Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Nature ; 627(8002): 149-156, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38418876

RESUMEN

The glymphatic movement of fluid through the brain removes metabolic waste1-4. Noninvasive 40 Hz stimulation promotes 40 Hz neural activity in multiple brain regions and attenuates pathology in mouse models of Alzheimer's disease5-8. Here we show that multisensory gamma stimulation promotes the influx of cerebrospinal fluid and the efflux of interstitial fluid in the cortex of the 5XFAD mouse model of Alzheimer's disease. Influx of cerebrospinal fluid was associated with increased aquaporin-4 polarization along astrocytic endfeet and dilated meningeal lymphatic vessels. Inhibiting glymphatic clearance abolished the removal of amyloid by multisensory 40 Hz stimulation. Using chemogenetic manipulation and a genetically encoded sensor for neuropeptide signalling, we found that vasoactive intestinal peptide interneurons facilitate glymphatic clearance by regulating arterial pulsatility. Our findings establish novel mechanisms that recruit the glymphatic system to remove brain amyloid.


Asunto(s)
Enfermedad de Alzheimer , Amiloide , Encéfalo , Líquido Cefalorraquídeo , Líquido Extracelular , Ritmo Gamma , Sistema Glinfático , Animales , Ratones , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/patología , Enfermedad de Alzheimer/prevención & control , Amiloide/metabolismo , Acuaporina 4/metabolismo , Astrocitos/metabolismo , Encéfalo/citología , Encéfalo/metabolismo , Encéfalo/patología , Líquido Cefalorraquídeo/metabolismo , Modelos Animales de Enfermedad , Líquido Extracelular/metabolismo , Sistema Glinfático/fisiología , Interneuronas/metabolismo , Péptido Intestinal Vasoactivo/metabolismo , Corteza Cerebral/citología , Corteza Cerebral/metabolismo , Corteza Cerebral/patología , Estimulación Eléctrica
2.
Nat Aging ; 4(1): 95-109, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-38066314

RESUMEN

Aging is a common risk factor in neurodegenerative disorders. Investigating neuronal aging in an isogenic background stands to facilitate analysis of the interplay between neuronal aging and neurodegeneration. Here we perform direct neuronal reprogramming of longitudinally collected human fibroblasts to reveal genetic pathways altered at different ages. Comparative transcriptome analysis of longitudinally aged striatal medium spiny neurons (MSNs) in Huntington's disease identified pathways involving RCAN1, a negative regulator of calcineurin. Notably, RCAN1 protein increased with age in reprogrammed MSNs as well as in human postmortem striatum and RCAN1 knockdown rescued patient-derived MSNs of Huntington's disease from degeneration. RCAN1 knockdown enhanced chromatin accessibility of genes involved in longevity and autophagy, mediated through enhanced calcineurin activity, leading to TFEB's nuclear localization by dephosphorylation. Furthermore, G2-115, an analog of glibenclamide with autophagy-enhancing activities, reduced the RCAN1-calcineurin interaction, phenocopying the effect of RCAN1 knockdown. Our results demonstrate that targeting RCAN1 genetically or pharmacologically can increase neuronal resilience in Huntington's disease.


Asunto(s)
Calcineurina , Enfermedad de Huntington , Humanos , Anciano , Calcineurina/genética , Enfermedad de Huntington/genética , Envejecimiento/genética , Factores de Transcripción/metabolismo , Cuerpo Estriado/metabolismo , Proteínas de Unión al ADN/metabolismo , Proteínas Musculares/genética , Factores de Transcripción Básicos con Cremalleras de Leucinas y Motivos Hélice-Asa-Hélice/metabolismo
3.
Cell ; 186(20): 4365-4385.e27, 2023 09 28.
Artículo en Inglés | MEDLINE | ID: mdl-37774677

RESUMEN

Alzheimer's disease (AD) is the most common cause of dementia worldwide, but the molecular and cellular mechanisms underlying cognitive impairment remain poorly understood. To address this, we generated a single-cell transcriptomic atlas of the aged human prefrontal cortex covering 2.3 million cells from postmortem human brain samples of 427 individuals with varying degrees of AD pathology and cognitive impairment. Our analyses identified AD-pathology-associated alterations shared between excitatory neuron subtypes, revealed a coordinated increase of the cohesin complex and DNA damage response factors in excitatory neurons and in oligodendrocytes, and uncovered genes and pathways associated with high cognitive function, dementia, and resilience to AD pathology. Furthermore, we identified selectively vulnerable somatostatin inhibitory neuron subtypes depleted in AD, discovered two distinct groups of inhibitory neurons that were more abundant in individuals with preserved high cognitive function late in life, and uncovered a link between inhibitory neurons and resilience to AD pathology.


Asunto(s)
Enfermedad de Alzheimer , Encéfalo , Anciano , Humanos , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/patología , Encéfalo/metabolismo , Encéfalo/patología , Cognición , Disfunción Cognitiva/metabolismo , Neuronas/metabolismo
4.
Cell ; 186(20): 4386-4403.e29, 2023 09 28.
Artículo en Inglés | MEDLINE | ID: mdl-37774678

RESUMEN

Altered microglial states affect neuroinflammation, neurodegeneration, and disease but remain poorly understood. Here, we report 194,000 single-nucleus microglial transcriptomes and epigenomes across 443 human subjects and diverse Alzheimer's disease (AD) pathological phenotypes. We annotate 12 microglial transcriptional states, including AD-dysregulated homeostatic, inflammatory, and lipid-processing states. We identify 1,542 AD-differentially-expressed genes, including both microglia-state-specific and disease-stage-specific alterations. By integrating epigenomic, transcriptomic, and motif information, we infer upstream regulators of microglial cell states, gene-regulatory networks, enhancer-gene links, and transcription-factor-driven microglial state transitions. We demonstrate that ectopic expression of our predicted homeostatic-state activators induces homeostatic features in human iPSC-derived microglia-like cells, while inhibiting activators of inflammation can block inflammatory progression. Lastly, we pinpoint the expression of AD-risk genes in microglial states and differential expression of AD-risk genes and their regulators during AD progression. Overall, we provide insights underlying microglial states, including state-specific and AD-stage-specific microglial alterations at unprecedented resolution.


Asunto(s)
Enfermedad de Alzheimer , Microglía , Humanos , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/patología , Regulación de la Expresión Génica , Inflamación/patología , Microglía/metabolismo , Factores de Transcripción/metabolismo , Transcriptoma , Epigenoma
5.
Cell ; 186(20): 4422-4437.e21, 2023 09 28.
Artículo en Inglés | MEDLINE | ID: mdl-37774680

RESUMEN

Recent work has identified dozens of non-coding loci for Alzheimer's disease (AD) risk, but their mechanisms and AD transcriptional regulatory circuitry are poorly understood. Here, we profile epigenomic and transcriptomic landscapes of 850,000 nuclei from prefrontal cortexes of 92 individuals with and without AD to build a map of the brain regulome, including epigenomic profiles, transcriptional regulators, co-accessibility modules, and peak-to-gene links in a cell-type-specific manner. We develop methods for multimodal integration and detecting regulatory modules using peak-to-gene linking. We show AD risk loci are enriched in microglial enhancers and for specific TFs including SPI1, ELF2, and RUNX1. We detect 9,628 cell-type-specific ATAC-QTL loci, which we integrate alongside peak-to-gene links to prioritize AD variant regulatory circuits. We report differential accessibility of regulatory modules in late AD in glia and in early AD in neurons. Strikingly, late-stage AD brains show global epigenome dysregulation indicative of epigenome erosion and cell identity loss.


Asunto(s)
Enfermedad de Alzheimer , Encéfalo , Regulación de la Expresión Génica , Humanos , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/patología , Encéfalo/patología , Epigenoma , Epigenómica , Estudio de Asociación del Genoma Completo
6.
Res Sq ; 2023 May 09.
Artículo en Inglés | MEDLINE | ID: mdl-37214956

RESUMEN

Aging is a common risk factor in neurodegenerative disorders and the ability to investigate aging of neurons in an isogenic background would facilitate discovering the interplay between neuronal aging and onset of neurodegeneration. Here, we perform direct neuronal reprogramming of longitudinally collected human fibroblasts to reveal genetic pathways altered at different ages. Comparative transcriptome analysis of longitudinally aged striatal medium spiny neurons (MSNs), a primary neuronal subtype affected in Huntington's disease (HD), identified pathways associated with RCAN1, a negative regulator of calcineurin. Notably, RCAN1 undergoes age-dependent increase at the protein level detected in reprogrammed MSNs as well as in human postmortem striatum. In patient-derived MSNs of adult-onset HD (HD-MSNs), counteracting RCAN1 by gene knockdown (KD) rescued HD-MSNs from degeneration. The protective effect of RCAN1 KD was associated with enhanced chromatin accessibility of genes involved in longevity and autophagy, mediated through enhanced calcineurin activity, which in turn dephosphorylates and promotes nuclear localization of TFEB transcription factor. Furthermore, we reveal that G2-115 compound, an analog of glibenclamide with autophagy-enhancing activities, reduces the RCAN1-Calcineurin interaction, phenocopying the effect of RCAN1 KD. Our results demonstrate that RCAN1 is a potential genetic or pharmacological target whose reduction-of-function increases neuronal resilience to neurodegeneration in HD through chromatin reconfiguration.

7.
Cell Metab ; 34(9): 1227-1229, 2022 09 06.
Artículo en Inglés | MEDLINE | ID: mdl-36070677

RESUMEN

In this issue of Cell Metabolism, Traxler et al. report a metabolic switch in neurons that hinges on the critical balance of pyruvate kinase M (PKM) isoform ratios. Using directly induced neurons (iNs) derived from patients with Alzheimer's disease (AD), the work delineates the impact of an AD-specific switch from PKM1 to PKM2 onto neuronal resilience against neurodegeneration.


Asunto(s)
Neuronas , Piruvato Quinasa , Enfermedad de Alzheimer/patología , Humanos , Neuronas/metabolismo , Piruvato Quinasa/metabolismo
8.
Sci Adv ; 8(39): eabo4662, 2022 09 30.
Artículo en Inglés | MEDLINE | ID: mdl-36170369

RESUMEN

DNA double-strand breaks (DSBs) are linked to neurodegeneration and senescence. However, it is not clear how DSB-bearing neurons influence neuroinflammation associated with neurodegeneration. Here, we characterize DSB-bearing neurons from the CK-p25 mouse model of neurodegeneration using single-nucleus, bulk, and spatial transcriptomic techniques. DSB-bearing neurons enter a late-stage DNA damage response marked by nuclear factor κB (NFκB)-activated senescent and antiviral immune pathways. In humans, Alzheimer's disease pathology is closely associated with immune activation in excitatory neurons. Spatial transcriptomics reveal that regions of CK-p25 brain tissue dense with DSB-bearing neurons harbor signatures of inflammatory microglia, which is ameliorated by NFκB knockdown in neurons. Inhibition of NFκB in DSB-bearing neurons also reduces microglia activation in organotypic mouse brain slice culture. In conclusion, DSBs activate immune pathways in neurons, which in turn adopt a senescence-associated secretory phenotype to elicit microglia activation. These findings highlight a previously unidentified role for neurons in the mechanism of disease-associated neuroinflammation.


Asunto(s)
Roturas del ADN de Doble Cadena , Microglía , Animales , Antivirales/metabolismo , ADN/metabolismo , Humanos , Ratones , Microglía/metabolismo , FN-kappa B/metabolismo , Neuronas/metabolismo
9.
Cell Stem Cell ; 29(8): 1197-1212.e8, 2022 08 04.
Artículo en Inglés | MEDLINE | ID: mdl-35931030

RESUMEN

Apolipoprotein E4 (APOE4) is the greatest known genetic risk factor for developing sporadic Alzheimer's disease. How the interaction of APOE4 microglia with neurons differs from microglia expressing the disease-neutral APOE3 allele remains unknown. Here, we employ CRISPR-edited induced pluripotent stem cells (iPSCs) to dissect the impact of APOE4 in neuron-microglia communication. Our results reveal that APOE4 induces a lipid-accumulated state that renders microglia weakly responsive to neuronal activity. By examining the transcriptional signatures of APOE3 versus APOE4 microglia in response to neuronal conditioned media, we established that neuronal cues differentially induce a lipogenic program in APOE4 microglia that exacerbates pro-inflammatory signals. Through decreased uptake of extracellular fatty acids and lipoproteins, we identified that APOE4 microglia disrupts the coordinated activity of neuronal ensembles. These findings suggest that abnormal neuronal network-level disturbances observed in Alzheimer's disease patients harboring APOE4 may in part be triggered by impairment in lipid homeostasis in non-neuronal cells.


Asunto(s)
Enfermedad de Alzheimer , Apolipoproteína E4 , Apolipoproteína E3/genética , Apolipoproteína E4/genética , Humanos , Microglía , Neuronas
10.
Nat Rev Neurol ; 18(1): 25-39, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34750588

RESUMEN

Alzheimer disease (AD) is the most prevalent type of dementia. It is marked by severe memory loss and cognitive decline, and currently has limited effective treatment options. Although individuals with AD have common neuropathological hallmarks, emerging data suggest that the disease has a complex polygenic aetiology, and more than 25 genetic loci have been linked to an elevated risk of AD and dementia. Nevertheless, our ability to decipher the cellular and molecular mechanisms that underlie genetic susceptibility to AD, and its progression and severity, remains limited. Here, we discuss ongoing efforts to leverage genomic data from patients using cellular reprogramming technologies to recapitulate complex brain systems and build in vitro discovery platforms. Much attention has already been given to methodologies to derive major brain cell types from pluripotent stem cells. We therefore focus on technologies that combine multiple cell types to recreate anatomical and physiological properties of human brain tissue in vitro. We discuss the advances in the field for modelling four domains that have come into view as key contributors to the pathogenesis of AD: the blood-brain barrier, myelination, neuroinflammation and neuronal circuits. We also highlight opportunities for the field to further interrogate the complex genetic and environmental factors of AD using in vitro models.


Asunto(s)
Enfermedad de Alzheimer/patología , Encéfalo/patología , Modelos Anatómicos , Modelos Neurológicos , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/fisiopatología , Animales , Encéfalo/fisiología , Encéfalo/fisiopatología , Predisposición Genética a la Enfermedad , Humanos
13.
Nat Med ; 26(6): 952-963, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32514169

RESUMEN

In Alzheimer's disease, amyloid deposits along the brain vasculature lead to a condition known as cerebral amyloid angiopathy (CAA), which impairs blood-brain barrier (BBB) function and accelerates cognitive degeneration. Apolipoprotein (APOE4) is the strongest risk factor for CAA, yet the mechanisms underlying this genetic susceptibility are unknown. Here we developed an induced pluripotent stem cell-based three-dimensional model that recapitulates anatomical and physiological properties of the human BBB in vitro. Similarly to CAA, our in vitro BBB displayed significantly more amyloid accumulation in APOE4 compared to APOE3. Combinatorial experiments revealed that dysregulation of calcineurin-nuclear factor of activated T cells (NFAT) signaling and APOE in pericyte-like mural cells induces APOE4-associated CAA pathology. In the human brain, APOE and NFAT are selectively dysregulated in pericytes of APOE4 carriers, and inhibition of calcineurin-NFAT signaling reduces APOE4-associated CAA pathology in vitro and in vivo. Our study reveals the role of pericytes in APOE4-mediated CAA and highlights calcineurin-NFAT signaling as a therapeutic target in CAA and Alzheimer's disease.


Asunto(s)
Apolipoproteína E4/genética , Barrera Hematoencefálica/metabolismo , Calcineurina/metabolismo , Angiopatía Amiloide Cerebral/genética , Factores de Transcripción NFATC/genética , Pericitos/metabolismo , Péptidos beta-Amiloides/metabolismo , Apolipoproteína E3/genética , Apolipoproteína E3/metabolismo , Apolipoproteína E4/metabolismo , Barrera Hematoencefálica/citología , Humanos , Técnicas In Vitro , Células Madre Pluripotentes Inducidas , Factores de Transcripción NFATC/metabolismo , Permeabilidad , RNA-Seq , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
14.
Neuron ; 105(5): 813-821.e6, 2020 03 04.
Artículo en Inglés | MEDLINE | ID: mdl-31899071

RESUMEN

Despite being an autosomal dominant disorder caused by a known coding mutation in the gene HTT, Huntington's disease (HD) patients with similar trinucleotide repeat mutations can have an age of onset that varies by decades. One likely contributing factor is the genetic heterogeneity of patients that might modify their vulnerability to disease. We report that although the heterozygous depletion of the autophagy adaptor protein Alfy/Wdfy3 has no consequence in control mice, it significantly accelerates age of onset and progression of HD pathogenesis. Alfy is required in the adult brain for the autophagy-dependent clearance of proteinaceous deposits, and its depletion in mice and neurons derived from patient fibroblasts accelerates the aberrant accumulation of this pathological hallmark shared across adult-onset neurodegenerative diseases. These findings indicate that selectively compromising the ability to eliminate aggregated proteins is a pathogenic driver, and the selective elimination of aggregates may confer disease resistance.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Relacionadas con la Autofagia/genética , Enfermedad de Huntington/genética , Macroautofagia/genética , Neuronas/metabolismo , Agregación Patológica de Proteínas/genética , Edad de Inicio , Animales , Muerte Celular/genética , Modelos Animales de Enfermedad , Femenino , Fibroblastos , Humanos , Proteína Huntingtina/genética , Proteína Huntingtina/metabolismo , Enfermedad de Huntington/metabolismo , Enfermedad de Huntington/fisiopatología , Masculino , Ratones , Ratones Noqueados , Agregación Patológica de Proteínas/metabolismo , Agregación Patológica de Proteínas/fisiopatología
15.
Bioinformatics ; 34(13): i422-i428, 2018 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-29950021

RESUMEN

Motivation: Reprogramming somatic cells into neurons holds great promise to model neuronal development and disease. The efficiency and success rate of neuronal reprogramming, however, may vary between different conversion platforms and cell types, thereby necessitating an unbiased, systematic approach to estimate neuronal identity of converted cells. Recent studies have demonstrated that long genes (>100 kb from transcription start to end) are highly enriched in neurons, which provides an opportunity to identify neurons based on the expression of these long genes. Results: We have developed a versatile R package, LONGO, to analyze gene expression based on gene length. We propose a systematic analysis of long gene expression (LGE) with a metric termed the long gene quotient (LQ) that quantifies LGE in RNA-seq or microarray data to validate neuronal identity at the single-cell and population levels. This unique feature of neurons provides an opportunity to utilize measurements of LGE in transcriptome data to quickly and easily distinguish neurons from non-neuronal cells. By combining this conceptual advancement and statistical tool in a user-friendly and interactive software package, we intend to encourage and simplify further investigation into LGE, particularly as it applies to validating and improving neuronal differentiation and reprogramming methodologies. Availability and implementation: LONGO is freely available for download at https://github.com/biohpc/longo. Supplementary information: Supplementary data are available at Bioinformatics online.


Asunto(s)
Reprogramación Celular , Perfilación de la Expresión Génica/métodos , Neuronas/metabolismo , Programas Informáticos , Anciano , Femenino , Humanos , Masculino , Persona de Mediana Edad , Neuronas/fisiología , Análisis de Secuencia por Matrices de Oligonucleótidos/métodos , Análisis de Secuencia de ARN/métodos , Transcriptoma
16.
Nat Neurosci ; 21(3): 341-352, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-29403030

RESUMEN

In Huntington's disease (HD), expansion of CAG codons in the huntingtin gene (HTT) leads to the aberrant formation of protein aggregates and the differential degeneration of striatal medium spiny neurons (MSNs). Modeling HD using patient-specific MSNs has been challenging, as neurons differentiated from induced pluripotent stem cells are free of aggregates and lack an overt cell death phenotype. Here we generated MSNs from HD patient fibroblasts through microRNA-based direct neuronal conversion, bypassing the induction of pluripotency and retaining age signatures of the original fibroblasts. We found that patient MSNs consistently exhibited mutant HTT (mHTT) aggregates, mHTT-dependent DNA damage, mitochondrial dysfunction and spontaneous degeneration in culture over time. We further provide evidence that erasure of age stored in starting fibroblasts or neuronal conversion of presymptomatic HD patient fibroblasts results in differential manifestation of cellular phenotypes associated with HD, highlighting the importance of age in modeling late-onset neurological disorders.


Asunto(s)
Envejecimiento/patología , Fibroblastos/patología , Enfermedad de Huntington/patología , Neostriado/patología , Neuronas/patología , Daño del ADN , Potenciales Postsinápticos Excitadores/genética , Perfilación de la Expresión Génica , Humanos , Proteína Huntingtina/biosíntesis , Proteína Huntingtina/genética , MicroARNs/genética , Enfermedades Mitocondriales/patología , Enfermedades Neurodegenerativas/patología , Estrés Oxidativo , Fenotipo , Células Madre Pluripotentes
17.
Elife ; 52016 09 20.
Artículo en Inglés | MEDLINE | ID: mdl-27644593

RESUMEN

Aging is a major risk factor in many forms of late-onset neurodegenerative disorders. The ability to recapitulate age-related characteristics of human neurons in culture will offer unprecedented opportunities to study the biological processes underlying neuronal aging. Here, we show that using a recently demonstrated microRNA-based cellular reprogramming approach, human fibroblasts from postnatal to near centenarian donors can be efficiently converted into neurons that maintain multiple age-associated signatures. Application of an epigenetic biomarker of aging (referred to as epigenetic clock) to DNA methylation data revealed that the epigenetic ages of fibroblasts were highly correlated with corresponding age estimates of reprogrammed neurons. Transcriptome and microRNA profiles reveal genes differentially expressed between young and old neurons. Further analyses of oxidative stress, DNA damage and telomere length exhibit the retention of age-associated cellular properties in converted neurons from corresponding fibroblasts. Our results collectively demonstrate the maintenance of age after neuronal conversion.


Asunto(s)
Envejecimiento , Transdiferenciación Celular , Fibroblastos/fisiología , MicroARNs/metabolismo , Neuronas/fisiología , Técnicas Citológicas , Daño del ADN , Epigénesis Genética , Perfilación de la Expresión Génica , Humanos , Estrés Oxidativo , Telómero
18.
Nat Protoc ; 10(10): 1543-55, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26379228

RESUMEN

The ability to generate human neurons of specific subtypes of clinical importance offers experimental platforms that may be instrumental for disease modeling. We recently published a study demonstrating the use of neuronal microRNAs (miRNAs) and transcription factors to directly convert human fibroblasts to a highly enriched population of striatal medium spiny neurons (MSNs), a neuronal subpopulation that has a crucial role in motor control and harbors selective susceptibility to cell death in Huntington's disease. Here we describe a stepwise protocol for the generation of MSNs by direct neuronal conversion of human fibroblasts in 30 d. We provide descriptions of cellular behaviors during reprogramming and crucial steps involved in gene delivery, cell adhesion and culturing conditions that promote cell survival. Our protocol offers a unique approach to combine microRNAs and transcription factors to guide the neuronal conversion of human fibroblasts toward a specific neuronal subtype.


Asunto(s)
Técnicas de Cultivo de Célula/métodos , Diferenciación Celular , Fibroblastos/citología , MicroARNs/metabolismo , Neuronas/citología , Animales , Adhesión Celular , Humanos , MicroARNs/genética
19.
Neuron ; 84(2): 311-23, 2014 Oct 22.
Artículo en Inglés | MEDLINE | ID: mdl-25374357

RESUMEN

The promise of using reprogrammed human neurons for disease modeling and regenerative medicine relies on the ability to induce patient-derived neurons with high efficiency and subtype specificity. We have previously shown that ectopic expression of brain-enriched microRNAs (miRNAs), miR-9/9* and miR-124 (miR-9/9*-124), promoted direct conversion of human fibroblasts into neurons. Here we show that coexpression of miR-9/9*-124 with transcription factors enriched in the developing striatum, BCL11B (also known as CTIP2), DLX1, DLX2, and MYT1L, can guide the conversion of human postnatal and adult fibroblasts into an enriched population of neurons analogous to striatal medium spiny neurons (MSNs). When transplanted in the mouse brain, the reprogrammed human cells persisted in situ for over 6 months, exhibited membrane properties equivalent to native MSNs, and extended projections to the anatomical targets of MSNs. These findings highlight the potential of exploiting the synergism between miR-9/9*-124 and transcription factors to generate specific neuronal subtypes.


Asunto(s)
Diferenciación Celular/fisiología , Cuerpo Estriado/metabolismo , Fibroblastos/citología , Fibroblastos/metabolismo , MicroARNs/metabolismo , Neostriado/citología , Neuronas/metabolismo , Animales , Células Cultivadas , Humanos , Ratones , Factores de Transcripción/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...