Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
1.
Cytotherapy ; 20(3): 461-476, 2018 03.
Artigo em Inglês | MEDLINE | ID: mdl-29398624

RESUMO

A summary of the First Signature Series Event, "Advancements in Cellular Therapies and Regenerative Medicine for Digestive Diseases," held on May 3, 2017, in London, United Kingdom, is presented. Twelve speakers from three continents covered major topics in the areas of cellular therapy and regenerative medicine applied to liver and gastrointestinal medicine as well as to diabetes mellitus. Highlights from their presentations, together with an overview of the global impact of digestive diseases and a proposal for a shared online collection and data-monitoring platform tool, are included in this proceedings. Although growing evidence demonstrate the feasibility and safety of exploiting cell-based technologies for the treatment of digestive diseases, regulatory and methodological obstacles will need to be overcome before the successful implementation in the clinic of these novel attractive therapeutic strategies.


Assuntos
Terapia Baseada em Transplante de Células e Tecidos/métodos , Gastroenteropatias/terapia , Medicina Regenerativa/métodos , Terapia Baseada em Transplante de Células e Tecidos/tendências , Diabetes Mellitus/patologia , Diabetes Mellitus/terapia , Gastroenteropatias/patologia , Humanos , Hepatopatias/patologia , Hepatopatias/terapia , Medicina Regenerativa/tendências
2.
Proc Natl Acad Sci U S A ; 110(36): 14723-8, 2013 Sep 03.
Artigo em Inglês | MEDLINE | ID: mdl-23946427

RESUMO

Pancreatic endocrine cells expand rapidly during embryogenesis by neogenesis and proliferation, but during adulthood, islet cells have a very slow turnover. Disruption of murine retinoblastoma tumor suppressor protein (Rb) in mature pancreatic ß-cells has a limited effect on cell proliferation. Here we show that deletion of Rb during embryogenesis in islet progenitors leads to an increase in the neurogenin 3-expressing precursor cell population, which persists in the postnatal period and is associated with increased ß-cell mass in adults. In contrast, Rb-deficient islet precursors, through repression of the cell fate factor aristaless related homeobox, result in decreased α-cell mass. The opposing effect on survival of Rb-deficient α- and ß-cells was a result of opposing effects on p53 in these cell types. As a consequence, loss of Rb in islet precursors led to a reduced α- to ß-cell ratio, leading to improved glucose homeostasis and protection against diabetes.


Assuntos
Células Secretoras de Glucagon/metabolismo , Células Secretoras de Insulina/metabolismo , Ilhotas Pancreáticas/metabolismo , Proteína do Retinoblastoma/metabolismo , Células-Tronco/metabolismo , Animais , Animais Recém-Nascidos , Sequência de Bases , Diferenciação Celular/genética , Linhagem Celular Tumoral , Sobrevivência Celular/genética , Diabetes Mellitus Experimental/genética , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Experimental/patologia , Fator de Transcrição E2F1/genética , Fator de Transcrição E2F1/metabolismo , Feminino , Células Secretoras de Glucagon/citologia , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/metabolismo , Imuno-Histoquímica , Células Secretoras de Insulina/citologia , Ilhotas Pancreáticas/citologia , Masculino , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Interferência de RNA , Proteína do Retinoblastoma/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Homologia de Sequência do Ácido Nucleico , Células-Tronco/citologia , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Proteína Supressora de Tumor p53/genética , Proteína Supressora de Tumor p53/metabolismo
3.
J Biol Chem ; 289(25): 17854-71, 2014 Jun 20.
Artigo em Inglês | MEDLINE | ID: mdl-24798330

RESUMO

The TGFß family member Nodal is central to control pluripotent stem cell fate, but its use as a stem cell differentiation factor is limited by low specific activity. During development, Nodal depends on growth and differentiation factor (Gdf)-1 and on the shared co-receptor Cryptic to specify visceral left-right axis asymmetry. We therefore asked whether the functionality of Nodal can be augmented by Gdf1. Because Nodal and Gdf1 coimmunoprecipitate each other, they were predicted to form heterodimers, possibly to facilitate diffusion or to increase the affinity for signaling receptors. Here, we report that Gdf1 suppresses an unexpected dependence of Nodal on serum proteins and that it is critically required for non-autonomous signaling in cells expressing Cryptic. Nodal, Gdf1, and their cleaved propeptides copurified as a heterodimeric low molecular weight complex that stimulated Activin receptor (Acvr) signaling far more potently than Nodal alone. Although heterodimerization with Gdf1 did not increase binding of Nodal to Fc fusions of co-receptors or Acvr extracellular domains, it was essential for soluble Acvr2 to inhibit Nodal signaling. This implies that Gdf1 potentiates Nodal activity by stabilizing a low molecular weight fraction that is susceptible to neutralization by soluble Acvr2. Finally, in differentiating human ES cells, endodermal markers were more efficiently induced by Nodal·Gdf1 than by Nodal, suggesting that Nodal·Gdf1 is an attractive new reagent to direct stem cell differentiation.


Assuntos
Diferenciação Celular/fisiologia , Embrião de Mamíferos/metabolismo , Células-Tronco Embrionárias/metabolismo , Endoderma/metabolismo , Fator 1 de Diferenciação de Crescimento/metabolismo , Proteína Nodal/metabolismo , Multimerização Proteica/fisiologia , Transdução de Sinais/fisiologia , Receptores de Activinas Tipo II/genética , Receptores de Activinas Tipo II/metabolismo , Animais , Embrião de Mamíferos/citologia , Células-Tronco Embrionárias/citologia , Endoderma/citologia , Fator 1 de Diferenciação de Crescimento/genética , Células HEK293 , Células Hep G2 , Humanos , Camundongos , Camundongos Knockout , Proteína Nodal/genética , Estrutura Terciária de Proteína
4.
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
5.
Biotechnol Bioeng ; 109(4): 853-66, 2012 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-22139975

RESUMO

We present a predictive bioprocess design strategy employing cell- and molecular-level analysis of rate-limiting steps in human pluripotent stem cell (hPSC) expansion and differentiation, and apply it to produce definitive endoderm (DE) progenitors using a scalable directed-differentiation technology. We define a bioprocess optimization parameter (L; targeted cell Loss) and, with quantitative cell division tracking and fate monitoring, identify and overcome key suspension bioprocess bottlenecks. Adapting process operating conditions to pivotal parameters (single cell survival and growth rate) in a cell-line-specific manner enabled adherent-equivalent expansion of hPSCs in feeder- and matrix-free defined-medium suspension culture. Predominantly instructive differentiation mechanisms were found to underlie a subsequent 18-fold expansion, during directed differentiation, to high-purity DE competent for further commitment along pancreatic and hepatic lineages. This study demonstrates that iPSC expansion and differentiation conditions can be prospectively specified to guide the enhanced production of target cells in a scale-free directed differentiation system.


Assuntos
Técnicas de Cultura Celular por Lotes , Endoderma/citologia , Células-Tronco Pluripotentes/citologia , Engenharia Tecidual/métodos , Animais , Antígenos de Diferenciação/análise , Agregação Celular , Ciclo Celular , Diferenciação Celular , Divisão Celular , Linhagem Celular/citologia , Linhagem Celular/efeitos dos fármacos , Técnicas de Cocultura , Meios de Cultura , Fibroblastos/metabolismo , Hepatócitos/citologia , Humanos , Camundongos , Pâncreas/citologia , Suspensões
6.
Stem Cell Reports ; 17(4): 964-978, 2022 04 12.
Artigo em Inglês | MEDLINE | ID: mdl-35364010

RESUMO

Human pluripotent stem cell (hPSC)-derived pancreatic progenitors (PPs) can be differentiated into beta-like cells in vitro and in vivo and therefore have therapeutic potential for type 1 diabetes (T1D) treatment. However, the purity of PPs varies across different hPSC lines, differentiation protocols, and laboratories. The uncommitted cells may give rise to non-pancreatic endodermal, mesodermal, or ectodermal derivatives in vivo, hampering the safety of hPSC-derived PPs for clinical applications and their differentiation efficiency in research settings. Recently, proteomics and transcriptomics analyses identified glycoprotein 2 (GP2) as a PP-specific cell surface marker. The GP2-enriched PPs generate higher percentages of beta-like cells in vitro, but their potential in vivo remains to be elucidated. Here, we demonstrate that the GP2-enriched-PPs give rise to all pancreatic cells in vivo, including functional beta-like cells. Remarkably, GP2 enrichment eliminates the risk of teratomas, which establishes GP2 sorting as an effective method for PP purification and safe pancreatic differentiation.


Assuntos
Células Secretoras de Insulina , Células-Tronco Pluripotentes , Teratoma , Diferenciação Celular/fisiologia , Endoderma , Humanos , Células Secretoras de Insulina/metabolismo , Pâncreas , Células-Tronco Pluripotentes/metabolismo , Teratoma/etiologia , Teratoma/metabolismo
7.
Cell Stem Cell ; 28(11): 1936-1949.e8, 2021 11 04.
Artigo em Inglês | MEDLINE | ID: mdl-34480863

RESUMO

Islet transplantation is a promising treatment for type 1 diabetes (T1D), yet the low donor pool, poor islet engraftment, and life-long immunosuppression prevent it from becoming the standard of care. Human embryonic stem cell (hESC)-derived pancreatic cells could eliminate donor shortages, but interventions to improve graft survival are needed. Here, we enhanced subcutaneous engraftment by employing a unique vascularization strategy based on ready-made microvessels (MVs) isolated from the adipose tissue. This resulted in improved cell survival and effective glucose response of both human islets and hESC-derived pancreatic cells, which ameliorated preexisting diabetes in three mouse models of T1D.


Assuntos
Diabetes Mellitus Tipo 1 , Células-Tronco Embrionárias Humanas , Transplante das Ilhotas Pancreáticas , Ilhotas Pancreáticas , Animais , Diabetes Mellitus Tipo 1/terapia , Humanos , Camundongos , Microvasos
8.
Cell Stem Cell ; 26(4): 471-473, 2020 04 02.
Artigo em Inglês | MEDLINE | ID: mdl-32243804

RESUMO

The existence of an endocrine progenitor in the adult mouse pancreas has been controversial. Recently in Cell, Wang et al. (2020) use the cell-surface marker Procr to define a population of cells within the adult islet of Langerhans that is capable of generating all endocrine cells and is amenable to in vitro expansion.


Assuntos
Ilhotas Pancreáticas , Organoides , Animais , Diferenciação Celular , Receptor de Proteína C Endotelial , Camundongos , Pâncreas
9.
Nat Commun ; 10(1): 4647, 2019 10 11.
Artigo em Inglês | MEDLINE | ID: mdl-31604927

RESUMO

Human embryonic stem cell-derived beta cells offer a promising cell-based therapy for diabetes. However, efficient stem cell to beta cell differentiation has proven difficult, possibly due to the lack of cross-talk with the appropriate mesenchymal niche. To define organ-specific niche signals, we isolated pancreatic and gastrointestinal stromal cells, and analyzed their gene expression during development. Our genetic studies reveal the importance of tightly regulated Hedgehog signaling in the pancreatic mesenchyme: inactivation of mesenchymal signaling leads to annular pancreas, whereas stroma-specific activation of signaling via loss of Hedgehog regulators, Sufu and Spop, impairs pancreatic growth and beta cell genesis. Genetic rescue and transcriptome analyses show that these Sufu and Spop knockout defects occur through Gli2-mediated activation of gastrointestinal stromal signals such as Wnt ligands. Importantly, inhibition of Wnt signaling in organoid and human stem cell cultures significantly promotes insulin-producing cell generation, altogether revealing the requirement for organ-specific regulation of stromal niche signals.


Assuntos
Células-Tronco Embrionárias/citologia , Proteínas Hedgehog/metabolismo , Células Secretoras de Insulina/citologia , Proteínas Nucleares/fisiologia , Proteínas Repressoras/fisiologia , Técnicas de Cultura de Células , Diferenciação Celular , Terapia Baseada em Transplante de Células e Tecidos/métodos , Diabetes Mellitus/terapia , Regulação para Baixo , Humanos , Células Secretoras de Insulina/transplante , Proteínas Nucleares/metabolismo , Organoides/citologia , Proteínas Repressoras/metabolismo , Proteínas Wnt/metabolismo
10.
J Mol Endocrinol ; 61(1): 1-12, 2018 07.
Artigo em Inglês | MEDLINE | ID: mdl-29678908

RESUMO

Pancreatic ß-cell Tcf7l2 deletion or its functional knockdown suggested the essential role of this Wnt pathway effector in controlling insulin secretion, glucose homeostasis and ß-cell gene expression. As the LIM homeodomain protein ISL1 is a suggested Wnt pathway downstream target, we hypothesize that it mediates metabolic functions of TCF7L2. We aimed to determine the role of ISL1 in mediating the function of TCF7L2 and the incretin hormone GLP-1 in pancreatic ß-cells. The effect of dominant negative TCF7L2 (TCF7L2DN) mediated Wnt pathway functional knockdown on Isl1 expression was determined in ßTCFDN mouse islets and in the rat insulinoma cell line INS-1 832/13. Luciferase reporter assay and chromatin immunoprecipitation were utilized to determine whether Isl1 is a direct downstream target of Tcf7l2 TCF7L2DN adenovirus infection and siRNA-mediated Isl1 knockdown on ß-cell gene expression were compared. Furthermore, Isl1 knockdown on GLP-1 stimulated ß-catenin S675 phosphorylation and insulin secretion was determined. We found that TCF7L2DN repressed ISL1 levels in ßTCFDN islets and the INS-1 832/13 cell line. Wnt stimulators enhanced Isl1 promoter activity and binding of TCF7L2 on Isl1 promoter. TCF7L2DN adenovirus infection and Isl1 knockdown generated similar repression on expression of ß-cell genes, including the ones that encode GLUT2 and GLP-1 receptor. Either TCF7L2DN adenovirus infection or Isl1 knockdown attenuated GLP-1-stimulated ß-catenin S675 phosphorylation in INS-1 832/13 cells or mouse islets and GLP-1 stimulated insulin secretion in INS-1 832/13 or MIN6 cells. Our observations support the existence of TCF7L2-ISL1 transcriptional network, and we suggest that this network also mediates ß-cell function of GLP-1.


Assuntos
Células Secretoras de Insulina/efeitos dos fármacos , Células Secretoras de Insulina/metabolismo , Proteínas com Homeodomínio LIM/metabolismo , Proteína 2 Semelhante ao Fator 7 de Transcrição/metabolismo , Animais , Linhagem Celular , Imunoprecipitação da Cromatina , Peptídeo 1 Semelhante ao Glucagon/farmacologia , Insulina/metabolismo , Proteínas com Homeodomínio LIM/genética , Masculino , Camundongos , Fosforilação/efeitos dos fármacos , Regiões Promotoras Genéticas/genética , Ligação Proteica , Ratos , Proteína 2 Semelhante ao Fator 7 de Transcrição/genética
11.
J Vis Exp ; (121)2017 03 07.
Artigo em Inglês | MEDLINE | ID: mdl-28362406

RESUMO

Pluripotent stem cells have the ability to self renew and differentiate to multiple lineages, making them an attractive source for the generation of pancreatic progenitor cells that can be used for the study of and future treatment of diabetes. This article outlines a four-stage differentiation protocol designed to generate pancreatic progenitor cells from human embryonic stem cells (hESCs). This protocol can be applied to a number of human pluripotent stem cell (hPSC) lines. The approach taken to generate pancreatic progenitor cells is to differentiate hESCs to accurately model key stages of pancreatic development. This begins with the induction of the definitive endoderm, which is achieved by culturing the cells in the presence of Activin A, basic Fibroblast Growth Factor (bFGF) and CHIR990210. Further differentiation and patterning with Fibroblast Growth Factor 10 (FGF10) and Dorsomorphin generates cells resembling the posterior foregut. The addition of Retinoic Acid, NOGGIN, SANT-1 and FGF10 differentiates posterior foregut cells into cells characteristic of pancreatic endoderm. Finally, the combination of Epidermal Growth Factor (EGF), Nicotinamide and NOGGIN leads to the efficient generation of PDX1+/NKX6-1+ cells. Flow cytometry is performed to confirm the expression of specific markers at key stages of pancreatic development. The PDX1+/NKX6-1+ pancreatic progenitors at the end of stage 4 are capable of generating mature ß cells upon transplantation into immunodeficient mice and can be further differentiated to generate insulin-producing cells in vitro. Thus, the efficient generation of PDX1+/NKX6-1+ pancreatic progenitors, as demonstrated in this protocol, is of great importance as it provides a platform to study human pancreatic development in vitro and provides a source of cells with the potential of differentiating to ß cells that could eventually be used for the treatment of diabetes.


Assuntos
Células-Tronco Embrionárias/citologia , Proteínas de Homeodomínio/biossíntese , Células Secretoras de Insulina/citologia , Pâncreas/embriologia , Células-Tronco Pluripotentes/citologia , Animais , Diferenciação Celular , Linhagem Celular , Células-Tronco Embrionárias/metabolismo , Citometria de Fluxo , Humanos , Células Secretoras de Insulina/metabolismo , Camundongos , Organogênese , Células-Tronco Pluripotentes/metabolismo
12.
Nat Commun ; 8(1): 331, 2017 08 24.
Artigo em Inglês | MEDLINE | ID: mdl-28835709

RESUMO

PDX1+/NKX6-1+ pancreatic progenitors (PPs) give rise to endocrine cells both in vitro and in vivo. This cell population can be successfully differentiated from human pluripotent stem cells (hPSCs) and hold the potential to generate an unlimited supply of ß cells for diabetes treatment. However, the efficiency of PP generation in vitro is highly variable, negatively impacting reproducibility and validation of in vitro and in vivo studies, and consequently, translation to the clinic. Here, we report the use of a proteomics approach to phenotypically characterize hPSC-derived PPs and distinguish these cells from non-PP populations during differentiation. Our analysis identifies the pancreatic secretory granule membrane major glycoprotein 2 (GP2) as a PP-specific cell surface marker. Remarkably, GP2 is co-expressed with NKX6-1 and PTF1A in human developing pancreata, indicating that it marks the multipotent pancreatic progenitors in vivo. Finally, we show that isolated hPSC-derived GP2+ cells generate ß-like cells (C-PEPTIDE+/NKX6-1+) more efficiently compared to GP2- and unsorted populations, underlining the potential therapeutic applications of GP2.Pancreatic progenitors (PPs) can be derived from human pluripotent stem cells in vitro but efficiency of differentiation varies, making it hard to sort for insulin-producing cells. Here, the authors use a proteomic approach to identify the secretory granule membrane glycoprotein 2 as a marker for PDX1+/NKX6-1+ PPs.


Assuntos
Biomarcadores Tumorais/metabolismo , Membrana Celular/metabolismo , Pâncreas/metabolismo , Células-Tronco/metabolismo , Diferenciação Celular , Células Cultivadas , Proteínas Ligadas por GPI , Proteínas de Homeodomínio/metabolismo , Humanos , Células Secretoras de Insulina/metabolismo , Espectrometria de Massas , Pâncreas/citologia , Células-Tronco Pluripotentes/citologia , Células-Tronco Pluripotentes/metabolismo , Proteômica/métodos , Transativadores/metabolismo , Fatores de Transcrição/metabolismo
13.
Exp Hematol ; 33(9): 955-64, 2005 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-16140142

RESUMO

Embryonic stem (ES) cells have the potential to develop into all cell types of the adult body. This capability provides the basis for considering the ES cell system as a novel and unlimited source of cells for replacement therapies for the treatment of a wide range of diseases. Before the cell-based therapy potential of ES cells can be realized, a better understanding of the pathways regulating lineage-specific differentiation is required. Current studies suggest that the bone morphogenic protein, transforming growth factor-beta, Wnt, and fibroblast growth factor pathways that are required for gastrulation and germ layer induction in the embryo are also essential for differentiation of ES cells in culture. The current understanding of how these factors influence germ layer induction in both the embryo and in the ES cell differentiation system is addressed in this review.


Assuntos
Desenvolvimento Embrionário , Indução Embrionária , Camadas Germinativas/citologia , Células-Tronco/fisiologia , Animais , Camadas Germinativas/fisiologia , Substâncias de Crescimento/fisiologia , Camundongos , Transdução de Sinais , Células-Tronco/citologia
14.
Stem Cell Reports ; 4(4): 591-604, 2015 Apr 14.
Artigo em Inglês | MEDLINE | ID: mdl-25843049

RESUMO

Human pluripotent stem cells (hPSCs) represent a renewable source of pancreatic beta cells for both basic research and therapeutic applications. Given this outstanding potential, significant efforts have been made to identify the signaling pathways that regulate pancreatic development in hPSC differentiation cultures. In this study, we demonstrate that the combination of epidermal growth factor (EGF) and nicotinamide signaling induces the generation of NKX6-1(+) progenitors from all hPSC lines tested. Furthermore, we show that the size of the NKX6-1(+) population is regulated by the duration of treatment with retinoic acid, fibroblast growth factor 10 (FGF10), and inhibitors of bone morphogenetic protein (BMP) and hedgehog signaling pathways. When transplanted into NOD scid gamma (NSG) recipients, these progenitors differentiate to give rise to exocrine and endocrine cells, including monohormonal insulin(+) cells. Together, these findings provide an efficient and reproducible strategy for generating highly enriched populations of hPSC-derived beta cell progenitors for studies aimed at further characterizing their developmental potential in vivo and deciphering the pathways that regulate their maturation in vitro.


Assuntos
Diferenciação Celular , Proteínas de Homeodomínio/metabolismo , Pâncreas/citologia , Pâncreas/metabolismo , Células-Tronco Pluripotentes/citologia , Células-Tronco Pluripotentes/metabolismo , Ativinas/metabolismo , Animais , Proteínas de Transporte/metabolismo , Linhagem Celular , Fator de Crescimento Epidérmico/metabolismo , Fator de Crescimento Epidérmico/farmacologia , Regulação da Expressão Gênica , Proteínas de Homeodomínio/genética , Humanos , Imunofenotipagem , Ilhotas Pancreáticas/citologia , Ilhotas Pancreáticas/metabolismo , Camundongos , Modelos Biológicos , Niacinamida/farmacologia , Organogênese/efeitos dos fármacos , Organogênese/genética , Transdução de Sinais/efeitos dos fármacos
15.
Mol Metab ; 4(4): 344-52, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25830097

RESUMO

OBJECTIVE: Disruption of TCF7L2 in mouse pancreatic ß-cells has generated different outcomes in several investigations. Here we aim to clarify role of ß-cell TCF7L2 and Wnt signaling using a functional-knockdown approach. METHODS: Adenovirus-mediated dominant negative TCF7L2 (TCF7L2DN) expression was conducted in Ins-1 cells. The fusion gene in which TCF7L2DN expression is driven by P TRE3G was utilized to generate the transgenic mouse line TCF7L2DN Tet . The double transgenic line was created by mating TCF7L2DN Tet with Ins2-rtTA, designated as ßTCFDN. ß-cell specific TCF7L2DN expression was induced in ßTCFDN by doxycycline feeding. RESULTS: TCF7L2DN expression in Ins-1 cells reduced GSIS, cell proliferation and expression of a battery of genes including incretin receptors and ß-cell transcription factors. Inducing TCF7L2DN expression in ßTCFDN during adulthood or immediately after weaning generated no or very modest metabolic defect, while its expression during embryonic development by doxycycline feeding in pregnant mothers resulted in significant glucose intolerance associated with altered ß-cell gene expression and reduced ß-cell mass. CONCLUSIONS: Our observations support a cell autonomous role for TCF7L2 in pancreatic ß-cells suggested by most, though not all, investigations. ßTCFDN is a novel model for further exploring the role of TCF7L2 in ß-cell genesis and metabolic homeostasis.

16.
Cell Stem Cell ; 10(4): 371-84, 2012 Apr 06.
Artigo em Inglês | MEDLINE | ID: mdl-22482503

RESUMO

The use of human pluripotent stem cells for laboratory studies and cell-based therapies is hampered by their tumor-forming potential and limited ability to generate pure populations of differentiated cell types in vitro. To address these issues, we established endodermal progenitor (EP) cell lines from human embryonic and induced pluripotent stem cells. Optimized growth conditions were established that allow near unlimited (>10(16)) EP cell self-renewal in which they display a morphology and gene expression pattern characteristic of definitive endoderm. Upon manipulation of their culture conditions in vitro or transplantation into mice, clonally derived EP cells differentiate into numerous endodermal lineages, including monohormonal glucose-responsive pancreatic ß-cells, hepatocytes, and intestinal epithelia. Importantly, EP cells are nontumorigenic in vivo. Thus, EP cells represent a powerful tool to study endoderm specification and offer a potentially safe source of endodermal-derived tissues for transplantation therapies.


Assuntos
Linhagem Celular/citologia , Linhagem Celular/metabolismo , Endoderma/citologia , Endoderma/embriologia , Células-Tronco Pluripotentes/citologia , Células-Tronco Pluripotentes/metabolismo , Animais , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Humanos , Camundongos , Transplante de Células-Tronco , Transplante Heterólogo
17.
Cell Stem Cell ; 2(1): 60-71, 2008 Jan 10.
Artigo em Inglês | MEDLINE | ID: mdl-18371422

RESUMO

The embryonic stem cell differentiation system was used to define the roles of the Activin/Nodal, BMP, and canonical Wnt signaling pathways at three distinct developmental stages during hematopoietic ontogeny: induction of a primitive streak-like population, formation of Flk1(+) mesoderm, and induction of hematopoietic progenitors. Activin/Nodal and Wnt, but not BMP, signaling are required for the induction of the primitive streak. Although BMP is not required for primitive streak induction, it displays a strong posteriorizing effect on this population. All three signaling pathways regulate induction of Flk1(+) mesoderm. The specification of Flk1(+) mesoderm to the hematopoietic lineages requires VEGF and Wnt, but not BMP or Activin/Nodal signaling. Specifically, Wnt signaling is essential for commitment of the primitive erythroid, but not the definitive lineages. These findings highlight dynamic changes in signaling requirements during blood cell development and identify a role for Wnt signaling in the establishment of the primitive erythroid lineage.


Assuntos
Ativinas/fisiologia , Proteína Morfogenética Óssea 4/fisiologia , Diferenciação Celular , Linhagem da Célula , Células-Tronco Embrionárias/fisiologia , Regulação da Expressão Gênica no Desenvolvimento , Células-Tronco Hematopoéticas/fisiologia , Transdução de Sinais , Proteínas Wnt/fisiologia , Animais , Indução Embrionária , Células-Tronco Embrionárias/citologia , Hematopoese , Células-Tronco Hematopoéticas/citologia , Camundongos , Linha Primitiva/citologia , Linha Primitiva/fisiologia
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa