Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
bioRxiv ; 2024 May 12.
Artículo en Inglés | MEDLINE | ID: mdl-38766140

RESUMEN

Midbrain dopamine neurons (DNs) respond to a first exposure to addictive drugs and play key roles in chronic drug usage1-3. As the synaptic and transcriptional changes that follow an acute cocaine exposure are mostly resolved within a few days4,5, the molecular changes that encode the long-term cellular memory of the exposure within DNs remain unknown. To investigate whether a single cocaine exposure induces long-term changes in the 3D genome structure of DNs, we applied Genome Architecture Mapping and single nucleus transcriptomic analyses in the mouse midbrain. We found extensive rewiring of 3D genome architecture at 24 hours past exposure which remains or worsens by 14 days, outlasting transcriptional responses. The cocaine-induced chromatin rewiring occurs at all genomic scales and affects genes with major roles in cocaine-induced synaptic changes. A single cocaine exposure triggers extensive long-lasting changes in chromatin condensation in post-synaptic and post-transcriptional regulatory genes, for example the unfolding of Rbfox1 which becomes most prominent 14 days post exposure. Finally, structurally remodeled genes are most expressed in a specific DN sub-type characterized by low expression of the dopamine auto-receptor Drd2, a key feature of highly cocaine-sensitive cells. These results reveal an important role for long-lasting 3D genome remodelling in the cellular memory of a single cocaine exposure, providing new hypotheses for understanding the inception of drug addiction and 3D genome plasticity.

2.
Sci Adv ; 8(41): eabn8367, 2022 10 14.
Artículo en Inglés | MEDLINE | ID: mdl-36223459

RESUMEN

Schizophrenia is one of the most widespread and complex mental disorders. To characterize the impact of schizophrenia, we performed single-nucleus RNA sequencing (snRNA-seq) of >220,000 neurons from the dorsolateral prefrontal cortex of patients with schizophrenia and matched controls. In addition, >115,000 neurons were analyzed topographically by immunohistochemistry. Compositional analysis of snRNA-seq data revealed a reduction in abundance of GABAergic neurons and a concomitant increase in principal neurons, most pronounced for upper cortical layer subtypes, which was substantiated by histological analysis. Many neuronal subtypes showed extensive transcriptomic changes, the most marked in upper-layer GABAergic neurons, including down-regulation in energy metabolism and up-regulation in neurotransmission. Transcription factor network analysis demonstrated a developmental origin of transcriptomic changes. Last, Visium spatial transcriptomics further corroborated upper-layer neuron vulnerability in schizophrenia. Overall, our results point toward general network impairment within upper cortical layers as a core substrate associated with schizophrenia symptomatology.


Asunto(s)
Esquizofrenia , Neuronas GABAérgicas/metabolismo , Humanos , Corteza Prefrontal/metabolismo , ARN Nuclear Pequeño/metabolismo , Esquizofrenia/patología , Factores de Transcripción/metabolismo
3.
Front Cell Dev Biol ; 10: 976549, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36046338

RESUMEN

Stellate cells are principal neurons in the entorhinal cortex that contribute to spatial processing. They also play a role in the context of Alzheimer's disease as they accumulate Amyloid beta early in the disease. Producing human stellate cells from pluripotent stem cells would allow researchers to study early mechanisms of Alzheimer's disease, however, no protocols currently exist for producing such cells. In order to develop novel stem cell protocols, we characterize at high resolution the development of the porcine medial entorhinal cortex by tracing neuronal and glial subtypes from mid-gestation to the adult brain to identify the transcriptomic profile of progenitor and adult stellate cells. Importantly, we could confirm the robustness of our data by extracting developmental factors from the identified intermediate stellate cell cluster and implemented these factors to generate putative intermediate stellate cells from human induced pluripotent stem cells. Six transcription factors identified from the stellate cell cluster including RUNX1T1, SOX5, FOXP1, MEF2C, TCF4, EYA2 were overexpressed using a forward programming approach to produce neurons expressing a unique combination of RELN, SATB2, LEF1 and BCL11B observed in stellate cells. Further analyses of the individual transcription factors led to the discovery that FOXP1 is critical in the reprogramming process and omission of RUNX1T1 and EYA2 enhances neuron conversion. Our findings contribute not only to the profiling of cell types within the developing and adult brain's medial entorhinal cortex but also provides proof-of-concept for using scRNAseq data to produce entorhinal intermediate stellate cells from human pluripotent stem cells in-vitro.

4.
NPJ Breast Cancer ; 8(1): 81, 2022 Jul 12.
Artículo en Inglés | MEDLINE | ID: mdl-35821504

RESUMEN

Normal breast luminal epithelial progenitors have been implicated as cell of origin in basal-like breast cancer, but their anatomical localization remains understudied. Here, we combine collection under the microscope of organoids from reduction mammoplasties and single-cell mRNA sequencing (scRNA-seq) of FACS-sorted luminal epithelial cells with multicolor imaging to profile ducts and terminal duct lobular units (TDLUs) and compare them with breast cancer subtypes. Unsupervised clustering reveals eleven distinct clusters and a differentiation trajectory starting with keratin 15+ (K15+) progenitors enriched in ducts. Spatial mapping of luminal progenitors is confirmed at the protein level by staining with critical duct markers. Comparison of the gene expression profiles of normal luminal cells with those of breast cancer subtypes suggests a strong correlation between normal breast ductal progenitors and basal-like breast cancer. We propose that K15+ basal-like breast cancers originate in ductal progenitors, which emphasizes the importance of not only lineages but also cellular position within the ductal-lobular tree.

5.
Cell Rep ; 37(8): 110050, 2021 11 23.
Artículo en Inglés | MEDLINE | ID: mdl-34818537

RESUMEN

Germ cells have evolved unique mechanisms to ensure the transmission of genetically and nongenetically encoded information, whose alteration compromises germ cell immortality. Chromatin factors play fundamental roles in these mechanisms. H3K36 and H3K27 methyltransferases shape and propagate a pattern of histone methylation essential for C. elegans germ cell maintenance, but the role of respective histone demethylases remains unexplored. Here, we show that jmjd-5 regulates H3K36me2 and H3K27me3 levels, preserves germline immortality, and protects germ cell identity by controlling gene expression. The transcriptional and biological effects of jmjd-5 loss can be hindered by the removal of H3K27demethylases, indicating that H3K36/K27 demethylases act in a transcriptional framework and promote the balance between H3K36 and H3K27 methylation required for germ cell immortality. Furthermore, we find that in wild-type, but not in jmjd-5 mutants, alterations of H3K36 methylation and transcription occur at high temperature, suggesting a role for jmjd-5 in adaptation to environmental changes.


Asunto(s)
Células Germinativas/metabolismo , Histona Demetilasas/metabolismo , Animales , Caenorhabditis elegans/metabolismo , Proteínas de Caenorhabditis elegans/metabolismo , Cromatina/metabolismo , N-Metiltransferasa de Histona-Lisina/metabolismo , Histonas/metabolismo , Histona Demetilasas con Dominio de Jumonji/metabolismo , Metilación
6.
Genes Chromosomes Cancer ; 60(7): 504-524, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-33611828

RESUMEN

The ability to capture alterations in the genome or transcriptome by next-generation sequencing has provided critical insight into molecular changes and programs underlying cancer biology. With the rapid technological development in single-cell sequencing, it has become possible to study individual cells at the transcriptional, genetic, epigenetic, and protein level. Using single-cell analysis, an increased resolution of fundamental processes underlying cancer development is obtained, providing comprehensive insights otherwise lost by sequencing of entire (bulk) samples, in which molecular signatures of individual cells are averaged across the entire cell population. Here, we provide a concise overview on the application of single-cell analysis of different modalities within cancer research by highlighting key articles of their respective fields. We furthermore examine the potential of existing technologies to meet clinical diagnostic needs and discuss current challenges associated with this translation.


Asunto(s)
Pruebas Genéticas/métodos , Neoplasias/genética , RNA-Seq/métodos , Análisis de la Célula Individual/métodos , Investigación Biomédica Traslacional/métodos , Animales , Pruebas Genéticas/normas , Humanos , Neoplasias/diagnóstico , RNA-Seq/normas , Análisis de la Célula Individual/normas , Investigación Biomédica Traslacional/normas
8.
Nat Commun ; 11(1): 5038, 2020 10 07.
Artículo en Inglés | MEDLINE | ID: mdl-33028830

RESUMEN

Epilepsy is one of the most common neurological disorders, yet its pathophysiology is poorly understood due to the high complexity of affected neuronal circuits. To identify dysfunctional neuronal subtypes underlying seizure activity in the human brain, we have performed single-nucleus transcriptomics analysis of >110,000 neuronal transcriptomes derived from temporal cortex samples of multiple temporal lobe epilepsy and non-epileptic subjects. We found that the largest transcriptomic changes occur in distinct neuronal subtypes from several families of principal neurons (L5-6_Fezf2 and L2-3_Cux2) and GABAergic interneurons (Sst and Pvalb), whereas other subtypes in the same families were less affected. Furthermore, the subtypes with the largest epilepsy-related transcriptomic changes may belong to the same circuit, since we observed coordinated transcriptomic shifts across these subtypes. Glutamate signaling exhibited one of the strongest dysregulations in epilepsy, highlighted by layer-wise transcriptional changes in multiple glutamate receptor genes and strong upregulation of genes coding for AMPA receptor auxiliary subunits. Overall, our data reveal a neuronal subtype-specific molecular phenotype of epilepsy.


Asunto(s)
Epilepsia Refractaria/genética , Epilepsia del Lóbulo Temporal/genética , Neuronas/patología , Lóbulo Temporal/patología , Transcriptoma/genética , Adolescente , Adulto , Biopsia , Estudios de Casos y Controles , Núcleo Celular/genética , Núcleo Celular/metabolismo , Conjuntos de Datos como Asunto , Epilepsia Refractaria/diagnóstico , Epilepsia Refractaria/patología , Epilepsia Refractaria/cirugía , Epilepsia del Lóbulo Temporal/diagnóstico , Epilepsia del Lóbulo Temporal/patología , Epilepsia del Lóbulo Temporal/cirugía , Femenino , Ácido Glutámico/metabolismo , Humanos , Imagen por Resonancia Magnética , Masculino , Microdisección , Persona de Mediana Edad , Modelos Genéticos , Red Nerviosa/metabolismo , Red Nerviosa/patología , Neuronas/citología , Neuronas/metabolismo , RNA-Seq , Receptores AMPA/genética , Receptores AMPA/metabolismo , Receptores de Glutamato/genética , Receptores de Glutamato/metabolismo , Transducción de Señal/genética , Análisis de la Célula Individual , Lóbulo Temporal/citología , Lóbulo Temporal/diagnóstico por imagen , Lóbulo Temporal/cirugía , Transcripción Genética , Regulación hacia Arriba , Adulto Joven
9.
EMBO J ; 39(18): e105759, 2020 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-32744742

RESUMEN

Parvalbumin-positive (PV+ ) fast-spiking interneurons are essential to control the firing activity of principal neuron ensembles, thereby regulating cognitive processes. The high firing frequency activity of PV+ interneurons imposes high-energy demands on their metabolism that must be supplied by distinctive machinery for energy generation. Exploring single-cell transcriptomic data for the mouse cortex, we identified a metabolism-associated gene with highly restricted expression to PV+ interneurons: Cox6a2, which codes for an isoform of a cytochrome c oxidase subunit. Cox6a2 deletion in mice disrupts perineuronal nets and enhances oxidative stress in PV+ interneurons, which in turn impairs the maturation of their morphological and functional properties. Such dramatic effects were likely due to an essential role of COX6A2 in energy balance of PV+ interneurons, underscored by a decrease in the ATP-to-ADP ratio in Cox6a2-/- PV+ interneurons. Energy disbalance and aberrant maturation likely hinder the integration of PV+ interneurons into cortical neuronal circuits, leading to behavioral alterations in mice. Additionally, in a human patient bearing mutations in COX6A2, we found a potential association of the mutations with mental/neurological abnormalities.


Asunto(s)
Complejo IV de Transporte de Electrones/metabolismo , Metabolismo Energético , Interneuronas/enzimología , Proteínas Musculares/metabolismo , Estrés Oxidativo , Adenosina Difosfato/genética , Adenosina Difosfato/metabolismo , Adenosina Trifosfato/genética , Adenosina Trifosfato/metabolismo , Anciano , Animales , Complejo IV de Transporte de Electrones/genética , Femenino , Humanos , Masculino , Ratones , Ratones Noqueados , Persona de Mediana Edad , Proteínas Musculares/genética
10.
JCI Insight ; 5(4)2020 02 27.
Artículo en Inglés | MEDLINE | ID: mdl-31999645

RESUMEN

Cell therapy raises hopes high for better treatment of brain disorders. However, the majority of transplanted cells often die soon after transplantation, and those that survive initially continue to die in the subacute phase, diminishing the impact of transplantations. In this study, we genetically modified transplanted human neural stem cells (hNSCs), from 2 distant embryonic stem cell lines (H9 and RC17), to express 1 of 4 prosurvival factors - Hif1a, Akt1, Bcl-2, or Bcl-xl - and studied how these modifications improve short- and long-term survival of transplanted hNSCs. All genetic modifications dramatically increased survival of the transplanted hNSCs. Importantly, 3 out of 4 modifications also enhanced the exit of hNSCs from the cell cycle, thus avoiding aberrant growth of the transplants. Bcl-xl expression provided the strongest protection of transplanted cells, reducing both immediate and delayed cell death, and stimulated hNSC differentiation toward neuronal and oligodendroglial lineages. By designing hNSCs with drug-controlled expression of Bcl-xl, we demonstrated that short-term expression of a prosurvival factor can ensure the long-term survival of transplanted cells. Importantly, transplantation of Bcl-xl-expressing hNSCs into mice suffering from stroke improved behavioral outcome and recovery of motor activity in mice.


Asunto(s)
Diferenciación Celular , Supervivencia Celular/genética , Células-Madre Neurales/citología , Trasplante de Células Madre , Accidente Cerebrovascular/patología , Animales , Ciclo Celular , Humanos , Ratones , Ratones Endogámicos NOD , Ratones SCID , Actividad Motora , Accidente Cerebrovascular/metabolismo , Resultado del Tratamiento , Proteína bcl-X/genética
11.
Mol Psychiatry ; 25(10): 2313-2329, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-31595033

RESUMEN

Severe infections during pregnancy are one of the major risk factors for cognitive impairment in the offspring. It has been suggested that maternal inflammation leads to dysfunction of cortical GABAergic interneurons that in turn underlies cognitive impairment of the affected offspring. However, the evidence comes largely from studies of adult or mature brains and how the impairment of inhibitory circuits arises upon maternal inflammation is unknown. Here we show that maternal inflammation affects multiple steps of cortical GABAergic interneuron development, i.e., proliferation of precursor cells, migration and positioning of neuroblasts, as well as neuronal maturation. Importantly, the development of distinct subtypes of cortical GABAergic interneurons was discretely impaired as a result of maternal inflammation. This translated into a reduction in cell numbers, redistribution across cortical regions and layers, and changes in morphology and cellular properties. Furthermore, selective vulnerability of GABAergic interneuron subtypes was associated with the stage of brain development. Thus, we propose that maternally derived insults have developmental stage-dependent effects, which contribute to the complex etiology of cognitive impairment in the affected offspring.


Asunto(s)
Corteza Cerebral , Inflamación , Interneuronas , Madres , Neurogénesis , Animales , Movimiento Celular , Proliferación Celular , Corteza Cerebral/patología , Disfunción Cognitiva/etiología , Disfunción Cognitiva/patología , Femenino , Neuronas GABAérgicas/patología , Interneuronas/clasificación , Interneuronas/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Embarazo , Efectos Tardíos de la Exposición Prenatal/patología
12.
Cell Stem Cell ; 21(6): 791-805.e9, 2017 Dec 07.
Artículo en Inglés | MEDLINE | ID: mdl-29174331

RESUMEN

Reprogramming of cellular identity using exogenous expression of transcription factors (TFs) is a powerful and exciting tool for tissue engineering, disease modeling, and regenerative medicine. However, generation of desired cell types using this approach is often plagued by inefficiency, slow conversion, and an inability to produce mature functional cells. Here, we show that expression of constitutively active SMAD2/3 significantly improves the efficiency of induced pluripotent stem cell (iPSC) generation by the Yamanaka factors. Mechanistically, SMAD3 interacts with reprogramming factors and co-activators and co-occupies OCT4 target loci during reprogramming. Unexpectedly, active SMAD2/3 also markedly enhances three other TF-mediated direct reprogramming conversions, from B cells to macrophages, myoblasts to adipocytes, and human fibroblasts to neurons, highlighting broad and general roles for SMAD2/3 as cell-reprogramming potentiators. Our results suggest that co-expression of active SMAD2/3 could enhance multiple types of TF-based cell identity conversion and therefore be a powerful tool for cellular engineering.


Asunto(s)
Reprogramación Celular , Células Madre Pluripotentes Inducidas/metabolismo , Proteína Smad2/metabolismo , Proteína smad3/metabolismo , Factores de Transcripción/metabolismo , Línea Celular , Humanos , Factores de Transcripción/genética
13.
Cell Death Dis ; 8(3): e2643, 2017 03 02.
Artículo en Inglés | MEDLINE | ID: mdl-28252642

RESUMEN

Neurogenic regions of mammalian brain produce many more neurons that will eventually survive and reach a mature stage. Developmental cell death affects both embryonically produced immature neurons and those immature neurons that are generated in regions of adult neurogenesis. Removal of substantial numbers of neurons that are not yet completely integrated into the local circuits helps to ensure that maturation and homeostatic function of neuronal networks in the brain proceed correctly. External signals from brain microenvironment together with intrinsic signaling pathways determine whether a particular neuron will die. To accommodate this signaling, immature neurons in the brain express a number of transmembrane factors as well as intracellular signaling molecules that will regulate the cell survival/death decision, and many of these factors cease being expressed upon neuronal maturation. Furthermore, pro-survival factors and intracellular responses depend on the type of neuron and region of the brain. Thus, in addition to some common neuronal pro-survival signaling, different types of neurons possess a variety of 'neuron type-specific' pro-survival constituents that might help them to adapt for survival in a certain brain region. This review focuses on how immature neurons survive during normal and impaired brain development, both in the embryonic/neonatal brain and in brain regions associated with adult neurogenesis, and emphasizes neuron type-specific mechanisms that help to survive for various types of immature neurons. Importantly, we mainly focus on in vivo data to describe neuronal survival specifically in the brain, without extrapolating data obtained in the PNS or spinal cord, and thus emphasize the influence of the complex brain environment on neuronal survival during development.


Asunto(s)
Encéfalo/fisiología , Supervivencia Celular/fisiología , Neuronas/fisiología , Animales , Humanos , Neurogénesis/fisiología , Transducción de Señal/fisiología
14.
Stem Cells Int ; 2017: 5718608, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-29317869

RESUMEN

Direct conversion of human somatic cells to induced neurons (iNs), using lineage-specific transcription factors has opened new opportunities for cell therapy in a number of neurological diseases, including epilepsy. In most severe cases of epilepsy, seizures often originate in the hippocampus, where populations of inhibitory interneurons degenerate. Thus, iNs could be of potential use to replace these lost interneurons. It is not known, however, if iNs survive and maintain functional neuronal properties for prolonged time periods in in vivo. We transplanted human fibroblast-derived iNs into the adult rat hippocampus and observed a progressive morphological differentiation, with more developed dendritic arborisation at six months as compared to one month. This was accompanied by mature electrophysiological properties and fast high amplitude action potentials at six months after transplantation. This proof-of-principle study suggests that human iNs can be developed as a candidate source for cell replacement therapy in temporal lobe epilepsy.

15.
Sci Rep ; 6: 38290, 2016 12 05.
Artículo en Inglés | MEDLINE | ID: mdl-27917895

RESUMEN

The generation of human induced neurons (hiNs) via exogenous delivery of neural transcription factors represents a novel technique to obtain disease and patient specific neurons. These cells have the potential to be used for disease modeling, diagnostics and drug screening, and also to be further developed for brain repair. In the present study, we utilized hiNs to develop an unbiased screening assay for small molecules that increase the conversion efficiency. Using this assay, we screened 307 compounds from five annotated libraries and identified six compounds that were very potent in potentiating the reprogramming process. When combined in an optimal combination and dose, these compounds increased the reprogramming efficiency of human fibroblasts more than 6-fold. Global gene expression and CellNet analysis at different timepoints during the reprogramming process revealed that neuron-specific genes and gene regulatory networks (GRNs) became progressively more activated while converting cells shut down fibroblast-specific GRNs. Further bioinformatics analysis revealed that the addition of the six compound resulted in the accelerated upregulation of a subset of neuronal genes, and also increased expression of genes associated with transcriptional activity and mediation of cellular stress response.


Asunto(s)
Reprogramación Celular/efectos de los fármacos , Fibroblastos/efectos de los fármacos , Proteínas del Tejido Nervioso/genética , Neuronas/efectos de los fármacos , Bibliotecas de Moléculas Pequeñas/farmacología , Factores de Transcripción/genética , Anilidas/farmacología , Benzazepinas/farmacología , Línea Celular , AMP Cíclico/metabolismo , Dinoprostona/farmacología , Feto , Fibroblastos/citología , Fibroblastos/metabolismo , Regulación de la Expresión Génica , Vectores Genéticos/química , Vectores Genéticos/metabolismo , Glucógeno Sintasa Quinasa 3 beta/antagonistas & inhibidores , Glucógeno Sintasa Quinasa 3 beta/genética , Glucógeno Sintasa Quinasa 3 beta/metabolismo , Humanos , Indoles/farmacología , Lentivirus/genética , Lentivirus/metabolismo , Proteínas del Tejido Nervioso/agonistas , Proteínas del Tejido Nervioso/metabolismo , Neuronas/citología , Neuronas/metabolismo , Piridinas/farmacología , Pirimidinas/farmacología , Resveratrol , Estilbenos/farmacología , Factores de Transcripción/metabolismo , Transducción Genética
16.
Exp Neurol ; 282: 78-85, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-27235932

RESUMEN

Stem cell therapies for neurological disorders are rapidly moving towards use in clinical trials. Before initiation of clinical trials, extensive pre-clinical validation in appropriate animal models is essential. However, grafts of human cells into the rodent brain are rejected within weeks after transplantation and the standard methods of immune-suppression for the purpose of studying human xenografts are not always sufficient for the long-term studies needed for transplanted human neurons to maturate, integrate and provide functional benefits in the host brain. Neonatal injections in rat pups using human fetal brain cells have been shown to desensitise the host to accept human tissue grafts as adults, whilst not compromising their immune system. Here, we show that differentiated human embryonic stem cells (hESCs) can be used for desensitisation to achieve long-term graft survival of human stem cell-derived neurons in a xenograft setting, surpassing the time of conventional pharmacological immune-suppressive treatments. The use of hESCs for desensitisation opens up for a widespread use of the technique, which will be of great value when performing pre-clinical evaluation of stem cell-derived neurons in animal models.


Asunto(s)
Lesiones Encefálicas/cirugía , Desensibilización Inmunológica , Rechazo de Injerto/prevención & control , Trasplante de Células Madre/métodos , Trasplante Heterólogo/métodos , Animales , Animales Recién Nacidos , Antígenos CD/metabolismo , Barrera Hematoencefálica/fisiopatología , Lesiones Encefálicas/inducido químicamente , Lesiones Encefálicas/inmunología , Línea Celular Transformada , Ciclosporina/farmacología , Modelos Animales de Enfermedad , Femenino , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Humanos , Terapia de Inmunosupresión , Inmunosupresores/farmacología , Proteínas del Tejido Nervioso/metabolismo , Moléculas de Adhesión de Célula Nerviosa/metabolismo , Oxidopamina/toxicidad , Embarazo , Ratas , Ratas Sprague-Dawley , Simpaticolíticos/toxicidad , Factores de Tiempo
17.
Sci Rep ; 4: 6330, 2014 Sep 11.
Artículo en Inglés | MEDLINE | ID: mdl-25208484

RESUMEN

Induced neurons (iNs) offer a novel source of human neurons that can be explored for applications of disease modelling, diagnostics, drug screening and cell replacement therapy. Here we present a protocol for highly efficient generation of functional iNs from fetal human fibroblasts, and also demonstrate the ability of these converted human iNs (hiNs) to survive transplantation and maintain their phenotype in the adult rat brain. The protocol encompasses a delay in transgene activation after viral transduction that resulted in a significant increase in conversion efficiency. Combining this approach with treatment of small molecules that inhibit SMAD signalling and activate WNT signalling provides a further increase in the conversion efficiency and neuronal purity, resulting in a protocol that provides a highly efficient method for the generation of large numbers of functional and transplantable iNs from human fibroblasts without the use of a selection step. When transplanting the converted neurons from different stages of in vitro culture into the brain of adult rats, we observed robust survival and maintenance of neuronal identity four weeks post-transplantation. Interestingly, the positive effect of small molecule treatment observed in vitro did not result in a higher yield of iNs surviving transplantation.


Asunto(s)
Encéfalo/citología , Reprogramación Celular/fisiología , Células Madre Pluripotentes Inducidas/citología , Neuronas/trasplante , Trasplante Heterólogo/métodos , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Diferenciación Celular/fisiología , Proliferación Celular , Supervivencia Celular , Células Cultivadas , Dopamina/biosíntesis , Femenino , Fibroblastos/citología , Proteínas de Homeodominio/genética , Humanos , Proteínas Asociadas a Microtúbulos/biosíntesis , Proteínas del Tejido Nervioso/genética , Neuronas/citología , Factores del Dominio POU/genética , Ratas , Ratas Sprague-Dawley , Proteínas Smad/antagonistas & inhibidores , Factores de Transcripción/genética , Transgenes/genética , Tubulina (Proteína)/biosíntesis , Vía de Señalización Wnt/efectos de los fármacos
18.
Acta Neuropathol ; 127(2): 283-95, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24292008

RESUMEN

Frontotemporal lobar degeneration (FTLD) consists of a group of neurodegenerative diseases characterized by behavioural and executive impairment, language disorders and motor dysfunction. About 20-30% of cases are inherited in a dominant manner. Mutations in the microtubule-associated protein tau gene (MAPT) cause frontotemporal dementia and parkinsonism linked to chromosome 17 (FTDP-17T). Here we report a novel MAPT mutation (K298E) in exon 10 in a patient with FTDP-17T. Neuropathological studies of post-mortem brain showed widespread neuronal loss and gliosis and abundant deposition of hyperphosphorylated tau in neurons and glia. Molecular studies demonstrated that the K298E mutation affects both protein function and alternative mRNA splicing. Fibroblasts from a skin biopsy of the proband taken at post-mortem were directly induced into neurons (iNs) and expressed both 3-repeat and 4-repeat tau isoforms. As well as contributing new knowledge on MAPT mutations in FTDP-17T, this is the first example of the successful generation of iNs from skin cells retrieved post-mortem.


Asunto(s)
Encéfalo/patología , Exones/genética , Mutación/genética , Neuronas/metabolismo , Tauopatías/genética , Proteínas tau/metabolismo , Anciano , Autopsia , Biopsia , Cromosomas Humanos Par 17/genética , Femenino , Fibroblastos/metabolismo , Fibroblastos/patología , Degeneración Lobar Frontotemporal/genética , Degeneración Lobar Frontotemporal/metabolismo , Degeneración Lobar Frontotemporal/mortalidad , Humanos , Neuronas/patología , Proteínas tau/genética
19.
Proc Natl Acad Sci U S A ; 110(17): 7038-43, 2013 Apr 23.
Artículo en Inglés | MEDLINE | ID: mdl-23530235

RESUMEN

Cellular reprogramming is a new and rapidly emerging field in which somatic cells can be turned into pluripotent stem cells or other somatic cell types simply by the expression of specific combinations of genes. By viral expression of neural fate determinants, it is possible to directly reprogram mouse and human fibroblasts into functional neurons, also known as induced neurons. The resulting cells are nonproliferating and present an alternative to induced pluripotent stem cells for obtaining patient- and disease-specific neurons to be used for disease modeling and for development of cell therapy. In addition, because the cells do not pass a stem cell intermediate, direct neural conversion has the potential to be performed in vivo. In this study, we show that transplanted human fibroblasts and human astrocytes, which are engineered to express inducible forms of neural reprogramming genes, convert into neurons when reprogramming genes are activated after transplantation. Using a transgenic mouse model to specifically direct expression of reprogramming genes to parenchymal astrocytes residing in the striatum, we also show that endogenous mouse astrocytes can be directly converted into neural nuclei (NeuN)-expressing neurons in situ. Taken together, our data provide proof of principle that direct neural conversion can take place in the adult rodent brain when using transplanted human cells or endogenous mouse cells as a starting cell for neural conversion.


Asunto(s)
Astrocitos/trasplante , Diferenciación Celular/fisiología , Reprogramación Celular/fisiología , Fibroblastos/trasplante , Neuronas/citología , Animales , Astrocitos/citología , Reprogramación Celular/efectos de los fármacos , Cuerpo Estriado/citología , Doxiciclina/farmacología , Fibroblastos/citología , Citometría de Flujo , Vectores Genéticos/genética , Proteínas Fluorescentes Verdes , Humanos , Lentivirus , Ratones , Ratones Transgénicos , Neuronas/fisiología , Ratas , Ratas Sprague-Dawley
20.
Cell Cycle ; 10(19): 3311-6, 2011 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-21934358

RESUMEN

Cellular reprogramming is a rapidly developing technology by which somatic cells are turned into pluripotent stem cells or other somatic cell types through expression of specific combinations of genes. This allows for the generation of patient-specific cell lines that can serve as tools for understanding disease pathogenesis, for drug screens and, potentially, for cell replacement therapies. Several cellular models of neurological disorders based on induced pluripotent cells (iPS cells) have been developed, and iPS-derived neurons are being explored as candidates for transplantation. Recent findings show that neurons can also be induced directly from embryonic and postnatal somatic cells by expression of defined combinations of genes. This conversion does not occur through a pluripotent stem cell stage, which eliminates the risk for tumor formation. Here, we demonstrate for the first time that functional neurons can be generated via direct conversion of fibroblasts also from adult individuals. Thus, this technology is an attractive alternative to iPS cells for generating patient- and disease-specific neurons suitable for disease modeling and autologous transplantation.


Asunto(s)
Fibroblastos/citología , Neuronas/citología , Actinas/metabolismo , Adulto , Anciano , Células Cultivadas , Medios de Cultivo , Femenino , Humanos , Masculino , Persona de Mediana Edad , Trasplante Autólogo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA