Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
iScience ; 27(5): 109777, 2024 May 17.
Artículo en Inglés | MEDLINE | ID: mdl-38711458

RESUMEN

Although adeno-associated virus 9 (AAV9) has been highly exploited as delivery platform for gene-based therapies, its efficacy is hampered by low efficiency in crossing the adult blood-brain barrier (BBB) and pronounced targeting to the liver upon intravenous delivery. We generated a new galactose binding-deficient AAV9 peptide display library and selected two new AAV9 engineered capsids with enhanced targeting in mouse and marmoset brains after intravenous delivery. Interestingly, the loss of galactose binding greatly reduced undesired targeting to peripheral organs, particularly the liver, while not compromising transduction of the brain vasculature. However, the galactose binding was necessary to efficiently infect non-endothelial brain cells. Thus, the combinatorial actions of the galactose-binding domain and the incorporated displayed peptide are crucial to enhance BBB crossing along with brain cell transduction. This study describes two novel capsids with high brain endothelial infectivity and extremely low liver targeting based on manipulating the AAV9 galactose-binding domain.

2.
Nat Commun ; 14(1): 3962, 2023 07 05.
Artículo en Inglés | MEDLINE | ID: mdl-37407555

RESUMEN

Huntington's disease (HD) is a neurodegenerative disorder caused by CAG-repeat expansions in the huntingtin (HTT) gene. The resulting mutant HTT (mHTT) protein induces toxicity and cell death via multiple mechanisms and no effective therapy is available. Here, we employ a genome-wide screening in pluripotent mouse embryonic stem cells (ESCs) to identify suppressors of mHTT toxicity. Among the identified suppressors, linked to HD-associated processes, we focus on Metal response element binding transcription factor 1 (Mtf1). Forced expression of Mtf1 counteracts cell death and oxidative stress caused by mHTT in mouse ESCs and in human neuronal precursor cells. In zebrafish, Mtf1 reduces malformations and apoptosis induced by mHTT. In R6/2 mice, Mtf1 ablates motor defects and reduces mHTT aggregates and oxidative stress. Our screening strategy enables a quick in vitro identification of promising suppressor genes and their validation in vivo, and it can be applied to other monogenic diseases.


Asunto(s)
Enfermedad de Huntington , Enfermedades Neurodegenerativas , Ratones , Animales , Humanos , Modelos Animales de Enfermedad , Pez Cebra/genética , Pez Cebra/metabolismo , Enfermedad de Huntington/metabolismo , Neuronas/metabolismo , Enfermedades Neurodegenerativas/metabolismo , Proteína Huntingtina/genética , Proteína Huntingtina/metabolismo
3.
Blood ; 141(19): 2316-2329, 2023 05 11.
Artículo en Inglés | MEDLINE | ID: mdl-36790505

RESUMEN

Adeno-associated virus (AAV) vectors have been successfully exploited in gene therapy applications for the treatment of several genetic disorders. AAV is considered an episomal vector, but it has been shown to integrate within the host cell genome after the generation of double-strand DNA breaks or nicks. Although AAV integration raises some safety concerns, it can also provide therapeutic benefit; the direct intrathymic injection of an AAV harboring a therapeutic transgene results in integration in T-cell progenitors and long-term T-cell immunity. To assess the mechanisms of AAV integration, we retrieved and analyzed hundreds of AAV integration sites from lymph node-derived mature T cells and compared these with liver and brain tissue from treated mice. Notably, we found that although AAV integrations in the liver and brain were distributed across the entire mouse genome, >90% of the integrations in T cells were clustered within the T-cell receptor α, ß, and γ genes. More precisely, the insertion mapped to DNA breaks created by the enzymatic activity of recombination activating genes (RAGs) during variable, diversity, and joining recombination. Our data indicate that RAG activity during T-cell receptor maturation induces a site-specific integration of AAV genomes and opens new therapeutic avenues for achieving long-term AAV-mediated gene transfer in dividing cells.


Asunto(s)
Terapia Genética , Vectores Genéticos , Ratones , Animales , Vectores Genéticos/genética , Transgenes , Plásmidos , Terapia Genética/métodos , Receptores de Antígenos de Linfocitos T/genética , Dependovirus/genética , Integración Viral
4.
Elife ; 122023 01 16.
Artículo en Inglés | MEDLINE | ID: mdl-36645345

RESUMEN

Wolfram syndrome 1 (WS1) is a rare genetic disorder caused by mutations in the WFS1 gene leading to a wide spectrum of clinical dysfunctions, among which blindness, diabetes, and neurological deficits are the most prominent. WFS1 encodes for the endoplasmic reticulum (ER) resident transmembrane protein wolframin with multiple functions in ER processes. However, the WFS1-dependent etiopathology in retinal cells is unknown. Herein, we showed that Wfs1 mutant mice developed early retinal electrophysiological impairments followed by marked visual loss. Interestingly, axons and myelin disruption in the optic nerve preceded the degeneration of the retinal ganglion cell bodies in the retina. Transcriptomics at pre-degenerative stage revealed the STAT3-dependent activation of proinflammatory glial markers with reduction of the homeostatic and pro-survival factors glutamine synthetase and BDNF. Furthermore, label-free comparative proteomics identified a significant reduction of the monocarboxylate transport isoform 1 (MCT1) and its partner basigin that are highly enriched on retinal glia and myelin-forming oligodendrocytes in optic nerve together with wolframin. Loss of MCT1 caused a failure in lactate transfer from glial to neuronal cell bodies and axons leading to a chronic hypometabolic state. Thus, this bioenergetic impairment is occurring concurrently both within the axonal regions and cell bodies of the retinal ganglion cells, selectively endangering their survival while impacting less on other retinal cells. This metabolic dysfunction occurs months before the frank RGC degeneration suggesting an extended time-window for intervening with new therapeutic strategies focused on boosting retinal and optic nerve bioenergetics in WS1.


Asunto(s)
Atrofia Óptica , Síndrome de Wolfram , Animales , Ratones , Degeneración Nerviosa/metabolismo , Enfermedades Neuroinflamatorias , Células Ganglionares de la Retina/metabolismo , Síndrome de Wolfram/genética , Síndrome de Wolfram/metabolismo
5.
Cell Death Dis ; 13(10): 881, 2022 10 19.
Artículo en Inglés | MEDLINE | ID: mdl-36261424

RESUMEN

Triplication of the SNCA gene, encoding the protein alpha-Synuclein (αSyn), is a rare cause of aggressive and early-onset parkinsonism. Herein, we generated iPSCs from two siblings with a recently described compact SNCA gene triplication and suffering from severe motor impairments, psychiatric symptoms, and cognitive deterioration. Using CRISPR/Cas9 gene editing, each SNCA copy was inactivated by targeted indel mutations generating a panel of isogenic iPSCs with a decremental number from 4 down to none of functional SNCA gene alleles. We differentiated these iPSC lines in midbrain dopaminergic (DA) neuronal cultures to characterize αSyn aggregation in native and seeded conditions and evaluate its associated cellular dysfunctions. Utilizing a new nanobody-based biosensor combined with super-resolved imaging, we were able to visualize and measure αSyn aggregates in early DA neurons in unstimulated conditions. Calcium dysregulation and mitochondrial alterations were the first pathological signs detectable in early differentiated DA neuronal cultures. Accelerated αSyn aggregation was induced by exposing neurons to structurally well-characterized synthetic αSyn fibrils. 4xSNCA DA neurons showed the highest vulnerability, which was associated with high levels of oxidized DA and amplified by TAX1BP1 gene disruption. Seeded DA neurons developed large αSyn deposits whose morphology and internal constituents resembled Lewy bodies commonly observed in Parkinson's disease (PD) patient brain tissues. These findings provide strong evidence that this isogenic panel of iPSCs with SNCA multiplications offers a remarkable cellular platform to investigate mechanisms of PD and validate candidate inhibitors of native and seeded αSyn aggregation.


Asunto(s)
Células Madre Pluripotentes Inducidas , Enfermedad de Parkinson , Humanos , alfa-Sinucleína/genética , alfa-Sinucleína/metabolismo , Neuronas Dopaminérgicas/metabolismo , Células Madre Pluripotentes Inducidas/metabolismo , Calcio/metabolismo , Enfermedad de Parkinson/metabolismo
6.
Nat Commun ; 13(1): 161, 2022 01 10.
Artículo en Inglés | MEDLINE | ID: mdl-35013317

RESUMEN

Dravet syndrome is a severe epileptic encephalopathy caused primarily by haploinsufficiency of the SCN1A gene. Repetitive seizures can lead to endurable and untreatable neurological deficits. Whether this severe pathology is reversible after symptom onset remains unknown. To address this question, we generated a Scn1a conditional knock-in mouse model (Scn1a Stop/+) in which Scn1a expression can be re-activated on-demand during the mouse lifetime. Scn1a gene disruption leads to the development of seizures, often associated with sudden unexpected death in epilepsy (SUDEP) and behavioral alterations including hyperactivity, social interaction deficits and cognitive impairment starting from the second/third week of age. However, we showed that Scn1a gene re-activation when symptoms were already manifested (P30) led to a complete rescue of both spontaneous and thermic inducible seizures, marked amelioration of behavioral abnormalities and normalization of hippocampal fast-spiking interneuron firing. We also identified dramatic gene expression alterations, including those associated with astrogliosis in Dravet syndrome mice, that, accordingly, were rescued by Scn1a gene expression normalization at P30. Interestingly, regaining of Nav1.1 physiological level rescued seizures also in adult Dravet syndrome mice (P90) after months of repetitive attacks. Overall, these findings represent a solid proof-of-concept highlighting that disease phenotype reversibility can be achieved when Scn1a gene activity is efficiently reconstituted in brain cells.


Asunto(s)
Disfunción Cognitiva/genética , Epilepsias Mioclónicas/genética , Hipocampo/metabolismo , Interneuronas/metabolismo , Canal de Sodio Activado por Voltaje NAV1.1/genética , Muerte Súbita e Inesperada en la Epilepsia/prevención & control , Potenciales de Acción/fisiología , Animales , Cerebelo/metabolismo , Cerebelo/fisiopatología , Corteza Cerebral/metabolismo , Corteza Cerebral/fisiopatología , Disfunción Cognitiva/metabolismo , Disfunción Cognitiva/fisiopatología , Disfunción Cognitiva/prevención & control , Cuerpo Estriado/metabolismo , Cuerpo Estriado/fisiopatología , Dependovirus/genética , Dependovirus/metabolismo , Modelos Animales de Enfermedad , Epilepsias Mioclónicas/metabolismo , Epilepsias Mioclónicas/fisiopatología , Epilepsias Mioclónicas/prevención & control , Técnicas de Sustitución del Gen , Terapia Genética/métodos , Hipocampo/fisiopatología , Humanos , Interneuronas/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Canal de Sodio Activado por Voltaje NAV1.1/deficiencia , Muerte Súbita e Inesperada en la Epilepsia/patología
7.
Cereb Cortex ; 32(13): 2885-2894, 2022 06 16.
Artículo en Inglés | MEDLINE | ID: mdl-34791112

RESUMEN

Rett syndrome (RTT) is characterized by dysfunction in neuronal excitation/inhibition (E/I) balance, potentially impacting seizure susceptibility via deficits in K+/Cl- cotransporter 2 (KCC2) function. Mice lacking the Methyl-CpG binding protein 2 (MeCP2) recapitulate many symptoms of RTT, and recombinant human insulin-like growth factor-1 (rhIGF-1) restores KCC2 expression and E/I balance in MeCP2 KO mice. However, clinical trial outcomes of rhIGF-1 in RTT have been variable, and increasing its therapeutic efficacy is highly desirable. To this end, the neuropeptide oxytocin (OXT) is promising, as it also critically modulates KCC2 function during early postnatal development. We measured basal KCC2 expression levels in MeCP2 KO mice and identified 3 key frontal brain regions showing KCC2 alterations in young adult mice, but not in postnatal P10 animals. We hypothesized that deficits in an IGF-1/OXT signaling crosstalk modulating KCC2 may occur in RTT during postnatal development. Consistently, we detected alterations of IGF-1 receptor and OXT receptor levels in those brain areas. rhIGF-1 and OXT treatments in KO mice rescued KCC2 expression in a region-specific and complementary manner. These results suggest that region-selective combinatorial pharmacotherapeutic strategies could be most effective at normalizing E/I balance in key brain regions subtending the RTT pathophysiology.


Asunto(s)
Síndrome de Rett , Simportadores , Animales , Modelos Animales de Enfermedad , Factor I del Crecimiento Similar a la Insulina/metabolismo , Proteína 2 de Unión a Metil-CpG/metabolismo , Ratones , Oxitocina/metabolismo , Síndrome de Rett/tratamiento farmacológico , Síndrome de Rett/genética , Síndrome de Rett/metabolismo , Simportadores/genética , Simportadores/metabolismo
9.
Nat Commun ; 12(1): 6237, 2021 10 29.
Artículo en Inglés | MEDLINE | ID: mdl-34716339

RESUMEN

Recent findings in human samples and animal models support the involvement of inflammation in the development of Parkinson's disease. Nevertheless, it is currently unknown whether microglial activation constitutes a primary event in neurodegeneration. We generated a new mouse model by lentiviral-mediated selective α-synuclein (αSYN) accumulation in microglial cells. Surprisingly, these mice developed progressive degeneration of dopaminergic (DA) neurons without endogenous αSYN aggregation. Transcriptomics and functional assessment revealed that αSYN-accumulating microglial cells developed a strong reactive state with phagocytic exhaustion and excessive production of oxidative and proinflammatory molecules. This inflammatory state created a molecular feed-forward vicious cycle between microglia and IFNγ-secreting immune cells infiltrating the brain parenchyma. Pharmacological inhibition of oxidative and nitrosative molecule production was sufficient to attenuate neurodegeneration. These results suggest that αSYN accumulation in microglia induces selective DA neuronal degeneration by promoting phagocytic exhaustion, an excessively toxic environment and the selective recruitment of peripheral immune cells.


Asunto(s)
Neuronas Dopaminérgicas/patología , Microglía/metabolismo , Degeneración Nerviosa/patología , Fagocitosis/fisiología , alfa-Sinucleína/metabolismo , Inmunidad Adaptativa/fisiología , Animales , Receptor 1 de Quimiocinas CX3C/genética , Receptor 1 de Quimiocinas CX3C/metabolismo , Encefalitis/metabolismo , Encefalitis/patología , Expresión Génica , Inmunidad Innata/fisiología , Masculino , Ratones Endogámicos C57BL , Ratones Transgénicos , Microglía/efectos de los fármacos , Microglía/patología , Óxido Nítrico/metabolismo , Óxido Nítrico/toxicidad , Enfermedad de Parkinson/patología , Especies Reactivas de Oxígeno/metabolismo , Sustancia Negra/metabolismo , Sustancia Negra/patología , alfa-Sinucleína/genética
10.
Nat Commun ; 12(1): 4050, 2021 06 30.
Artículo en Inglés | MEDLINE | ID: mdl-34193871

RESUMEN

The investigation of genetic forms of juvenile neurodegeneration could shed light on the causative mechanisms of neuronal loss. Schinzel-Giedion syndrome (SGS) is a fatal developmental syndrome caused by mutations in the SETBP1 gene, inducing the accumulation of its protein product. SGS features multi-organ involvement with severe intellectual and physical deficits due, at least in part, to early neurodegeneration. Here we introduce a human SGS model that displays disease-relevant phenotypes. We show that SGS neural progenitors exhibit aberrant proliferation, deregulation of oncogenes and suppressors, unresolved DNA damage, and resistance to apoptosis. Mechanistically, we demonstrate that high SETBP1 levels inhibit P53 function through the stabilization of SET, which in turn hinders P53 acetylation. We find that the inheritance of unresolved DNA damage in SGS neurons triggers the neurodegenerative process that can be alleviated either by PARP-1 inhibition or by NAD + supplementation. These results implicate that neuronal death in SGS originates from developmental alterations mainly in safeguarding cell identity and homeostasis.


Asunto(s)
Anomalías Múltiples/patología , Proteínas Portadoras/metabolismo , Anomalías Craneofaciales/patología , Daño del ADN , Deformidades Congénitas de la Mano/patología , Trastornos Heredodegenerativos del Sistema Nervioso/patología , Discapacidad Intelectual/patología , Mutación , Uñas Malformadas/patología , Células-Madre Neurales/patología , Proteínas Nucleares/metabolismo , Proteína p53 Supresora de Tumor/antagonistas & inhibidores , Anomalías Múltiples/genética , Anomalías Múltiples/metabolismo , Proteínas Portadoras/genética , Células Cultivadas , Anomalías Craneofaciales/genética , Anomalías Craneofaciales/metabolismo , Deformidades Congénitas de la Mano/genética , Deformidades Congénitas de la Mano/metabolismo , Trastornos Heredodegenerativos del Sistema Nervioso/genética , Trastornos Heredodegenerativos del Sistema Nervioso/metabolismo , Humanos , Discapacidad Intelectual/genética , Discapacidad Intelectual/metabolismo , Uñas Malformadas/genética , Uñas Malformadas/metabolismo , Células-Madre Neurales/metabolismo , Proteínas Nucleares/genética , Organoides
11.
Nat Commun ; 11(1): 4178, 2020 08 21.
Artículo en Inglés | MEDLINE | ID: mdl-32826895

RESUMEN

Friedreich's ataxia (FRDA) is an autosomal-recessive neurodegenerative and cardiac disorder which occurs when transcription of the FXN gene is silenced due to an excessive expansion of GAA repeats into its first intron. Herein, we generate dorsal root ganglia organoids (DRG organoids) by in vitro differentiation of human iPSCs. Bulk and single-cell RNA sequencing show that DRG organoids present a transcriptional signature similar to native DRGs and display the main peripheral sensory neuronal and glial cell subtypes. Furthermore, when co-cultured with human intrafusal muscle fibers, DRG organoid sensory neurons contact their peripheral targets and reconstitute the muscle spindle proprioceptive receptors. FRDA DRG organoids model some molecular and cellular deficits of the disease that are rescued when the entire FXN intron 1 is removed, and not with the excision of the expanded GAA tract. These results strongly suggest that removal of the repressed chromatin flanking the GAA tract might contribute to rescue FXN total expression and fully revert the pathological hallmarks of FRDA DRG neurons.


Asunto(s)
Ataxia de Friedreich/genética , Ataxia de Friedreich/patología , Ganglios Espinales/metabolismo , Edición Génica/métodos , Proteínas de Unión a Hierro/genética , Organoides/metabolismo , Células Receptoras Sensoriales/metabolismo , Antioxidantes/farmacología , Sistemas CRISPR-Cas , Diferenciación Celular , Cromatina/metabolismo , Ataxia de Friedreich/tratamiento farmacológico , Ganglios Espinales/efectos de los fármacos , Ganglios Espinales/patología , Predisposición Genética a la Enfermedad/genética , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Intrones , Mitocondrias/metabolismo , Organoides/efectos de los fármacos , Organoides/patología , Células Receptoras Sensoriales/patología , Análisis de Secuencia de ARN , Transcriptoma , Frataxina
12.
Elife ; 92020 03 24.
Artículo en Inglés | MEDLINE | ID: mdl-32207685

RESUMEN

Rett syndrome is an incurable neurodevelopmental disorder caused by mutations in the gene encoding for methyl-CpG binding-protein 2 (MeCP2). Gene therapy for this disease presents inherent hurdles since MECP2 is expressed throughout the brain and its duplication leads to severe neurological conditions as well. Herein, we use the AAV-PHP.eB to deliver an instability-prone Mecp2 (iMecp2) transgene cassette which, increasing RNA destabilization and inefficient protein translation of the viral Mecp2 transgene, limits supraphysiological Mecp2 protein levels. Intravenous injections of the PHP.eB-iMecp2 virus in symptomatic Mecp2 mutant mice significantly improved locomotor activity, lifespan and gene expression normalization. Remarkably, PHP.eB-iMecp2 administration was well tolerated in female Mecp2 mutant or in wild-type animals. In contrast, we observed a strong immune response to the transgene in treated male Mecp2 mutant mice that was overcome by immunosuppression. Overall, PHP.eB-mediated delivery of iMecp2 provided widespread and efficient gene transfer maintaining physiological Mecp2 protein levels in the brain.


Asunto(s)
Encéfalo/metabolismo , Proteína 2 de Unión a Metil-CpG/genética , Mutación/genética , Síndrome de Rett/genética , Animales , Modelos Animales de Enfermedad , Expresión Génica/fisiología , Regulación de la Expresión Génica , Terapia Genética/métodos , Ratones Transgénicos , Transgenes/genética
13.
Cell Stem Cell ; 23(1): 13-14, 2018 07 05.
Artículo en Inglés | MEDLINE | ID: mdl-29979987

RESUMEN

In a recent issue of Nature, Tsunemoto et al. (2018) perform a systematic screening to identify several transcription factor pairs able to generate a variety of different induced neuronal cell populations that share a core neuronal signature, yet differ for specific molecular features.


Asunto(s)
Neuronas , Factores de Transcripción/genética , Regulación de la Expresión Génica
14.
Hum Mol Genet ; 27(5): 761-779, 2018 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-29281027

RESUMEN

P23H is the most common mutation in the RHODOPSIN (RHO) gene leading to a dominant form of retinitis pigmentosa (RP), a rod photoreceptor degeneration that invariably causes vision loss. Specific disruption of the disease P23H RHO mutant while preserving the wild-type (WT) functional allele would be an invaluable therapy for this disease. However, various technologies tested in the past failed to achieve effective changes and consequently therapeutic benefits. We validated a CRISPR/Cas9 strategy to specifically inactivate the P23H RHO mutant, while preserving the WT allele in vitro. We, then, translated this approach in vivo by delivering the CRISPR/Cas9 components in murine Rho+/P23H mutant retinae. Targeted retinae presented a high rate of cleavage in the P23H but not WT Rho allele. This gene manipulation was sufficient to slow photoreceptor degeneration and improve retinal functions. To improve the translational potential of our approach, we tested intravitreal delivery of this system by means of adeno-associated viruses (AAVs). To this purpose, the employment of the AAV9-PHP.B resulted the most effective in disrupting the P23H Rho mutant. Finally, this approach was translated successfully in human cells engineered with the homozygous P23H RHO gene mutation. Overall, this is a significant proof-of-concept that gene allele specific targeting by CRISPR/Cas9 technology is specific and efficient and represents an unprecedented tool for treating RP and more broadly dominant genetic human disorders affecting the eye, as well as other tissues.


Asunto(s)
Marcación de Gen/métodos , Vectores Genéticos , Retina/fisiología , Degeneración Retiniana/terapia , Rodopsina/genética , Alelos , Animales , Sistemas CRISPR-Cas , Electroporación/métodos , Fibroblastos , Terapia Genética/métodos , Células HEK293 , Humanos , Ratones Endogámicos C57BL , Ratones Mutantes , Ratones Transgénicos , Mutación , ARN Guía de Kinetoplastida , Retina/patología , Degeneración Retiniana/genética
15.
Mol Ther ; 25(12): 2727-2742, 2017 Dec 06.
Artículo en Inglés | MEDLINE | ID: mdl-28882452

RESUMEN

The lack of technology for direct global-scale targeting of the adult mouse nervous system has hindered research on brain processing and dysfunctions. Currently, gene transfer is normally achieved by intraparenchymal viral injections, but these injections target a restricted brain area. Herein, we demonstrated that intravenous delivery of adeno-associated virus (AAV)-PHP.B viral particles permeated and diffused throughout the neural parenchyma, targeting both the central and the peripheral nervous system in a global pattern. We then established multiple procedures of viral transduction to control gene expression or inactivate gene function exclusively in the adult nervous system and assessed the underlying behavioral effects. Building on these results, we established an effective gene therapy strategy to counteract the widespread accumulation of α-synuclein deposits throughout the forebrain in a mouse model of synucleinopathy. Transduction of A53T-SCNA transgenic mice with AAV-PHP.B-GBA1 restored physiological levels of the enzyme, reduced α-synuclein pathology, and produced significant behavioral recovery. Finally, we provided evidence that AAV-PHP.B brain penetration does not lead to evident dysfunctions in blood-brain barrier integrity or permeability. Altogether, the AAV-PHP.B viral platform enables non-invasive, widespread, and long-lasting global neural expression of therapeutic genes, such as GBA1, providing an invaluable approach to treat neurodegenerative diseases with diffuse brain pathology such as synucleinopathies.


Asunto(s)
Dependovirus/genética , Expresión Génica , Vectores Genéticos/genética , beta-Glucosidasa/metabolismo , Animales , Barrera Hematoencefálica/metabolismo , Encéfalo/metabolismo , Modelos Animales de Enfermedad , Electroencefalografía , Activación Enzimática , Orden Génico , Técnicas de Transferencia de Gen , Terapia Genética , Vectores Genéticos/administración & dosificación , Humanos , Ratones , Ratones Transgénicos , Neuronas/metabolismo , Transducción Genética , Proteína 1 del Complejo de la Esclerosis Tuberosa , Proteínas Supresoras de Tumor/genética , Proteínas Supresoras de Tumor/metabolismo , alfa-Sinucleína/genética , alfa-Sinucleína/metabolismo
16.
Sci Rep ; 6: 37540, 2016 11 18.
Artículo en Inglés | MEDLINE | ID: mdl-27857203

RESUMEN

The CRISPR/Cas9 system is a rapid and customizable tool for gene editing in mammalian cells. In particular, this approach has widely opened new opportunities for genetic studies in neurological disease. Human neurons can be differentiated in vitro from hPSC (human Pluripotent Stem Cells), hNPCs (human Neural Precursor Cells) or even directly reprogrammed from fibroblasts. Here, we described a new platform which enables, rapid and efficient CRISPR/Cas9-mediated genome targeting simultaneously with three different paradigms for in vitro generation of neurons. This system was employed to inactivate two genes associated with neurological disorder (TSC2 and KCNQ2) and achieved up to 85% efficiency of gene targeting in the differentiated cells. In particular, we devised a protocol that, combining the expression of the CRISPR components with neurogenic factors, generated functional human neurons highly enriched for the desired genome modification in only 5 weeks. This new approach is easy, fast and that does not require the generation of stable isogenic clones, practice that is time consuming and for some genes not feasible.


Asunto(s)
Diferenciación Celular/genética , Reprogramación Celular/genética , Células Madre Pluripotentes Inducidas/citología , Células-Madre Neurales/clasificación , Sistemas CRISPR-Cas/genética , Fibroblastos/citología , Fibroblastos/metabolismo , Silenciador del Gen , Vectores Genéticos , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Células-Madre Neurales/citología , Células-Madre Neurales/metabolismo , Neuronas/citología , Neuronas/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...