Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
1.
Stem Cell Reports ; 19(1): 41-53, 2024 01 09.
Artículo en Inglés | MEDLINE | ID: mdl-38101401

RESUMEN

While studied extensively in model systems, human gastrulation remains obscure. The scarcity of fetal biological material as well as ethical considerations limit our understanding of this process. In vitro attachment of natural blastocysts shed light on aspects of the second week of human development in the absence of the morphological manifestation of gastrulation. Stem cell-derived blastocyst models, blastoids, provide the opportunity to reconstitute pre- to post-implantation development in vitro. Here we show that upon in vitro attachment, human blastoids self-organize a BRA+ population and undergo gastrulation. Single-cell RNA sequencing of these models replicates the transcriptomic signature of the human gastrula. Analysis of developmental timing reveals that in both blastoid models and natural human embryos, the onset of gastrulation as defined by molecular markers, can be traced to timescales equivalent to 12 days post fertilization. In all, natural human embryos and blastoid models self-organize primitive streak and mesoderm derivatives upon in vitro attachment.


Asunto(s)
Gástrula , Gastrulación , Humanos , Desarrollo Embrionario , Blastocisto , Mesodermo
2.
iScience ; 26(9): 107525, 2023 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-37646018

RESUMEN

The hypothalamus is a region of the brain that plays an important role in regulating body functions and behaviors. There is a growing interest in human pluripotent stem cells (hPSCs) for modeling diseases that affect the hypothalamus. Here, we established an hPSC-derived hypothalamus organoid differentiation protocol to model the cellular diversity of this brain region. Using an hPSC line with a tyrosine hydroxylase (TH)-TdTomato reporter for dopaminergic neurons (DNs) and other TH-expressing cells, we interrogated DN-specific pathways and functions in electrophysiologically active hypothalamus organoids. Single-cell RNA sequencing (scRNA-seq) revealed diverse neuronal and non-neuronal cell types in mature hypothalamus organoids. We identified several molecularly distinct hypothalamic DN subtypes that demonstrated different developmental maturities. Our in vitro 3D hypothalamus differentiation protocol can be used to study the development of this critical brain structure and can be applied to disease modeling to generate novel therapeutic approaches for disorders centered around the hypothalamus.

3.
Elife ; 122023 01 16.
Artículo en Inglés | MEDLINE | ID: mdl-36645408

RESUMEN

Infantile neuroaxonal dystrophy (INAD) is caused by recessive variants in PLA2G6 and is a lethal pediatric neurodegenerative disorder. Loss of the Drosophila homolog of PLA2G6, leads to ceramide accumulation, lysosome expansion, and mitochondrial defects. Here, we report that retromer function, ceramide metabolism, the endolysosomal pathway, and mitochondrial morphology are affected in INAD patient-derived neurons. We show that in INAD mouse models, the same features are affected in Purkinje cells, arguing that the neuropathological mechanisms are evolutionary conserved and that these features can be used as biomarkers. We tested 20 drugs that target these pathways and found that Ambroxol, Desipramine, Azoramide, and Genistein alleviate neurodegenerative phenotypes in INAD flies and INAD patient-derived neural progenitor cells. We also develop an AAV-based gene therapy approach that delays neurodegeneration and prolongs lifespan in an INAD mouse model.


Asunto(s)
Proteínas de Drosophila , Distrofias Neuroaxonales , Trastornos Parkinsonianos , Ratones , Animales , Neuronas/metabolismo , Trastornos Parkinsonianos/metabolismo , Drosophila/metabolismo , Ceramidas/metabolismo , Distrofias Neuroaxonales/genética , Distrofias Neuroaxonales/metabolismo , Distrofias Neuroaxonales/patología , Fosfolipasas A2 Grupo VI/metabolismo , Proteínas del Ojo/metabolismo , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo
4.
Cell Mol Life Sci ; 79(2): 78, 2022 Jan 19.
Artículo en Inglés | MEDLINE | ID: mdl-35044538

RESUMEN

Three-dimensional (3D) in vitro culture systems using human induced pluripotent stem cells (hiPSCs) are useful tools to model neurodegenerative disease biology in physiologically relevant microenvironments. Though many successful biomaterials-based 3D model systems have been established for other neurogenerative diseases, such as Alzheimer's disease, relatively few exist for Parkinson's disease (PD) research. We employed tissue engineering approaches to construct a 3D silk scaffold-based platform for the culture of hiPSC-dopaminergic (DA) neurons derived from healthy individuals and PD patients harboring LRRK2 G2019S or GBA N370S mutations. We then compared results from protein, gene expression, and metabolic analyses obtained from two-dimensional (2D) and 3D culture systems. The 3D platform enabled the formation of dense dopamine neuronal network architectures and developed biological profiles both similar and distinct from 2D culture systems in healthy and PD disease lines. PD cultures developed in 3D platforms showed elevated levels of α-synuclein and alterations in purine metabolite profiles. Furthermore, computational network analysis of transcriptomic networks nominated several novel molecular interactions occurring in neurons from patients with mutations in LRRK2 and GBA. We conclude that the brain-like 3D system presented here is a realistic platform to interrogate molecular mechanisms underlying PD biology.


Asunto(s)
Neuronas Dopaminérgicas/patología , Enfermedad de Parkinson/patología , Bioingeniería , Técnicas de Cultivo Tridimensional de Células , Células Cultivadas , Neuronas Dopaminérgicas/citología , Humanos , Células Madre Pluripotentes Inducidas/citología , Células Madre Pluripotentes Inducidas/patología , Neurogénesis , Seda/química , Andamios del Tejido/química
5.
Development ; 148(6)2021 03 29.
Artículo en Inglés | MEDLINE | ID: mdl-33782043

RESUMEN

Rostro-caudal patterning of vertebrates depends on the temporally progressive activation of HOX genes within axial stem cells that fuel axial embryo elongation. Whether the pace of sequential activation of HOX genes, the 'HOX clock', is controlled by intrinsic chromatin-based timing mechanisms or by temporal changes in extrinsic cues remains unclear. Here, we studied HOX clock pacing in human pluripotent stem cell-derived axial progenitors differentiating into diverse spinal cord motor neuron subtypes. We show that the progressive activation of caudal HOX genes is controlled by a dynamic increase in FGF signaling. Blocking the FGF pathway stalled induction of HOX genes, while a precocious increase of FGF, alone or with GDF11 ligand, accelerated the HOX clock. Cells differentiated under accelerated HOX induction generated appropriate posterior motor neuron subtypes found along the human embryonic spinal cord. The pacing of the HOX clock is thus dynamically regulated by exposure to secreted cues. Its manipulation by extrinsic factors provides synchronized access to multiple human neuronal subtypes of distinct rostro-caudal identities for basic and translational applications.This article has an associated 'The people behind the papers' interview.


Asunto(s)
Relojes Circadianos , Proteínas de Homeodominio/metabolismo , Neuronas Motoras/metabolismo , Células Madre Pluripotentes/metabolismo , Benzamidas/farmacología , Proteínas Morfogenéticas Óseas/genética , Proteínas Morfogenéticas Óseas/metabolismo , Proteínas Morfogenéticas Óseas/farmacología , Diferenciación Celular , Relojes Circadianos/efectos de los fármacos , Difenilamina/análogos & derivados , Difenilamina/farmacología , Embrión de Mamíferos/citología , Embrión de Mamíferos/metabolismo , Desarrollo Embrionario , Factores de Crecimiento de Fibroblastos/antagonistas & inhibidores , Factores de Crecimiento de Fibroblastos/metabolismo , Factores de Crecimiento de Fibroblastos/farmacología , Regulación del Desarrollo de la Expresión Génica , Factores de Diferenciación de Crecimiento/genética , Factores de Diferenciación de Crecimiento/metabolismo , Factores de Diferenciación de Crecimiento/farmacología , Proteínas de Homeodominio/genética , Humanos , Neuronas Motoras/citología , Células Madre Pluripotentes/citología , Pirimidinas/farmacología , Transducción de Señal/efectos de los fármacos , Médula Espinal/metabolismo
6.
Neuron ; 107(3): 436-453.e12, 2020 08 05.
Artículo en Inglés | MEDLINE | ID: mdl-32485136

RESUMEN

New methods for investigating human astrocytes are urgently needed, given their critical role in the central nervous system. Here we show that CD49f is a novel marker for human astrocytes, expressed in fetal and adult brains from healthy and diseased individuals. CD49f can be used to purify fetal astrocytes and human induced pluripotent stem cell (hiPSC)-derived astrocytes. We provide single-cell and bulk transcriptome analyses of CD49f+ hiPSC-astrocytes and demonstrate that they perform key astrocytic functions in vitro, including trophic support of neurons, glutamate uptake, and phagocytosis. Notably, CD49f+ hiPSC-astrocytes respond to inflammatory stimuli, acquiring an A1-like reactive state, in which they display impaired phagocytosis and glutamate uptake and fail to support neuronal maturation. Most importantly, we show that conditioned medium from human reactive A1-like astrocytes is toxic to human and rodent neurons. CD49f+ hiPSC-astrocytes are thus a valuable resource for investigating human astrocyte function and dysfunction in health and disease.


Asunto(s)
Astrocitos/metabolismo , Células Madre Pluripotentes Inducidas/metabolismo , Integrina alfa6/metabolismo , Enfermedad de Alzheimer/metabolismo , Animales , Astrocitos/fisiología , Biomarcadores/metabolismo , Citometría de Flujo , Perfilación de la Expresión Génica , Ácido Glutámico/metabolismo , Humanos , Inflamación/metabolismo , Inflamación/fisiopatología , Ratones , Técnicas de Placa-Clamp , Fagocitosis/fisiología , RNA-Seq , Análisis de la Célula Individual
7.
Nat Cell Biol ; 21(7): 900-910, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-31263269

RESUMEN

Breaking the anterior-posterior symmetry in mammals occurs at gastrulation. Much of the signalling network underlying this process has been elucidated in the mouse; however, there is no direct molecular evidence of events driving axis formation in humans. Here, we use human embryonic stem cells to generate an in vitro three-dimensional model of a human epiblast whose size, cell polarity and gene expression are similar to a day 10 human epiblast. A defined dose of BMP4 spontaneously breaks axial symmetry, and induces markers of the primitive streak and epithelial-to-mesenchymal transition. We show that WNT signalling and its inhibitor DKK1 play key roles in this process downstream of BMP4. Our work demonstrates that a model human epiblast can break axial symmetry despite the absence of asymmetry in the initial signal and of extra-embryonic tissues or maternal cues. Our three-dimensional model is an assay for the molecular events underlying human axial symmetry breaking.


Asunto(s)
Proteína Morfogenética Ósea 4/metabolismo , Regulación del Desarrollo de la Expresión Génica/fisiología , Estratos Germinativos/metabolismo , Línea Primitiva/metabolismo , Técnicas de Cultivo de Tejidos , Polaridad Celular/fisiología , Transición Epitelial-Mesenquimal , Gastrulación/fisiología , Humanos , Línea Primitiva/embriología , Transducción de Señal/fisiología
8.
Elife ; 72018 08 22.
Artículo en Inglés | MEDLINE | ID: mdl-30132757

RESUMEN

Piezo1 is a mechanosensitive (MS) ion channel with characteristic fast-inactivation kinetics. We found a slowly-inactivating MS current in mouse embryonic stem (mES) cells and characterized it throughout their differentiation into motor-neurons to investigate its components. MS currents were large and slowly-inactivating in the stem-cell stage, and became smaller and faster-inactivating throughout the differentiation. We found that Piezo1 is expressed in mES cells, and its knockout abolishes MS currents, indicating that the slowly-inactivating current in mES cells is carried by Piezo1. To further investigate its slow inactivation in these cells, we cloned Piezo1 cDNA from mES cells and found that it displays fast-inactivation kinetics in heterologous expression, indicating that sources of modulation other than the aminoacid sequence determine its slow kinetics in mES cells. Finally, we report that Piezo1 knockout ES cells showed a reduced rate of proliferation but no significant differences in other markers of pluripotency and differentiation.


Asunto(s)
Activación del Canal Iónico , Canales Iónicos/metabolismo , Mecanotransducción Celular , Células Madre Embrionarias de Ratones/metabolismo , Animales , Secuencia de Bases , Diferenciación Celular , Proliferación Celular , Forma de la Célula , ADN Complementario/genética , Gastrulación , Células HEK293 , Humanos , Cinética , Ratones , Ratones Noqueados , Neuronas Motoras/citología , Neuronas Motoras/metabolismo , Células Madre Embrionarias de Ratones/citología , Mutación/genética , Fenotipo , Células Madre Pluripotentes/citología , Células Madre Pluripotentes/metabolismo
9.
Development ; 145(2)2018 01 29.
Artículo en Inglés | MEDLINE | ID: mdl-29378824

RESUMEN

Huntington's disease (HD) is a fatal neurodegenerative disease caused by expansion of CAG repeats in the Huntingtin gene (HTT). Neither its pathogenic mechanisms nor the normal functions of HTT are well understood. To model HD in humans, we engineered a genetic allelic series of isogenic human embryonic stem cell (hESC) lines with graded increases in CAG repeat length. Neural differentiation of these lines unveiled a novel developmental HD phenotype: the appearance of giant multinucleated telencephalic neurons at an abundance directly proportional to CAG repeat length, generated by a chromosomal instability and failed cytokinesis over multiple rounds of DNA replication. We conclude that disrupted neurogenesis during development is an important, unrecognized aspect of HD pathogenesis. To address the function of normal HTT protein we generated HTT+/- and HTT-/- lines. Surprisingly, the same phenotype emerged in HTT-/- but not HTT+/- lines. We conclude that HD is a developmental disorder characterized by chromosomal instability that impairs neurogenesis, and that HD represents a genetic dominant-negative loss of function, contrary to the prevalent gain-of-toxic-function hypothesis. The consequences of developmental alterations should be considered as a new target for HD therapies.


Asunto(s)
Inestabilidad Cromosómica , Proteína Huntingtina/genética , Enfermedad de Huntington/genética , Neurogénesis/genética , Alelos , Diferenciación Celular/genética , Línea Celular , Células Madre Embrionarias/metabolismo , Células Madre Embrionarias/patología , Humanos , Proteína Huntingtina/deficiencia , Proteína Huntingtina/metabolismo , Enfermedad de Huntington/etiología , Enfermedad de Huntington/patología , Modelos Biológicos , Fenotipo , Huso Acromático/patología , Expansión de Repetición de Trinucleótido
10.
Reprod Biol Endocrinol ; 15(1): 33, 2017 Apr 27.
Artículo en Inglés | MEDLINE | ID: mdl-28449669

RESUMEN

BACKGROUND: It has become increasingly apparent that the trophectoderm (TE) at blastocyst stage is much more mosaic than has been appreciated. Whether preimplantation genetic screening (PGS), utilizing a single TE biopsy (TEB), can reliably determine embryo ploidy has, therefore, increasingly been questioned in parallel. METHODS: We for that reason here established 2 mathematical models to assess probabilities of false-negative and false-positive results of an on average 6-cell biopsy from an approximately 300-cell TE. This study was a collaborative effort between investigators at The Center for Human Reproduction in New York City and the Center for Studies in Physics and Biology and the Brivanlou Laboratory of Stem Cell Biology and Molecular Embryology, the latter two both at Rockefeller University in New York City. RESULTS: Both models revealed that even under best case scenario, assuming even distribution of mosaicism in TE (since mosaicism is usually clonal, a highly unlikely scenario), a biopsy of at least 27 TE cells would be required to reach minimal diagnostic predictability from a single TEB. CONCLUSIONS: As currently performed, a single TEB is, therefore, mathematically incapable of reliably determining whether an embryo can be transferred or should be discarded. Since a single TEB, as currently performed, apparently is not representative of the complete TE, this study, thus, raises additional concern about the clinical utilization of PGS.


Asunto(s)
Blastocisto , Fase de Segmentación del Huevo , Ectodermo/patología , Ploidias , Diagnóstico Preimplantación/métodos , Trofoblastos/patología , Aneuploidia , Biopsia , Blastocisto/metabolismo , Blastocisto/patología , Fase de Segmentación del Huevo/metabolismo , Fase de Segmentación del Huevo/patología , Implantación del Embrión/genética , Femenino , Humanos , Modelos Teóricos , Embarazo , Diagnóstico Preimplantación/normas , Reproducibilidad de los Resultados
11.
Cell Rep ; 16(2): 545-558, 2016 07 12.
Artículo en Inglés | MEDLINE | ID: mdl-27373155

RESUMEN

Suboptimal axonal regeneration contributes to the consequences of nervous system trauma and neurodegenerative disease, but the intrinsic mechanisms that regulate axon growth remain unclear. We screened 50,400 small molecules for their ability to promote axon outgrowth on inhibitory substrata. The most potent hits were the statins, which stimulated growth of all mouse- and human-patient-derived neurons tested, both in vitro and in vivo, as did combined inhibition of the protein prenylation enzymes farnesyltransferase (PFT) and geranylgeranyl transferase I (PGGT-1). Compensatory sprouting of motor axons may delay clinical onset of amyotrophic lateral sclerosis (ALS). Accordingly, elevated levels of PGGT1B, which would be predicted to reduce sprouting, were found in motor neurons of early- versus late-onset ALS patients postmortem. The mevalonate-prenylation pathway therefore constitutes an endogenous brake on axonal growth, and its inhibition provides a potential therapeutic approach to accelerate neuronal regeneration in humans.


Asunto(s)
Neuritas/fisiología , Prenilación de Proteína , Esclerosis Amiotrófica Lateral/patología , Animales , Aumento de la Célula , Células Cultivadas , Humanos , Inhibidores de Hidroximetilglutaril-CoA Reductasas/farmacología , Ácido Mevalónico/metabolismo , Ratones , Neuronas Motoras/fisiología , Regeneración Nerviosa
12.
Nature ; 533(7602): 251-4, 2016 05 12.
Artículo en Inglés | MEDLINE | ID: mdl-27144363

RESUMEN

Implantation of the blastocyst is a developmental milestone in mammalian embryonic development. At this time, a coordinated program of lineage diversification, cell-fate specification, and morphogenetic movements establishes the generation of extra-embryonic tissues and the embryo proper, and determines the conditions for successful pregnancy and gastrulation. Despite its basic and clinical importance, this process remains mysterious in humans. Here we report the use of a novel in vitro system to study the post-implantation development of the human embryo. We unveil the self-organizing abilities and autonomy of in vitro attached human embryos. We find human-specific molecular signatures of early cell lineage, timing, and architecture. Embryos display key landmarks of normal development, including epiblast expansion, lineage segregation, bi-laminar disc formation, amniotic and yolk sac cavitation, and trophoblast diversification. Our findings highlight the species-specificity of these developmental events and provide a new understanding of early human embryonic development beyond the blastocyst stage. In addition, our study establishes a new model system relevant to early human pregnancy loss. Finally, our work will also assist in the rational design of differentiation protocols of human embryonic stem cells to specific cell types for disease modelling and cell replacement therapy.


Asunto(s)
Embrión de Mamíferos/citología , Embrión de Mamíferos/embriología , Desarrollo Embrionario , Amnios/citología , Amnios/embriología , Animales , Diferenciación Celular , Linaje de la Célula , Pérdida del Embrión/patología , Embrión de Mamíferos/anatomía & histología , Células Madre Embrionarias/citología , Células Madre Embrionarias/patología , Células Madre Embrionarias/trasplante , Estratos Germinativos/citología , Estratos Germinativos/embriología , Humanos , Técnicas In Vitro , Ratones , Modelos Biológicos , Especificidad de la Especie , Trofoblastos/citología , Saco Vitelino/citología , Saco Vitelino/embriología
13.
Cell Rep ; 12(2): 335-45, 2015 Jul 14.
Artículo en Inglés | MEDLINE | ID: mdl-26146077

RESUMEN

Neurodegenerative phenotypes reflect complex, time-dependent molecular processes whose elucidation may reveal neuronal class-specific therapeutic targets. The current focus in neurodegeneration has been on individual genes and pathways. In contrast, we assembled a genome-wide regulatory model (henceforth, "interactome"), whose unbiased interrogation revealed 23 candidate causal master regulators of neurodegeneration in an in vitro model of amyotrophic lateral sclerosis (ALS), characterized by a loss of spinal motor neurons (MNs). Of these, eight were confirmed as specific MN death drivers in our model of familial ALS, including NF-κB, which has long been considered a pro-survival factor. Through an extensive array of molecular, pharmacological, and biochemical approaches, we have confirmed that neuronal NF-κB drives the degeneration of MNs in both familial and sporadic models of ALS, thus providing proof of principle that regulatory network analysis is a valuable tool for studying cell-specific mechanisms of neurodegeneration.


Asunto(s)
Modelos Biológicos , Neuronas Motoras/metabolismo , FN-kappa B/metabolismo , Esclerosis Amiotrófica Lateral/metabolismo , Esclerosis Amiotrófica Lateral/patología , Animales , Apoptosis/efectos de los fármacos , Astrocitos/citología , Astrocitos/metabolismo , Células Cultivadas , Medios de Cultivo Condicionados/farmacología , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Humanos , Ratones , Ratones Transgénicos , Neuronas Motoras/citología , Neuronas Motoras/efectos de los fármacos , Mutación , Interferencia de ARN , ARN Interferente Pequeño/metabolismo , Superóxido Dismutasa/antagonistas & inhibidores , Superóxido Dismutasa/genética , Superóxido Dismutasa/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Transcriptoma/efectos de los fármacos
14.
PLoS One ; 10(5): e0127687, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26010866

RESUMEN

Huntington's disease (HD) is a devastating neurological disorder that is caused by an expansion of the poly-Q tract in exon 1 of the Huntingtin gene (HTT). HTT is an evolutionarily conserved and ubiquitously expressed protein that has been linked to a variety of functions including transcriptional regulation, mitochondrial function, and vesicle transport. This large protein has numerous caspase and calpain cleavage sites and can be decorated with several post-translational modifications such as phosphorylations, acetylations, sumoylations, and palmitoylations. However, the exact function of HTT and the role played by its modifications in the cell are still not well understood. Scrutiny of HTT function has been focused on a single, full length mRNA. In this study, we report the discovery of 5 novel HTT mRNA splice isoforms that are expressed in normal and HTT-expanded human embryonic stem cell (hESC) lines as well as in cortical neurons differentiated from hESCs. Interestingly, none of the novel isoforms generates a truncated protein. Instead, 4 of the 5 new isoforms specifically eliminate domains and modifications to generate smaller HTT proteins. The fifth novel isoform incorporates a previously unreported additional exon, dubbed 41b, which is hominid-specific and introduces a potential phosphorylation site in the protein. The discovery of this hominid-specific isoform may shed light on human-specific pathogenic mechanisms of HTT, which could not be investigated with current mouse models of the disease.


Asunto(s)
Exones , Enfermedad de Huntington , Proteínas del Tejido Nervioso , Animales , Células Madre Embrionarias Humanas/metabolismo , Células Madre Embrionarias Humanas/patología , Humanos , Proteína Huntingtina , Enfermedad de Huntington/genética , Enfermedad de Huntington/metabolismo , Enfermedad de Huntington/patología , Ratones , Ratones Noqueados , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Especificidad de la Especie
15.
Cancer Res ; 74(20): 5914-24, 2014 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-25164012

RESUMEN

The Hedgehog (Hh) signaling pathway regulates normal development and cell proliferation in metazoan organisms, but its aberrant activation can promote tumorigenesis. Hh-induced tumors arise from various tissues and they may be indolent or aggressive, as is the case with skin basal cell carcinoma (BCC) or cerebellar medulloblastoma, respectively. Little is known about common cell-intrinsic factors that control the development of such diverse Hh-dependent tumors. Transcription factor Zfx is required for the self-renewal of hematopoietic and embryonic stem cells, as well as for the propagation of acute myeloid and T-lymphoblastic leukemias. We report here that Zfx facilitates the development of experimental BCC and medulloblastoma in mice initiated by deletion of the Hh inhibitory receptor Ptch1. Simultaneous deletion of Zfx along with Ptch1 prevented BCC formation and delayed medulloblastoma development. In contrast, Zfx was dispensable for tumorigenesis in a mouse model of glioblastoma. We used genome-wide expression and chromatin-binding analysis in a human medulloblastoma cell line to characterize direct, evolutionarily conserved targets of Zfx, identifying Dis3L and Ube2j1 as two targets required for the growth of the human medulloblastoma cells. Our results establish Zfx as a common cell-intrinsic regulator of diverse Hh-induced tumors, with implications for the definition of new therapeutic targets in these malignancies.


Asunto(s)
Carcinogénesis/genética , Proteínas Hedgehog/metabolismo , Factores de Transcripción de Tipo Kruppel/fisiología , Animales , Carcinoma Basocelular/genética , Carcinoma Basocelular/metabolismo , Carcinoma Basocelular/patología , Línea Celular Tumoral , Proliferación Celular , Neoplasias Cerebelosas/genética , Neoplasias Cerebelosas/metabolismo , Técnicas de Inactivación de Genes , Humanos , Masculino , Meduloblastoma/genética , Meduloblastoma/metabolismo , Ratones Noqueados , Receptores Patched , Receptor Patched-1 , Receptores de Superficie Celular/genética , Ribonucleasas/metabolismo , Transducción de Señal , Neoplasias Cutáneas/genética , Neoplasias Cutáneas/metabolismo , Neoplasias Cutáneas/patología , Enzimas Ubiquitina-Conjugadoras/metabolismo
16.
PLoS One ; 8(7): e69208, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23861962

RESUMEN

The Aicda gene encodes Activation-Induced cytidine Deaminase (AID), an enzyme essential for remodeling antibody genes in mature B lymphocytes. AID is also responsible for DNA damage at oncogenes, leading to their mutation and cancer-associated chromosome translocation in lymphoma. We used fate mapping and AID(GFP) reporter mice to determine if AID expression in the mouse extends beyond lymphocytes. We discovered that AID(cre) tags a small fraction of non-lymphoid cells starting at 10.5 days post conception (dpc), and that AID(GFP+) cells are detectable at dpc 11.5 and 12.5. Embryonic cells are tagged by AID(cre) in the submandibular region, where conditional deletion of the tumor suppressor PTEN causes squamous papillomas. AID(cre) also tags non-lymphoid cells in the embryonic central nervous system. Finally, in the adult mouse brain, AID(cre) marks a small fraction of diverse neurons and distinct neuronal populations, including pyramidal cells in cortical layer IV.


Asunto(s)
Linaje de la Célula , Citidina Desaminasa/metabolismo , Linfocitos/citología , Linfocitos/enzimología , Envejecimiento/metabolismo , Animales , Encéfalo/enzimología , Encéfalo/patología , Desarrollo Embrionario , Integrasas/metabolismo , Ratones , Fosfohidrolasa PTEN/metabolismo , Papiloma/patología , Piel/metabolismo
17.
J Neurosci ; 33(2): 574-86, 2013 Jan 09.
Artículo en Inglés | MEDLINE | ID: mdl-23303937

RESUMEN

Human pluripotent stem cells are a promising source of differentiated cells for developmental studies, cell transplantation, disease modeling, and drug testing. However, their widespread use even for intensely studied cell types like spinal motor neurons is hindered by the long duration and low yields of existing protocols for in vitro differentiation and by the molecular heterogeneity of the populations generated. We report a combination of small molecules that within 3 weeks induce motor neurons at up to 50% abundance and with defined subtype identities of relevance to neurodegenerative disease. Despite their accelerated differentiation, motor neurons expressed combinations of HB9, ISL1, and column-specific markers that mirror those observed in vivo in human embryonic spinal cord. They also exhibited spontaneous and induced activity, and projected axons toward muscles when grafted into developing chick spinal cord. Strikingly, this novel protocol preferentially generates motor neurons expressing markers of limb-innervating lateral motor column motor neurons (FOXP1(+)/LHX3(-)). Access to high-yield cultures of human limb-innervating motor neuron subtypes will facilitate in-depth study of motor neuron subtype-specific properties, disease modeling, and development of large-scale cell-based screening assays.


Asunto(s)
Extremidades/inervación , Neuronas Motoras/fisiología , Células-Madre Neurales/fisiología , Animales , Axones/fisiología , Calcio/fisiología , Señalización del Calcio/fisiología , Diferenciación Celular/fisiología , Células Cultivadas , Embrión de Pollo , ADN Complementario/biosíntesis , ADN Complementario/genética , Femenino , Factores de Transcripción Forkhead/biosíntesis , Factores de Transcripción Forkhead/genética , Proteínas de Homeodominio/genética , Humanos , Inmunohistoquímica , Proteínas con Homeodominio LIM/genética , Masculino , Ratones , Neuronas Motoras/metabolismo , Células-Madre Neurales/metabolismo , Técnicas de Placa-Clamp , Complejo Silenciador Inducido por ARN , Proteínas Represoras/biosíntesis , Proteínas Represoras/genética , Médula Espinal/citología , Médula Espinal/embriología , Trasplante de Células Madre/métodos , Factores de Transcripción/genética
18.
PLoS One ; 7(7): e40154, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22802953

RESUMEN

Our understanding of motor neuron biology in humans is derived mainly from investigation of human postmortem tissue and more indirectly from live animal models such as rodents. Thus generation of motor neurons from human embryonic stem cells and human induced pluripotent stem cells is an important new approach to model motor neuron function. To be useful models of human motor neuron function, cells generated in vitro should develop mature properties that are the hallmarks of motor neurons in vivo such as elaborated neuronal processes and mature electrophysiological characteristics. Here we have investigated changes in morphological and electrophysiological properties associated with maturation of neurons differentiated from human embryonic stem cells expressing GFP driven by a motor neuron specific reporter (Hb9::GFP) in culture. We observed maturation in cellular morphology seen as more complex neurite outgrowth and increased soma area over time. Electrophysiological changes included decreasing input resistance and increasing action potential firing frequency over 13 days in vitro. Furthermore, these human embryonic stem cell derived motor neurons acquired two physiological characteristics that are thought to underpin motor neuron integrated function in motor circuits; spike frequency adaptation and rebound action potential firing. These findings show that human embryonic stem cell derived motor neurons develop functional characteristics typical of spinal motor neurons in vivo and suggest that they are a relevant and useful platform for studying motor neuron development and function and for modeling motor neuron diseases.


Asunto(s)
Potenciales de Acción/fisiología , Células Madre Embrionarias/citología , Neuronas Motoras/citología , Neuronas Motoras/fisiología , Neurogénesis , Diferenciación Celular/fisiología , Células Cultivadas , Humanos , Factores de Transcripción/fisiología
19.
Cell ; 144(3): 439-52, 2011 Feb 04.
Artículo en Inglés | MEDLINE | ID: mdl-21295703

RESUMEN

The developmental potential of human pluripotent stem cells suggests that they can produce disease-relevant cell types for biomedical research. However, substantial variation has been reported among pluripotent cell lines, which could affect their utility and clinical safety. Such cell-line-specific differences must be better understood before one can confidently use embryonic stem (ES) or induced pluripotent stem (iPS) cells in translational research. Toward this goal we have established genome-wide reference maps of DNA methylation and gene expression for 20 previously derived human ES lines and 12 human iPS cell lines, and we have measured the in vitro differentiation propensity of these cell lines. This resource enabled us to assess the epigenetic and transcriptional similarity of ES and iPS cells and to predict the differentiation efficiency of individual cell lines. The combination of assays yields a scorecard for quick and comprehensive characterization of pluripotent cell lines.


Asunto(s)
Metilación de ADN , Células Madre Embrionarias/fisiología , Perfilación de la Expresión Génica/normas , Células Madre Pluripotentes Inducidas/fisiología , Diferenciación Celular , Línea Celular , Células Madre Embrionarias/citología , Humanos , Células Madre Pluripotentes Inducidas/citología
20.
Nat Biotechnol ; 29(3): 279-86, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21293464

RESUMEN

Human induced pluripotent stem cells (iPSCs) present exciting opportunities for studying development and for in vitro disease modeling. However, reported variability in the behavior of iPSCs has called their utility into question. We established a test set of 16 iPSC lines from seven individuals of varying age, sex and health status, and extensively characterized the lines with respect to pluripotency and the ability to terminally differentiate. Under standardized procedures in two independent laboratories, 13 of the iPSC lines gave rise to functional motor neurons with a range of efficiencies similar to that of human embryonic stem cells (ESCs). Although three iPSC lines were resistant to neural differentiation, early neuralization rescued their performance. Therefore, all 16 iPSC lines passed a stringent test of differentiation capacity despite variations in karyotype and in the expression of early pluripotency markers and transgenes. This iPSC and ESC test set is a robust resource for those interested in the basic biology of stem cells and their applications.


Asunto(s)
Técnicas de Cultivo de Célula/métodos , Fibroblastos/citología , Células Madre Pluripotentes/citología , Piel/citología , Ingeniería de Tejidos/métodos , Diferenciación Celular , Células Cultivadas , Humanos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA