRNAs (miRNAs) are increasingly recognized as important posttranscriptional regulators of gene expression, and changes in their actions can contribute to disease states. Little is understood regarding miRNA functions in the airway epithelium under normal or diseased conditions. We profiled miRNA expression in well-differentiated primary cultures of human cystic fibrosis (CF) and non-CF airway epithelia, and discovered that miR-509-3p and miR-494 concentrations were increased in CF epithelia. Human non-CF airway epithelia, transfected with the mimics of miR-509-3p or miR-494, showed decreased cystic fibrosis transmembrane conductance regulator (CFTR) expression, whereas their respective anti-miRs exerted the opposite effect. Interestingly, the two miRNAs acted cooperatively in regulating CFTR expression. Upon infecting non-CF airway epithelial cells with Staphylococcus aureus, or upon stimulating them with the proinflammatory cytokines TNF-α or IL-1ß, we observed an increased expression of both miRNAs and a concurrent decrease in CFTR expression and function, suggesting that inflammatory mediators may regulate these miRNAs. Transfecting epithelia with anti-miRs for miR-509-3p and miR-494, or inhibiting NF-κB signaling before stimulating cells with TNFα or IL-1ß, suppressed these responses, suggesting that the expression of both miRNAs was responsive to NF-κB signaling. Thus, miR-509-3p and miR-494 are dynamic regulators of CFTR abundance and function in normal, non-CF airway epithelia.


Asunto(s)
Regulador de Conductancia de Transmembrana de Fibrosis Quística/metabolismo , Fibrosis Quística/metabolismo , MicroARNs/metabolismo , Procesamiento Postranscripcional del ARN , Línea Celular , Fibrosis Quística/genética , Regulador de Conductancia de Transmembrana de Fibrosis Quística/genética , Células Epiteliales/metabolismo , Expresión Génica , Humanos , Mediadores de Inflamación/metabolismo , Interleucina-1beta/genética , Interleucina-1beta/metabolismo , MicroARNs/genética , FN-kappa B/genética , FN-kappa B/metabolismo , Mucosa Respiratoria/metabolismo , Factor de Necrosis Tumoral alfa/genética , Factor de Necrosis Tumoral alfa/metabolismo
10.
Am J Physiol Lung Cell Mol Physiol ; 305(1): L23-32, 2013 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-23624792

RESUMEN

Polarized and pseudostratified primary airway epithelia present barriers that significantly reduce their transfection efficiency and the efficacy of RNA interference oligonucleotides. This creates an impediment in studies of the airway epithelium, diminishing the utility of loss-of-function as a research tool. Here we outline methods to introduce RNAi oligonucleotides into primary human and porcine airway epithelia grown at an air-liquid interface and difficult-to-transfect transformed epithelial cell lines grown on plastic. At the time of plating, we reverse transfect small-interfering RNA (siRNA), Dicer-substrate siRNA, or microRNA oligonucleotides into cells by use of lipid or peptide transfection reagents. Using this approach we achieve significant knockdown in vitro of hypoxanthine-guanine phosphoribosyltransferase, IL-8, and CFTR expression at the mRNA and protein levels in 1-3 days. We also attain significant reduction of secreted IL-8 in polarized primary pig airway epithelia 3 days posttransfection and inhibition of CFTR-mediated Cl⁻ conductance in polarized air-liquid interface cultures of human airway epithelia 2 wk posttransfection. These results highlight an efficient means to deliver RNA interference reagents to airway epithelial cells and achieve significant knockdown of target gene expression and function. The ability to reliably conduct loss-of-function assays in polarized primary airway epithelia offers benefits to research in studies of epithelial cell homeostasis, candidate gene function, gene-based therapeutics, microRNA biology, and targeting the replication of respiratory viruses.


Asunto(s)
Polaridad Celular , Neoplasias del Colon/patología , Regulador de Conductancia de Transmembrana de Fibrosis Quística/genética , Células Epiteliales/metabolismo , Oligonucleótidos/farmacología , ARN Interferente Pequeño/genética , Mucosa Respiratoria/efectos de los fármacos , Adenocarcinoma/genética , Adenocarcinoma/patología , Animales , Western Blotting , Células Cultivadas , Neoplasias del Colon/genética , Electrofisiología , Citometría de Flujo , Humanos , Técnicas para Inmunoenzimas , Técnicas In Vitro , Interleucina-8/metabolismo , Masculino , ARN Mensajero/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Mucosa Respiratoria/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Porcinos
11.
Nat Commun ; 14(1): 902, 2023 02 17.
Artículo en Inglés | MEDLINE | ID: mdl-36804928

RESUMEN

CRISPR-mediated transcriptional activation (CRISPRa) is a powerful technology for inducing gene expression from endogenous loci with exciting applications in high throughput gain-of-function genomic screens and the engineering of cell-based models. However, current strategies for generating potent, stable, CRISPRa-competent cell lines present limitations for the broad utility of this approach. Here, we provide a high-efficiency, self-selecting CRISPRa enrichment strategy, which combined with piggyBac transposon technology enables rapid production of CRISPRa-ready cell populations compatible with a variety of downstream assays. We complement this with an optimized guide RNA scaffold that significantly enhances CRISPRa functionality. Finally, we describe a synthetic guide RNA tool set that enables transient, population-wide gene activation when used with the self-selecting CRISPRa system. Taken together, this versatile platform greatly enhances the potential for CRISPRa across a wide variety of cellular contexts.


Asunto(s)
Genoma , ARN , Activación Transcripcional , Línea Celular , Sistemas CRISPR-Cas/genética
12.
Mol Ther Methods Clin Dev ; 25: 311-330, 2022 Jun 09.
Artículo en Inglés | MEDLINE | ID: mdl-35573047

RESUMEN

Chimeric antigen receptor (CAR) redirected T cells are potent therapeutic options against hematological malignancies. The current dominant manufacturing approach for CAR T cells depends on retroviral transduction. With the advent of gene editing, insertion of a CD19-CAR into the T cell receptor (TCR) alpha constant (TRAC) locus using adeno-associated viruses for gene transfer was demonstrated, and these CD19-CAR T cells showed improved functionality over their retrovirally transduced counterparts. However, clinical-grade production of viruses is complex and associated with extensive costs. Here, we optimized a virus-free genome-editing method for efficient CAR insertion into the TRAC locus of primary human T cells via nuclease-assisted homology-directed repair (HDR) using CRISPR-Cas and double-stranded template DNA (dsDNA). We evaluated DNA-sensor inhibition and HDR enhancement as two pharmacological interventions to improve cell viability and relative CAR knockin rates, respectively. While the toxicity of transfected dsDNA was not fully prevented, the combination of both interventions significantly increased CAR knockin rates and CAR T cell yield. Resulting TRAC-replaced CD19-CAR T cells showed antigen-specific cytotoxicity and cytokine production in vitro and slowed leukemia progression in a xenograft mouse model. Amplicon sequencing did not reveal significant indel formation at potential off-target sites with or without exposure to DNA-repair-modulating small molecules. With TRAC-integrated CAR+ T cell frequencies exceeding 50%, this study opens new perspectives to exploit pharmacological interventions to improve non-viral gene editing in T cells.

13.
Mol Ther Methods Clin Dev ; 21: 478-491, 2021 Jun 11.
Artículo en Inglés | MEDLINE | ID: mdl-33981780

RESUMEN

CRISPR systems enable targeted genome editing in a wide variety of organisms by introducing single- or double-strand DNA breaks, which are repaired using endogenous molecular pathways. Characterization of on- and off-target editing events from CRISPR proteins can be evaluated using targeted genome resequencing. We characterized DNA repair fingerprints that result from non-homologous end joining (NHEJ) after double-stranded breaks (DSBs) were introduced by Cas9 or Cas12a for >500 paired treatment/control experiments. We found that building biological understanding of the repair into a novel analysis tool (CRISPAltRations) improved the quality of the results. We validated our software using simulated, targeted amplicon sequencing data (11 guide RNAs [gRNAs] and 603 on- and off-target locations) and demonstrated that CRISPAltRations outperforms other publicly available software tools in accurately annotating CRISPR-associated indels and homology-directed repair (HDR) events. We enable non-bioinformaticians to use CRISPAltRations by developing a web-accessible, cloud-hosted deployment, which allows rapid batch processing of samples in a graphical user interface (GUI) and complies with HIPAA security standards. By ensuring that our software is thoroughly tested, version controlled, and supported with a user interface (UI), we enable resequencing analysis of CRISPR genome editing experiments to researchers no matter their skill in bioinformatics.

14.
Sci Adv ; 6(47)2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-33208369

RESUMEN

Harnessing CRISPR-Cas9 technology for cancer therapeutics has been hampered by low editing efficiency in tumors and potential toxicity of existing delivery systems. Here, we describe a safe and efficient lipid nanoparticle (LNP) for the delivery of Cas9 mRNA and sgRNAs that use a novel amino-ionizable lipid. A single intracerebral injection of CRISPR-LNPs against PLK1 (sgPLK1-cLNPs) into aggressive orthotopic glioblastoma enabled up to ~70% gene editing in vivo, which caused tumor cell apoptosis, inhibited tumor growth by 50%, and improved survival by 30%. To reach disseminated tumors, cLNPs were also engineered for antibody-targeted delivery. Intraperitoneal injections of EGFR-targeted sgPLK1-cLNPs caused their selective uptake into disseminated ovarian tumors, enabled up to ~80% gene editing in vivo, inhibited tumor growth, and increased survival by 80%. The ability to disrupt gene expression in vivo in tumors opens new avenues for cancer treatment and research and potential applications for targeted gene editing of noncancerous tissues.


Asunto(s)
Nanopartículas , Neoplasias , Sistemas CRISPR-Cas , Edición Génica , Técnicas de Transferencia de Gen , Liposomas , Neoplasias/genética , Neoplasias/terapia
15.
Mol Ther Methods Clin Dev ; 17: 1097-1107, 2020 Jun 12.
Artículo en Inglés | MEDLINE | ID: mdl-32478125

RESUMEN

Genome editing of human cluster of differentiation 34+ (CD34+) hematopoietic stem and progenitor cells (HSPCs) holds great therapeutic potential. This study aimed to optimize on-target, ex vivo genome editing using the CRISPR-Cas9 system in CD34+ HSPCs and to create a clear workflow for precise identification of off-target effects. Modified synthetic guide RNAs (gRNAs), either 2-part gRNA or single-guide RNA (sgRNA), were delivered to CD34+ HSPCs as part of ribonucleoprotein (RNP) complexes, targeting therapeutically relevant genes. The addition of an Alt-R electroporation enhancer (EE), a short, single-stranded oligodeoxynucleotide (ssODN), significantly increased editing efficiency in CD34+ HSPCs. Notably, similar editing improvement was observed when excess gRNA over Cas9 protein was used, providing a DNA-free alternative suitable for therapeutic applications. Furthermore, we demonstrated that sgRNA may be preferable over 2-part gRNA in a locus-specific manner. Finally, we present a clear experimental framework suitable for the unbiased identification of bona fide off-target sites by Genome-Wide, Unbiased Identification of Double-Strand Breaks (DSBs) Enabled by Sequencing (GUIDE-seq), as well as subsequent editing quantification in CD34+ HSPCs using rhAmpSeq. These findings may facilitate the implementation of genome editing in CD34+ HSPCs for research and therapy and can be adapted for other hematopoietic cells.

16.
Dev Cell ; 51(5): 645-657.e4, 2019 12 02.
Artículo en Inglés | MEDLINE | ID: mdl-31708433

RESUMEN

Inconsistent activity limits the use of CRISPR-Cas9 in zebrafish. We show supernumerary guanine nucleotides at the 5' ends of single guide RNAs (sgRNAs) account for diminished CRISPR-Cas9 activity in zebrafish embryos. Genomic sequences can be targeted consistently with extremely high efficiency using Cas9 ribonucleoproteins (RNPs) containing either a sgRNA molecule or a synthetic crRNA:tracrRNA duplex that perfectly matches the protospacer target site. Following injection of zebrafish eggs with such RNPs, virtually every copy of a targeted locus harbors an induced indel mutation. Loss of gene function is often complete, as F0 embryos closely resemble true null mutants without detectable non-specific effects. Mosaicism is sufficiently low in F0 embryos that cell non-autonomous gene functions can be probed effectively and redundant activities of genes can be uncovered when two genes are targeted simultaneously. Finally, heritable deletion mutations of at least 50 kbp can be readily induced using pairs of duplex guide RNPs targeted to a single chromosome.


Asunto(s)
Sistemas CRISPR-Cas , Eliminación de Gen , Edición Génica/métodos , Pez Cebra/genética , Animales , Embrión no Mamífero/metabolismo , Mutación con Pérdida de Función
17.
Elife ; 72018 04 11.
Artículo en Inglés | MEDLINE | ID: mdl-29638216

RESUMEN

CRISPR/Cas9 can be used for precise genetic knock-in of epitope tags into endogenous genes, simplifying experimental analysis of protein function. However, Cas9-assisted epitope tagging in primary mammalian cell cultures is often inefficient and reliant on plasmid-based selection strategies. Here, we demonstrate improved knock-in efficiencies of diverse tags (V5, 3XFLAG, Myc, HA) using co-delivery of Cas9 protein pre-complexed with two-part synthetic modified RNAs (annealed crRNA:tracrRNA) and single-stranded oligodeoxynucleotide (ssODN) repair templates. Knock-in efficiencies of ~5-30%, were achieved without selection in embryonic stem (ES) cells, neural stem (NS) cells, and brain-tumor-derived stem cells. Biallelic-tagged clonal lines were readily derived and used to define Olig2 chromatin-bound interacting partners. Using our novel web-based design tool, we established a 96-well format pipeline that enabled V5-tagging of 60 different transcription factors. This efficient, selection-free and scalable epitope tagging pipeline enables systematic surveys of protein expression levels, subcellular localization, and interactors across diverse mammalian stem cells.


Asunto(s)
Proteína 9 Asociada a CRISPR/metabolismo , Sistemas CRISPR-Cas , Mapeo Epitopo/métodos , Ensayos Analíticos de Alto Rendimiento , Ribonucleoproteínas/metabolismo , Células Madre/citología , Factores de Transcripción/metabolismo , Animales , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/patología , Proteína 9 Asociada a CRISPR/genética , Células Cultivadas , Células Madre Embrionarias/citología , Células Madre Embrionarias/metabolismo , Edición Génica , Humanos , Ratones , Células Madre Neoplásicas/metabolismo , Células Madre Neoplásicas/patología , Células-Madre Neurales/citología , Células-Madre Neurales/metabolismo , Oligodesoxirribonucleótidos/genética , ARN Guía de Kinetoplastida , Ribonucleoproteínas/genética , Células Madre/metabolismo , Factores de Transcripción/genética
18.
Nat Med ; 24(8): 1216-1224, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-30082871

RESUMEN

Translation of the CRISPR-Cas9 system to human therapeutics holds high promise. However, specificity remains a concern especially when modifying stem cell populations. We show that existing rationally engineered Cas9 high-fidelity variants have reduced on-target activity when using the therapeutically relevant ribonucleoprotein (RNP) delivery method. Therefore, we devised an unbiased bacterial screen to isolate variants that retain activity in the RNP format. Introduction of a single point mutation, p.R691A, in Cas9 (high-fidelity (HiFi) Cas9) retained the high on-target activity of Cas9 while reducing off-target editing. HiFi Cas9 induces robust AAV6-mediated gene targeting at five therapeutically relevant loci (HBB, IL2RG, CCR5, HEXB, and TRAC) in human CD34+ hematopoietic stem and progenitor cells (HSPCs) as well as primary T cells. We also show that HiFi Cas9 mediates high-level correction of the sickle cell disease (SCD)-causing p.E6V mutation in HSPCs derived from patients with SCD. We anticipate that HiFi Cas9 will have wide utility for both basic science and therapeutic genome-editing applications.


Asunto(s)
Proteína 9 Asociada a CRISPR/genética , Edición Génica , Células Madre Hematopoyéticas/metabolismo , Mutación/genética , Ribonucleoproteínas/metabolismo , Anemia de Células Falciformes/genética , Anemia de Células Falciformes/terapia , Antígenos CD34/metabolismo , Secuencia de Bases , Escherichia coli , Células HEK293 , Humanos
19.
Genome Biol ; 18(1): 92, 2017 05 17.
Artículo en Inglés | MEDLINE | ID: mdl-28511701

RESUMEN

BACKGROUND: Conditional knockout mice and transgenic mice expressing recombinases, reporters, and inducible transcriptional activators are key for many genetic studies and comprise over 90% of mouse models created. Conditional knockout mice are generated using labor-intensive methods of homologous recombination in embryonic stem cells and are available for only ~25% of all mouse genes. Transgenic mice generated by random genomic insertion approaches pose problems of unreliable expression, and thus there is a need for targeted-insertion models. Although CRISPR-based strategies were reported to create conditional and targeted-insertion alleles via one-step delivery of targeting components directly to zygotes, these strategies are quite inefficient. RESULTS: Here we describe Easi-CRISPR (Efficient additions with ssDNA inserts-CRISPR), a targeting strategy in which long single-stranded DNA donors are injected with pre-assembled crRNA + tracrRNA + Cas9 ribonucleoprotein (ctRNP) complexes into mouse zygotes. We show for over a dozen loci that Easi-CRISPR generates correctly targeted conditional and insertion alleles in 8.5-100% of the resulting live offspring. CONCLUSIONS: Easi-CRISPR solves the major problem of animal genome engineering, namely the inefficiency of targeted DNA cassette insertion. The approach is robust, succeeding for all tested loci. It is versatile, generating both conditional and targeted insertion alleles. Finally, it is highly efficient, as treating an average of only 50 zygotes is sufficient to produce a correctly targeted allele in up to 100% of live offspring. Thus, Easi-CRISPR offers a comprehensive means of building large-scale Cre-LoxP animal resources.


Asunto(s)
Repeticiones Palindrómicas Cortas Agrupadas y Regularmente Espaciadas , Edición Génica/métodos , Ratones Transgénicos/genética , Mutagénesis Insercional/métodos , Ribonucleoproteínas/genética , Animales , ADN de Cadena Simple/genética , ADN de Cadena Simple/metabolismo , Endonucleasas/genética , Endonucleasas/metabolismo , Efecto Fundador , Genes Reporteros , Sitios Genéticos , Integrasas/genética , Integrasas/metabolismo , Ratones , Ratones Transgénicos/crecimiento & desarrollo , Microinyecciones , ARN Guía de Kinetoplastida/genética , ARN Guía de Kinetoplastida/metabolismo , Reparación del ADN por Recombinación , Ribonucleoproteínas/metabolismo , Cigoto/crecimiento & desarrollo , Cigoto/metabolismo
20.
Cell Signal ; 27(8): 1597-608, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25916507

RESUMEN

Much is known about the how Gßγ subunits regulate effectors in response to G protein-coupled receptor stimulation. However, there is still a lot we don't know about how specific combinations of Gß and Gγ are wired into different signalling pathways. Here, using an siRNA screen for different Gß and Gγ subunits, we examined an endogenous M3 muscarinic receptor signalling pathway in HEK 293 cells. We observed that Gß(4) subunits were critical for calcium signalling and a downstream surrogate measured as ERK1/2 MAP kinase activity. A number of Gγ subunits could partner with Gß(4) but the best coupling was seen via Gß(4)γ(1). Intriguingly, knocking down Gß(1) actually increased signalling through the M3-mAChR most likely via an increase in Gß(4) levels. We noted that Gß(1) occupies the promoter of Gß(4) and may participate in maturation of its mRNA. This highlights a new role for Gßγ signalling beyond their canonical roles in cellular signalling.


Asunto(s)
Subunidades beta de la Proteína de Unión al GTP/metabolismo , Subunidades gamma de la Proteína de Unión al GTP/metabolismo , Receptores Muscarínicos/metabolismo , Transducción de Señal , Sitios de Unión , Señalización del Calcio , Carbacol/farmacología , Agonistas Colinérgicos/farmacología , Relación Dosis-Respuesta a Droga , Subunidades beta de la Proteína de Unión al GTP/genética , Subunidades gamma de la Proteína de Unión al GTP/genética , Regulación de la Expresión Génica , Células HEK293 , Humanos , Sistema de Señalización de MAP Quinasas , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Regiones Promotoras Genéticas , Multimerización de Proteína , Interferencia de ARN , ARN Mensajero/metabolismo , Receptor Muscarínico M3 , Receptores Muscarínicos/efectos de los fármacos , Receptores Muscarínicos/genética , Transducción de Señal/efectos de los fármacos , Transcripción Genética , Transfección
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA