Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 197
Filtrar
1.
Toxicol In Vitro ; 98: 105836, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38702034

RESUMEN

Nanoparticles have unique properties that make them useful in biomedicine. However, their extensive use raises concerns about potential hazards to the body. Therefore, it is crucial to establish effective and robust toxicology models to evaluate the developmental and functional toxicity of nanoparticles on the body. This article discusses the use of stem cells to study the developmental and functional toxicity of organs of endodermal origin due to nanoparticles. The study discovered that various types of nanoparticles have varying effects on stem cells. The application of stem cell models can provide a possibility for studying the effects of nanoparticles on organ development and function, as they can more accurately reflect the toxic mechanisms of different types of nanoparticles. However, stem cell toxicology systems currently cannot fully reflect the effects of nanoparticles on entire organs. Therefore, the establishment of organoid models and other advanced assessment models is expected to address this issue.


Asunto(s)
Endodermo , Nanopartículas , Células Madre , Animales , Nanopartículas/toxicidad , Humanos , Células Madre/efectos de los fármacos , Endodermo/efectos de los fármacos , Endodermo/citología
2.
Cell Commun Signal ; 22(1): 300, 2024 May 30.
Artículo en Inglés | MEDLINE | ID: mdl-38816763

RESUMEN

Optimizing the efficiency of definitive endoderm (DE) differentiation is necessary for the generation of diverse organ-like structures. In this study, we used the small molecule inhibitor saracatinib (SAR) to enhance DE differentiation of human embryonic stem cells and induced pluripotent stem cells. SAR significantly improved DE differentiation efficiency at low concentrations. The interaction between SAR and Focal Adhesion Kinase (FAK) was explored through RNA-seq and molecular docking simulations, which further supported the inhibition of DE differentiation by p-FAK overexpression in SAR-treated cells. In addition, we found that SAR inhibited the nuclear translocation of Yes-associated protein (YAP), a downstream effector of FAK, which promoted DE differentiation. Moreover, the addition of SAR enabled a significant reduction in activin A (AA) from 50 to 10 ng/mL without compromising DE differentiation efficiency. For induction of the pancreatic lineage, 10 ng/ml AA combined with SAR at the DE differentiation stage yielded a comparative number of PDX1+/NKX6.1+ pancreatic progenitor cells to those obtained by 50 ng/ml AA treatment. Our study highlights SAR as a potential modulator that facilitates the cost-effective generation of DE cells and provides insight into the orchestration of cell fate determination.


Asunto(s)
Benzodioxoles , Diferenciación Celular , Endodermo , Quinazolinas , Transducción de Señal , Humanos , Diferenciación Celular/efectos de los fármacos , Endodermo/efectos de los fármacos , Endodermo/citología , Endodermo/metabolismo , Benzodioxoles/farmacología , Transducción de Señal/efectos de los fármacos , Quinazolinas/farmacología , Factores de Transcripción/metabolismo , Células Madre Pluripotentes Inducidas/efectos de los fármacos , Células Madre Pluripotentes Inducidas/metabolismo , Células Madre Pluripotentes Inducidas/citología , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Proteínas Señalizadoras YAP/metabolismo , Quinasa 1 de Adhesión Focal/metabolismo , Quinasa 1 de Adhesión Focal/genética , Células Madre Embrionarias Humanas/efectos de los fármacos , Células Madre Embrionarias Humanas/metabolismo , Células Madre Embrionarias Humanas/citología , Activinas/metabolismo , Simulación del Acoplamiento Molecular
3.
Stem Cells ; 39(4): 429-442, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33400835

RESUMEN

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.


Asunto(s)
Técnicas de Cultivo de Célula , Endodermo/citología , Células Madre Pluripotentes Inducidas/citología , Intestinos/citología , Hígado/citología , Páncreas/citología , Proyectos de Investigación , Biomarcadores/metabolismo , Ácido Butírico/farmacología , Factor de Transcripción CDX2/genética , Factor de Transcripción CDX2/metabolismo , Diferenciación Celular/efectos de los fármacos , Línea Celular , Linaje de la Célula/efectos de los fármacos , Endodermo/efectos de los fármacos , Endodermo/metabolismo , Análisis Factorial , Factor 2 de Crecimiento de Fibroblastos/farmacología , Expresión Génica , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Humanos , Células Madre Pluripotentes Inducidas/efectos de los fármacos , Células Madre Pluripotentes Inducidas/metabolismo , Intestinos/efectos de los fármacos , Intestinos/metabolismo , Hígado/efectos de los fármacos , Hígado/metabolismo , Factor 3 de Transcripción de Unión a Octámeros/genética , Factor 3 de Transcripción de Unión a Octámeros/metabolismo , Páncreas/efectos de los fármacos , Páncreas/metabolismo , Transducción de Señal , Transactivadores/genética , Transactivadores/metabolismo , Tretinoina/farmacología , alfa-Fetoproteínas/genética , alfa-Fetoproteínas/metabolismo
4.
Ann Clin Lab Sci ; 50(4): 468-473, 2020 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-32826243

RESUMEN

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.


Asunto(s)
Activinas/farmacología , Diferenciación Celular/efectos de los fármacos , Proteína Wnt3A/farmacología , Células Cultivadas , Células Madre Embrionarias/citología , Células Madre Embrionarias/efectos de los fármacos , Células Madre Embrionarias/metabolismo , Endodermo/citología , Endodermo/efectos de los fármacos , Endodermo/metabolismo , Humanos , Transducción de Señal/efectos de los fármacos , Proteína Wnt3A/metabolismo
5.
Biochem Biophys Res Commun ; 527(3): 811-817, 2020 06 30.
Artículo en Inglés | MEDLINE | ID: mdl-32446562

RESUMEN

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.


Asunto(s)
Endodermo/citología , Células Madre Embrionarias Humanas/citología , Células Madre Embrionarias Humanas/efectos de los fármacos , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Quinasas p38 Activadas por Mitógenos/antagonistas & inhibidores , Diferenciación Celular/efectos de los fármacos , Línea Celular , Endodermo/efectos de los fármacos , Células Madre Embrionarias Humanas/metabolismo , Humanos , Páncreas/citología , Páncreas/efectos de los fármacos , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
6.
Thyroid ; 30(4): 609-620, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-31801416

RESUMEN

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.


Asunto(s)
Diferenciación Celular/efectos de los fármacos , Células Madre Embrionarias/efectos de los fármacos , Endodermo/efectos de los fármacos , Epigénesis Genética/efectos de los fármacos , Expresión Génica/efectos de los fármacos , Yoduro de Sodio/farmacología , Glándula Tiroides/efectos de los fármacos , Animales , Células Madre Embrionarias/citología , Endodermo/citología , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Ratones , Glándula Tiroides/citología
7.
Cell Death Dis ; 10(10): 763, 2019 10 10.
Artículo en Inglés | MEDLINE | ID: mdl-31601782

RESUMEN

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.


Asunto(s)
Técnicas de Cultivo de Célula/métodos , Medios de Cultivo/química , Endodermo/citología , Hepatocitos/citología , Células Madre Embrionarias Humanas/citología , Activinas/farmacología , Animales , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/genética , Sistema Enzimático del Citocromo P-450/metabolismo , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Endodermo/efectos de los fármacos , Endodermo/metabolismo , Regulación del Desarrollo de la Expresión Génica/genética , Hepatocitos/metabolismo , Células Madre Embrionarias Humanas/efectos de los fármacos , Células Madre Embrionarias Humanas/metabolismo , Humanos , Subunidad gamma Común de Receptores de Interleucina/genética , Subunidad gamma Común de Receptores de Interleucina/metabolismo , Hígado/citología , Fallo Hepático/terapia , Regeneración Hepática , Masculino , Ratones , Ratones Noqueados , Familia de Multigenes , Piridinas/farmacología , Pirimidinas/farmacología , Trasplante de Células Madre , Transcriptoma/genética
8.
Exp Cell Res ; 384(2): 111642, 2019 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-31562862

RESUMEN

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.


Asunto(s)
Amnios/efectos de los fármacos , Diferenciación Celular/efectos de los fármacos , Diabetes Mellitus Experimental/terapia , Células Epiteliales/efectos de los fármacos , Ácido Hialurónico/farmacología , Activinas/metabolismo , Amnios/metabolismo , Animales , Técnicas de Cultivo de Célula/métodos , Tratamiento Basado en Trasplante de Células y Tejidos/métodos , Células Cultivadas , Modelos Animales de Enfermedad , Células Madre Embrionarias/efectos de los fármacos , Células Madre Embrionarias/metabolismo , Endodermo/efectos de los fármacos , Endodermo/metabolismo , Células Epiteliales/metabolismo , Humanos , Inflamación/metabolismo , Insulina/metabolismo , Células Secretoras de Insulina/efectos de los fármacos , Células Secretoras de Insulina/metabolismo , Masculino , Ratones , Páncreas/efectos de los fármacos , Páncreas/metabolismo
9.
Methods Mol Biol ; 1994: 41-53, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31124103

RESUMEN

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.


Asunto(s)
Técnicas de Cultivo de Célula/métodos , Diferenciación Celular , Endodermo/citología , Endodermo/metabolismo , Hepatocitos/citología , Células Madre Pluripotentes Inducidas/citología , Diferenciación Celular/efectos de los fármacos , Separación Celular/métodos , Células Cultivadas , Endodermo/efectos de los fármacos , Proteínas Fetales/metabolismo , Citometría de Flujo , Hepatocitos/metabolismo , Humanos , Células Madre Pluripotentes Inducidas/efectos de los fármacos , Células Madre Pluripotentes Inducidas/metabolismo , Receptores CXCR4/metabolismo , Albúmina Sérica Humana/metabolismo
10.
Biochem Biophys Res Commun ; 512(2): 399-404, 2019 04 30.
Artículo en Inglés | MEDLINE | ID: mdl-30902394

RESUMEN

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.


Asunto(s)
Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Células Madre Embrionarias de Ratones/efectos de los fármacos , Células Madre Embrionarias de Ratones/metabolismo , Inhibidores de Proteínas Quinasas/farmacología , Animales , Benzamidas/farmacología , Diferenciación Celular/efectos de los fármacos , Células Cultivadas , Difenilamina/análogos & derivados , Difenilamina/farmacología , Doxiciclina/farmacología , Endodermo/citología , Endodermo/efectos de los fármacos , Factor de Transcripción GATA4/antagonistas & inhibidores , Factor de Transcripción GATA4/genética , Factor de Transcripción GATA4/metabolismo , Glucógeno Sintasa Quinasa 3 beta/antagonistas & inhibidores , Factor 4 Similar a Kruppel , Ratones , Proteína Quinasa 1 Activada por Mitógenos/antagonistas & inhibidores , Proteína Quinasa 3 Activada por Mitógenos/antagonistas & inhibidores , Células Madre Embrionarias de Ratones/citología , Fosforilación , Piridinas/farmacología , Pirimidinas/farmacología
11.
Proc Natl Acad Sci U S A ; 116(4): 1384-1393, 2019 01 22.
Artículo en Inglés | MEDLINE | ID: mdl-30606801

RESUMEN

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.


Asunto(s)
Diferenciación Celular/fisiología , Células Madre Pluripotentes Inducidas/metabolismo , Células Madre Pluripotentes Inducidas/fisiología , Interferón Tipo I/metabolismo , Antivirales/farmacología , Biomarcadores/metabolismo , Diferenciación Celular/efectos de los fármacos , Células Cultivadas , Reprogramación Celular/fisiología , Ectodermo/efectos de los fármacos , Ectodermo/metabolismo , Ectodermo/fisiología , Ectodermo/virología , Endodermo/efectos de los fármacos , Endodermo/metabolismo , Endodermo/fisiología , Endodermo/virología , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Fibroblastos/fisiología , Fibroblastos/virología , Estratos Germinativos/efectos de los fármacos , Estratos Germinativos/metabolismo , Estratos Germinativos/fisiología , Estratos Germinativos/virología , Humanos , Células Madre Pluripotentes Inducidas/efectos de los fármacos , Células Madre Pluripotentes Inducidas/virología , Factor 4 Similar a Kruppel , ARN Viral/genética , Factores de Transcripción/metabolismo , Regulación hacia Arriba/efectos de los fármacos , Regulación hacia Arriba/fisiología
12.
J Hazard Mater ; 364: 581-590, 2019 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-30388642

RESUMEN

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.


Asunto(s)
Cadmio/química , Lípidos/biosíntesis , Raíces de Plantas/efectos de los fármacos , Silicio/farmacología , Triticum/efectos de los fármacos , Adsorción , Biomasa , Cadmio/metabolismo , Pared Celular/efectos de los fármacos , Pared Celular/metabolismo , Endodermo/efectos de los fármacos , Endodermo/metabolismo , Raíces de Plantas/química , Raíces de Plantas/crecimiento & desarrollo , Raíces de Plantas/metabolismo , Triticum/química , Triticum/crecimiento & desarrollo , Triticum/metabolismo
13.
Nat Protoc ; 14(1): 28-50, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30470820

RESUMEN

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.


Asunto(s)
Técnicas de Cultivo de Célula , Endodermo/citología , Células Epiteliales/citología , Fundus Gástrico/citología , Organoides/citología , Células Madre Pluripotentes/citología , Antro Pilórico/citología , Activinas/farmacología , Benzamidas/farmacología , Proteínas Portadoras/farmacología , Diferenciación Celular , Colágeno/química , Medios de Cultivo/química , Medios de Cultivo/farmacología , Difenilamina/análogos & derivados , Difenilamina/farmacología , Combinación de Medicamentos , Endodermo/efectos de los fármacos , Endodermo/metabolismo , Factor de Crecimiento Epidérmico/farmacología , Células Epiteliales/efectos de los fármacos , Células Epiteliales/metabolismo , Factor 10 de Crecimiento de Fibroblastos/farmacología , Factor 4 de Crecimiento de Fibroblastos/farmacología , Fundus Gástrico/metabolismo , Humanos , Laminina/química , Especificidad de Órganos , Organoides/efectos de los fármacos , Organoides/metabolismo , Células Madre Pluripotentes/efectos de los fármacos , Células Madre Pluripotentes/metabolismo , Proteoglicanos/química , Antro Pilórico/metabolismo , Piridinas/farmacología , Pirimidinas/farmacología , Tretinoina/farmacología , Vía de Señalización Wnt/efectos de los fármacos
14.
Nature ; 564(7735): 219-224, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30518857

RESUMEN

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.


Asunto(s)
Linaje de la Célula , Rastreo Celular/métodos , Reprogramación Celular , Células Clonales/citología , Análisis de la Célula Individual/métodos , Animales , Linaje de la Célula/efectos de los fármacos , Separación Celular , Reprogramación Celular/efectos de los fármacos , Células Clonales/efectos de los fármacos , Endodermo/citología , Endodermo/efectos de los fármacos , Fibroblastos/citología , Fibroblastos/efectos de los fármacos , Células HEK293 , Humanos , Metiltransferasas/metabolismo , Ratones , Células Madre/citología , Células Madre/efectos de los fármacos , Factores de Tiempo
15.
Differentiation ; 101: 16-24, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29626713

RESUMEN

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.


Asunto(s)
Diferenciación Celular/efectos de los fármacos , Hepatocitos/efectos de los fármacos , Células Madre Pluripotentes/efectos de los fármacos , Piridinas/farmacología , Pirimidinas/farmacología , Endodermo/citología , Endodermo/efectos de los fármacos , Expresión Génica/efectos de los fármacos , Hepatocitos/citología , Humanos , Hígado/efectos de los fármacos , Células Madre Pluripotentes/citología
16.
Exp Cell Res ; 362(1): 227-234, 2018 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-29157981

RESUMEN

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.


Asunto(s)
Proteína Forkhead Box O1/metabolismo , Células Madre Embrionarias Humanas/metabolismo , Células Secretoras de Insulina/metabolismo , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/fisiología , Línea Celular , Endodermo/efectos de los fármacos , Endodermo/metabolismo , Glucosa/metabolismo , Proteínas de Homeodominio/metabolismo , Células Madre Embrionarias Humanas/efectos de los fármacos , Humanos , Insulina/metabolismo , Células Secretoras de Insulina/efectos de los fármacos , Islotes Pancreáticos/efectos de los fármacos , Islotes Pancreáticos/metabolismo , Quinolonas/farmacología , Transactivadores/metabolismo
17.
J Cell Physiol ; 233(4): 3578-3589, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29044512

RESUMEN

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.


Asunto(s)
Activinas/farmacología , Ácido Butírico/farmacología , Células Madre Embrionarias/efectos de los fármacos , Endodermo/efectos de los fármacos , Hepatocitos/efectos de los fármacos , Diferenciación Celular/efectos de los fármacos , Línea Celular , Células Cultivadas , Células Madre Embrionarias/citología , Endodermo/citología , Hepatocitos/metabolismo , Humanos , Células Madre Pluripotentes/citología , Células Madre Pluripotentes/efectos de los fármacos
18.
Nat Cell Biol ; 19(10): 1164-1177, 2017 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-28945231

RESUMEN

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.


Asunto(s)
Diferenciación Celular/efectos de los fármacos , Autorrenovación de las Células/efectos de los fármacos , Células Madre Embrionarias/efectos de los fármacos , Endodermo/efectos de los fármacos , Insulina/farmacología , Células Madre Pluripotentes/efectos de los fármacos , Activinas/farmacología , Animales , Línea Celular , Linaje de la Célula , Técnicas de Cultivo de Embriones , Células Madre Embrionarias/metabolismo , Endodermo/citología , Endodermo/metabolismo , Regulación del Desarrollo de la Expresión Génica , Edad Gestacional , Factor Inhibidor de Leucemia/farmacología , Ratones Endogámicos C57BL , Proteína Nodal/farmacología , Células Madre Pluripotentes/metabolismo , Factores de Tiempo , Transfección , Vía de Señalización Wnt/efectos de los fármacos , Proteína Wnt3A/farmacología
19.
Development ; 144(9): 1629-1634, 2017 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-28289133

RESUMEN

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.


Asunto(s)
Endodermo/embriología , Proteínas de Homeodominio/metabolismo , Desarrollo de Músculos , Músculos Faríngeos/embriología , Faringe/embriología , Animales , Ciona intestinalis , Endodermo/efectos de los fármacos , Retroalimentación Fisiológica/efectos de los fármacos , Desarrollo de Músculos/efectos de los fármacos , Músculos Faríngeos/efectos de los fármacos , Faringe/efectos de los fármacos , Tretinoina/farmacología
20.
Biochem Biophys Res Commun ; 486(2): 481-485, 2017 04 29.
Artículo en Inglés | MEDLINE | ID: mdl-28322785

RESUMEN

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.


Asunto(s)
Endodermo/efectos de los fármacos , Lentivirus/genética , Células Madre Embrionarias de Ratones/efectos de los fármacos , Plásmidos/metabolismo , Puromicina/farmacología , ARN Interferente Pequeño/genética , Animales , Artefactos , Diferenciación Celular/efectos de los fármacos , Línea Celular , Endodermo/citología , Endodermo/metabolismo , Expresión Génica/efectos de los fármacos , Biblioteca de Genes , Silenciador del Gen , Lentivirus/metabolismo , Ratones , Células Madre Embrionarias de Ratones/citología , Células Madre Embrionarias de Ratones/metabolismo , Plásmidos/química , ARN Interferente Pequeño/metabolismo , Transfección
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA