Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
1.
Development ; 143(10): 1721-31, 2016 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-26965372

RESUMEN

LIM homeodomain factors regulate the development of many cell types. However, transcriptional coactivators that mediate their developmental function remain poorly defined. To address these, we examined how two related NLI-dependent LIM complexes, which govern the development of spinal motor neurons and V2a interneurons, activate the transcription in the embryonic spinal cord. We found that single-stranded DNA-binding proteins are recruited to these LIM complexes via NLI, and enhance their transcriptional activation potential. Ssdp1 and Ssdp2 (Ssdp1/2) are highly expressed in the neural tube and promote motor neuron differentiation in the embryonic spinal cord and P19 stem cells. Inhibition of Ssdp1/2 activity in mouse and chick embryos suppresses the generation of motor neurons and V2a interneurons. Furthermore, Ssdp1/2 recruit histone-modifying enzymes to the motor neuron-specifying LIM complex and trigger acetylation and lysine 4 trimethylation of histone H3, which are well-established chromatin marks for active transcription. Our results suggest that Ssdp1/2 function as crucial transcriptional coactivators for LIM complexes to specify spinal neuronal identities during development.


Asunto(s)
Cromatina/metabolismo , Proteínas de Unión al ADN/metabolismo , Proteínas con Homeodominio LIM/metabolismo , Neuronas/metabolismo , Médula Espinal/citología , Activación Transcripcional/genética , Secuencia de Aminoácidos , Animales , Tipificación del Cuerpo , Embrión de Pollo , Proteínas de Unión al ADN/química , Proteínas de Unión al ADN/genética , Embrión de Mamíferos/metabolismo , Humanos , Interneuronas/metabolismo , Ratones , Neuronas Motoras/citología , Neuronas Motoras/metabolismo , Mutación/genética , Neuronas/citología , Unión Proteica/genética , Ratas , Médula Espinal/embriología , Médula Espinal/metabolismo , Transactivadores/metabolismo
2.
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
3.
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
4.
Mol Cell Biol ; 23(21): 7585-99, 2003 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-14560005

RESUMEN

The TAL1 (or SCL) gene, originally identified from its involvement by a recurrent chromosomal translocation, encodes a basic helix-loop-helix transcription factor essential for erythropoiesis. Although presumed to regulate transcription, its target genes are largely unknown. We show here that a nuclear complex containing TAL1, its DNA-binding partner E47, zinc finger transcription factor GATA-1, LIM domain protein LMO2, and LIM domain-binding protein Ldb1 transactivates the protein 4.2 (P4.2) gene through two E box GATA elements in its proximal promoter. Binding of this complex to DNA was dependent on the integrity of both E box and GATA sites and was demonstrated to occur on the P4.2 promoter in cells. Maximal transcription in transiently transfected cells required both E box GATA elements and expression of all five components of the complex. This complex was shown, in addition, to be capable of linking in solution double-stranded oligonucleotides corresponding to the two P4.2 E box GATA elements. This DNA-linking activity required Ldb1 and increased with dimethyl sulfoxide-induced differentiation of murine erythroleukemia (MEL) cells. In contrast, enforced expression in MEL cells of dimerization-defective mutant Ldb1, as well as wild-type Ldb1, significantly decreased E box GATA DNA-binding activities, P4.2 promoter activity, and accumulation of P4.2 and beta-globin mRNAs. These studies define a physiologic target for a TAL1- and GATA-1-containing ternary complex and reveal a positive role for Ldb1 in erythroid gene expression and differentiation.


Asunto(s)
Proteínas Sanguíneas/metabolismo , Diferenciación Celular/fisiología , Proteínas del Citoesqueleto/metabolismo , Proteínas de Unión al ADN/metabolismo , Regulación de la Expresión Génica , Proteínas Proto-Oncogénicas/metabolismo , Factores de Transcripción/metabolismo , Transcripción Genética , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico , Proteínas del Citoesqueleto/genética , Proteínas de Unión al ADN/genética , Eritrocitos/metabolismo , Factores de Unión al ADN Específico de las Células Eritroides , Factor de Transcripción GATA1 , Genes Reguladores , Secuencias Hélice-Asa-Hélice , Proteínas con Dominio LIM , Leucemia Eritroblástica Aguda , Proteínas de la Membrana , Ratones , Regiones Promotoras Genéticas , Proteínas Proto-Oncogénicas/genética , Proteína 1 de la Leucemia Linfocítica T Aguda , Factores de Transcripción/genética , Células Tumorales Cultivadas
5.
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
6.
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
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.
Development ; 130(9): 1915-25, 2003 May.
Artículo en Inglés | MEDLINE | ID: mdl-12642495

RESUMEN

LIM-homeodomain transcription factors control a variety of developmental processes, and are assembled into functional complexes with the LIM-binding co-factor Ldb1 (in mouse) or Chip (in Drosophila). We describe the identification and characterization of members of the Ssdp family of proteins, which we show to interact with Ldb1 and Chip. The N terminus of Ssdp is highly conserved among species and binds a highly conserved domain within Ldb1/Chip that is distinct from the domains required for LIM binding and self-dimerization. In Drosophila, Ssdp is expressed in the developing nervous system and imaginal tissues, and it is capable of modifying the in vivo activity of complexes comprised of Chip and the LIM-homeodomain protein Apterous. Null mutations of the ssdp gene are cell-lethal in clones of cells within the developing wing disc. However, clones mutant for a hypomorphic allele give rise to ectopic margins, wing outgrowth and cell identity defects similar to those produced by mutant clones of Chip or apterous. Ssdp and Ldb/Chip each show structural similarity to two Arabidopsis proteins that cooperate with one another to regulate gene expression during flower development, suggesting that the molecular interactions between Ssdp and Ldb/Chip proteins are evolutionarily ancient and supply a fundamental function in the regulated control of transcription.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Proteínas de Homeodominio/metabolismo , Proteínas Nucleares/metabolismo , Animales , Proteínas de Unión al ADN/genética , Drosophila/embriología , Drosophila/metabolismo , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Proteínas de Homeodominio/genética , Proteínas con Dominio LIM , Proteínas con Homeodominio LIM , Ratones , Mutación , Sistema Nervioso/embriología , Estructura Terciaria de Proteína , Análisis de Secuencia de ADN , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Alas de Animales/anomalías , Alas de Animales/embriología
9.
Development ; 130(3): 495-505, 2003 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-12490556

RESUMEN

The LIM domain-binding protein 1 (Ldb1) is found in multi-protein complexes containing various combinations of LIM-homeodomain, LIM-only, bHLH, GATA and Otx transcription factors. These proteins exert key functions during embryogenesis. Here we show that targeted deletion of the Ldb1 gene in mice results in a pleiotropic phenotype. There is no heart anlage and head structures are truncated anterior to the hindbrain. In about 40% of the mutants, posterior axis duplication is observed. There are also severe defects in mesoderm-derived extraembryonic structures, including the allantois, blood islands of the yolk sack, primordial germ cells and the amnion. Abnormal organizer gene expression during gastrulation may account for the observed axis defects in Ldb1 mutant embryos. The expression of several Wnt inhibitors is curtailed in the mutant, suggesting that Wnt pathways may be involved in axial patterning regulated by Ldb1.


Asunto(s)
Tipificación del Cuerpo/genética , Proteínas de Unión al ADN/deficiencia , Proteínas de Unión al ADN/genética , Gástrula/citología , Proteínas de Pez Cebra , Animales , Secuencia de Bases , Tipificación del Cuerpo/fisiología , ADN/genética , Proteínas de Unión al ADN/fisiología , Femenino , Corazón Fetal/embriología , Gástrula/metabolismo , Marcación de Gen , Proteínas con Dominio LIM , Masculino , Mesodermo/citología , Ratones , Ratones Noqueados , Oocitos/metabolismo , Fenotipo , Embarazo , Proteínas Proto-Oncogénicas/antagonistas & inhibidores , Proteínas Proto-Oncogénicas/fisiología , ARN Mensajero/genética , ARN Mensajero/metabolismo , Proteínas Wnt
10.
Proc Natl Acad Sci U S A ; 99(14): 9521-6, 2002 Jul 09.
Artículo en Inglés | MEDLINE | ID: mdl-12093930

RESUMEN

Persephin (Pspn), a recently cloned member of the transforming growth factor-beta superfamily (TGF-beta) and glial cell line-derived neurotrophic factor (GDNF) subfamily, is distributed throughout the nervous system at extremely low levels and is thought to function as a survival factor for midbrain dopaminergic and spinal motor neurons in vivo. Here, we report that mice lacking Pspn by homologous recombination show normal development and behavior, but are hypersensitive to cerebral ischemia. A 300% increase in infarction volume was observed after middle cerebral artery occlusion. We find that glutamate-induced Ca(2+) influx, thought to be a major component of ischemic neuronal cell death, can be regulated directly by the Persephin protein (PSP) and that PSP can reduce hypoxia/reperfusion cell death in vitro. Neuronal cell death can be prevented or markedly attenuated by administration of recombinant human PSP in vivo before ischemia in both mouse and rat models. Taken together, these data indicate that PSP is a potent modulator of excitotoxicity in the central nervous system with pronounced neuroprotective activity. Our findings support the view that PSP signaling can exert an important control function in the context of stroke and glutamate-mediated neurotoxicity, and also suggest that future therapeutic approaches may involve this novel trophic protein.


Asunto(s)
Isquemia Encefálica/etiología , Factores de Crecimiento Nervioso/deficiencia , Proteínas del Tejido Nervioso/deficiencia , Animales , Conducta Animal , Isquemia Encefálica/patología , Isquemia Encefálica/fisiopatología , Isquemia Encefálica/prevención & control , Señalización del Calcio/efectos de los fármacos , Muerte Celular/efectos de los fármacos , Ácido Glutámico/farmacología , Humanos , Técnicas In Vitro , Ratones , Ratones Noqueados , Factores de Crecimiento Nervioso/genética , Factores de Crecimiento Nervioso/fisiología , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/farmacología , Proteínas del Tejido Nervioso/fisiología , Ratas , Proteínas Recombinantes/farmacología , Accidente Cerebrovascular/etiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA