Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
3.
Cell Stem Cell ; 25(1): 120-136.e10, 2019 07 03.
Artigo em Inglês | MEDLINE | ID: mdl-31155483

RESUMO

Current challenges in capturing naive human pluripotent stem cells (hPSCs) suggest that the factors regulating human naive versus primed pluripotency remain incompletely defined. Here we demonstrate that the widely used Essential 8 minimal medium (E8) captures hPSCs at a naive-to-primed intermediate state of pluripotency expressing several naive-like developmental, bioenergetic, and epigenomic features despite providing primed-state-sustaining growth factor conditions. Transcriptionally, E8 hPSCs are marked by activated lipid biosynthesis and suppressed MAPK/TGF-ß gene expression, resulting in endogenous ERK inhibition. These features are dependent on lipid-free culture conditions and are lost upon lipid exposure, whereas short-term pharmacological ERK inhibition restores naive-to-primed intermediate traits even in the presence of lipids. Finally, we identify de novo lipogenesis as a common transcriptional signature of E8 hPSCs and the pre-implantation human epiblast in vivo. These findings implicate exogenous lipid availability in regulating human pluripotency and define E8 hPSCs as a stable, naive-to-primed intermediate (NPI) pluripotent state.


Assuntos
Blastocisto/citologia , Camadas Germinativas/citologia , Células-Tronco Pluripotentes/fisiologia , Diferenciação Celular , Células Cultivadas , Meios de Cultura Livres de Soro , Células-Tronco Embrionárias , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Humanos , Metabolismo dos Lipídeos , Transdução de Sinais , Fator de Crescimento Transformador beta/metabolismo
4.
Genes Dev ; 32(17-18): 1161-1174, 2018 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-30115631

RESUMO

Alternative splicing (AS) plays important roles in embryonic stem cell (ESC) differentiation. In this study, we first identified transcripts that display specific AS patterns in pluripotent human ESCs (hESCs) relative to differentiated cells. One of these encodes T-cell factor 3 (TCF3), a transcription factor that plays important roles in ESC differentiation. AS creates two TCF3 isoforms, E12 and E47, and we identified two related splicing factors, heterogeneous nuclear ribonucleoproteins (hnRNPs) H1 and F (hnRNP H/F), that regulate TCF3 splicing. We found that hnRNP H/F levels are high in hESCs, leading to high E12 expression, but decrease during differentiation, switching splicing to produce elevated E47 levels. Importantly, hnRNP H/F knockdown not only recapitulated the switch in TCF3 AS but also destabilized hESC colonies and induced differentiation. Providing an explanation for this, we show that expression of known TCF3 target E-cadherin, critical for maintaining ESC pluripotency, is repressed by E47 but not by E12.


Assuntos
Processamento Alternativo , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Caderinas/metabolismo , Células-Tronco Embrionárias/metabolismo , Ribonucleoproteínas Nucleares Heterogêneas Grupo F-H/metabolismo , Antígenos CD , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Caderinas/genética , Diferenciação Celular/genética , Linhagem Celular , Células-Tronco Embrionárias/citologia , Éxons , Regulação da Expressão Gênica , Humanos , Precursores de RNA/química , RNA Mensageiro/química , Sequências Reguladoras de Ácido Ribonucleico
5.
Methods Mol Biol ; 1307: 329-43, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-24301074

RESUMO

The neural crest (NC) is a transient population of multipotent cells giving rise to the peripheral nervous system, skin pigmentation, heart, and facial mesenchyme. The broad cell fate potential of NC makes it an attractive cell fate to derive from human pluripotent stem cells (hPSCs) for exploring embryonic development, modeling disease, and generating cells for transplantation. Here, we discuss recent publications and methods for efficiently differentiating hPSCs into NC. We also provide methods to direct NC into two different terminal fates: melanocytes and sensory neurons.


Assuntos
Técnicas de Cultura de Células/métodos , Crista Neural/citologia , Células-Tronco Pluripotentes/citologia , Proteínas Smad/antagonistas & inibidores , Proteínas Wnt/metabolismo , Animais , Contagem de Células , Diferenciação Celular , Linhagem da Célula , Células Cultivadas , Humanos , Melanócitos/citologia , Camundongos , Células-Tronco Pluripotentes/metabolismo , Células Receptoras Sensoriais/citologia , Proteínas Smad/metabolismo
6.
Cell Stem Cell ; 13(4): 377-8, 2013 Oct 03.
Artigo em Inglês | MEDLINE | ID: mdl-24094317

RESUMO

A major barrier in understanding nervous system development is modeling the cellular interactions that form the human brain. Recently, in the journal Nature, Lancaster et al. (2013) established a protocol for culturing pluripotent stem cell (PSC)-derived "cerebral organoids" that mimics the developing human brain's cellular organization, segregates into distinct brain regions, and models microcephaly.


Assuntos
Encéfalo/crescimento & desenvolvimento , Encéfalo/patologia , Microcefalia/patologia , Modelos Biológicos , Organoides/citologia , Organoides/crescimento & desenvolvimento , Técnicas de Cultura de Tecidos/métodos , Animais , Humanos
7.
Cell Rep ; 3(4): 1140-52, 2013 Apr 25.
Artigo em Inglês | MEDLINE | ID: mdl-23583175

RESUMO

Melanocytes are pigment-producing cells of neural crest (NC) origin that are responsible for protecting the skin against UV irradiation. Pluripotent stem cell (PSC) technology offers a promising approach for studying human melanocyte development and disease. Here, we report that timed exposure to activators of WNT, BMP, and EDN3 signaling triggers the sequential induction of NC and melanocyte precursor fates under dual-SMAD-inhibition conditions. Using a SOX10::GFP human embryonic stem cell (hESC) reporter line, we demonstrate that the temporal onset of WNT activation is particularly critical for human NC induction. Subsequent maturation of hESC-derived melanocytes yields pure populations that match the molecular and functional properties of adult melanocytes. Melanocytes from Hermansky-Pudlak syndrome and Chediak-Higashi syndrome patient-specific induced PSCs (iPSCs) faithfully reproduce the ultrastructural features of disease-associated pigmentation defects. Our data define a highly specific requirement for WNT signaling during NC induction and enable the generation of pure populations of human iPSC-derived melanocytes for faithful modeling of pigmentation disorders.


Assuntos
Células-Tronco Embrionárias/citologia , Células-Tronco Pluripotentes Induzidas/citologia , Melanócitos/citologia , Modelos Biológicos , Crista Neural/citologia , Proteínas Morfogenéticas Ósseas/metabolismo , Diferenciação Celular , Linhagem da Célula , Síndrome de Chediak-Higashi/metabolismo , Síndrome de Chediak-Higashi/patologia , Células-Tronco Embrionárias/metabolismo , Endotelina-3/metabolismo , Quinase 3 da Glicogênio Sintase/antagonistas & inibidores , Quinase 3 da Glicogênio Sintase/metabolismo , Glicogênio Sintase Quinase 3 beta , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Síndrome de Hermanski-Pudlak/metabolismo , Síndrome de Hermanski-Pudlak/patologia , Humanos , Células-Tronco Pluripotentes Induzidas/metabolismo , Células-Tronco Pluripotentes Induzidas/ultraestrutura , Melanócitos/metabolismo , Crista Neural/metabolismo , Pigmentação , Fatores de Transcrição SOXE/genética , Fatores de Transcrição SOXE/metabolismo , Transdução de Sinais , Proteínas Wnt/metabolismo
8.
PLoS One ; 7(8): e42302, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22879936

RESUMO

Embryonic stem cells (ESCs) and induced pluripotent stem cells (iPSCs) offer great promise in regenerative medicine and disease modeling due to their unlimited self-renewal and broad differentiation capacity. There is evidence that the growth properties and critical signaling pathways differ between murine and human ESCs; therefore, it is essential to perform functional studies to test the putatively conserved mechanisms of pluripotent stem cell self-renewal between species. Previously, we identified the transcription factor Zfx as a key regulator of self-renewal in murine ESCs. Here we extend those findings to human ESCs. ZFX knockdown in hESCs hindered clonal growth and decreased colony size after serial replating. ZFX overexpression enhanced clone formation in the presence of Y-27632, increased colony size at low density and decreased expression of differentiation-related genes in human ESCs. ZFX-overexpressing hESCs resisted spontaneous differentiation but could be directed to differentiate into endodermal and neural cell fates when provided with the appropriate cues. Thus, ZFX acts as a molecular rheostat regulating the balance between self-renewal and differentiation in hESCs, revealing the close evolutionary conservation of the self-renewal mechanisms in murine and human ESCs.


Assuntos
Células-Tronco Embrionárias/citologia , Células-Tronco Embrionárias/metabolismo , Fatores de Transcrição Kruppel-Like/metabolismo , Animais , Diferenciação Celular/genética , Linhagem Celular , Proliferação de Células , Tamanho Celular , Cromossomos Artificiais Bacterianos/genética , Células Clonais , Endoderma/citologia , Endoderma/metabolismo , Perfilação da Expressão Gênica , Regulação da Expressão Gênica , Técnicas de Silenciamento de Genes , Humanos , Fatores de Transcrição Kruppel-Like/genética , Camundongos , Transgenes/genética
9.
Nat Biotechnol ; 30(7): 715-20, 2012 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-22750882

RESUMO

Considerable progress has been made in identifying signaling pathways that direct the differentiation of human pluripotent stem cells (hPSCs) into specialized cell types, including neurons. However, differentiation of hPSCs with extrinsic factors is a slow, step-wise process, mimicking the protracted timing of human development. Using a small-molecule screen, we identified a combination of five small-molecule pathway inhibitors that yield hPSC-derived neurons at >75% efficiency within 10 d of differentiation. The resulting neurons express canonical markers and functional properties of human nociceptors, including tetrodotoxin (TTX)-resistant, SCN10A-dependent sodium currents and response to nociceptive stimuli such as ATP and capsaicin. Neuronal fate acquisition occurs about threefold faster than during in vivo development, suggesting that use of small-molecule pathway inhibitors could become a general strategy for accelerating developmental timing in vitro. The quick and high-efficiency derivation of nociceptors offers unprecedented access to this medically relevant cell type for studies of human pain.


Assuntos
Diferenciação Celular , Nociceptores , Células-Tronco Pluripotentes , Bibliotecas de Moléculas Pequenas , Acetanilidas/farmacologia , Ácidos Cafeicos/farmacologia , Técnicas de Cultura de Células , Diferenciação Celular/efeitos dos fármacos , Linhagem Celular , Regulação da Expressão Gênica no Desenvolvimento/efeitos dos fármacos , Humanos , Dados de Sequência Molecular , Canal de Sódio Disparado por Voltagem NAV1.8/metabolismo , Nociceptores/citologia , Nociceptores/efeitos dos fármacos , Nociceptores/metabolismo , Dor/metabolismo , Dor/fisiopatologia , Células-Tronco Pluripotentes/citologia , Células-Tronco Pluripotentes/efeitos dos fármacos , Células-Tronco Pluripotentes/metabolismo , Piridinas/farmacologia , Pirimidinas/farmacologia , Pirróis/farmacologia , Transdução de Sinais/efeitos dos fármacos , Bibliotecas de Moléculas Pequenas/química , Bibliotecas de Moléculas Pequenas/farmacologia , Tetrodotoxina/farmacologia
10.
Hum Mol Genet ; 21(18): 4104-14, 2012 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-22723015

RESUMO

Exposure to the antiepileptic drug valproic acid (VPA) during gestation causes neurofunctional and anatomic deficits in later life. At present, there are little human data on how early neural development is affected by chemicals. We used human embryonic stem cells, differentiating to neuroectodermal precursors, as a model to investigate the modes of action of VPA. Microarray expression profiling, qPCR of specific marker genes, immunostaining and the expression of green fluorescent protein under the control of the promoter of the canonical neural precursor cell marker HES5 were used as readouts. Exposure to VPA resulted in distorted marker gene expression, characterized by a relative increase in NANOG and OCT4 and a reduction in PAX6. A similar response pattern was observed with trichostatin A, a potent and specific histone deacetylase inhibitor (HDACi), but not with several other toxicants. Differentiation markers were disturbed by prolonged, but not by acute treatment with HDACi, and the strongest disturbance of differentiation was observed by toxicant exposure during early neural fate decision. The increased acetylation of histones observed in the presence of HDACi may explain the up-regulation of some genes. However, to understand the down-regulation of PAX6 and the overall complex transcript changes, we examined further epigenetic markers. Alterations in the methylation of lysines 4 and 27 of histone H3 were detected in the promoter region of PAX6 and OCT4. The changes in these activating and silencing histone marks provide a more general mechanistic rational for the regulation of developmentally important genes at non-cytotoxic drug concentrations.


Assuntos
Anormalidades Induzidas por Medicamentos/genética , Células-Tronco Embrionárias/metabolismo , Epigênese Genética/efeitos dos fármacos , Placa Neural/embriologia , Anormalidades Induzidas por Medicamentos/patologia , Antígenos de Diferenciação/genética , Antígenos de Diferenciação/metabolismo , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Diferenciação Celular/efeitos dos fármacos , Células Cultivadas , Células-Tronco Embrionárias/fisiologia , Proteínas do Olho/genética , Proteínas do Olho/metabolismo , Inibidores de Histona Desacetilases/farmacologia , Histonas/metabolismo , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/metabolismo , Humanos , Ácidos Hidroxâmicos/farmacologia , Metilação , Proteína Homeobox Nanog , Placa Neural/patologia , Células-Tronco Neurais/metabolismo , Células-Tronco Neurais/fisiologia , Células Neuroepiteliais/metabolismo , Células Neuroepiteliais/fisiologia , Fator 3 de Transcrição de Octâmero/genética , Fator 3 de Transcrição de Octâmero/metabolismo , Análise de Sequência com Séries de Oligonucleotídeos , Fator de Transcrição PAX6 , Fatores de Transcrição Box Pareados/genética , Fatores de Transcrição Box Pareados/metabolismo , Análise de Componente Principal , Regiões Promotoras Genéticas , Processamento de Proteína Pós-Traducional , Proteínas Repressoras/genética , Proteínas Repressoras/metabolismo , Transcrição Gênica , Transcriptoma , Ácido Valproico/efeitos adversos
11.
Methods Mol Biol ; 793: 87-97, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21913095

RESUMO

Human embryonic stem cells (hESCs) and the related induced pluripotent stem cells (hiPSCs) have attracted considerable attention since they can provide an unlimited source of many different tissue types. One challenge of using pluripotent cells is directing their broad differentiation potential into one specific tissue or cell fate. The cell fate choices of extraembryonic, endoderm, mesoderm, and ectoderm (including neural) lineages represent the earliest decisions. We found that pluripotent cells efficiently neuralize by blocking the signaling pathways required for alternative cell fate decisions. In this chapter, we detail methods to direct hESCs or hiPSCs into early neural cells and subsequently postmitotic neurons.


Assuntos
Diferenciação Celular , Técnicas Citológicas/métodos , Células-Tronco Embrionárias/citologia , Células-Tronco Pluripotentes Induzidas/citologia , Modelos Biológicos , Doenças Neurodegenerativas/patologia , Neurônios/citologia , Animais , Linhagem Celular , Humanos , Camundongos
12.
Blood ; 118(6): 1525-33, 2011 Aug 11.
Artigo em Inglês | MEDLINE | ID: mdl-21633090

RESUMO

The IFN-inducible immunity-related p47 GTPase Irgm1 has been linked to Crohn disease as well as susceptibility to tuberculosis. Previously we demonstrated that HSC quiescence and function are aberrant in mice lacking Irgm1. To investigate the molecular basis for these defects, we conducted microarray expression profiling of Irgm1-deficient HSCs. Cell-cycle and IFN-response genes are up-regulated in Irgm1(-/-) HSCs, consistent with dysregulated IFN signaling. To test the hypothesis that Irgm1 normally down-regulates IFN signaling in HSCs, we generated Irgm1(-/-)Ifngr1(-/-) and Irgm1(-/-)Stat1(-/-) double-knockout animals. Strikingly, hyperproliferation, self-renewal, and autophagy defects in Irgm1(-/-) HSCs were normalized in double-knockout animals. These defects were also abolished in Irgm1(-/-)Irgm3(-/-) double-knockout animals, indicating that Irgm1 may regulate Irgm3 activity. Furthermore, the number of HSCs was reduced in aged Irgm1(-/-) animals, suggesting that negative feedback inhibition of IFN signaling by Irgm1 is necessary to prevent hyperproliferation and depletion of the stem cell compartment. Collectively, our results indicate that Irgm1 is a powerful negative regulator of IFN-dependent stimulation in HSCs, with an essential role in preserving HSC number and function. The deleterious effects of excessive IFN signaling may explain how hematologic abnormalities arise in patients with inflammatory conditions.


Assuntos
Proteínas de Ligação ao GTP/genética , Células-Tronco Hematopoéticas/metabolismo , Receptores de Interferon/genética , Transdução de Sinais/genética , Animais , Autofagia/efeitos dos fármacos , Autofagia/genética , Transplante de Medula Óssea , Ciclo Celular/efeitos dos fármacos , Ciclo Celular/genética , Proliferação de Células/efeitos dos fármacos , Citometria de Fluxo , Proteínas de Ligação ao GTP/deficiência , Expressão Gênica/efeitos dos fármacos , Perfilação da Expressão Gênica , Células HEK293 , Humanos , Imuno-Histoquímica , Interferons/genética , Interferons/metabolismo , Interferons/farmacologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Análise de Sequência com Séries de Oligonucleotídeos , Receptores de Interferon/deficiência , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fator de Transcrição STAT1/deficiência , Fator de Transcrição STAT1/genética , Receptor de Interferon gama
13.
Cell ; 145(6): 827-30, 2011 Jun 10.
Artigo em Inglês | MEDLINE | ID: mdl-21663788

RESUMO

Building on the discovery that MyoD expression reprograms fibroblasts into muscle, three papers (Vierbuchen et al., 2010; Ieda et al., 2010; Szabo et al., 2010) recently reported the reprogramming of fibroblasts into neurons, cardiomyocytes, and blood cell progenitors without first passing the cells through a pluripotent state. Here we discuss the advantages and challenges of harnessing this direct reprogramming method for regenerative medicine.


Assuntos
Reprogramação Celular , Células-Tronco Pluripotentes Induzidas/citologia , Medicina Regenerativa , Animais , Diferenciação Celular , Fibroblastos/citologia , Humanos , Fatores de Transcrição/metabolismo
14.
Nat Protoc ; 5(4): 688-701, 2010 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-20360764

RESUMO

Human pluripotent stem cell (hPSC)-derived neural crest (NC) cells present a valuable tool for modeling aspects of human NC development, including cell fate specification, multipotency and cell migration. hPSC-derived NC cells are also suitable for modeling human disease and as a renewable cell source for applications in regenerative medicine. Here we provide protocols for the step-wise differentiation of human embryonic stem cells (hESCs) or human induced pluripotent stem cells (hiPSCs) into neuroectodermal and NC cells using either the MS5 coculture system or a novel defined culture method based on pharmacological inhibition of bone morphogenetic protein and transforming growth factor-beta signaling pathways. Furthermore, we present protocols for the purification and propagation of hPSC-NC cells using flow cytometry and defined in vitro culture conditions. Our protocol has been validated in multiple independent hESC and hiPSC lines. The average time required for generating purified hPSC-NC precursors using this protocol is 2-5 weeks.


Assuntos
Técnicas de Cultura de Células/métodos , Crista Neural/citologia , Células-Tronco Pluripotentes/citologia , Diferenciação Celular , Proliferação de Células , Separação Celular , Cromossomos Artificiais Bacterianos/genética , Técnicas de Cocultura , Células-Tronco Embrionárias/citologia , Células-Tronco Embrionárias/metabolismo , Citometria de Fluxo , Humanos , Modelos Neurológicos , Crista Neural/metabolismo , Células-Tronco Pluripotentes/metabolismo , Células de Schwann/citologia
15.
Cell Stem Cell ; 6(4): 336-347, 2010 Apr 02.
Artigo em Inglês | MEDLINE | ID: mdl-20362538

RESUMO

The floor plate (FP) is a critical signaling center during neural development located along the ventral midline of the embryo. Little is known about human FP development because of the lack of tissue accessibility. Here we report the efficient derivation of human embryonic stem cell (hESC)-derived FP tissue capable of secreting Netrin-1 and SHH and patterning primary and hESC derived tissues. FP induction in hESCs is dependent on early SHH exposure and occurs at the expense of anterior neurectoderm (AN). Global gene expression and functional studies identify SHH-mediated inhibition of Dkk-1 as key factor in FP versus AN specification. hESC-derived FP tissue is shown to be of anterior SIX6+ character but is responsive to caudalizing factors suppressing SIX6 expression and inducing a shift in usage of region-specific SHH enhancers. These data define the early signals that drive human FP versus AN specification and determine regional identity in hESC-derived FP.


Assuntos
Células-Tronco Embrionárias/citologia , Sistema Nervoso/citologia , Animais , Padronização Corporal/genética , Diferenciação Celular/genética , Linhagem da Célula/genética , Células-Tronco Embrionárias/metabolismo , Fatores de Transcrição Forkhead/metabolismo , Perfilação da Expressão Gênica , Regulação da Expressão Gênica no Desenvolvimento , Proteínas Hedgehog/metabolismo , Fator 3-beta Nuclear de Hepatócito/metabolismo , Humanos , Peptídeos e Proteínas de Sinalização Intercelular/genética , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Camundongos , Proteínas do Tecido Nervoso/metabolismo , Sistema Nervoso/metabolismo , Placa Neural/embriologia , Placa Neural/metabolismo , Fatores de Tempo , Transcrição Gênica
16.
Cell Stem Cell ; 6(3): 265-78, 2010 Mar 05.
Artigo em Inglês | MEDLINE | ID: mdl-20207229

RESUMO

The traditional view of hematopoiesis has been that all the cells of the peripheral blood are the progeny of a unitary homogeneous pool of hematopoietic stem cells (HSCs). Recent evidence suggests that the hematopoietic system is actually maintained by a consortium of HSC subtypes with distinct functional characteristics. We show here that myeloid-biased HSCs (My-HSCs) and lymphoid-biased HSCs (Ly-HSCs) can be purified according to their capacity for Hoechst dye efflux in combination with canonical HSC markers. These phenotypes are stable under natural (aging) or artificial (serial transplantation) stress and are exacerbated in the presence of competing HSCs. My- and Ly-HSCs respond differently to TGF-beta1, presenting a possible mechanism for differential regulation of HSC subtype activation. This study demonstrates definitive isolation of lineage-biased HSC subtypes and contributes to the fundamental change in view that the hematopoietic system is maintained by a continuum of HSC subtypes, rather than a functionally uniform pool.


Assuntos
Linhagem da Célula , Células-Tronco Hematopoéticas/citologia , Células-Tronco Hematopoéticas/metabolismo , Fator de Crescimento Transformador beta1/metabolismo , Envelhecimento , Animais , Diferenciação Celular , Proliferação de Células , Transplante de Células , Camundongos , Fenótipo , Transcrição Gênica
17.
Nature ; 461(7262): 402-6, 2009 Sep 17.
Artigo em Inglês | MEDLINE | ID: mdl-19693009

RESUMO

The isolation of human induced pluripotent stem cells (iPSCs) offers a new strategy for modelling human disease. Recent studies have reported the derivation and differentiation of disease-specific human iPSCs. However, a key challenge in the field is the demonstration of disease-related phenotypes and the ability to model pathogenesis and treatment of disease in iPSCs. Familial dysautonomia (FD) is a rare but fatal peripheral neuropathy, caused by a point mutation in the IKBKAP gene involved in transcriptional elongation. The disease is characterized by the depletion of autonomic and sensory neurons. The specificity to the peripheral nervous system and the mechanism of neuron loss in FD are poorly understood owing to the lack of an appropriate model system. Here we report the derivation of patient-specific FD-iPSCs and the directed differentiation into cells of all three germ layers including peripheral neurons. Gene expression analysis in purified FD-iPSC-derived lineages demonstrates tissue-specific mis-splicing of IKBKAP in vitro. Patient-specific neural crest precursors express particularly low levels of normal IKBKAP transcript, suggesting a mechanism for disease specificity. FD pathogenesis is further characterized by transcriptome analysis and cell-based assays revealing marked defects in neurogenic differentiation and migration behaviour. Furthermore, we use FD-iPSCs for validating the potency of candidate drugs in reversing aberrant splicing and ameliorating neuronal differentiation and migration. Our study illustrates the promise of iPSC technology for gaining new insights into human disease pathogenesis and treatment.


Assuntos
Disautonomia Familiar/patologia , Disautonomia Familiar/terapia , Modelos Biológicos , Células-Tronco Pluripotentes/metabolismo , Células-Tronco Pluripotentes/transplante , Adolescente , Processamento Alternativo/efeitos dos fármacos , Processamento Alternativo/genética , Animais , Proteínas de Transporte/genética , Desdiferenciação Celular , Diferenciação Celular , Linhagem da Célula , Movimento Celular , Células Cultivadas , Criança , Disautonomia Familiar/tratamento farmacológico , Disautonomia Familiar/genética , Feminino , Fibroblastos/citologia , Fibroblastos/metabolismo , Perfilação da Expressão Gênica , Humanos , Cinetina/farmacologia , Cinetina/uso terapêutico , Masculino , Camundongos , Crista Neural/citologia , Crista Neural/efeitos dos fármacos , Especificidade de Órgãos , Fenótipo , Células-Tronco Pluripotentes/citologia , Células-Tronco Pluripotentes/efeitos dos fármacos , Fatores de Elongação da Transcrição
18.
Proc Natl Acad Sci U S A ; 106(31): 12759-64, 2009 Aug 04.
Artigo em Inglês | MEDLINE | ID: mdl-19549847

RESUMO

Human-induced pluripotent stem cells (hiPSCs) are generated from somatic cells by ectopic expression of the 4 reprogramming factors (RFs) Oct-4, Sox2, Klf4, and c-Myc. To better define the stoichiometric requirements and dynamic expression patterns required for successful hiPSC induction, we generated 4 bicistronic lentiviral vectors encoding the 4 RFs co-expressed with discernable fluorescent proteins. Using this system, we define the optimal stoichiometry of RF expression to be highly sensitive to Oct4 dosage, and we demonstrate the impact that variations in the relative ratios of RF expression exert on the efficiency of hiPSC induction. Monitoring of expression of each individual RF in single cells during the course of reprogramming revealed that vector silencing follows acquisition of pluripotent cell markers. Pronounced lentiviral vector silencing was a characteristic of successfully reprogrammed hiPSC clones, but lack of complete silencing did not hinder hiPSC induction, maintenance, or directed differentiation. The vector system described here presents a powerful tool for mechanistic studies of reprogramming and the optimization of hiPSC generation.


Assuntos
Genes myc/fisiologia , Fatores de Transcrição Kruppel-Like/fisiologia , Fator 3 de Transcrição de Octâmero/fisiologia , Células-Tronco Pluripotentes/citologia , Fatores de Transcrição SOXB1/fisiologia , Diferenciação Celular , Epigênese Genética , Humanos , Fator 4 Semelhante a Kruppel , Fatores de Transcrição Kruppel-Like/genética , Fator 3 de Transcrição de Octâmero/genética , Fatores de Transcrição SOXB1/genética
19.
Nat Biotechnol ; 27(3): 275-80, 2009 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-19252484

RESUMO

Current neural induction protocols for human embryonic stem (hES) cells rely on embryoid body formation, stromal feeder co-culture or selective survival conditions. Each strategy has considerable drawbacks, such as poorly defined culture conditions, protracted differentiation and low yield. Here we report that the synergistic action of two inhibitors of SMAD signaling, Noggin and SB431542, is sufficient to induce rapid and complete neural conversion of >80% of hES cells under adherent culture conditions. Temporal fate analysis reveals the appearance of a transient FGF5(+) epiblast-like stage followed by PAX6(+) neural cells competent to form rosettes. Initial cell density determines the ratio of central nervous system and neural crest progeny. Directed differentiation of human induced pluripotent stem (hiPS) cells into midbrain dopamine and spinal motoneurons confirms the robustness and general applicability of the induction protocol. Noggin/SB431542-based neural induction should facilitate the use of hES and hiPS cells in regenerative medicine and disease modeling and obviate the need for protocols based on stromal feeders or embryoid bodies.


Assuntos
Diferenciação Celular , Células-Tronco Embrionárias/citologia , Neurônios/citologia , Células-Tronco Pluripotentes/citologia , Transdução de Sinais/efeitos dos fármacos , Proteínas Smad/efeitos dos fármacos , Proteínas de Transporte/farmacologia , Técnicas de Cultura de Células , Humanos , Mesencéfalo/citologia , Proteínas Smad/metabolismo
20.
Blood ; 111(4): 2444-51, 2008 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-18055867

RESUMO

Hematopoietic stem cells (HSCs) are a self-renewing population of bone marrow cells that replenish the cellular elements of blood throughout life. HSCs represent a paradigm for the study of stem-cell biology, because robust methods for prospective isolation of HSCs have facilitated rigorous characterization of these cells. Recently, a new isolation method was reported, using the SLAM family of cell-surface markers, including CD150 (SlamF1), to offer potential advantages over established protocols. We examined the overlap between SLAM family member expression with an established isolation scheme based on Hoechst dye efflux (side population; SP) in conjunction with canonical HSC cell-surface markers (Sca-1, c-Kit, and lineage markers). Importantly, we find that stringent gating of SLAM markers is essential to achieving purity in HSC isolation and that the inclusion of canonical HSC markers in the SLAM scheme can greatly augment HSC purity. Furthermore, we observe that both CD150(+) and CD150(-) cells can be found within the SP population and that both populations can contribute to long-term multilineage reconstitution. Thus, using SLAM family markers to isolate HSCs excludes a substantial fraction of the marrow HSC compartment. Interestingly, these 2 subpopulations are functionally distinct, with respect to lineage output as well as proliferative status.


Assuntos
Antígenos CD/análise , Transplante de Células-Tronco Hematopoéticas , Células-Tronco Hematopoéticas/citologia , Receptores de Superfície Celular/análise , Animais , Antígenos CD/genética , Biomarcadores/análise , Bromodesoxiuridina , Separação Celular , Células-Tronco Hematopoéticas/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Receptores de Superfície Celular/deficiência , Receptores de Superfície Celular/genética , Membro 1 da Família de Moléculas de Sinalização da Ativação Linfocitária , Quimeras de Transplante
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...