Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Nat Med ; 21(11): 1364-71, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26501191

RESUMO

There are few in vitro models of exocrine pancreas development and primary human pancreatic adenocarcinoma (PDAC). We establish three-dimensional culture conditions to induce the differentiation of human pluripotent stem cells into exocrine progenitor organoids that form ductal and acinar structures in culture and in vivo. Expression of mutant KRAS or TP53 in progenitor organoids induces mutation-specific phenotypes in culture and in vivo. Expression of TP53(R175H) induces cytosolic SOX9 localization. In patient tumors bearing TP53 mutations, SOX9 was cytoplasmic and associated with mortality. We also define culture conditions for clonal generation of tumor organoids from freshly resected PDAC. Tumor organoids maintain the differentiation status, histoarchitecture and phenotypic heterogeneity of the primary tumor and retain patient-specific physiological changes, including hypoxia, oxygen consumption, epigenetic marks and differences in sensitivity to inhibition of the histone methyltransferase EZH2. Thus, pancreatic progenitor organoids and tumor organoids can be used to model PDAC and for drug screening to identify precision therapy strategies.


Assuntos
Antimetabólitos Antineoplásicos/farmacologia , Carcinoma Ductal Pancreático/tratamento farmacológico , Desoxicitidina/análogos & derivados , Organoides/efeitos dos fármacos , Pâncreas/efeitos dos fármacos , Neoplasias Pancreáticas/tratamento farmacológico , Células-Tronco Pluripotentes , Animais , Carcinoma Ductal Pancreático/genética , Desoxicitidina/farmacologia , Ensaios de Seleção de Medicamentos Antitumorais/métodos , Humanos , Camundongos , Modelos Biológicos , Mutação , Organoides/patologia , Organoides/ultraestrutura , Pâncreas/patologia , Pâncreas/ultraestrutura , Neoplasias Pancreáticas/genética , Proteínas Proto-Oncogênicas/genética , Proteínas Proto-Oncogênicas p21(ras) , Fatores de Transcrição SOX9/metabolismo , Técnicas de Cultura de Tecidos , Proteína Supressora de Tumor p53/genética , Proteínas ras/genética , Gencitabina
2.
Development ; 142(24): 4253-65, 2015 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-26493401

RESUMO

The efficient generation of hepatocytes from human pluripotent stem cells (hPSCs) requires the induction of a proper endoderm population, broadly characterized by the expression of the cell surface marker CXCR4. Strategies to identify and isolate endoderm subpopulations predisposed to the liver fate do not exist. In this study, we generated mouse monoclonal antibodies against human embryonic stem cell-derived definitive endoderm with the goal of identifying cell surface markers that can be used to track the development of this germ layer and its specification to a hepatic fate. Through this approach, we identified two endoderm-specific antibodies, HDE1 and HDE2, which stain different stages of endoderm development and distinct derivative cell types. HDE1 marks a definitive endoderm population with high hepatic potential, whereas staining of HDE2 tracks with developing hepatocyte progenitors and hepatocytes. When used in combination, the staining patterns of these antibodies enable one to optimize endoderm induction and hepatic specification from any hPSC line.


Assuntos
Biomarcadores/metabolismo , Diferenciação Celular , Endoderma/citologia , Hepatócitos/citologia , Células-Tronco Pluripotentes/citologia , Animais , Anticorpos/metabolismo , Linhagem Celular , Separação Celular , Ectoderma/citologia , Hepatócitos/metabolismo , Células-Tronco Embrionárias Humanas/citologia , Células-Tronco Embrionárias Humanas/metabolismo , Humanos , Cinética , Mesoderma/citologia , Camundongos Endogâmicos BALB C , Pâncreas/embriologia , Células-Tronco Pluripotentes/metabolismo , Coloração e Rotulagem
3.
Acta Biomater ; 22: 103-10, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-25913222

RESUMO

This study aimed to develop a three dimensional culture platform for aggregates of human embryonic stem cell (hESC)-derived pancreatic progenitors that enables long-term culture, maintains aggregate size and morphology, does not adversely affect differentiation and provides a means for aggregate recovery. A platform was developed with poly(ethylene glycol) hydrogels containing collagen type I, for cell-matrix interactions, and peptide crosslinkers, for facile recovery of aggregates. The platform was first demonstrated with RIN-m5F cells, showing encapsulation and subsequent release of single cells and aggregates without adversely affecting viability. Aggregates of hESC-derived pancreatic progenitors with an effective diameter of 82 (15)µm were either encapsulated in hydrogels or cultured in suspension for 28 days. At day 14, aggregate viability was maintained in the hydrogels, but significantly reduced (88%) in suspension culture. However by day 28, viability was reduced under both culture conditions. Aggregate size was maintained in the hydrogels, but in suspension was significantly higher (∼ 2-fold) by day 28. The ability to release aggregates followed by a second enzyme treatment to achieve single cells enabled assessment by flow cytometry. Prior to encapsulation, there were 39% Pdx1(+)/Nkx6.1(+) cells, key endocrine markers required for ß-cell maturation. The fraction of doubly positive cells was not affected in hydrogels but was slightly and significantly lower in suspension culture by 28 days. In conclusion, we demonstrate that a MMP-sensitive PEG hydrogel containing collagen type I is a promising platform for hESC-derived pancreatic progenitors that maintains viable aggregates, aggregate size, and progenitor state and offers facile recovery of aggregates.


Assuntos
Técnicas de Cultura de Células/métodos , Enzimas/metabolismo , Células-Tronco Embrionárias Humanas/citologia , Hidrogéis/química , Pâncreas/citologia , Polietilenoglicóis/química , Sequência de Aminoácidos , Animais , Agregação Celular , Linhagem Celular Tumoral , Tamanho Celular , Sobrevivência Celular , Citometria de Fluxo , Humanos , Camundongos , Microscopia Confocal , Dados de Sequência Molecular , Peptídeos/química , Polimerização , Ratos , Fatores de Transcrição/metabolismo
4.
PLoS One ; 7(10): e46844, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23056483

RESUMO

Using the zebrafish model we describe a previously unrecognized requirement for the transcription factor gata4 controlling embryonic angiogenesis. The development of a vascular plexus in the embryonic tail, the caudal hematopoietic tissue (CHT), fails in embryos depleted of gata4. Rather than forming a normal vascular plexus, the CHT of gata4 morphants remains fused, and cells in the CHT express high levels of osteogenic markers ssp1 and runx1. Definitive progenitors emerge from the hemogenic aortic endothelium, but fail to colonize the poorly vascularized CHT. We also found abnormal patterns and levels for the chemokine sdf1a in gata4 morphants, which was found to be functionally relevant, since the embryos also show defects in development of the lateral line, a mechano-sensory organ system highly dependent on a gradient of sdf1a levels. Reduction of sdf1a levels was sufficient to rescue lateral line development, circulation, and CHT morphology. The result was surprising since neither gata4 nor sdf1a is obviously expressed in the CHT. Therefore, we generated transgenic fish that conditionally express a dominant-negative gata4 isoform, and determined that gata4 function is required during gastrulation, when it is co-expressed with sdf1a in lateral mesoderm. Our study shows that the gata4 gene regulates sdf1a levels during early embryogenesis, which impacts embryonic patterning and subsequently the development of the caudal vascular plexus.


Assuntos
Vasos Sanguíneos/metabolismo , Quimiocina CXCL12/metabolismo , Fator de Transcrição GATA4/metabolismo , Peixe-Zebra/metabolismo , Animais , Vasos Sanguíneos/citologia , Vasos Sanguíneos/fisiologia , Quimiocina CXCL12/deficiência , Quimiocina CXCL12/genética , Embrião não Mamífero/citologia , Embrião não Mamífero/embriologia , Embrião não Mamífero/metabolismo , Fator de Transcrição GATA4/deficiência , Gastrulação , Regulação da Expressão Gênica no Desenvolvimento , Técnicas de Silenciamento de Genes , Hematopoese , Neovascularização Fisiológica , Transdução de Sinais , Peixe-Zebra/embriologia , Proteínas de Peixe-Zebra/deficiência , Proteínas de Peixe-Zebra/genética , Proteínas de Peixe-Zebra/metabolismo
5.
Development ; 138(5): 861-71, 2011 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-21270052

RESUMO

The generation of insulin-producing ß-cells from human pluripotent stem cells is dependent on efficient endoderm induction and appropriate patterning and specification of this germ layer to a pancreatic fate. In this study, we elucidated the temporal requirements for TGFß family members and canonical WNT signaling at these developmental stages and show that the duration of nodal/activin A signaling plays a pivotal role in establishing an appropriate definitive endoderm population for specification to the pancreatic lineage. WNT signaling was found to induce a posterior endoderm fate and at optimal concentrations enhanced the development of pancreatic lineage cells. Inhibition of the BMP signaling pathway at specific stages was essential for the generation of insulin-expressing cells and the extent of BMP inhibition required varied widely among the cell lines tested. Optimal stage-specific manipulation of these pathways resulted in a striking 250-fold increase in the levels of insulin expression and yielded populations containing up to 25% C-peptide+ cells.


Assuntos
Células Secretoras de Insulina/citologia , Pâncreas/citologia , Células-Tronco Pluripotentes/fisiologia , Fator de Crescimento Transformador beta/metabolismo , Proteínas Wnt/metabolismo , Ativinas/metabolismo , Padronização Corporal , Proteínas Morfogenéticas Ósseas/antagonistas & inibidores , Peptídeo C , Linhagem Celular , Linhagem da Célula , Endoderma , Humanos , Insulina/biossíntese , Transdução de Sinais/fisiologia
6.
Dev Biol ; 337(1): 63-73, 2010 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-19850025

RESUMO

The transcription factor Gata4 is essential for normal heart morphogenesis and regulates the survival, growth, and proliferation of cardiomyocytes. We tested if Gata4 can specify cardiomyocyte fate from an uncommitted stem or progenitor cell population, by developing a system for conditional expression of Gata4 in embryonic stem cells. We find that in embryoid body cultures containing even a low ratio of these cells, expression of Gata4 is sufficient to enhance significantly the generation of cardiomyocytes, via a non-cell-autonomous mechanism. The Gata4-expressing cells do not generate cardiac or other mesoderm derivatives. Rather, Gata4 expression directs the development of two types of Sox17+ endoderm. This includes an epCam+Dpp4+ subtype of visceral endoderm. In addition, Gata4 generates similar amounts of epCam+Dpp4- definitive endoderm enriched for Cxcr4, FoxA2, FoxA3, Dlx5 and other characteristic transcripts. Both types of endoderm express cardiac-inducing factors, including WNT antagonists Dkk1 and Sfrp5, although the visceral endoderm subtype has much higher cardiac-inducing activity correlating with relatively enhanced levels of transcripts encoding BMPs. The Gata4-expressing cells eventually express differentiation markers showing commitment to liver development, even under conditions that normally support mesoderm development. The results suggest that Gata4 is capable of specifying endoderm fates that facilitate, with temporal and spatial specificity, the generation of cardiomyocyte progenitors from associated mesoderm.


Assuntos
Células-Tronco Embrionárias/citologia , Endoderma/citologia , Fator de Transcrição GATA4/fisiologia , Coração/embriologia , Animais , Proteínas Morfogenéticas Ósseas/fisiologia , Diferenciação Celular , Linhagem Celular , Peptídeos e Proteínas de Sinalização Intercelular/fisiologia , Fígado/embriologia , Camundongos , Transdução de Sinais , Proteínas Wnt/antagonistas & inibidores , Proteínas Wnt/fisiologia
7.
Dev Biol ; 312(2): 613-22, 2007 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-17950269

RESUMO

An outstanding problem in vertebrate development has been to define the genetic program that specifies the cardiomyocyte lineage. It has been a challenge to define the transcription factors that control specification, since candidate gene knockouts typically cause rather complex morphogenetic defects. In contrast, Drosophila genetics identified single transcription factors that are essential for specification of cardiomyocytes from uncommitted mesoderm. For those vertebrate orthologs, it has been considered that paralogous family members might compensate for the loss-of-function of individual genes. However, this hypothesis had not been formally tested. In zebrafish, defects in gata5 can lead to a loss of myocardial tissue, but most embryos depleted for any single vertebrate Gata4/5/6 transcription factor develop a cardiac morphogenetic defect, and cardiomyocytes are specified and differentiate. Here we show that in zebrafish the gata5 and gata6 genes are redundant for specification of cardiomyocytes. Embryos depleted of these two gene products are heartless. Restoring either gene product is sufficient to rescue cardiomyocyte specification. In contrast, embryos depleted of Gata4 and Gata6, or Gata4 and Gata5, develop defective heart tubes. Our study identifies a specific pair of vertebrate transcription factor paralogs that is essential for cardiomyocyte specification.


Assuntos
Fatores de Transcrição GATA/fisiologia , Fator de Transcrição GATA5/fisiologia , Fator de Transcrição GATA6/fisiologia , Miócitos Cardíacos/metabolismo , Proteínas de Peixe-Zebra/fisiologia , Animais , Diferenciação Celular/genética , Embrião não Mamífero/metabolismo , Fatores de Transcrição GATA/genética , Fatores de Transcrição GATA/metabolismo , Fator de Transcrição GATA4/genética , Fator de Transcrição GATA4/metabolismo , Fator de Transcrição GATA5/genética , Fator de Transcrição GATA5/metabolismo , Fator de Transcrição GATA6/genética , Fator de Transcrição GATA6/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Fenótipo , Peixe-Zebra/embriologia , Peixe-Zebra/genética , Proteínas de Peixe-Zebra/genética , Proteínas de Peixe-Zebra/metabolismo
8.
Development ; 132(17): 4005-14, 2005 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-16079152

RESUMO

We have developed a loss-of-function model for Gata4 in zebrafish, in order to examine broadly its requirement for organogenesis. We show that the function of Gata4 in zebrafish heart development is well conserved with that in mouse, and that, in addition, Gata4 is required for development of the intestine, liver, pancreas and swim bladder. Therefore, a single transcription factor regulates the formation of many organs. Gata6 is a closely related transcription factor with an overlapping expression pattern. We show that zebrafish depleted of Gata6 show defects in liver bud growth similar to mouse Gata6 mutants and zebrafish Gata4 morphants, and that zebrafish embryos depleted of both Gata4 and Gata6 display an earlier block in liver development, and thus completely lack liver buds. Therefore, Gata4 and Gata6 have distinct non-redundant functions in cardiac morphogenesis, but are redundant for an early step of liver development. In addition, both Gata4 and Gata6 are essential and non-redundant for liver growth following initial budding.


Assuntos
Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Organogênese , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Peixe-Zebra/embriologia , Peixe-Zebra/metabolismo , Animais , Proteínas de Ligação a DNA/deficiência , Embrião não Mamífero/embriologia , Embrião não Mamífero/metabolismo , Fator de Transcrição GATA4 , Fator de Transcrição GATA6 , Trato Gastrointestinal/embriologia , Trato Gastrointestinal/metabolismo , Expressão Gênica , Coração/embriologia , Modelos Biológicos , Miocárdio/metabolismo , Organogênese/genética , Fatores de Transcrição/deficiência , Peixe-Zebra/anatomia & histologia , Peixe-Zebra/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...