Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
1.
Differentiation ; 81(5): 292-8, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21306817

RESUMEN

Human parthenogenetic stem cells (hpSC) hold great promise as a source of pluripotent stem cells for cell-based transplantation therapy due to their ethical method of derivation as well as the enhanced capacity for immunomatching with significant segments of the human population. We report here the directed differentiation of hpSC to produce enriched populations of definitive endoderm. Moreover, we find that treatment of undifferentiated hpSC by trichostatin A (TSA) before applying the directed differentiation protocol significantly increases the proportion of definitive endoderm cells in the final population. TSA-pretreated as well as non-TSA-treated hpSC undergoing differentiation toward definitive endoderm demonstrate a similar temporal sequence of gene expression to that which occurs in the course of definitive endoderm differentiation during vertebrate gastrulation and for differentiation of hESCs to definitive endoderm. Creation of the definitive endoderm lineages from hpSC represents the critical first step toward the development of hpSC-based cellular therapies for diseases of the liver or pancreas.


Asunto(s)
Diferenciación Celular/efectos de los fármacos , Células Madre Embrionarias/metabolismo , Endodermo/citología , Endodermo/metabolismo , Ácidos Hidroxámicos/farmacología , Partenogénesis/genética , Técnicas de Cultivo de Célula/métodos , Diferenciación Celular/genética , Linaje de la Célula , Células Cultivadas , Células Madre Embrionarias/citología , Humanos , Células Madre Pluripotentes/citología , Células Madre Pluripotentes/metabolismo , Células Madre/citología , Células Madre/metabolismo
2.
Cell Rep Med ; 2(12): 100466, 2021 12 21.
Artículo en Inglés | MEDLINE | ID: mdl-35028608

RESUMEN

These preliminary data from an ongoing first-in-human phase 1/2, open-label study provide proof-of-concept that pluripotent stem cell-derived pancreatic endoderm cells (PEC-01) engrafted in type 1 diabetes patients become islet cells releasing insulin in a physiologically regulated fashion. In this study of 17 subjects aged 22-57 with type 1 diabetes, PEC-01 cells were implanted subcutaneously in VC-02 macroencapsulation devices, allowing for direct vascularization of the cells. Engraftment and insulin expression were observed in 63% of VC-02 units explanted from subjects at 3-12 months post-implant. Six of 17 subjects (35.3%) demonstrated positive C-peptide as early as 6 months post-implant. Most reported adverse events were related to surgical implant or explant procedures (27.9%) or to side-effects of immunosuppression (33.7%). Initial data suggest that pluripotent stem cells, which can be propagated to the desired biomass and differentiated into pancreatic islet-like tissue, may offer a scalable, renewable alternative to pancreatic islet transplants.


Asunto(s)
Péptido C/metabolismo , Células Inmovilizadas/citología , Diabetes Mellitus Tipo 1/terapia , Endodermo/citología , Insulina/metabolismo , Páncreas/citología , Trasplante de Células Madre , Células Madre/citología , Adolescente , Adulto , Anciano , Diabetes Mellitus Tipo 1/metabolismo , Femenino , Humanos , Masculino , Persona de Mediana Edad , Adulto Joven
3.
Nature ; 430(6997): 350-6, 2004 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-15254537

RESUMEN

Somatic stem cells have been claimed to possess an unexpectedly broad differentiation potential (referred to here as plasticity) that could be induced by exposing stem cells to the extracellular developmental signals of other lineages in mixed-cell cultures. Recently, this and other experimental evidence supporting the existence of stem-cell plasticity have been refuted because stem cells have been shown to adopt the functional features of other lineages by means of cell-fusion-mediated acquisition of lineage-specific determinants (chromosomal DNA) rather than by signal-mediated differentiation. In this study we co-cultured mouse neural stem cells (NSCs), which are committed to become neurons and glial cells, with human endothelial cells, which form the lining of blood vessels. We show that in the presence of endothelial cells six per cent of the NSC population converted to cells that did not express neuronal or glial markers, but instead showed the stable expression of multiple endothelial markers and the capacity to form capillary networks. This was surprising because NSCs and endothelial cells are believed to develop from the ectoderm and mesoderm, respectively. Experiments in which endothelial cells were killed by fixation before co-culture with live NSCs (to prevent cell fusion) and karyotyping analyses, revealed that NSCs had differentiated into endothelial-like cells independently of cell fusion. We conclude that stem-cell plasticity is a true characteristic of NSCs and that the conversion of NSCs to unanticipated cell types can be accomplished without cell fusion.


Asunto(s)
Antígenos CD , Diferenciación Celular , Linaje de la Célula , Células Endoteliales/citología , Endotelio Vascular/citología , Moléculas de Adhesión de Célula Nerviosa , Neuronas/citología , Células Madre/citología , Animales , Biomarcadores/análisis , Antígeno CD146 , Capilares/citología , Capilares/metabolismo , Fusión Celular , Células Cultivadas , Células Clonales/citología , Células Clonales/metabolismo , Técnicas de Cocultivo , Células Endoteliales/metabolismo , Endotelio Vascular/metabolismo , Humanos , Cariotipificación , Glicoproteínas de Membrana/metabolismo , Ratones , Neuronas/metabolismo , Células Madre/metabolismo
4.
Nat Biotechnol ; 24(11): 1392-401, 2006 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-17053790

RESUMEN

Of paramount importance for the development of cell therapies to treat diabetes is the production of sufficient numbers of pancreatic endocrine cells that function similarly to primary islets. We have developed a differentiation process that converts human embryonic stem (hES) cells to endocrine cells capable of synthesizing the pancreatic hormones insulin, glucagon, somatostatin, pancreatic polypeptide and ghrelin. This process mimics in vivo pancreatic organogenesis by directing cells through stages resembling definitive endoderm, gut-tube endoderm, pancreatic endoderm and endocrine precursor--en route to cells that express endocrine hormones. The hES cell-derived insulin-expressing cells have an insulin content approaching that of adult islets. Similar to fetal beta-cells, they release C-peptide in response to multiple secretory stimuli, but only minimally to glucose. Production of these hES cell-derived endocrine cells may represent a critical step in the development of a renewable source of cells for diabetes cell therapy.


Asunto(s)
Diferenciación Celular/fisiología , Células Madre Embrionarias/metabolismo , Células Enteroendocrinas/fisiología , Islotes Pancreáticos/crecimiento & desarrollo , Hormonas Pancreáticas/biosíntesis , Hormonas Peptídicas/biosíntesis , Células Cultivadas , Ghrelina , Humanos , Islotes Pancreáticos/citología , Islotes Pancreáticos/metabolismo , Páncreas/citología , Hormonas Pancreáticas/aislamiento & purificación
5.
Nat Biotechnol ; 23(12): 1534-41, 2005 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-16258519

RESUMEN

The potential of human embryonic stem (hES) cells to differentiate into cell types of a variety of organs has generated much excitement over the possible use of hES cells in therapeutic applications. Of great interest are organs derived from definitive endoderm, such as the pancreas. We have focused on directing hES cells to the definitive endoderm lineage as this step is a prerequisite for efficient differentiation to mature endoderm derivatives. Differentiation of hES cells in the presence of activin A and low serum produced cultures consisting of up to 80% definitive endoderm cells. This population was further enriched to near homogeneity using the cell-surface receptor CXCR4. The process of definitive endoderm formation in differentiating hES cell cultures includes an apparent epithelial-to-mesenchymal transition and a dynamic gene expression profile that are reminiscent of vertebrate gastrulation. These findings may facilitate the use of hES cells for therapeutic purposes and as in vitro models of development.


Asunto(s)
Técnicas de Cultivo de Célula/métodos , Endodermo/citología , Endodermo/fisiología , Células Madre/citología , Células Madre/fisiología , Ingeniería de Tejidos/métodos , Animales , Diferenciación Celular , Proliferación Celular , Células Cultivadas , Humanos , Ratones
7.
J Neurosci ; 24(24): 5459-66, 2004 Jun 16.
Artículo en Inglés | MEDLINE | ID: mdl-15201317

RESUMEN

Choline acetyltransferase (ChAT), the enzyme that synthesizes the neurotransmitter acetylcholine (ACh), is thought to be present in kinetic excess in cholinergic neurons. The rate-limiting factor in ACh production is the provision of choline to ChAT. Cholinergic neurons are relatively unique in their expression of the choline transporter 1 (CHT1), which exhibits high-affinity for choline and catalyzes its uptake from the extracellular space to the neuron. Multiple lines of evidence indicate that the activity of CHT1 is a key determinant of choline supply for ACh synthesis. We examined the interaction of ChAT and ChT activity using mice heterozygous for a null mutation in the Chat gene (Chat+/-). In these mice, brain ChAT activity was reduced by 40-50% relative to the wild type, but brain ACh levels as well as ACh content and depolarization-evoked ACh release in hippocampal slices were normal. However, the amount of choline taken up by CHT1 and ACh synthesized de novo from choline transported by CHT1 in hippocampal slices, as well as levels of CHT1 mRNA in the septum and CHT1 protein in several regions of the CNS, were 50-100% higher in Chat+/- than in Chat+/+ mice. Thus, haploinsufficiency of ChAT leads to an increased expression of CHT1. Increased ChT activity may compensate for the reduced ChAT activity in Chat+/- mice, contributing to the maintenance of apparently normal cholinergic function as reflected by normal performance of these mice in several behavioral assays.


Asunto(s)
Encéfalo/metabolismo , Colina O-Acetiltransferasa/genética , Proteínas de Transporte de Membrana/biosíntesis , Acetilcolina/metabolismo , Animales , Conducta Animal , Transporte Biológico , Colina O-Acetiltransferasa/biosíntesis , Colina O-Acetiltransferasa/deficiencia , Hipocampo/metabolismo , Técnicas In Vitro , Proteínas de Transporte de Membrana/genética , Ratones , Ratones Mutantes , ARN Mensajero/biosíntesis , Tabique del Cerebro/metabolismo , Regulación hacia Arriba
8.
J Neurosci ; 23(2): 539-49, 2003 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-12533614

RESUMEN

In this study we examined the developmental roles of acetylcholine (ACh) by establishing and analyzing mice lacking choline acetyltransferase (ChAT), the biosynthetic enzyme for ACh. As predicted, ChAT-deficient embryos lack both spontaneous and nerve-evoked postsynaptic potentials in muscle and die at birth. In mutant embryos, abnormally increased nerve branching occurs on contact with muscle, and hyperinnervation continues throughout subsequent prenatal development. Postsynaptically, ACh receptor clusters are markedly increased in number and occupy a broader muscle territory in the mutants. Concomitantly, the mutants have significantly more motor neurons than normal. At an ultrastructural level, nerve terminals are smaller in mutant neuromuscular junctions, and they make fewer synaptic contacts to the postsynaptic muscle membrane, although all of the typical synaptic components are present in the mutant. These results indicate that ChAT is uniquely essential for the patterning and formation of mammalian neuromuscular synapses.


Asunto(s)
Colina O-Acetiltransferasa/deficiencia , Enfermedades Neuromusculares/patología , Unión Neuromuscular/patología , Animales , Recuento de Células , Supervivencia Celular , Colina O-Acetiltransferasa/genética , Diafragma/embriología , Diafragma/inervación , Diafragma/patología , Potenciales Postsinápticos Excitadores/genética , Marcación de Gen , Técnicas In Vitro , Ratones , Ratones Endogámicos C57BL , Ratones Mutantes , Neuronas Motoras/patología , Enfermedades Neuromusculares/congénito , Enfermedades Neuromusculares/genética , Unión Neuromuscular/ultraestructura , ARN Mensajero/biosíntesis , Agregación de Receptores , Receptores Colinérgicos/genética , Receptores Colinérgicos/metabolismo , Sinapsis/metabolismo , Sinapsis/ultraestructura , Transmisión Sináptica/genética , Sinaptofisina/biosíntesis
9.
Stem Cells Transl Med ; 4(10): 1214-22, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26304037

RESUMEN

UNLABELLED: The PEC-01 cell population, differentiated from human embryonic stem cells (hESCs), contains pancreatic progenitors (PPs) that, when loaded into macroencapsulation devices (to produce the VC-01 candidate product) and transplanted into mice, can mature into glucose-responsive insulin-secreting cells and other pancreatic endocrine cells involved in glucose metabolism. We modified the protocol for making PEC-01 cells such that 73%-80% of the cell population consisted of PDX1-positive (PDX1+) and NKX6.1+ PPs. The PPs were further differentiated to islet-like cells (ICs) that reproducibly contained 73%-89% endocrine cells, of which approximately 40%-50% expressed insulin. A large fraction of these insulin-positive cells were single hormone-positive and expressed the transcription factors PDX1 and NKX6.1. To preclude a significant contribution of progenitors to the in vivo function of ICs, we used a simple enrichment process to remove remaining PPs, yielding aggregates that contained 93%-98% endocrine cells and 1%-3% progenitors. Enriched ICs, when encapsulated and implanted into mice, functioned similarly to the VC-01 candidate product, demonstrating conclusively that in vitro-produced hESC-derived insulin-producing cells can mature and function in vivo in devices. A scaled version of our suspension culture was used, and the endocrine aggregates could be cryopreserved and retain functionality. Although ICs expressed multiple important ß cell genes, the cells contained relatively low levels of several maturity-associated markers. Correlating with this, the time to function of ICs was similar to PEC-01 cells, indicating that ICs required cell-autonomous maturation after delivery in vivo, which would occur concurrently with graft integration into the host. SIGNIFICANCE: Type 1 diabetes (T1D) affects approximately 1.25 million people in the U.S. alone and is deadly if not managed with insulin injections. This paper describes the production of insulin-producing cells in vitro and a new protocol for producing the cells, representing another potential cell source for a diabetes cell therapy. These cells can be loaded into a protective device that is implanted under the skin. The device is designed to protect the cells from immune rejection by the implant recipient. The implant can engraft and respond to glucose by secreting insulin, thus potentially replacing the ß cells lost in patients with T1D.


Asunto(s)
Células Madre Embrionarias Humanas/citología , Células Secretoras de Insulina/citología , Insulina/biosíntesis , Animales , Biomarcadores , Glucemia/análisis , Diferenciación Celular , Separación Celular/métodos , Células Cultivadas , Células Inmovilizadas/trasplante , Criopreservación , Perfilación de la Expresión Génica , Proteínas de Homeodominio/biosíntesis , Células Madre Embrionarias Humanas/metabolismo , Humanos , Células Secretoras de Insulina/metabolismo , Células Secretoras de Insulina/trasplante , Ratones , Proinsulina/metabolismo , Procesamiento Proteico-Postraduccional , Reproducibilidad de los Resultados , Transactivadores/biosíntesis
10.
Cell Stem Cell ; 12(2): 224-37, 2013 Feb 07.
Artículo en Inglés | MEDLINE | ID: mdl-23318056

RESUMEN

Embryonic development is characterized by dynamic changes in gene expression, yet the role of chromatin remodeling in these cellular transitions remains elusive. To address this question, we profiled the transcriptome and select chromatin modifications at defined stages during pancreatic endocrine differentiation of human embryonic stem cells. We identify removal of Polycomb group (PcG)-mediated repression on stage-specific genes as a key mechanism for the induction of developmental regulators. Furthermore, we discover that silencing of transitory genes during lineage progression associates with reinstatement of PcG-dependent repression. Significantly, in vivo- but not in vitro-differentiated endocrine cells exhibit close similarity to primary human islets in regard to transcriptome and chromatin structure. We further demonstrate that endocrine cells produced in vitro do not fully eliminate PcG-mediated repression on endocrine-specific genes, probably contributing to their malfunction. These studies reveal dynamic chromatin remodeling during developmental lineage progression and identify possible strategies for improving cell differentiation in culture.


Asunto(s)
Ensamble y Desensamble de Cromatina/fisiología , Células Madre Embrionarias/citología , Páncreas/citología , Proteínas del Grupo Polycomb/metabolismo , Animales , Diferenciación Celular/genética , Diferenciación Celular/fisiología , Ensamble y Desensamble de Cromatina/genética , Células Madre Embrionarias/metabolismo , Células Endocrinas/citología , Células Endocrinas/metabolismo , Endodermo/citología , Endodermo/metabolismo , Humanos , Ratones , Proteínas del Grupo Polycomb/genética
11.
PLoS One ; 7(5): e37004, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22623968

RESUMEN

Development of a human embryonic stem cell (hESC)-based therapy for type 1 diabetes will require the translation of proof-of-principle concepts into a scalable, controlled, and regulated cell manufacturing process. We have previously demonstrated that hESC can be directed to differentiate into pancreatic progenitors that mature into functional glucose-responsive, insulin-secreting cells in vivo. In this study we describe hESC expansion and banking methods and a suspension-based differentiation system, which together underpin an integrated scalable manufacturing process for producing pancreatic progenitors. This system has been optimized for the CyT49 cell line. Accordingly, qualified large-scale single-cell master and working cGMP cell banks of CyT49 have been generated to provide a virtually unlimited starting resource for manufacturing. Upon thaw from these banks, we expanded CyT49 for two weeks in an adherent culture format that achieves 50-100 fold expansion per week. Undifferentiated CyT49 were then aggregated into clusters in dynamic rotational suspension culture, followed by differentiation en masse for two weeks with a four-stage protocol. Numerous scaled differentiation runs generated reproducible and defined population compositions highly enriched for pancreatic cell lineages, as shown by examining mRNA expression at each stage of differentiation and flow cytometry of the final population. Islet-like tissue containing glucose-responsive, insulin-secreting cells was generated upon implantation into mice. By four- to five-months post-engraftment, mature neo-pancreatic tissue was sufficient to protect against streptozotocin (STZ)-induced hyperglycemia. In summary, we have developed a tractable manufacturing process for the generation of functional pancreatic progenitors from hESC on a scale amenable to clinical entry.


Asunto(s)
Técnicas de Cultivo Celular por Lotes/métodos , Diferenciación Celular/fisiología , Diabetes Mellitus Tipo 1/terapia , Células Madre Embrionarias/citología , Células Madre Embrionarias/trasplante , Células Secretoras de Insulina/citología , Análisis de Varianza , Animales , Criopreservación/métodos , Células Madre Embrionarias/fisiología , Citometría de Flujo , Técnica del Anticuerpo Fluorescente , Perfilación de la Expresión Génica , Humanos , Masculino , Ratones , Ratones SCID , Estreptozocina
12.
Nat Biotechnol ; 29(8): 750-6, 2011 Jul 31.
Artículo en Inglés | MEDLINE | ID: mdl-21804561

RESUMEN

Using a flow cytometry-based screen of commercial antibodies, we have identified cell-surface markers for the separation of pancreatic cell types derived from human embryonic stem (hES) cells. We show enrichment of pancreatic endoderm cells using CD142 and of endocrine cells using CD200 and CD318. After transplantation into mice, enriched pancreatic endoderm cells give rise to all the pancreatic lineages, including functional insulin-producing cells, demonstrating that they are pancreatic progenitors. In contrast, implanted, enriched polyhormonal endocrine cells principally give rise to glucagon cells. These antibodies will aid investigations that use pancreatic cells generated from pluripotent stem cells to study diabetes and pancreas biology.


Asunto(s)
Antígenos CD/metabolismo , Biomarcadores/metabolismo , Separación Celular/métodos , Células Madre Embrionarias/citología , Páncreas/citología , Animales , Anticuerpos/metabolismo , Células Cultivadas , Células Madre Embrionarias/metabolismo , Endodermo/citología , Citometría de Flujo , Humanos , Ratones , Ratones SCID , Microscopía Fluorescente , Trasplante Heterólogo
13.
Stem Cell Res ; 3(2-3): 73-87, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-19766074

RESUMEN

The potential of pluripotent human cells, such as human embryonic stem cells (hESCs) and induced pluripotent stem (iPS) cells, to differentiate into any adult cell type makes them ideally suited for the generation of various somatic cells and tissues in vitro. This remarkable differentiation capacity permits analyzing aspects of human embryonic development in the laboratory, as well as generating specialized adult human cells for screening drugs, and for replacing tissues damaged by injury or degenerative diseases, such as diabetes. Understanding and controlling the fundamental processes that drive the differentiation of specialized cells are the keys to the eventual application of this technology to patients. In this review, we discuss the different protocols developed that are aimed at deriving beta-cells from hESCs. Despite many differences, successful strategies share a general adherence to the normal differentiation pathway through definitive endoderm. Mimicking normal pancreagenesis offers the best strategy for producing glucose-responsive insulin-producing cells in vitro for people with diabetes.


Asunto(s)
Células Madre Embrionarias/citología , Células Secretoras de Insulina/citología , Diferenciación Celular , Diabetes Mellitus/etiología , Diabetes Mellitus/terapia , Endodermo/citología , Humanos , Insulina/genética , Insulina/metabolismo , Factores de Transcripción/metabolismo
14.
Nat Biotechnol ; 26(4): 443-52, 2008 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-18288110

RESUMEN

Development of a cell therapy for diabetes would be greatly aided by a renewable supply of human beta-cells. Here we show that pancreatic endoderm derived from human embryonic stem (hES) cells efficiently generates glucose-responsive endocrine cells after implantation into mice. Upon glucose stimulation of the implanted mice, human insulin and C-peptide are detected in sera at levels similar to those of mice transplanted with approximately 3,000 human islets. Moreover, the insulin-expressing cells generated after engraftment exhibit many properties of functional beta-cells, including expression of critical beta-cell transcription factors, appropriate processing of proinsulin and the presence of mature endocrine secretory granules. Finally, in a test of therapeutic potential, we demonstrate that implantation of hES cell-derived pancreatic endoderm protects against streptozotocin-induced hyperglycemia. Together, these data provide definitive evidence that hES cells are competent to generate glucose-responsive, insulin-secreting cells.


Asunto(s)
Técnicas de Cultivo de Célula/tendencias , Células Madre Embrionarias/citología , Glucosa/metabolismo , Células Secretoras de Insulina/citología , Células Secretoras de Insulina/metabolismo , Insulina/metabolismo , Ingeniería de Tejidos/tendencias , Animales , Diferenciación Celular , Proliferación Celular , Células Cultivadas , Células Madre Embrionarias/metabolismo , Células Madre Embrionarias/trasplante , Endodermo/citología , Endodermo/metabolismo , Humanos , Células Secretoras de Insulina/trasplante , Ratones , Páncreas Artificial/tendencias
15.
Cell Stem Cell ; 1(5): 515-28, 2007 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-18371391

RESUMEN

To characterize the properties of adult neural stem cells (NSCs), we generated and analyzed Sox2-GFP transgenic mice. Sox2-GFP cells in the subgranular zone (SGZ) express markers specific for progenitors, but they represent two morphologically distinct populations that differ in proliferation levels. Lentivirus- and retrovirus-mediated fate-tracing studies showed that Sox2+ cells in the SGZ have potential to give rise to neurons and astrocytes, revealing their multipotency at the population as well as at a single-cell level. A subpopulation of Sox2+ cells gives rise to cells that retain Sox2, highlighting Sox2+ cells as a primary source for adult NSCs. In response to mitotic signals, increased proliferation of Sox2+ cells is coupled with the generation of Sox2+ NSCs as well as neuronal precursors. An asymmetric contribution of Sox2+ NSCs may play an important role in maintaining the constant size of the NSC pool and producing newly born neurons during adult neurogenesis.


Asunto(s)
Células Madre Adultas/metabolismo , Astrocitos/metabolismo , Linaje de la Célula , Proliferación Celular , Proteínas de Unión al ADN/metabolismo , Giro Dentado/metabolismo , Proteínas HMGB/metabolismo , Células Madre Multipotentes/metabolismo , Neuronas/metabolismo , Factores de Transcripción/metabolismo , Animales , Diferenciación Celular , Forma de la Célula , Células Cultivadas , Proteínas de Unión al ADN/genética , Giro Dentado/citología , Técnicas de Transferencia de Gen , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Proteínas HMGB/genética , Lentivirus/genética , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Microscopía Fluorescente , Esfuerzo Físico , Regiones Promotoras Genéticas , Retroviridae/genética , Factores de Transcripción SOXB1 , Factores de Tiempo , Factores de Transcripción/genética
16.
Stem Cells ; 25(1): 29-38, 2007 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-17204604

RESUMEN

Human ESCs (hESCs) respond to signals that determine their pluripotency, proliferation, survival, and differentiation status. In this report, we demonstrate that phosphatidylinositol 3-kinase (PI3K) antagonizes the ability of hESCs to differentiate in response to transforming growth factor beta family members such as Activin A and Nodal. Inhibition of PI3K signaling efficiently promotes differentiation of hESCs into mesendoderm and then definitive endoderm (DE) by allowing them to be specified by Activin/Nodal signals present in hESC cultures. Under conditions where hESCs are grown in mouse embryo fibroblast-conditioned medium under feeder-free conditions, approximately 70%-80% are converted into DE following 5 days of treatment with inhibitors of the PI3K pathway, such as LY 294002 and AKT1-II. Microarray and quantitative polymerase chain reaction-based gene expression profiling demonstrates that definitive endoderm formation under these conditions closely parallels that following specification with elevated Activin A and low fetal calf serum (FCS)/knockout serum replacement (KSR). Reduced insulin/insulin-like growth factor (IGF) signaling was found to be critical for cell fate commitment into DE. Levels of insulin/IGF present in FCS/KSR, normally used to promote self-renewal of hESCs, antagonized differentiation. In summary, we show that generation of hESC-DE requires two conditions: signaling by Activin/Nodal family members and release from inhibitory signals generated by PI3K through insulin/IGF. These findings have important implications for our understanding of hESC self-renewal and early cell fate decisions.


Asunto(s)
Activinas/fisiología , Diferenciación Celular/fisiología , Células Madre Embrionarias/citología , Células Madre Embrionarias/fisiología , Endodermo/fisiología , Fosfatidilinositol 3-Quinasas/fisiología , Animales , Medios de Cultivo Condicionados , Endodermo/citología , Citometría de Flujo , Perfilación de la Expresión Génica , Humanos , Ratones , Análisis de Secuencia por Matrices de Oligonucleótidos , Inhibidores de las Quinasa Fosfoinosítidos-3 , Transducción de Señal , Ensayo de Capsula Subrrenal , Transcripción Genética
17.
Blood ; 110(12): 4111-9, 2007 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-17761519

RESUMEN

Despite progress in developing defined conditions for human embryonic stem cell (hESC) cultures, little is known about the cell-surface receptors that are activated under conditions supportive of hESC self-renewal. A simultaneous interrogation of 42 receptor tyrosine kinases (RTKs) in hESCs following stimulation with mouse embryonic fibroblast (MEF) conditioned medium (CM) revealed rapid and prominent tyrosine phosphorylation of insulin receptor (IR) and insulin-like growth factor-1 receptor (IGF1R); less prominent tyrosine phosphorylation of epidermal growth factor receptor (EGFR) family members, including ERBB2 and ERBB3; and trace phosphorylation of fibroblast growth factor receptors. Intense IGF1R and IR phosphorylation occurred in the absence of MEF conditioning (NCM) and was attributable to high concentrations of insulin in the proprietary KnockOut Serum Replacer (KSR). Inhibition of IGF1R using a blocking antibody or lentivirus-delivered shRNA reduced hESC self-renewal and promoted differentiation, while disruption of ERBB2 signaling with the selective inhibitor AG825 severely inhibited hESC proliferation and promoted apoptosis. A simple defined medium containing an IGF1 analog, heregulin-1beta (a ligand for ERBB2/ERBB3), fibroblast growth factor-2 (FGF2), and activin A supported long-term growth of multiple hESC lines. These studies identify previously unappreciated RTKs that support hESC proliferation and self-renewal, and provide a rationally designed medium for the growth and maintenance of pluripotent hESCs.


Asunto(s)
Proliferación Celular , Células Madre Embrionarias/metabolismo , Células Madre Pluripotentes/metabolismo , Receptor ErbB-2/metabolismo , Receptor IGF Tipo 2/metabolismo , Transducción de Señal/fisiología , Animales , Anticuerpos Monoclonales/farmacología , Apoptosis/efectos de los fármacos , Apoptosis/fisiología , Benzotiazoles/farmacología , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/fisiología , Línea Celular , Proliferación Celular/efectos de los fármacos , Medios de Cultivo Condicionados , Células Madre Embrionarias/citología , Factor 2 de Crecimiento de Fibroblastos/farmacología , Fibroblastos/citología , Fibroblastos/metabolismo , Humanos , Ratones , Neurregulina-1/farmacología , Fosforilación/efectos de los fármacos , Células Madre Pluripotentes/citología , Receptor ErbB-2/antagonistas & inhibidores , Receptor ErbB-3/antagonistas & inhibidores , Receptor ErbB-3/metabolismo , Receptor IGF Tipo 2/antagonistas & inhibidores , Receptor de Insulina/antagonistas & inhibidores , Receptor de Insulina/metabolismo , Transducción de Señal/efectos de los fármacos , Tirfostinos/farmacología
18.
Proc Natl Acad Sci U S A ; 100 Suppl 1: 11866-72, 2003 Sep 30.
Artículo en Inglés | MEDLINE | ID: mdl-12923297

RESUMEN

Stem cells (SCs) are functionally defined by their abilities to self-renew and generate differentiated cells. Although much effort has been focused on defining the common characteristics among various types of SCs, the genetic and functional differences between multipotent and pluripotent SCs have garnered less attention. We report a direct genetic and functional comparison of molecularly defined and clonally related populations of neural SCs (NSCs) and embryonic SCs (ESCs), using the Sox2 promoter for isolation of purified populations by fluorescence-activated cell sorting. A stringent expression profile comparison of promoter-defined NSCs and ESCs revealed a striking dissimilarity, and subsequent chimera analyses confirmed the fundamental differences in cellular potency between these populations. This direct comparison elucidates the molecular basis for the functional differences in pluripotent ESCs and multipotent NSCs.


Asunto(s)
Células Madre Multipotentes/citología , Células Madre Multipotentes/fisiología , Neuronas/química , Neuronas/fisiología , Células Madre Pluripotentes/citología , Células Madre Pluripotentes/fisiología , Animales , Secuencia de Bases , ADN Complementario/genética , Proteínas de Unión al ADN/genética , Perfilación de la Expresión Génica , Proteínas Fluorescentes Verdes , Proteínas HMGB , Proteínas Luminiscentes/genética , Ratones , Ratones Transgénicos , Proteínas Nucleares/genética , Regiones Promotoras Genéticas , Proteínas Recombinantes de Fusión/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factores de Transcripción SOXB1 , Factores de Transcripción
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA