Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
1.
Cell ; 136(5): 964-77, 2009 Mar 06.
Artículo en Inglés | MEDLINE | ID: mdl-19269371

RESUMEN

Induced pluripotent stem cells (iPSCs) derived from somatic cells of patients represent a powerful tool for biomedical research and may provide a source for replacement therapies. However, the use of viruses encoding the reprogramming factors represents a major limitation of the current technology since even low vector expression may alter the differentiation potential of the iPSCs or induce malignant transformation. Here, we show that fibroblasts from five patients with idiopathic Parkinson's disease can be efficiently reprogrammed and subsequently differentiated into dopaminergic neurons. Moreover, we derived hiPSCs free of reprogramming factors using Cre-recombinase excisable viruses. Factor-free hiPSCs maintain a pluripotent state and show a global gene expression profile, more closely related to hESCs than to hiPSCs carrying the transgenes. Our results indicate that residual transgene expression in virus-carrying hiPSCs can affect their molecular characteristics and that factor-free hiPSCs therefore represent a more suitable source of cells for modeling of human disease.


Asunto(s)
Enfermedad de Parkinson/metabolismo , Células Madre Pluripotentes/patología , Diferenciación Celular , Reprogramación Celular , Dopamina/metabolismo , Fibroblastos/metabolismo , Humanos , Neuronas/metabolismo
2.
Mol Cell Neurosci ; 45(3): 258-66, 2010 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-20603216

RESUMEN

The cardinal motor symptoms of Parkinson's disease (PD) are caused by the vulnerability to dysfunction and degeneration of ventral midbrain (VM) dopaminergic (DA) neurons. A major limitation for experimental studies of current ES/iPS cell differentiation protocols is the lack of VM DA neurons with a stable phenotype as defined by an expression marker code of FOXA2/TH/ß-tubulin. Here we demonstrate a combination of three modifications that were required to produce VM DA neurons. Firstly, early and specific exposure to 10(-)(8)M (low dose) retinoic acid improved the regional identity of neural progenitor cells derived from human ES cells, PD or healthy subject-specific iPS cells. Secondly, a high activity form of human sonic hedgehog established a sizeable FOXA2(+) neural progenitor cell population in vitro. Thirdly, early exposure to FGF8a, rather than Fgf8b, and WNT1 was required for robust differentiation of the FOXA2(+) floor plate-like human neural progenitor cells into FOXA2(+) DA neurons. FOXA2(+) DA neurons were also generated when this protocol was adapted to feeder-free conditions. In summary, this new human ES and iPS cell differentiation protocol using FGF8a, WNT1, low dose retinoic acid and a high activity form of SHH can generate human VM DA neurons that are required for relevant new bioassays, drug discovery and cell based therapies for PD.


Asunto(s)
Diferenciación Celular/efectos de los fármacos , Dopamina/metabolismo , Células Madre Embrionarias/citología , Factor 8 de Crecimiento de Fibroblastos/farmacología , Proteínas Hedgehog/metabolismo , Neuronas/citología , Células Madre Pluripotentes/citología , Tretinoina/farmacología , Animales , Técnicas de Cultivo de Célula , Diferenciación Celular/fisiología , Células Cultivadas , Células Madre Embrionarias/efectos de los fármacos , Células Madre Embrionarias/fisiología , Proteínas Hedgehog/genética , Factor Nuclear 3-beta del Hepatocito/metabolismo , Humanos , Mesencéfalo/citología , Ratones , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Enfermedad de Parkinson , Células Madre Pluripotentes/efectos de los fármacos , Células Madre Pluripotentes/fisiología , Proteína Wnt1/farmacología
3.
Stem Cells ; 27(12): 2928-40, 2009 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-19725119

RESUMEN

Identification and use of cell surface cluster of differentiation (CD) biomarkers have enabled much scientific and clinical progress. We identify a CD surface antigen code for the neural lineage based on combinatorial flow cytometric analysis of three distinct populations derived from human embryonic stem cells: (1) CD15(+)/CD29(HI)/CD24(LO) surface antigen expression defined neural stem cells; (2) CD15(-)/CD29(HI)/CD24(LO) revealed neural crest-like and mesenchymal phenotypes; and (3) CD15(-)/CD29(LO)/CD24(HI) selected neuroblasts and neurons. Fluorescence-activated cell sorting (FACS) for the CD15(-)/CD29(LO)/CD24(HI) profile reduced proliferative cell types in human embryonic stem cell differentiation. This eliminated tumor formation in vivo, resulting in pure neuronal grafts. In conclusion, combinatorial CD15/CD24/CD29 marker profiles define neural lineage development of neural stem cell, neural crest, and neuronal populations from human stem cells. We believe this set of biomarkers enables analysis and selection of neural cell types for developmental studies and pharmacological and therapeutic applications.


Asunto(s)
Antígeno CD24/metabolismo , Diferenciación Celular , Membrana Celular/metabolismo , Fucosiltransferasas/metabolismo , Integrina beta1/metabolismo , Antígeno Lewis X/metabolismo , Neuronas/metabolismo , Células Madre/metabolismo , Animales , Biomarcadores , Linaje de la Célula , Células Cultivadas , Femenino , Humanos , Ratones , Ratones Endogámicos C57BL , Neuronas/citología , Células Madre/citología
4.
J Neurosci ; 27(32): 8581-92, 2007 Aug 08.
Artículo en Inglés | MEDLINE | ID: mdl-17687036

RESUMEN

Fibroblast growth factors (FGFs) secreted from the midbrain-rhombomere 1 (r1) boundary instruct cell behavior in the surrounding neuroectoderm. For example, a combination of FGF and sonic hedgehog (SHH) can induce the development of the midbrain dopaminergic neurons, but the mechanisms behind the action and integration of these signals are unclear. We studied how FGF receptors (FGFRs) regulate cellular responses by analyzing midbrain-r1 development in mouse embryos, which carry different combinations of mutant Fgfr1, Fgfr2, and Fgfr3 alleles. Our results show that the FGFRs act redundantly to support cell survival in the dorsal neuroectoderm, promote r1 tissue identity, and regulate the production of ventral neuronal populations, including midbrain dopaminergic neurons. The compound Fgfr mutants have apparently normal WNT/SHH signaling and neurogenic gene expression in the ventral midbrain, but the number of proliferative neural progenitors is reduced as a result of precocious neuronal differentiation. Our results suggest a SoxB1 family member, Sox3, as a potential FGF-induced transcription factor promoting progenitor renewal. We propose a model for regulation of progenitor cell self-renewal and neuronal differentiation by combinatorial intercellular signals in the ventral midbrain.


Asunto(s)
Mesencéfalo/embriología , Neuronas/fisiología , Receptores de Factores de Crecimiento de Fibroblastos/fisiología , Rombencéfalo/embriología , Células Madre/fisiología , Animales , Diferenciación Celular/fisiología , Femenino , Regulación del Desarrollo de la Expresión Génica/fisiología , Mesencéfalo/citología , Mesencéfalo/fisiología , Ratones , Ratones Transgénicos , Neuronas/citología , Embarazo , Rombencéfalo/citología , Rombencéfalo/fisiología , Células Madre/citología
5.
PLoS One ; 10(5): e0127681, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25993409

RESUMEN

GABAergic neurons are the primary inhibitory cell type in the mature brain and their dysfunction is associated with important neurological conditions like schizophrenia and anxiety. We aimed to discover the underlying mechanisms for dorsal/ventral midbrain GABAergic neurogenesis. Previous work by us and others has provided crucial insights into the key function of Mgn and Mash1 genes in determining GABAergic neurotransmitter fate. Induction of dorsal midbrain GABAergic neurons does not take place at any time during development in either of the single mutant mice. However, GABAergic neurons in the ventral midbrain remained unchanged. Thus, the similarities in MB-GABAergic phenotype observed in the Mgn and Mash1 single mutants suggest the existence of other factors that take over the function of MGN and MASH1 in the ventral midbrain or the existence of different molecular mechanisms. We show that this process essentially depends on heterodimers and homodimers formed by MGN and MASH1 and deciphered the in vivo relevance of the interaction by phenotypic analysis of Mgn/Mash1 double knockout and compound mice. Furthermore, the combination of gain- and loss-of-function experiments in the developing midbrain showed co-operative roles for Mgn and Mash1 genes in determining GABAergic identity. Transcription factors belonging to the Enhancer-of-split-related and proneural families have long been believed to counterpart each other's function. This work uncovers a synergistic cooperation between these two families, and provides a novel paradigm for how these two families cooperate for the acquisition of MB-GABAergic neuronal identity. Understanding their molecular mechanisms is essential for cell therapy strategies to amend GABAergic deficits.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Neuronas GABAérgicas/metabolismo , Mesencéfalo/citología , Mesencéfalo/metabolismo , Neurogénesis , Proteínas Represoras/metabolismo , Animales , Neuronas GABAérgicas/citología , Inmunoprecipitación , Ratones , Mutación , Neurotransmisores/metabolismo , Unión Proteica , Multimerización de Proteína , Saccharomyces cerevisiae/metabolismo , Técnicas del Sistema de Dos Híbridos
6.
Stem Cells Transl Med ; 3(9): 1032-42, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-25024431

RESUMEN

Human embryonic and induced pluripotent stem cells are potential cell sources for regenerative approaches in Parkinson disease. Inductive differentiation protocols can generate midbrain dopamine neurons but result in heterogeneous cell mixtures. Therefore, selection strategies are necessary to obtain uniform dopamine cell populations. Here, we developed a selection approach using lentivirus vectors to express green fluorescent protein under the promoter region of FOXA2, a transcription factor that is expressed in the floor plate domain that gives rise to dopamine neurons during embryogenesis. We first validated the specificity of the vectors in human cell lines against a promoterless construct. We then selected FOXA2-positive neural progenitors from several human pluripotent stem cell lines, which demonstrated a gene expression profile typical for the ventral domain of the midbrain and floor plate, but failed to enrich for dopamine neurons. To investigate whether this was due to the selection approach, we overexpressed FOXA2 in neural progenitors derived from human pluripotent stem cell lines. FOXA2 forced expression resulted in an increased expression of floor plate but not mature neuronal markers. Furthermore, selection of the FOXA2 overexpressing fraction also failed to enrich for dopamine neurons. Collectively, our results suggest that FOXA2 is not sufficient to induce a dopaminergic fate in this system. On the other hand, our study demonstrates that a combined approach of promoter activation and lentivirus vector technology can be used as a versatile tool for the selection of a defined cell population from a variety of human pluripotent stem cell lines.


Asunto(s)
Separación Celular/métodos , Neuronas Dopaminérgicas/citología , Factor Nuclear 3-beta del Hepatocito/genética , Células-Madre Neurales/citología , Células Madre Pluripotentes/citología , Western Blotting , Citometría de Flujo , Técnica del Anticuerpo Fluorescente , Vectores Genéticos , Proteínas Fluorescentes Verdes/genética , Humanos , Lentivirus , Microscopía Confocal , Regiones Promotoras Genéticas , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción Genética
7.
Dev Biol ; 303(1): 231-43, 2007 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-17150206

RESUMEN

The mid-/hindbrain organizer (MHO) is characterized by the expression of a network of genes, which controls the patterning and development of the prospective midbrain and anterior hindbrain. One key molecule acting at the MHO is the fibroblast growth factor (Fgf) 8. Ectopic expression of Fgf8 induces genes that are normally expressed at the mid-/hindbrain boundary followed by the induction of midbrain and anterior hindbrain structures. Inactivation of the Fgf receptor (Fgfr) 1 gene, which was thought to be the primary transducer of the Fgf8 signal at the MHO, in the mid-/hindbrain region, leads to a deletion of dorsal structures of the mid-/hindbrain region, whereas ventral tissues are less severely affected. This suggests that other Fgfrs might be responsible for ventral mid-/hindbrain region development. Here we report the analysis of Fgfr2 conditional knockout mice, lacking the Fgfr2 in the mid-/hindbrain region and of Fgfr3 knockout mice with respect to the mid-/hindbrain region. In both homozygous mouse mutants, patterning of the mid-/hindbrain region is not altered, neuronal populations develop normal and are maintained into adulthood. This analysis shows that the Fgfr2 and the Fgfr3 on their own are dispensable for the development of the mid-/hindbrain region. We suggest functional redundancy of Fgf receptors in the mid-/hindbrain region.


Asunto(s)
Tipificación del Cuerpo/fisiología , Mesencéfalo/embriología , Receptor Tipo 2 de Factor de Crecimiento de Fibroblastos/metabolismo , Receptor Tipo 3 de Factor de Crecimiento de Fibroblastos/metabolismo , Rombencéfalo/embriología , Animales , Factor 8 de Crecimiento de Fibroblastos/metabolismo , Inmunohistoquímica , Hibridación in Situ , Ratones , Ratones Noqueados , Receptor Tipo 2 de Factor de Crecimiento de Fibroblastos/genética , Receptor Tipo 3 de Factor de Crecimiento de Fibroblastos/genética
8.
Dev Dyn ; 233(3): 1023-30, 2005 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-15830353

RESUMEN

Fibroblast growth factor 8 (FGF8) mediates the function of the midbrain-hindbrain organizer (MHO). FGF signals are transmitted by means of four known FGF receptors (FGFRs). Studies of Fgfr expression in early vertebrate development have shown that Fgfr1 is expressed along the entire neural tube, whereas Fgfr2 and Fgfr3 expression has been shown to spare the tissue adjacent to the MHO. The FGF8 signal from the MHO, therefore, was believed to be transmitted by FGFR1 exclusively. However, incongruent results from conditional mutants of Fgf8 and Fgfr1 in the midbrain-hindbrain (MHB) region contradict this hypothesis. Therefore, we reexamined the expression of the Fgfrs in this region. Fgfr1 is expressed all over the neural tube. Strikingly, Fgfr2 is expressed throughout the floor plate of the MHB region. In the basal plate, Fgfr2 directly abuts the Fgf8 expression domain at the MHO, anteriorly and posteriorly. Fgfr3 expression is in contact with the Fgf8 expression domain only in the rostroventral hindbrain. Based on these findings, we postulate a role for FGFR2 and FGFR3 in FGF signaling in the ventral midbrain and hindbrain.


Asunto(s)
Mesencéfalo/embriología , Mesencéfalo/metabolismo , Prosencéfalo/embriología , Prosencéfalo/metabolismo , Proteínas Tirosina Quinasas/genética , Proteínas Tirosina Quinasas Receptoras/genética , Receptores de Factores de Crecimiento de Fibroblastos/genética , Animales , Regulación del Desarrollo de la Expresión Génica/genética , Hibridación in Situ , Ratones , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos , Receptor Tipo 2 de Factor de Crecimiento de Fibroblastos , Receptor Tipo 3 de Factor de Crecimiento de Fibroblastos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA