Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
1.
Clin Sci (Lond) ; 118(2): 87-97, 2009 Oct 12.
Artículo en Inglés | MEDLINE | ID: mdl-19807695

RESUMEN

With the already heightened demand placed on organ donation, stem cell therapy has become a tantalizing idea to provide glucose-responsive insulin-producing cells to Type 1 diabetic patients as an alternative to islet transplantation. Multiple groups have developed varied approaches to create a population of cells with the appropriate characteristics. Both adult and embryonic stem cells have received an enormous amount of attention as possible sources of insulin-producing cells. Although adult stem cells lack the pluripotent nature of their embryonic counterparts, they appear to avoid the ethical debate that has centred around the latter. This may limit the eventual application of embryonic stem cells, which have already shown promise in early mouse models. One must also consider the potential of stem cells to form teratomas, a complication which would prove devastating in an immunologically compromised transplant recipient. The present review looks at the progress to date in both the adult and embryonic stem cells fields as potential treatments for diabetes. We also consider some of the limitations of stem cell therapy and the potential complications that may develop with their use.


Asunto(s)
Diabetes Mellitus Tipo 1/terapia , Trasplante de Células Madre/métodos , Adulto , Células Madre Adultas/trasplante , Células Madre Embrionarias/trasplante , Trasplante de Células Madre Hematopoyéticas/métodos , Humanos , Células Secretoras de Insulina/trasplante , Trasplante de Células Madre/efectos adversos , Trasplante de Células Madre/tendencias
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.
Artículo en Inglés | MEDLINE | ID: mdl-26764480

RESUMEN

Nutritional research is entering a paradigm shift which necessitates the modeling of complex interactions between diet, genetics, lifestyle, and environmental factors. This requires the development of analytical and processing capabilities for multiple data and information sources to be able to improve targeted and personalized nutritional approaches for the maintenance of health. Ideally, such knowledge will be employed to underpin the development of concepts that combine consumer and medical nutrition with diagnostic targeting for early intervention designed to maintain proper metabolic homeostasis and delay the onset of chronic diseases. Nutritional status is fundamental to any description of health, and when combined with other data on lifestyle, environment, and genetics, it can be used to drive stratified or even personalized nutritional strategies for health maintenance and preventive medicine. In this work, we will discuss the importance of developing new nutrient assessment methods and diagnostic capabilities for nutritional status to generate scientific hypotheses and actionable concepts from which to develop targeted and eventually personalized nutritional solutions for health protection. We describe efforts to develop algorithms for dietary nutrient intake and a holistic nutritional profiling platform as the basis of understanding the complex nutrition and health interactome.


Asunto(s)
Dieta , Salud , Estado Nutricional , Algoritmos , Biomarcadores , Humanos
5.
Cell Stem Cell ; 16(2): 148-57, 2015 Feb 05.
Artículo en Inglés | MEDLINE | ID: mdl-25533131

RESUMEN

Type 1 diabetes (T1D) is an autoimmune disease caused by T cell-mediated destruction of insulin-producing ß cells in the islets of Langerhans. In most cases, reversal of disease would require strategies combining islet cell replacement with immunotherapy that are currently available only for the most severely affected patients. Here, we demonstrate that immunotherapies that target T cell costimulatory pathways block the rejection of xenogeneic human embryonic-stem-cell-derived pancreatic endoderm (hESC-PE) in mice. The therapy allowed for long-term development of hESC-PE into islet-like structures capable of producing human insulin and maintaining normoglycemia. Moreover, short-term costimulation blockade led to robust immune tolerance that could be transferred independently of regulatory T cells. Importantly, costimulation blockade prevented the rejection of allogeneic hESC-PE by human PBMCs in a humanized model in vivo. These results support the clinical development of hESC-derived therapy, combined with tolerogenic treatments, as a sustainable alternative strategy for patients with T1D.


Asunto(s)
Diabetes Mellitus Tipo 1/terapia , Endodermo/citología , Endodermo/inmunología , Células Madre Embrionarias Humanas/trasplante , Tolerancia Inmunológica/inmunología , Páncreas/citología , Linfocitos T Reguladores/inmunología , Animales , Diabetes Mellitus Tipo 1/patología , Humanos , Ratones , Ratones Endogámicos NOD , Ratones Noqueados , Ratones SCID , Páncreas/inmunología , Trasplante Heterólogo
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.
Artículo en Inglés | MEDLINE | ID: mdl-26764481
8.
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
9.
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
10.
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
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA