Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 195
Filtrar
1.
Stem Cells ; 39(4): 429-442, 2021 04.
Artigo em Inglês | MEDLINE | ID: mdl-33400835

RESUMO

Tissues and cells derived from pluripotent stem cells (PSC) are likely to become widely used in disease modeling, drug screening, and regenerative medicine. For these applications, the in vitro PSC differentiation process must be elaborately investigated and controlled to reliably obtain the desired end products. However, because traditional experimental methods, such as one factor at a time or brute-force approaches, are impractical for detailed screening of complex PSC cultivation conditions, more strategic and effective screening based on statistical design of experiments (DOE) ought to be indispensable. Among various DOE approaches, we regard robust parameter design (RPD) as particularly suited for differentiation protocol optimization due to its suitability for multifactorial screening. We confirmed the adaptability of RPD for investigating human induced PSC lineage specification toward anterior-posterior gut tube endodermal cells and clarified both the contribution of each cell signaling pathway and the effect of cell signaling condition alteration on marker RNA expression levels, while increasing the efficiency of the screening in 243-fold (18 vs 4374) compared with that of a brute-force approach. Specific induction of anterior foregut, hepatic, pancreatic, or mid-hindgut cells was achieved using seven iPSC strains with the optimal culture protocols established on the basis of RPD analysis. RPD has the potential to enable efficient construction and optimization of PSC differentiation protocols, and its use is recommended from fundamental research to mass production of PSC-derived products.


Assuntos
Técnicas de Cultura de Células , Endoderma/citologia , Células-Tronco Pluripotentes Induzidas/citologia , Intestinos/citologia , Fígado/citologia , Pâncreas/citologia , Projetos de Pesquisa , Biomarcadores/metabolismo , Ácido Butírico/farmacologia , Fator de Transcrição CDX2/genética , Fator de Transcrição CDX2/metabolismo , Diferenciação Celular/efeitos dos fármacos , Linhagem Celular , Linhagem da Célula/efeitos dos fármacos , Endoderma/efeitos dos fármacos , Endoderma/metabolismo , Análise Fatorial , Fator 2 de Crescimento de Fibroblastos/farmacologia , Expressão Gênica , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/metabolismo , Humanos , Células-Tronco Pluripotentes Induzidas/efeitos dos fármacos , Células-Tronco Pluripotentes Induzidas/metabolismo , Intestinos/efeitos dos fármacos , Intestinos/metabolismo , Fígado/efeitos dos fármacos , Fígado/metabolismo , Fator 3 de Transcrição de Octâmero/genética , Fator 3 de Transcrição de Octâmero/metabolismo , Pâncreas/efeitos dos fármacos , Pâncreas/metabolismo , Transdução de Sinais , Transativadores/genética , Transativadores/metabolismo , Tretinoína/farmacologia , alfa-Fetoproteínas/genética , alfa-Fetoproteínas/metabolismo
2.
Ann Clin Lab Sci ; 50(4): 468-473, 2020 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-32826243

RESUMO

OBJECTIVE: This study aimed to investigate the effects of combined activin A and Wnt3a treatment on definitive endoderm (DE) differentiation from human parthenogenetic embryonic stem cells (hPESCs). METHODS: hPESCs on human foreskin fibroblast feeder layers were induced to differentiate into DE using a combination of 50 ng/ml activin A and 25 ng/ml Wnt3a. Expression of the DE markers CXCR4, E-cadherin (ECD), Sox17, and Goosecoid (Gsc) were examined using flow cytometry and real-time quantitative PCR. RESULTS: The combination of activin A and Wnt3a significantly enhanced the percentages of CXCR4+, ECD+, Sox17+, and Gsc+ cells, culminating on day 2 of induction. This combined use promoted DE differentiation from hPESCs in vitro. CONCLUSIONS: Through the combination treatment using activin A and Wnt3a, DE differentiation from hPESCs culminated at 48 h, which can be regarded as the optimal time-point to induce differentiation of endodermal cells such as pancreatic, liver, and intestinal cells.


Assuntos
Ativinas/farmacologia , Diferenciação Celular/efeitos dos fármacos , Proteína Wnt3A/farmacologia , Células Cultivadas , Células-Tronco Embrionárias/citologia , Células-Tronco Embrionárias/efeitos dos fármacos , Células-Tronco Embrionárias/metabolismo , Endoderma/citologia , Endoderma/efeitos dos fármacos , Endoderma/metabolismo , Humanos , Transdução de Sinais/efeitos dos fármacos , Proteína Wnt3A/metabolismo
3.
Biochem Biophys Res Commun ; 527(3): 811-817, 2020 06 30.
Artigo em Inglês | MEDLINE | ID: mdl-32446562

RESUMO

The ability of human embryonic stem cells (hESCs) to proliferate unlimitedly and give rise to all tissues makes these cells a promising source for cell replacement therapies. To realize the full potential of hESCs in cell therapy, it is necessary to interrogate regulatory pathways that influence hESC maintenance and commitment. Here, we reveal that pharmacological attenuation of p38 mitogen-activated protein kinase (p38-MAPK) in hESCs concomitantly augments some characteristics associated with pluripotency and the expressions of early lineage markers. Moreover, this blockage capacitates hESCs to differentiate towards an endoderm lineage at the expense of other lineages upon spontaneous hESC differentiation. Notably, hESCs pre-treated with p38-MAPK inhibitor exhibit significantly improved pancreatic progenitor directed differentiation. Together, our findings suggest a new approach to the robust endoderm differentiation of hESCs and potentially enables the facile derivation of various endoderm-derived lineages such as pancreatic cells.


Assuntos
Endoderma/citologia , Células-Tronco Embrionárias Humanas/citologia , Células-Tronco Embrionárias Humanas/efeitos dos fármacos , Inibidores de Proteínas Quinases/farmacologia , Proteínas Quinases p38 Ativadas por Mitógeno/antagonistas & inibidores , Diferenciação Celular/efeitos dos fármacos , Linhagem Celular , Endoderma/efeitos dos fármacos , Células-Tronco Embrionárias Humanas/metabolismo , Humanos , Pâncreas/citologia , Pâncreas/efeitos dos fármacos , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
4.
Thyroid ; 30(4): 609-620, 2020 04.
Artigo em Inglês | MEDLINE | ID: mdl-31801416

RESUMO

Background: Thyroid hormone (TH) synthesis is essential for the control of development, growth, and metabolism in vertebrates and depends on a sufficient dietary iodine intake. Importantly, both iodine deficiency and iodine excess (IE) impair TH synthesis, causing serious health problems especially during fetal/neonatal development. While it is known that IE disrupts thyroid function by inhibiting thyroid gene expression, its effects on thyroid development are less clear. Accordingly, this study sought to investigate the effects of IE during the embryonic development/differentiation of endoderm and the thyroid gland. Methods: We used the murine embryonic stem (ES) cell model of in vitro directed differentiation to assess the impact of IE on the generation of endoderm and thyroid cells. Additionally, we subjected endoderm and thyroid explants obtained during early gestation to IE and evaluated gene and protein expression of endodermal markers in both models. Results: ES cells were successfully differentiated into endoderm cells and, subsequently, into thyrocytes expressing the specific thyroid markers Tshr, Slc5a5, Tpo, and Tg. IE exposure decreased the messenger RNA (mRNA) levels of the main endoderm markers Afp, Crcx4, Foxa1, Foxa2, and Sox17 in both ES cell-derived endoderm cells and embryonic explants. Interestingly, IE also decreased the expression of the main thyroid markers in ES cell-derived thyrocytes and thyroid explants. Finally, we demonstrate that DNA methyltransferase expression was increased by exposure to IE, and this was accompanied by hypermethylation and hypoacetylation of histone H3, pointing to an association between the gene repression triggered by IE and the observed epigenetic changes. Conclusions: These data establish that IE treatment is deleterious for embryonic endoderm and thyroid gene expression.


Assuntos
Diferenciação Celular/efeitos dos fármacos , Células-Tronco Embrionárias/efeitos dos fármacos , Endoderma/efeitos dos fármacos , Epigênese Genética/efeitos dos fármacos , Expressão Gênica/efeitos dos fármacos , Iodeto de Sódio/farmacologia , Glândula Tireoide/efeitos dos fármacos , Animais , Células-Tronco Embrionárias/citologia , Endoderma/citologia , Regulação da Expressão Gênica no Desenvolvimento/efeitos dos fármacos , Camundongos , Glândula Tireoide/citologia
5.
Cell Death Dis ; 10(10): 763, 2019 10 10.
Artigo em Inglês | MEDLINE | ID: mdl-31601782

RESUMO

Hepatocytes have been successfully generated from human pluripotent stem cells (hPSCs). However, the cost-effective and clinical-grade generation of hepatocytes from hPSCs still need to be improved. In this study, we reported the production of functional hepatocytes from clinical-grade human embryonic stem cells (hESCs) under good manufacturing practice (GMP) requirements. We sequentially generated primitive streak (PS), definitive endoderm (DE), hepatoblasts and hepatocyte-like cells (HLCs) from hESCs in the different stages with completely defined reagents. During hepatoblast differentiation, dimethyl sulfoxide (DMSO), transferrin, L-ascorbic acid 2-phosphate sesquimagnesium salt hydrate (Vc-Mg), insulin, and sodium selenite were used instead of cytokines and FBS/KOSR. Then, hepatoblasts were differentiated into HLCs that had a typical hepatocyte morphology and possessed characteristics of mature hepatocytes, such as metabolic-related gene expression, albumin secretion, fat accumulation, glycogen storage, and inducible cytochrome P450 activity in vitro. HLCs integrated into the livers of Tet-uPA Rag2-/- Il2rg-/- (URG) mice, which partially recovered after transplantation. Furthermore, a series of biosafety-related experiments were performed to ensure future clinical applications. In conclusion, we developed a chemically defined system to generate qualified clinical-grade HLCs from hESCs under GMP conditions. HLCs have been proven to be safe and effective for treating liver failure. This efficient platform could facilitate the treatment of liver diseases using hESC-derived HLCs transplantation.


Assuntos
Técnicas de Cultura de Células/métodos , Meios de Cultura/química , Endoderma/citologia , Hepatócitos/citologia , Células-Tronco Embrionárias Humanas/citologia , Ativinas/farmacologia , Animais , Diferenciação Celular/efeitos dos fármacos , Diferenciação Celular/genética , Sistema Enzimático do Citocromo P-450/metabolismo , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Endoderma/efeitos dos fármacos , Endoderma/metabolismo , Regulação da Expressão Gênica no Desenvolvimento/genética , Hepatócitos/metabolismo , Células-Tronco Embrionárias Humanas/efeitos dos fármacos , Células-Tronco Embrionárias Humanas/metabolismo , Humanos , Subunidade gama Comum de Receptores de Interleucina/genética , Subunidade gama Comum de Receptores de Interleucina/metabolismo , Fígado/citologia , Falência Hepática/terapia , Regeneração Hepática , Masculino , Camundongos , Camundongos Knockout , Família Multigênica , Piridinas/farmacologia , Pirimidinas/farmacologia , Transplante de Células-Tronco , Transcriptoma/genética
6.
Exp Cell Res ; 384(2): 111642, 2019 11 15.
Artigo em Inglês | MEDLINE | ID: mdl-31562862

RESUMO

Our hypothesis is that hyaluronic acid may regulate the differentiation of human amniotic epithelial cells (hAECs) into insulin-producing cells and help the treatment of type 1 diabetes. Herein, a protocol for the stepwise in vitro differentiation of hAECs into functional insulin-producing cells was developed by mimicking the process of pancreas development. Treatment of hAECs with hyaluronic acid enhanced their differentiation of definitive endoderm and pancreatic progenitors. Endodermal markers Sox17 and Foxa2 and pancreatic progenitor markers Pax6, Nkx6.1, and Ngn3 were upregulated an enhanced gene expression in hAECs, but hAECs did not express the ß cell-specific transcription factor Pdx1. Interestingly, hyaluronic acid promoted the expression of major pancreatic development-related genes and proteins after combining with commonly used inducers of stem cells differentiation into insulin-producing cells. This indicated the potent synergistic effects of the combination on hAECs differentiation in vitro. By establishing a multiple injection transplantation strategy via tail vein injections, hAECs transplantation significantly reduced hyperglycemia symptoms, increased the plasma insulin content, and partially repaired the islet structure in type 1 diabetic mice. In particular, the combination of hAECs with hyaluronic acid exhibited a remarkable therapeutic effect compared to both the insulin group and the hAECs alone group. The hAECs' paracrine action and hyaluronic acid co-regulated the local immune response, improved the inflammatory microenvironment in the damaged pancreas of type 1 diabetic mice, and promoted the trans-differentiation of pancreatic α cells into ß cells. These findings suggest that hyaluronic acid is an efficient co-inducer of the differentiation of hAECs into functional insulin-producing cells, and hAECs treatment with hyaluronic acid may be a promising cell-replacement therapeutic approach for the treatment of type 1 diabetes.


Assuntos
Âmnio/efeitos dos fármacos , Diferenciação Celular/efeitos dos fármacos , Diabetes Mellitus Experimental/terapia , Células Epiteliais/efeitos dos fármacos , Ácido Hialurônico/farmacologia , Ativinas/metabolismo , Âmnio/metabolismo , Animais , Técnicas de Cultura de Células/métodos , Terapia Baseada em Transplante de Células e Tecidos/métodos , Células Cultivadas , Modelos Animais de Doenças , Células-Tronco Embrionárias/efeitos dos fármacos , Células-Tronco Embrionárias/metabolismo , Endoderma/efeitos dos fármacos , Endoderma/metabolismo , Células Epiteliais/metabolismo , Humanos , Inflamação/metabolismo , Insulina/metabolismo , Células Secretoras de Insulina/efeitos dos fármacos , Células Secretoras de Insulina/metabolismo , Masculino , Camundongos , Pâncreas/efeitos dos fármacos , Pâncreas/metabolismo
7.
Methods Mol Biol ; 1994: 41-53, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31124103

RESUMO

We have developed a method to bulk culture definitive endoderm cells generated from human iPSCs which can be stored and differentiated to hepatocytes. Human iPSC-derived definitive endoderm cells were sorted based on the expression of CXCR4. The sorted cells were able to proliferate for extended periods and can be cryopreserved. The definitive endoderm cells were subsequently utilized to generate functional hepatocytes expressing albumin and α-fetoprotein in different multiwell formats. This provides a method to reliably produce more consistent hepatocytes in greater quantities and has enabled the development of high-throughput screening strategies.


Assuntos
Técnicas de Cultura de Células/métodos , Diferenciação Celular , Endoderma/citologia , Endoderma/metabolismo , Hepatócitos/citologia , Células-Tronco Pluripotentes Induzidas/citologia , Diferenciação Celular/efeitos dos fármacos , Separação Celular/métodos , Células Cultivadas , Endoderma/efeitos dos fármacos , Proteínas Fetais/metabolismo , Citometria de Fluxo , Hepatócitos/metabolismo , Humanos , Células-Tronco Pluripotentes Induzidas/efeitos dos fármacos , Células-Tronco Pluripotentes Induzidas/metabolismo , Receptores CXCR4/metabolismo , Albumina Sérica Humana/metabolismo
8.
Biochem Biophys Res Commun ; 512(2): 399-404, 2019 04 30.
Artigo em Inglês | MEDLINE | ID: mdl-30902394

RESUMO

A combination of extracellular signal-regulated kinase 1/2 (ERK1/2) and glycogen synthase kinase 3ß (GSK3ß) inhibitors, called 2i, is widely used for maintaining the pluripotency of mouse embryonic stem cells (ESCs) in vitro. Without 2i, a few mouse ESCs spontaneously gives rise to primitive endoderm (PrE) cells, whereas 2i completely blocks this PrE cell differentiation. However, the molecular mechanisms underlying the inhibitory action of 2i on PrE cell differentiation remain unclear. Robust PrE cell induction is achieved by enforced expression of the transcription factor Gata4. Here, we analyzed how 2i inhibits the PrE cell differentiation using mouse ESCs carrying an inducible Gata4 expression cassette. We found that 2i effectively inhibited the Gata4-induced PrE cell differentiation and the ERK1/2 inhibitor was responsible for this effect. We further revealed that the transcriptional activation ability of Gata4 was necessary for PrE cell induction and its disruption by the ERK1/2 inhibitor. The phosphorylation of Ser105, Ser266, and Ser411 of the Gata4 protein was not involved in the PrE cell induction. Overexpression of Klf4, an ERK1/2 substrate, inhibited the Gata4-mediated transcriptional activation. Our data indicated that ERK1/2 supported the PrE cell induction via the indirect transcriptional activation of Gata4.


Assuntos
Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Células-Tronco Embrionárias Murinas/efeitos dos fármacos , Células-Tronco Embrionárias Murinas/metabolismo , Inibidores de Proteínas Quinases/farmacologia , Animais , Benzamidas/farmacologia , Diferenciação Celular/efeitos dos fármacos , Células Cultivadas , Difenilamina/análogos & derivados , Difenilamina/farmacologia , Doxiciclina/farmacologia , Endoderma/citologia , Endoderma/efeitos dos fármacos , Fator de Transcrição GATA4/antagonistas & inibidores , Fator de Transcrição GATA4/genética , Fator de Transcrição GATA4/metabolismo , Glicogênio Sintase Quinase 3 beta/antagonistas & inibidores , Fator 4 Semelhante a Kruppel , Camundongos , Proteína Quinase 1 Ativada por Mitógeno/antagonistas & inibidores , Proteína Quinase 3 Ativada por Mitógeno/antagonistas & inibidores , Células-Tronco Embrionárias Murinas/citologia , Fosforilação , Piridinas/farmacologia , Pirimidinas/farmacologia
9.
Proc Natl Acad Sci U S A ; 116(4): 1384-1393, 2019 01 22.
Artigo em Inglês | MEDLINE | ID: mdl-30606801

RESUMO

Upon virus infection, pluripotent stem cells neither induce nor respond to canonical type I interferons (IFN-I). To better understand this biology, we characterized induced pluripotent stem cells (iPSCs) as well as their differentiated parental or rederived counterparts. We confirmed that only iPSCs failed to respond to viral RNA, IFN-I, or viral infection. This lack of response could be phenocopied in fibroblasts with the expression of a reprogramming factor which repressed the capacity to induce canonical antiviral pathways. To ascertain the consequences of restoring the antiviral response in the context of pluripotency, we engineered a system to engage these defenses in iPSCs. Inducible expression of a recombinant virus-activated transcription factor resulted in the successful reconstitution of antiviral defenses through the direct up-regulation of IFN-I-stimulated genes. Induction of the antiviral signature in iPSCs, even for a short duration, resulted in the dysregulation of genes associated with all three germ layers despite maintaining pluripotency markers. Trilineage differentiation of these same cells showed that engagement of the antiviral defenses compromised ectoderm and endoderm formation and dysregulated the development of mesodermal sublineages. In all, these data suggest that the temporal induction of the antiviral response primes iPSCs away from pluripotency and induces numerous aberrant gene products upon differentiation. Together these results suggest that the IFN-I system and pluripotency may be incompatible with each other and thus explain why stem cells do not utilize the canonical antiviral system.


Assuntos
Diferenciação Celular/fisiologia , Células-Tronco Pluripotentes Induzidas/metabolismo , Células-Tronco Pluripotentes Induzidas/fisiologia , Interferon Tipo I/metabolismo , Antivirais/farmacologia , Biomarcadores/metabolismo , Diferenciação Celular/efeitos dos fármacos , Células Cultivadas , Reprogramação Celular/fisiologia , Ectoderma/efeitos dos fármacos , Ectoderma/metabolismo , Ectoderma/fisiologia , Ectoderma/virologia , Endoderma/efeitos dos fármacos , Endoderma/metabolismo , Endoderma/fisiologia , Endoderma/virologia , Fibroblastos/efeitos dos fármacos , Fibroblastos/metabolismo , Fibroblastos/fisiologia , Fibroblastos/virologia , Camadas Germinativas/efeitos dos fármacos , Camadas Germinativas/metabolismo , Camadas Germinativas/fisiologia , Camadas Germinativas/virologia , Humanos , Células-Tronco Pluripotentes Induzidas/efeitos dos fármacos , Células-Tronco Pluripotentes Induzidas/virologia , Fator 4 Semelhante a Kruppel , RNA Viral/genética , Fatores de Transcrição/metabolismo , Regulação para Cima/efeitos dos fármacos , Regulação para Cima/fisiologia
10.
Nat Protoc ; 14(1): 28-50, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-30470820

RESUMO

The human stomach contains two primary domains: the corpus, which contains the fundic epithelium, and the antrum. Each of these domains has distinct cell types and functions, and therefore each presents with unique disease pathologies. Here, we detail two protocols to differentiate human pluripotent stem cells (hPSCs) into human gastric organoids (hGOs) that recapitulate both domains. Both protocols begin with the differentiation of hPSCs into definitive endoderm (DE) using activin A, followed by the generation of free-floating 3D posterior foregut spheroids using FGF4, Wnt pathway agonist CHIR99021 (CHIR), BMP pathway antagonist Noggin, and retinoic acid. Embedding spheroids in Matrigel and continuing 3D growth in epidermal growth factor (EGF)-containing medium for 4 weeks results in antral hGOs (hAGOs). To obtain fundic hGOs (hFGOs), spheroids are additionally treated with CHIR and FGF10. Induced differentiation of acid-secreting parietal cells in hFGOs requires temporal treatment of BMP4 and the MEK inhibitor PD0325901 for 48 h on protocol day 30. In total, it takes ~34 d to generate hGOs from hPSCs. To date, this is the only approach that generates functional human differentiated gastric cells de novo from hPSCs.


Assuntos
Técnicas de Cultura de Células , Endoderma/citologia , Células Epiteliais/citologia , Fundo Gástrico/citologia , Organoides/citologia , Células-Tronco Pluripotentes/citologia , Antro Pilórico/citologia , Ativinas/farmacologia , Benzamidas/farmacologia , Proteínas de Transporte/farmacologia , Diferenciação Celular , Colágeno/química , Meios de Cultura/química , Meios de Cultura/farmacologia , Difenilamina/análogos & derivados , Difenilamina/farmacologia , Combinação de Medicamentos , Endoderma/efeitos dos fármacos , Endoderma/metabolismo , Fator de Crescimento Epidérmico/farmacologia , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/metabolismo , Fator 10 de Crescimento de Fibroblastos/farmacologia , Fator 4 de Crescimento de Fibroblastos/farmacologia , Fundo Gástrico/metabolismo , Humanos , Laminina/química , Especificidade de Órgãos , Organoides/efeitos dos fármacos , Organoides/metabolismo , Células-Tronco Pluripotentes/efeitos dos fármacos , Células-Tronco Pluripotentes/metabolismo , Proteoglicanas/química , Antro Pilórico/metabolismo , Piridinas/farmacologia , Pirimidinas/farmacologia , Tretinoína/farmacologia , Via de Sinalização Wnt/efeitos dos fármacos
11.
J Hazard Mater ; 364: 581-590, 2019 02 15.
Artigo em Inglês | MEDLINE | ID: mdl-30388642

RESUMO

Silicon (Si) can alleviate cadmium (Cd) toxicity in many plants, but mechanisms underlying this beneficial effect are still lacking. In this study, the roles of Si in time-dependent apoplastic and symplastic Cd absorption by roots of wheat plants were investigated. Results showed that, during short-term Cd exposure, the symplastic pathway of Cd in roots was not significantly affected by Si. Cell wall properties and cell wall-bound Cd regarding the apoplastic pathway were unaffected by Si either. Nevertheless, Cd concentrations in the apoplastic fluid of roots were decreased by Si. The reason could be that Si delayed endodermal suberization of roots resulting in promoted apoplastic Cd translocation to shoots, thus decreasing Cd in the apoplastic fluid of roots after short-term Cd stress. By contrast, after long-term Cd stress, cell wall properties and the expression of genes related to Cd influx and transport were unaffected. Intriguingly, Si up-regulated the expression of the Cd efflux-related gene TaTM20 and repressed apoplastic Cd translocation to shoots, which might contribute to decrease of Cd after long-term Cd exposure. Taken together, these results indicate that Si-dependent decrease in root Cd concentrations during short-term Cd exposure helps plants to mitigate Cd toxicity in the long-term.


Assuntos
Cádmio/química , Lipídeos/biossíntese , Raízes de Plantas/efeitos dos fármacos , Silício/farmacologia , Triticum/efeitos dos fármacos , Adsorção , Biomassa , Cádmio/metabolismo , Parede Celular/efeitos dos fármacos , Parede Celular/metabolismo , Endoderma/efeitos dos fármacos , Endoderma/metabolismo , Raízes de Plantas/química , Raízes de Plantas/crescimento & desenvolvimento , Raízes de Plantas/metabolismo , Triticum/química , Triticum/crescimento & desenvolvimento , Triticum/metabolismo
12.
Nature ; 564(7735): 219-224, 2018 12.
Artigo em Inglês | MEDLINE | ID: mdl-30518857

RESUMO

Direct lineage reprogramming involves the conversion of cellular identity. Single-cell technologies are useful for deconstructing the considerable heterogeneity that emerges during lineage conversion. However, lineage relationships are typically lost during cell processing, complicating trajectory reconstruction. Here we present 'CellTagging', a combinatorial cell-indexing methodology that enables parallel capture of clonal history and cell identity, in which sequential rounds of cell labelling enable the construction of multi-level lineage trees. CellTagging and longitudinal tracking of fibroblast to induced endoderm progenitor reprogramming reveals two distinct trajectories: one leading to successfully reprogrammed cells, and one leading to a 'dead-end' state, paths determined in the earliest stages of lineage conversion. We find that expression of a putative methyltransferase, Mettl7a1, is associated with the successful reprogramming trajectory; adding Mettl7a1 to the reprogramming cocktail increases the yield of induced endoderm progenitors. Together, these results demonstrate the utility of our lineage-tracing method for revealing the dynamics of direct reprogramming.


Assuntos
Linhagem da Célula , Rastreamento de Células/métodos , Reprogramação Celular , Células Clonais/citologia , Análise de Célula Única/métodos , Animais , Linhagem da Célula/efeitos dos fármacos , Separação Celular , Reprogramação Celular/efeitos dos fármacos , Células Clonais/efeitos dos fármacos , Endoderma/citologia , Endoderma/efeitos dos fármacos , Fibroblastos/citologia , Fibroblastos/efeitos dos fármacos , Células HEK293 , Humanos , Metiltransferases/metabolismo , Camundongos , Células-Tronco/citologia , Células-Tronco/efeitos dos fármacos , Fatores de Tempo
13.
Differentiation ; 101: 16-24, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29626713

RESUMO

A variety of approaches have been developed for the derivation of hepatocyte-like cells from pluripotent stem cells. Currently, most of these strategies employ step-wise differentiation approaches with recombinant growth-factors or small-molecule analogs to recapitulate developmental signaling pathways. Here, we tested the efficacy of a small-molecule based differentiation protocol for the generation of hepatocyte-like cells from human pluripotent stem cells. Quantitative gene-expression, immunohistochemical, and western blot analyses for SOX17, FOXA2, CXCR4, HNF4A, AFP, indicated the stage-specific differentiation into definitive endoderm, hepatoblast and hepatocyte-like derivatives. Furthermore, hepatocyte-like cells displayed morphological and functional features characteristic of primary hepatocytes, as indicated by the production of ALB (albumin) and α-1-antitrypsin (A1AT), as well as glycogen storage capacity by periodic acid-Schiff staining. Together, these data support that the small-molecule based hepatic differentiation protocol is a simple, reproducible, and inexpensive method to efficiently drive the differentiation of human pluripotent stem cells towards a hepatocyte-like phenotype, for downstream pharmacogenomic and regenerative medicine applications.


Assuntos
Diferenciação Celular/efeitos dos fármacos , Hepatócitos/efeitos dos fármacos , Células-Tronco Pluripotentes/efeitos dos fármacos , Piridinas/farmacologia , Pirimidinas/farmacologia , Endoderma/citologia , Endoderma/efeitos dos fármacos , Expressão Gênica/efeitos dos fármacos , Hepatócitos/citologia , Humanos , Fígado/efeitos dos fármacos , Células-Tronco Pluripotentes/citologia
14.
Exp Cell Res ; 362(1): 227-234, 2018 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-29157981

RESUMO

Insulin-producing cells (IPCs) derived from human embryonic stem cells (hESCs) hold great potential for cell transplantation therapy in diabetes. Tremendous progress has been made in inducing differentiation of hESCs into IPCs in vitro, of which definitive endoderm (DE) protocol mimicking foetal pancreatic development has been widely used. However, immaturity of the obtained IPCs limits their further applications in treating diabetes. Forkhead box O1 (FoxO1) is involved in the differentiation and functional maintenance of murine pancreatic ß cells, but its role in human ß cell differentiation is under elucidation. Here, we showed that although FoxO1 expression level remained consistent, cytoplasmic phosphorylated FoxO1 protein level increased during IPC differentiation of hESCs induced by DE protocol. Lentiviral silencing of FoxO1 in pancreatic progenitors upregulated the levels of pancreatic islet differentiation-related genes and improved glucose-stimulated insulin secretion response in their progeny IPCs, whereas overexpression of FoxO1 showed the opposite effects. Notably, treatment with the FoxO1 inhibitor AS1842856 displayed similar effects with FoxO1 knockdown in pancreatic progenitors. These effects were closely associated with the mutually exclusive nucleocytoplasmic shuttling of FoxO1 and Pdx1 in the AS1842856-treated pancreatic progenitors. Our data demonstrated a promising effect of FoxO1 inhibition by the small molecule on gene expression profile during the differentiation, and in turn, on determining IPC maturation via modulating subcellular location of FoxO1 and Pdx1. Therefore, we identify a novel role of FoxO1 inhibition in promoting IPC differentiation of hESCs, which may provide clues for induction of mature ß cells from hESCs and clinical applications in regenerative medicine.


Assuntos
Proteína Forkhead Box O1/metabolismo , Células-Tronco Embrionárias Humanas/metabolismo , Células Secretoras de Insulina/metabolismo , Diferenciação Celular/efeitos dos fármacos , Diferenciação Celular/fisiologia , Linhagem Celular , Endoderma/efeitos dos fármacos , Endoderma/metabolismo , Glucose/metabolismo , Proteínas de Homeodomínio/metabolismo , Células-Tronco Embrionárias Humanas/efeitos dos fármacos , Humanos , Insulina/metabolismo , Células Secretoras de Insulina/efeitos dos fármacos , Ilhotas Pancreáticas/efeitos dos fármacos , Ilhotas Pancreáticas/metabolismo , Quinolonas/farmacologia , Transativadores/metabolismo
15.
J Cell Physiol ; 233(4): 3578-3589, 2018 04.
Artigo em Inglês | MEDLINE | ID: mdl-29044512

RESUMO

Definitive endoderm (DE) is the first stage of human pluripotent stem cell (hPSC) differentiation into hepatocyte-like cells. Developing human liver cell models for pharmaceutical applications is highly demanding. Due to the vast number of existing protocols to generate DE cells from hPSCs, we aimed to compare the specificity and efficiency of selected published differentiation conditions. We differentiated two hPSC lines (induced PSC and embryonic stem cell) to DE cells on Matrigel matrix using growth factors (Activin A and Wnt-3a) and small molecules (sodium butyrate and IDE 1) in different combinations. By studying dynamic changes during 6 days in cell morphology and the expression of markers for pluripotency, DE, and other germ layer lineages, we found that Activin A is essential for DE differentiation, while Wnt-3a and sodium butyrate are dispensable. Although sodium butyrate exerted rapid DE differentiation kinetics, it caused massive cell death and could not generate sufficient cells for further differentiation and applications. We further discover that IDE 1 could not induce DE as reported previously. Hereby, we compared different conditions for DE induction and found an effective six day-protocol to obtain DE cells for the further differentiation and applications.


Assuntos
Ativinas/farmacologia , Ácido Butírico/farmacologia , Células-Tronco Embrionárias/efeitos dos fármacos , Endoderma/efeitos dos fármacos , Hepatócitos/efeitos dos fármacos , Diferenciação Celular/efeitos dos fármacos , Linhagem Celular , Células Cultivadas , Células-Tronco Embrionárias/citologia , Endoderma/citologia , Hepatócitos/metabolismo , Humanos , Células-Tronco Pluripotentes/citologia , Células-Tronco Pluripotentes/efeitos dos fármacos
16.
Nat Cell Biol ; 19(10): 1164-1177, 2017 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-28945231

RESUMO

Signalling downstream of Activin/Nodal (ActA) and Wnt can induce endoderm differentiation and also support self-renewal in pluripotent cells. Here we find that these apparently contradictory activities are fine-tuned by insulin. In the absence of insulin, the combination of these cytokines supports endoderm in a context-dependent manner. When applied to naive pluripotent cells that resemble peri-implantation embryos, ActA and Wnt induce extra-embryonic primitive endoderm (PrE), whereas when applied to primed pluripotent epiblast stem cells (EpiSC), these cytokines induce gastrulation-stage embryonic definitive endoderm. In naive embryonic stem cell culture, we find that insulin complements LIF signalling to support self-renewal; however, when it is removed, LIF, ActA and Wnt signalling not only induce PrE differentiation, but also support its expansion. Self-renewal of these PrE cultures is robust and, on the basis of gene expression, these cells resemble early blastocyst-stage PrE, a naive endoderm state able to make both visceral and parietal endoderm.


Assuntos
Diferenciação Celular/efeitos dos fármacos , Autorrenovação Celular/efeitos dos fármacos , Células-Tronco Embrionárias/efeitos dos fármacos , Endoderma/efeitos dos fármacos , Insulina/farmacologia , Células-Tronco Pluripotentes/efeitos dos fármacos , Ativinas/farmacologia , Animais , Linhagem Celular , Linhagem da Célula , Técnicas de Cultura Embrionária , Células-Tronco Embrionárias/metabolismo , Endoderma/citologia , Endoderma/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Idade Gestacional , Fator Inibidor de Leucemia/farmacologia , Camundongos Endogâmicos C57BL , Proteína Nodal/farmacologia , Células-Tronco Pluripotentes/metabolismo , Fatores de Tempo , Transfecção , Via de Sinalização Wnt/efeitos dos fármacos , Proteína Wnt3A/farmacologia
17.
Development ; 144(9): 1629-1634, 2017 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-28289133

RESUMO

The chordate pharynx, possessing gill slits and the endostyle, is a complex of multiple tissues that are highly organized along the anterior-posterior (AP) axis. Although Hox genes show AP coordinated expression in the pharyngeal endoderm, tissue-specific roles of these factors for establishing the regional identities within this tissue have not been demonstrated. Here, we show that Hox1 is essential for the establishment of AP axial identity of the endostyle, a major structure of the pharyngeal endoderm, in the ascidian Ciona intestinalis We found that knockout of Hox1 causes posterior-to-anterior transformation of the endostyle identity, and that Hox1 represses Otx expression and anterior identity, and vice versa. Furthermore, alteration of the regional identity of the endostyle disrupts the formation of body wall muscles, suggesting that the endodermal axial identity is essential for coordinated pharyngeal development. Our results demonstrate an essential role of Hox genes in establishment of the AP regional identity in the pharyngeal endoderm and reveal crosstalk between endoderm and mesoderm during development of chordate pharynx.


Assuntos
Endoderma/embriologia , Proteínas de Homeodomínio/metabolismo , Desenvolvimento Muscular , Músculos Faríngeos/embriologia , Faringe/embriologia , Animais , Ciona intestinalis , Endoderma/efeitos dos fármacos , Retroalimentação Fisiológica/efeitos dos fármacos , Desenvolvimento Muscular/efeitos dos fármacos , Músculos Faríngeos/efeitos dos fármacos , Faringe/efeitos dos fármacos , Tretinoína/farmacologia
18.
Biochem Biophys Res Commun ; 486(2): 481-485, 2017 04 29.
Artigo em Inglês | MEDLINE | ID: mdl-28322785

RESUMO

RNA silencing is used as a common method for investigating loss-of-function effects of genes of interest. In mammalian cells, RNA interference (RNAi) or RNA silencing can be achieved by transient siRNA (small or short interfering RNA) transfection or by stable shRNA (short hairpin RNA) systems. Various vectors are used for efficient delivery of shRNA. Lentiviral vectors offer an efficient delivery system for stable and long-term expression of the shRNA in mammalian cells. The widely used lentiviral pLKO.1 plasmid vector is very popular in RNAi studies. A large RNAi database, a TRC (the RNAi Consortium) library, was established based on the pLKO.1-TRC plasmid vector. This plasmid (also called pLKO.1-puro) has a puromycin-resistant gene for selection in mammalian cells along with designs for generating lentiviral particles as well for RNA silencing. While using the pLKO.1-puro TRC control shRNA plasmid for transfection in murine P19 embryonic stem (ES) cells, it was unexpectedly discovered that this plasmid vector induced robust endodermal differentiation. Since P19 ES cells are pluripotent and respond to external stimuli that have the potential to alter the phenotype and thus its stemness, other cell types used in RNA silencing studies do not display the obvious effect and therefore, may affect experiments in subtle ways that would go undetected. This study for the first time provides evidence that raises concern and warrants extreme caution while using the pLKO.1-puro control shRNA vector because of its unexpected non-specific effects on cellular integrity.


Assuntos
Endoderma/efeitos dos fármacos , Lentivirus/genética , Células-Tronco Embrionárias Murinas/efeitos dos fármacos , Plasmídeos/metabolismo , Puromicina/farmacologia , RNA Interferente Pequeno/genética , Animais , Artefatos , Diferenciação Celular/efeitos dos fármacos , Linhagem Celular , Endoderma/citologia , Endoderma/metabolismo , Expressão Gênica/efeitos dos fármacos , Biblioteca Gênica , Inativação Gênica , Lentivirus/metabolismo , Camundongos , Células-Tronco Embrionárias Murinas/citologia , Células-Tronco Embrionárias Murinas/metabolismo , Plasmídeos/química , RNA Interferente Pequeno/metabolismo , Transfecção
19.
PLoS One ; 12(2): e0170812, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28152080

RESUMO

Mouse F9 cells differentiate to primitive endoderm (PrE) when treated with retinoic acid (RA). Differentiation is accompanied by increased reactive oxygen species (ROS) levels, and while treating F9 cells with antioxidants attenuates differentiation, H2O2 treatment alone is sufficient to induce PrE. We identified the NADPH oxidase (NOX) complexes as candidates for the source of this endogenous ROS, and within this gene family, and over the course of differentiation, Nox1 and Nox 4 show the greatest upregulation induced by RA. Gata6, encoding a master regulator of extraembryonic endoderm is also up-regulated by RA and we provide evidence that NOX1 and NOX4 protein levels increase in F9 cells overexpressing Gata6. Pan-NOX and NOX1-specific inhibitors significantly reduced the ability of RA to induce PrE, and this was recapitulated using a genetic approach to knockdown Nox1 and/or Nox4 transcripts. Interestingly, overexpressing either gene in untreated F9 cells did not induce differentiation, even though each elevated ROS levels. Thus, the data suggests that ROS produced during PrE differentiation is dependent in part on increased NOX1 and NOX4 levels, which is under the control of GATA6. Furthermore, these results suggest that the combined activity of multiple NOX proteins is necessary for the differentiation of F9 cells to primitive endoderm.


Assuntos
Endoderma/citologia , Endoderma/metabolismo , NADH NADPH Oxirredutases/metabolismo , NADPH Oxidases/metabolismo , Animais , Diferenciação Celular/efeitos dos fármacos , Diferenciação Celular/genética , Diferenciação Celular/fisiologia , Linhagem Celular Tumoral , Endoderma/efeitos dos fármacos , Fator de Transcrição GATA6/genética , Fator de Transcrição GATA6/metabolismo , Técnicas de Silenciamento de Genes , Camundongos , NADH NADPH Oxirredutases/antagonistas & inibidores , NADH NADPH Oxirredutases/genética , NADPH Oxidase 1 , NADPH Oxidase 4 , NADPH Oxidases/antagonistas & inibidores , NADPH Oxidases/genética , Espécies Reativas de Oxigênio/metabolismo , Tretinoína/farmacologia , Via de Sinalização Wnt
20.
Stem Cell Reports ; 8(3): 589-604, 2017 03 14.
Artigo em Inglês | MEDLINE | ID: mdl-28196690

RESUMO

Induced pluripotent stem cells were created from a pancreas agenesis patient with a mutation in GATA6. Using genome-editing technology, additional stem cell lines with mutations in both GATA6 alleles were generated and demonstrated a severe block in definitive endoderm induction, which could be rescued by re-expression of several different GATA family members. Using the endodermal progenitor stem cell culture system to bypass the developmental block at the endoderm stage, cell lines with mutations in one or both GATA6 alleles could be differentiated into ß-like cells but with reduced efficiency. Use of suboptimal doses of retinoic acid during pancreas specification revealed a more severe phenotype, more closely mimicking the patient's disease. GATA6 mutant ß-like cells fail to secrete insulin upon glucose stimulation and demonstrate defective insulin processing. These data show that GATA6 plays a critical role in endoderm and pancreas specification and ß-like cell functionality in humans.


Assuntos
Endoderma/metabolismo , Fator de Transcrição GATA6/genética , Células Secretoras de Insulina/citologia , Células Secretoras de Insulina/metabolismo , Pâncreas/metabolismo , Apoptose/efeitos dos fármacos , Apoptose/genética , Biomarcadores , Diferenciação Celular/efeitos dos fármacos , Diferenciação Celular/genética , Linhagem Celular , Endoderma/efeitos dos fármacos , Endoderma/embriologia , Fator de Transcrição GATA6/metabolismo , Perfilação da Expressão Gênica , Humanos , Células-Tronco Pluripotentes Induzidas/citologia , Células-Tronco Pluripotentes Induzidas/metabolismo , Peptídeos e Proteínas de Sinalização Intercelular/farmacologia , Modelos Biológicos , Família Multigênica , Mutação , Pâncreas/embriologia , Fenótipo , Tretinoína/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...