Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Stem Cell Reports ; 12(4): 787-800, 2019 04 09.
Artículo en Inglés | MEDLINE | ID: mdl-30853374

RESUMEN

In type 1 diabetes, a renewable source of human pancreatic ß cells, in particular from human induced pluripotent stem cell (hiPSC) origin, would greatly benefit cell therapy. Earlier work showed that pancreatic progenitors differentiated from human embryonic stem cells in vitro can further mature to become glucose responsive following macroencapsulation and transplantation in mice. Here we took a similar approach optimizing the generation of pancreatic progenitors from hiPSCs. This work demonstrates that hiPSCs differentiated to pancreatic endoderm in vitro can be efficiently and robustly generated under large-scale conditions. The hiPSC-derived pancreatic endoderm cells (HiPECs) can further differentiate into glucose-responsive islet-like cells following macroencapsulation and in vivo implantation. The HiPECs can protect mice from streptozotocin-induced hyperglycemia and maintain normal glucose homeostasis and equilibrated plasma glucose concentrations at levels similar to the human set point. These results further validate the potential use of hiPSC-derived islet cells for application in clinical settings.


Asunto(s)
Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Experimental/terapia , Células Madre Pluripotentes Inducidas/citología , Células Secretoras de Insulina/citología , Células Secretoras de Insulina/metabolismo , Trasplante de Células Madre , Animales , Biomarcadores , Glucemia , Péptido C/sangre , Diferenciación Celular , Diabetes Mellitus Experimental/sangre , Diabetes Mellitus Experimental/etiología , Modelos Animales de Enfermedad , Endodermo/citología , Técnica del Anticuerpo Fluorescente , Humanos , Hiperglucemia/etiología , Hiperglucemia/metabolismo , Hiperglucemia/terapia , Inmunofenotipificación , Insulina/biosíntesis , Ratones , Modelos Biológicos , Resultado del Tratamiento
2.
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
3.
PLoS One ; 7(12): e52181, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-23251699

RESUMEN

The peptide hormone Urocortin 3 (Ucn 3) is abundantly and exclusively expressed in mouse pancreatic beta cells where it regulates insulin secretion. Here we demonstrate that Ucn 3 first appears at embryonic day (E) 17.5 and, from approximately postnatal day (p) 7 and onwards throughout adult life, becomes a unifying and exclusive feature of mouse beta cells. These observations identify Ucn 3 as a potential beta cell maturation marker. To determine whether Ucn 3 is similarly restricted to beta cells in humans, we conducted comprehensive immunohistochemistry and gene expression experiments on macaque and human pancreas and sorted primary human islet cells. This revealed that Ucn 3 is not restricted to the beta cell lineage in primates, but is also expressed in alpha cells. To substantiate these findings, we analyzed human embryonic stem cell (hESC)-derived pancreatic endoderm that differentiates into mature endocrine cells upon engraftment in mice. Ucn 3 expression in hESC-derived grafts increased robustly upon differentiation into mature endocrine cells and localized to both alpha and beta cells. Collectively, these observations confirm that Ucn 3 is expressed in adult beta cells in both mouse and human and appears late in beta cell differentiation. Expression of Pdx1, Nkx6.1 and PC1/3 in hESC-derived Ucn 3(+) beta cells supports this. However, the expression of Ucn 3 in primary and hESC-derived alpha cells demonstrates that human Ucn 3 is not exclusive to the beta cell lineage but is a general marker for both the alpha and beta cell lineages. Ucn 3(+) hESC-derived alpha cells do not express Nkx6.1, Pdx1 or PC1/3 in agreement with the presence of a separate population of Ucn 3(+) alpha cells. Our study highlights important species differences in Ucn 3 expression, which have implications for its utility as a marker to identify mature beta cells in (re)programming strategies.


Asunto(s)
Células Madre Embrionarias/metabolismo , Células Secretoras de Glucagón/metabolismo , Células Secretoras de Insulina/metabolismo , Urocortinas/metabolismo , Animales , Diferenciación Celular/genética , Linaje de la Célula , Células Madre Embrionarias/citología , Expresión Génica , Células Secretoras de Glucagón/citología , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Humanos , Inmunohistoquímica/métodos , Células Secretoras de Insulina/citología , Islotes Pancreáticos/citología , Islotes Pancreáticos/metabolismo , Ratones , Transactivadores/genética , Transactivadores/metabolismo , Urocortinas/genética
4.
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
5.
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
6.
Anat Rec (Hoboken) ; 292(3): 333-41, 2009 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-19248165

RESUMEN

The development of the cardiovascular system is a highly dynamic process dependent on multiple signaling pathways regulating proliferation, differentiation, migration, cell-cell and cell-matrix interactions. To characterize cell and tissue dynamics during the formation of the cardiovascular system in mice, we generated a novel transgenic mouse line, Tg(Flk1::myr-mCherry), in which endothelial cell membranes are brightly labeled with mCherry, a red fluorescent protein. Tg(Flk1::myr-mCherry) mice are viable, fertile, and do not exhibit any developmental abnormalities. High levels of mCherry are expressed in the embryonic endothelium and endocardium, and expression is also observed in capillaries in adult animals. Targeting of the fluorescent protein to the cell membrane allows for subcellular imaging and cell tracking. By acquiring confocal time lapses of live embryos cultured on the microscope stage, we demonstrate that the newly generated transgenic model beautifully highlights the sprouting behaviors of endothelial cells during vascular plexus formation. We have also used embryos from this line to imaging the endocardium in the beating embryonic mouse heart, showing that Tg(Flk1::myr-mCherry) mice are suitable for the characterization of cardio dynamics. Furthermore, when combined with the previously described Tg(Flk1::H2B-EYFP) line, cell number in addition to cell architecture is revealed, making it possible to determine how individual endothelial cells contribute to the structure of the vessel.


Asunto(s)
Vasos Sanguíneos/embriología , Sistema Cardiovascular/embriología , Membrana Celular/metabolismo , Embrión de Mamíferos/irrigación sanguínea , Células Endoteliales/metabolismo , Proteínas Luminiscentes/metabolismo , Animales , Vasos Sanguíneos/metabolismo , Sistema Cardiovascular/metabolismo , Núcleo Celular , Embrión de Mamíferos/metabolismo , Endocardio , Endotelio Vascular , Células HeLa , Humanos , Ratones , Ratones Transgénicos , Receptor 2 de Factores de Crecimiento Endotelial Vascular/fisiología , Proteína Fluorescente Roja
7.
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
8.
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
9.
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
10.
Genesis ; 42(3): 162-71, 2005 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-15986455

RESUMEN

We report the first endothelial lineage-specific transgenic mouse allowing live imaging at subcellular resolution. We generated an H2B-EYFP fusion protein which can be used for fluorescent labeling of nucleosomes and used it to specifically label endothelial cells in mice and in differentiating embryonic stem (ES) cells. A fusion cDNA encoding a human histone H2B tagged at its C-terminus with enhanced yellow fluorescent protein (EYFP) was expressed under the control of an Flk1 promoter and intronic enhancer. The Flk1::H2B-EYFP transgenic mice are viable and high levels of chromatin-localized reporter expression are maintained in endothelial cells of developing embryos and in adult animals upon breeding. The onset of fluorescence in differentiating ES cells and in embryos corresponds with the beginning of endothelial cell specification. These transgenic lines permit real-time imaging in normal and pathological vasculogenesis and angiogenesis to track individual cells and mitotic events at a level of detail that is unprecedented in the mouse.


Asunto(s)
Proteínas Bacterianas/metabolismo , Células Endoteliales/metabolismo , Histonas/metabolismo , Proteínas Luminiscentes/metabolismo , Células Madre/metabolismo , Envejecimiento/genética , Animales , Proteínas Bacterianas/genética , Diferenciación Celular , Núcleo Celular/metabolismo , Embrión de Mamíferos/irrigación sanguínea , Embrión de Mamíferos/metabolismo , Células Endoteliales/citología , Regulación del Desarrollo de la Expresión Génica , Genes Reporteros/genética , Histonas/genética , Proteínas Luminiscentes/genética , Ratones , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Células Madre/citología , Transgenes/genética
11.
Dev Cell ; 6(6): 801-13, 2004 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-15177029

RESUMEN

Exostosin1 (Ext1) belongs to a family of glycosyltransferases necessary for the synthesis of the heparan sulfate (HS) chains of proteoglycans, which regulate signaling of several growth factors. Loss of tout velu (ttv), the homolog of Ext1 in Drosophila, inhibits Hedgehog movement. In contrast, we show that reduced HS synthesis in mice carrying a hypomorphic mutation in Ext1 results in an elevated range of Indian hedgehog (Ihh) signaling during embryonic chondrocyte differentiation. Our data suggest a dual function for HS: First, HS is necessary to bind Hedgehog in the extracellular space. Second, HS negatively regulates the range of Hedgehog signaling in a concentration-dependent manner. Additionally, our data indicate that Ihh acts as a long-range morphogen, directly activating the expression of parathyroid hormone-like hormone. Finally, we propose that the development of exostoses in the human Hereditary Multiple Exostoses syndrome can be attributed to activation of Ihh signaling.


Asunto(s)
Huesos/embriología , Huesos/metabolismo , Heparitina Sulfato/metabolismo , N-Acetilglucosaminiltransferasas/metabolismo , Osteogénesis/genética , Transactivadores/metabolismo , Animales , Huesos/citología , Diferenciación Celular/genética , Condrocitos/citología , Condrocitos/metabolismo , Exostosis Múltiple Hereditaria/genética , Exostosis Múltiple Hereditaria/metabolismo , Exostosis Múltiple Hereditaria/fisiopatología , Factores de Crecimiento de Fibroblastos/genética , Factores de Crecimiento de Fibroblastos/metabolismo , Regulación del Desarrollo de la Expresión Génica/genética , Proteínas Hedgehog , Heparitina Sulfato/genética , Ratones , Ratones Transgénicos , Mutación/genética , N-Acetilglucosaminiltransferasas/genética , Proteína Relacionada con la Hormona Paratiroidea/genética , Proteína Relacionada con la Hormona Paratiroidea/metabolismo , Transducción de Señal/genética , Transactivadores/genética , Regulación hacia Arriba/genética
12.
Development ; 131(12): 2803-15, 2004 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-15142971

RESUMEN

Recent work has identified LDL receptor-related family members, Lrp5 and Lrp6, as co-receptors for the transduction of Wnt signals. Our analysis of mice carrying mutations in both Lrp5 and Lrp6 demonstrates that the functions of these genes are redundant and are essential for gastrulation. Lrp5;Lrp6 double homozygous mutants fail to establish a primitive streak, although the anterior visceral endoderm and anterior epiblast fates are specified. Thus, Lrp5 and Lrp6 are required for posterior patterning of the epiblast, consistent with a role in transducing Wnt signals in the early embryo. Interestingly, Lrp5(+/-);Lrp6(-/-) embryos die shortly after gastrulation and exhibit an accumulation of cells at the primitive streak and a selective loss of paraxial mesoderm. A similar phenotype is observed in Fgf8 and Fgfr1 mutant embryos and provides genetic evidence in support of a molecular link between the Fgf and Wnt signaling pathways in patterning nascent mesoderm. Lrp5(+/-);Lrp6(-/-) embryos also display an expansion of anterior primitive streak derivatives and anterior neurectoderm that correlates with increased Nodal expression in these embryos. The effect of reducing, but not eliminating, Wnt signaling in Lrp5(+/-);Lrp6(-/-) mutant embryos provides important insight into the interplay between Wnt, Fgf and Nodal signals in patterning the early mouse embryo.


Asunto(s)
Gástrula/fisiología , Proteínas Proto-Oncogénicas/fisiología , Receptores de LDL/fisiología , Animales , Secuencia de Bases , Tipificación del Cuerpo/genética , Diferenciación Celular , Cartilla de ADN , Endodermo/citología , Endodermo/fisiología , Factores de Crecimiento de Fibroblastos/fisiología , Regulación del Desarrollo de la Expresión Génica , Genes Letales , Proteínas Relacionadas con Receptor de LDL , Proteína-5 Relacionada con Receptor de Lipoproteína de Baja Densidad , Proteína-6 Relacionada a Receptor de Lipoproteína de Baja Densidad , Mesodermo/citología , Mesodermo/fisiología , Ratones , Ratones Noqueados , Mutación , Receptores de LDL/deficiencia , Receptores de LDL/genética , Proteínas Wnt
13.
J Biol Chem ; 279(31): 32134-41, 2004 Jul 30.
Artículo en Inglés | MEDLINE | ID: mdl-15161920

RESUMEN

Mutational defects in either EXT1 or EXT2 genes cause multiple exostoses, an autosomal hereditary human disorder. The EXT1 and EXT2 genes encode glycosyltransferases that play an essential role in heparan sulfate chain elongation. In this study, we have analyzed heparan sulfate synthesized by primary fibroblast cell cultures established from mice with a gene trap mutation in Ext1. The gene trap mutation results in embryonic lethality, and homozygous mice die around embryonic day 14. Metabolic labeling and immunohistochemistry revealed that Ext1 mutant fibroblasts still produced small amounts of heparan sulfate. The domain structure of the mutant heparan sulfate was conserved, and the disaccharide composition was similar to that of wild type heparan sulfate. However, a dramatic difference was seen in the polysaccharide chain length. The average molecular sizes of the heparan sulfate chains from wild type and Ext1 mutant embryonic fibroblasts were estimated to be around 70 and 20 kDa, respectively. These data suggest that not only the sulfation pattern but also the length of the heparan sulfate chains is a critical determinant of normal mouse development.


Asunto(s)
Técnicas Genéticas , Heparitina Sulfato/metabolismo , Mutación , N-Acetilglucosaminiltransferasas/genética , N-Acetilglucosaminiltransferasas/metabolismo , Animales , Aniones , Northern Blotting , Cromatografía en Gel , Cromatografía Líquida de Alta Presión , Análisis Mutacional de ADN , Disacáridos/química , Fibroblastos/metabolismo , Sustancias de Crecimiento/metabolismo , Heparitina Sulfato/química , Homocigoto , Inmunohistoquímica , Ratones , Modelos Biológicos , Modelos Genéticos , Mutagénesis , Ácido Nitroso/farmacología , Polisacáridos/química , Estructura Terciaria de Proteína , ARN Mensajero/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal , Factores de Tiempo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...