Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 51
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Diabetologia ; 67(6): 1066-1078, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38630142

RESUMEN

AIMS/HYPOTHESIS: Rodent pancreas development has been described in great detail. On the other hand, there are still gaps in our understanding of the developmental trajectories of pancreatic cells during human ontogenesis. Here, our aim was to map the spatial and chronological dynamics of human pancreatic cell differentiation and proliferation by using 3D imaging of cleared human embryonic and fetal pancreases. METHODS: We combined tissue clearing with light-sheet fluorescence imaging in human embryonic and fetal pancreases during the first trimester of pregnancy. In addition, we validated an explant culture system enabling in vitro proliferation of pancreatic progenitors to determine the mitogenic effect of candidate molecules. RESULTS: We detected the first insulin-positive cells as early as five post-conceptional weeks, two weeks earlier than previously observed. We observed few insulin-positive clusters at five post-conceptional weeks (mean ± SD 9.25±5.65) with a sharp increase to 11 post-conceptional weeks (4307±152.34). We identified a central niche as the location of onset of the earliest insulin cell production and detected extra-pancreatic loci within the adjacent developing gut. Conversely, proliferating pancreatic progenitors were located in the periphery of the epithelium, suggesting the existence of two separated pancreatic niches for differentiation and proliferation. Additionally, we observed that the proliferation ratio of progenitors ranged between 20% and 30%, while for insulin-positive cells it was 1%. We next unveiled a mitogenic effect of the platelet-derived growth factor AA isoform (PDGFAA) in progenitors acting through the pancreatic mesenchyme by increasing threefold the number of proliferating progenitors. CONCLUSIONS/INTERPRETATION: This work presents a first 3D atlas of the human developing pancreas, charting both endocrine and proliferating cells across early development.


Asunto(s)
Diferenciación Celular , Proliferación Celular , Imagenología Tridimensional , Páncreas , Humanos , Páncreas/embriología , Páncreas/citología , Páncreas/metabolismo , Diferenciación Celular/fisiología , Femenino , Células Madre/citología , Células Madre/metabolismo , Embarazo , Insulina/metabolismo
2.
Cell Stem Cell ; 30(1): 38-51.e8, 2023 01 05.
Artículo en Inglés | MEDLINE | ID: mdl-36563694

RESUMEN

MODY3 is a monogenic hereditary form of diabetes caused by mutations in the transcription factor HNF1A. The patients progressively develop hyperglycemia due to perturbed insulin secretion, but the pathogenesis is unknown. Using patient-specific hiPSCs, we recapitulate the insulin secretion sensitivity to the membrane depolarizing agent sulfonylurea commonly observed in MODY3 patients. Unexpectedly, MODY3 patient-specific HNF1A+/R272C ß cells hypersecrete insulin both in vitro and in vivo after transplantation into mice. Consistently, we identified a trend of increased birth weight in human HNF1A mutation carriers compared with healthy siblings. Reduced expression of potassium channels, specifically the KATP channel, in MODY3 ß cells, increased calcium signaling, and rescue of the insulin hypersecretion phenotype by pharmacological targeting ATP-sensitive potassium channels or low-voltage-activated calcium channels suggest that more efficient membrane depolarization underlies the hypersecretion of insulin in MODY3 ß cells. Our findings identify a pathogenic mechanism leading to ß cell failure in MODY3.


Asunto(s)
Diabetes Mellitus Tipo 2 , Células Secretoras de Insulina , Humanos , Ratones , Animales , Insulina/metabolismo , Células Secretoras de Insulina/metabolismo , Diabetes Mellitus Tipo 2/genética , Fenotipo
3.
STAR Protoc ; 2(4): 100806, 2021 12 17.
Artículo en Inglés | MEDLINE | ID: mdl-34632415

RESUMEN

We have developed a protocol to quantify the position of a cell in a branched structure based on two-dimensional microscopy images of tissue sections. Biological branched structures include organs such as the lungs, kidneys, and pancreas. In these organs, cell fate has been correlated with position, based on a qualitative estimate. However, a quantitative means of evaluating the cell position has been lacking. With this protocol, the correlation between cell fate and cell position was measured in mouse embryonic pancreas. For complete details on the use and execution of this protocol, please refer to Nyeng et al. (2019).


Asunto(s)
Procesamiento de Imagen Asistido por Computador/métodos , Inmunohistoquímica/métodos , Microscopía/métodos , Animales , Embrión de Mamíferos/química , Embrión de Mamíferos/citología , Femenino , Riñón/citología , Pulmón/citología , Masculino , Ratones , Páncreas/citología
4.
STAR Protoc ; 2(3): 100636, 2021 09 17.
Artículo en Inglés | MEDLINE | ID: mdl-34258596

RESUMEN

We here report a flow-cytometry-based protocol to measure single-cell protein expression in small samples. The protocol is optimized for simultaneous detection of fluorescent proteins and intracellular and surface antigens in the embryonic pancreas from the mouse. Owing to low cell numbers, current protocols for flow cytometric analysis of embryonic tissues rely on tissue pooling. Our protocol enables analysis of one pancreas per sample, thereby facilitating detection of biological variation and minimizing the number of experimental animals needed. For complete details on the use and execution of this protocol, please refer to Nyeng et al (2019).


Asunto(s)
Antígenos de Superficie/análisis , Antígenos/análisis , Embrión de Mamíferos/inmunología , Citometría de Flujo/métodos , Páncreas/inmunología , Animales , Femenino , Masculino , Ratones , Análisis de la Célula Individual/métodos
5.
Dev Cell ; 49(1): 31-47.e9, 2019 04 08.
Artículo en Inglés | MEDLINE | ID: mdl-30853440

RESUMEN

The mechanism of how organ shape emerges and specifies cell fate is not understood. Pancreatic duct and endocrine lineages arise in a spatially distinct domain from the acinar lineage. Whether these lineages are pre-determined or settle once these niches have been established remains unknown. Here, we reconcile these two apparently opposing models, demonstrating that pancreatic progenitors re-localize to establish the niche that will determine their ultimate fate. We identify a p120ctn-regulated mechanism for coordination of organ architecture and cellular fate mediated by differential E-cadherin based cell sorting. Reduced p120ctn expression is necessary and sufficient to re-localize a subset of progenitors to the peripheral tip domain, where they acquire an acinar fate. The same mechanism is used re-iteratively during endocrine specification, where it balances the choice between the alpha and beta cell fates. In conclusion, organ patterning is regulated by p120ctn-mediated cellular positioning, which precedes and determines pancreatic progenitor fate.


Asunto(s)
Tipificación del Cuerpo/genética , Cateninas/genética , Páncreas/crecimiento & desarrollo , Conductos Pancreáticos/crecimiento & desarrollo , Animales , Cadherinas/genética , Diferenciación Celular/genética , Linaje de la Célula/genética , Movimiento Celular/genética , Desarrollo Embrionario/genética , Citometría de Flujo , Regulación del Desarrollo de la Expresión Génica , Humanos , Islotes Pancreáticos/crecimiento & desarrollo , Islotes Pancreáticos/metabolismo , Ratones , Páncreas/metabolismo , Receptores Notch/genética , Transducción de Señal/genética , Células Madre/metabolismo , Catenina delta
6.
Nature ; 564(7734): 114-118, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30487608

RESUMEN

The pancreas originates from two epithelial evaginations of the foregut, which consist of multipotent epithelial progenitors that organize into a complex tubular epithelial network. The trunk domain of each epithelial branch consists of bipotent pancreatic progenitors (bi-PPs) that give rise to both duct and endocrine lineages, whereas the tips give rise to acinar cells1. Here we identify the extrinsic and intrinsic signalling mechanisms that coordinate the fate-determining transcriptional events underlying these lineage decisions1,2. Single-cell analysis of pancreatic bipotent pancreatic progenitors derived from human embryonic stem cells reveal that cell confinement is a prerequisite for endocrine specification, whereas spreading drives the progenitors towards a ductal fate. Mechanistic studies identify the interaction of extracellular matrix (ECM) with integrin α5 as the extracellular cue that cell-autonomously, via the F-actin-YAP1-Notch mechanosignalling axis, controls the fate of bipotent pancreatic progenitors. Whereas ECM-integrin α5 signalling promotes differentiation towards the duct lineage, endocrinogenesis is stimulated when this signalling cascade is disrupted. This cascade can be disrupted pharmacologically or genetically to convert bipotent pancreatic progenitors derived from human embryonic stem cells to hormone-producing islet cells. Our findings identify the cell-extrinsic and intrinsic mechanotransduction pathway that acts as gatekeeper in the fate decisions of bipotent pancreatic progenitors in the developing pancreas.


Asunto(s)
Diferenciación Celular , Linaje de la Célula , Integrinas/metabolismo , Organogénesis , Páncreas/citología , Transducción de Señal , Células Madre/citología , Células Madre/metabolismo , Actinas/metabolismo , Proteínas Adaptadoras Transductoras de Señales/deficiencia , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Animales , Proteínas de Ciclo Celular , Linaje de la Célula/genética , Forma de la Célula , Proteínas de Unión al ADN/metabolismo , Elementos de Facilitación Genéticos/genética , Femenino , Fibronectinas/metabolismo , Regulación del Desarrollo de la Expresión Génica , Humanos , Integrina alfa5beta1/metabolismo , Masculino , Ratones , Proteínas Musculares/metabolismo , Páncreas/embriología , Páncreas/metabolismo , Fosfoproteínas/deficiencia , Fosfoproteínas/genética , Fosfoproteínas/metabolismo , Receptor Notch1/genética , Análisis de la Célula Individual , Factores de Transcripción de Dominio TEA , Factor de Transcripción HES-1/genética , Factores de Transcripción/metabolismo , Transcripción Genética , Transgenes , Proteínas Señalizadoras YAP
8.
Nat Cell Biol ; 19(11): 1313-1325, 2017 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-29058721

RESUMEN

Apicobasal polarity is known to affect epithelial morphogenesis and cell differentiation, but it remains unknown how these processes are mechanistically orchestrated. We find that ligand-specific EGFR signalling via PI(3)K and Rac1 autonomously modulates apicobasal polarity to enforce the sequential control of morphogenesis and cell differentiation. Initially, EGF controls pancreatic tubulogenesis by negatively regulating apical polarity induction. Subsequently, betacellulin, working via inhibition of atypical protein kinase C (aPKC), causes apical domain constriction within neurogenin3+ endocrine progenitors, which results in reduced Notch signalling, increased neurogenin3 expression, and ß-cell differentiation. Notably, the ligand-specific EGFR output is not driven at the ligand level, but seems to have evolved in response to stage-specific epithelial influences. The EGFR-mediated control of ß-cell differentiation via apical polarity is also conserved in human neurogenin3+ cells. We provide insight into how ligand-specific EGFR signalling coordinates epithelial morphogenesis and cell differentiation via apical polarity dynamics.


Asunto(s)
Polaridad Celular/fisiología , Receptores ErbB/metabolismo , Células Secretoras de Insulina/metabolismo , Células Secretoras de Insulina/fisiología , Organogénesis/fisiología , Transducción de Señal/fisiología , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Diferenciación Celular/fisiología , Células Epiteliales/metabolismo , Ratones , Ratones Noqueados , Morfogénesis/fisiología , Proteínas del Tejido Nervioso/metabolismo , Neuropéptidos/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Proteína Quinasa C/metabolismo , Proteína de Unión al GTP rac1/metabolismo
9.
Elife ; 62017 07 21.
Artículo en Inglés | MEDLINE | ID: mdl-28731406

RESUMEN

Information remains scarce on human development compared to animal models. Here, we reconstructed human fetal pancreatic differentiation using cell surface markers. We demonstrate that at 7weeks of development, the glycoprotein 2 (GP2) marks a multipotent cell population that will differentiate into the acinar, ductal or endocrine lineages. Development towards the acinar lineage is paralleled by an increase in GP2 expression. Conversely, a subset of the GP2+ population undergoes endocrine differentiation by down-regulating GP2 and CD142 and turning on NEUROG3, a marker of endocrine differentiation. Endocrine maturation progresses by up-regulating SUSD2 and lowering ECAD levels. Finally, in vitro differentiation of pancreatic endocrine cells derived from human pluripotent stem cells mimics key in vivo events. Our work paves the way to extend our understanding of the origin of mature human pancreatic cell types and how such lineage decisions are regulated.


Asunto(s)
Biomarcadores/metabolismo , Diferenciación Celular , Linaje de la Célula , Feto/citología , Regulación del Desarrollo de la Expresión Génica , Páncreas/citología , Células Acinares/citología , Células Acinares/metabolismo , Células Cultivadas , Células Endocrinas/citología , Células Endocrinas/metabolismo , Femenino , Feto/metabolismo , Humanos , Páncreas/metabolismo , Conductos Pancreáticos/citología , Conductos Pancreáticos/metabolismo , Células Madre Pluripotentes/citología , Células Madre Pluripotentes/metabolismo , Transcriptoma
10.
Cell Rep ; 19(1): 36-49, 2017 04 04.
Artículo en Inglés | MEDLINE | ID: mdl-28380361

RESUMEN

Stem cell-based therapy for type 1 diabetes would benefit from implementation of a cell purification step at the pancreatic endoderm stage. This would increase the safety of the final cell product, allow the establishment of an intermediate-stage stem cell bank, and provide a means for upscaling ß cell manufacturing. Comparative gene expression analysis revealed glycoprotein 2 (GP2) as a specific cell surface marker for isolating pancreatic endoderm cells (PECs) from differentiated hESCs and human fetal pancreas. Isolated GP2+ PECs efficiently differentiated into glucose responsive insulin-producing cells in vitro. We found that in vitro PEC proliferation declines due to enhanced expression of the cyclin-dependent kinase (CDK) inhibitors CDKN1A and CDKN2A. However, we identified a time window when reducing CDKN1A or CDKN2A expression increased proliferation and yield of GP2+ PECs. Altogether, our results contribute tools and concepts toward the isolation and use of PECs as a source for the safe production of hPSC-derived ß cells.


Asunto(s)
Células Madre Embrionarias/metabolismo , Proteínas Ligadas a GPI/metabolismo , Glucosa/metabolismo , Células Secretoras de Insulina/citología , Biomarcadores/metabolismo , Diferenciación Celular , Proliferación Celular , Separación Celular/métodos , Células Cultivadas , Inhibidor p16 de la Quinasa Dependiente de Ciclina , Inhibidor p18 de las Quinasas Dependientes de la Ciclina/genética , Inhibidor p18 de las Quinasas Dependientes de la Ciclina/metabolismo , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/genética , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/metabolismo , Diabetes Mellitus Tipo 1/terapia , Células Madre Embrionarias/citología , Endodermo/citología , Proteínas Ligadas a GPI/genética , Regulación de la Expresión Génica , Estudio de Asociación del Genoma Completo , Humanos , Insulina/metabolismo , Secreción de Insulina , Células Secretoras de Insulina/metabolismo
11.
Endocrinology ; 157(12): 4615-4631, 2016 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-27740873

RESUMEN

Vitamin A-derived retinoic acid (RA) signals are critical for the development of several organs, including the pancreas. However, the tissue-specific control of RA synthesis in organ and cell lineage development has only poorly been addressed in vivo. Here, we show that retinol dehydrogenase-10 (Rdh10), a key enzyme in embryonic RA production, has important functions in pancreas organogenesis and endocrine cell differentiation. Rdh10 was expressed in the developing pancreas epithelium and surrounding mesenchyme. Rdh10 null mutant mouse embryos exhibited dorsal pancreas agenesis and a hypoplastic ventral pancreas with retarded tubulogenesis and branching. Conditional disruption of Rdh10 from the endoderm caused increased mortality, reduced body weight, and lowered blood glucose levels after birth. Endodermal Rdh10 deficiency led to a smaller dorsal pancreas with a reduced density of early glucagon+ and insulin+ cells. During the secondary transition, the reduction of Neurogenin3+ endocrine progenitors in the mutant dorsal pancreas accounted for fewer α- and ß-cells. Changes in the expression of α- and ß-cell-specific transcription factors indicated that Rdh10 might also participate in the terminal differentiation of endocrine cells. Together, our results highlight the importance of both mesenchymal and epithelial Rdh10 for pancreogenesis and the first wave of endocrine cell differentiation. We further propose a model in which the Rdh10-expressing exocrine tissue acts as an essential source of RA signals in the second wave of endocrine cell differentiation.


Asunto(s)
Oxidorreductasas de Alcohol/metabolismo , Diferenciación Celular/fisiología , Organogénesis/fisiología , Páncreas/embriología , Comunicación Paracrina/fisiología , Tretinoina/metabolismo , Oxidorreductasas de Alcohol/genética , Animales , Glucemia/metabolismo , Peso Corporal/genética , Anomalías Congénitas/genética , Anomalías Congénitas/metabolismo , Regulación del Desarrollo de la Expresión Génica , Células Secretoras de Insulina/metabolismo , Ratones , Ratones Noqueados , Páncreas/anomalías , Páncreas/metabolismo
12.
Cell Stem Cell ; 16(6): 639-52, 2015 Jun 04.
Artículo en Inglés | MEDLINE | ID: mdl-25921273

RESUMEN

Canonical Wnt and Nodal signaling are both required for induction of the primitive streak (PS), which guides organization of the early embryo. The Wnt effector ß-catenin is thought to function in these early lineage specification decisions via transcriptional activation of Nodal signaling. Here, we demonstrate a broader role for ß-catenin in PS formation by analyzing its genome-wide binding in a human embryonic stem cell model of PS induction. ß-catenin occupies regulatory regions in numerous PS and neural crest genes, and direct interactions between ß-catenin and the Nodal effectors SMAD2/SMAD3 are required at these regions for PS gene activation. Furthermore, OCT4 binding in proximity to these sites is likewise required for PS induction, suggesting a collaborative interaction between ß-catenin and OCT4. Induction of neural crest genes by ß-catenin is repressed by SMAD2/SMAD3, ensuring proper lineage specification. This study provides mechanistic insight into how Wnt signaling controls early cell lineage decisions.


Asunto(s)
Factor 3 de Transcripción de Unión a Octámeros/metabolismo , Línea Primitiva/metabolismo , Proteína Smad2/metabolismo , Proteína smad3/metabolismo , beta Catenina/metabolismo , Secuencia de Bases , Línea Celular , Linaje de la Célula , Regulación del Desarrollo de la Expresión Génica , Humanos , Modelos Biológicos , Datos de Secuencia Molecular , Cresta Neural/citología , Proteína Nodal/metabolismo , Regiones Promotoras Genéticas/genética , Unión Proteica , Células Madre/citología , Células Madre/metabolismo , Vía de Señalización Wnt/genética
13.
Development ; 141(3): 685-96, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24449844

RESUMEN

Delamination plays a pivotal role during normal development and cancer. Previous work has demonstrated that delamination and epithelial cell movement within the plane of an epithelium are associated with a change in cellular phenotype. However, how this positional change is linked to differentiation remains unknown. Using the developing mouse pancreas as a model system, we show that ß cell delamination and differentiation are two independent events, which are controlled by Cdc42/N-WASP signaling. Specifically, we show that expression of constitutively active Cdc42 in ß cells inhibits ß cell delamination and differentiation. These processes are normally associated with junctional actin and cell-cell junction disassembly and the expression of fate-determining transcription factors, such as Isl1 and MafA. Mechanistically, we demonstrate that genetic ablation of N-WASP in ß cells expressing constitutively active Cdc42 partially restores both delamination and ß cell differentiation. These findings elucidate how junctional actin dynamics via Cdc42/N-WASP signaling cell-autonomously control not only epithelial delamination but also cell differentiation during mammalian organogenesis.


Asunto(s)
Actinas/metabolismo , Diferenciación Celular , Células Secretoras de Insulina/metabolismo , Células Secretoras de Insulina/patología , Transducción de Señal , Proteína Neuronal del Síndrome de Wiskott-Aldrich/metabolismo , Proteína de Unión al GTP cdc42/metabolismo , Animales , Animales Recién Nacidos , Embrión de Mamíferos/metabolismo , Embrión de Mamíferos/patología , Epitelio/metabolismo , Humanos , Hiperglucemia/metabolismo , Hiperglucemia/patología , Uniones Intercelulares/metabolismo , Uniones Intercelulares/patología , Ratones , Ratas , Imagen de Lapso de Tiempo
14.
Development ; 140(21): 4452-62, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-24130330

RESUMEN

In the context of a cellular therapy for diabetes, methods for pancreatic progenitor expansion and subsequent differentiation into insulin-producing beta cells would be extremely valuable. Here we establish three-dimensional culture conditions in Matrigel that enable the efficient expansion of dissociated mouse embryonic pancreatic progenitors. By manipulating the medium composition we generate either hollow spheres, which are mainly composed of pancreatic progenitors, or complex organoids that spontaneously undergo pancreatic morphogenesis and differentiation. The in vitro maintenance and expansion of pancreatic progenitors require active Notch and FGF signaling, thus recapitulating in vivo niche signaling interactions. Our experiments reveal new aspects of pancreas development, such as a community effect by which small groups of cells better maintain progenitor properties and expand more efficiently than isolated cells, as well as the requirement for three-dimensionality. Finally, growth conditions in chemically defined biomaterials pave the way for testing the biophysical and biochemical properties of the niche that sustains pancreatic progenitors.


Asunto(s)
Técnicas de Cultivo de Célula/métodos , Diferenciación Celular/fisiología , Morfogénesis/fisiología , Páncreas/citología , Páncreas/crecimiento & desarrollo , Células Madre/citología , Animales , Colágeno , Combinación de Medicamentos , Hidrogel de Polietilenoglicol-Dimetacrilato , Inmunohistoquímica , Laminina , Ratones , Microscopía Fluorescente , Proteoglicanos , Reacción en Cadena en Tiempo Real de la Polimerasa , Transducción de Señal/fisiología , Estadísticas no Paramétricas , Imagen de Lapso de Tiempo
15.
Sci Rep ; 3: 1381, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23459332

RESUMEN

In embryonic stem cells, removal of oxidatively damaged proteins is triggered upon the first signs of cell fate specification but the underlying mechanism is not known. Here, we report that this phase of differentiation encompasses an unexpected induction of genes encoding the proteasome activator PA28αß (11S), subunits of the immunoproteasome (20Si), and the 20Si regulator TNFα. This induction is accompanied by assembly of mature PA28-20S(i) proteasomes and elevated proteasome activity. Inhibiting accumulation of PA28α using miRNA counteracted the removal of damaged proteins demonstrating that PA28αß has a hitherto unidentified role required for resetting the levels of protein damage at the transition from self-renewal to cell differentiation.


Asunto(s)
Células Madre Embrionarias/citología , Células Madre Embrionarias/metabolismo , Complejo de la Endopetidasa Proteasomal/metabolismo , Proteínas/metabolismo , Animales , Diferenciación Celular , Línea Celular , Linaje de la Célula , Ratones , Carbonilación Proteica , Subunidades de Proteína/metabolismo
16.
Methods ; 59(1): 59-70, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-22503774

RESUMEN

Characterization of directed differentiation of pluripotent stem cells towards therapeutically relevant cell types, including pancreatic beta-cells and hepatocytes, depends on molecular markers and assays that resolve the signature of individual cells. Pancreas and liver both have a common origin of anterior definitive endoderm (DE). Here, we differentiated human embryonic stem cells towards DE using three different activin A based treatments. Differentiation efficiencies were evaluated by gene expression profiling over time at cell population level. A panel of key markers was used to study DE formation. Final DE differentiation was also analyzed with immunocytochemistry and single-cell gene expression profiling. We found that cells treated with activin A in combination with sodium butyrate and B27 serum-free supplement medium generated the most mature DE cells. Cell population studies were useful to monitor the temporal expression of genes involved in primitive streak formation and endoderm formation, while single-cell analysis allowed us to study cell culture heterogeneity and fingerprint individual cells. In addition, single-cell analysis revealed distinct gene expression patterns for the three activin A based protocols applied. Our data provide novel insights in DE gene expression at the cellular level of in vitro differentiated human embryonic stem cells, and illustrate the power of using single-cell gene expression profiling to study differentiation heterogeneity and to characterize cell types and subpopulations.


Asunto(s)
Diferenciación Celular , Células Madre Embrionarias/metabolismo , Perfilación de la Expresión Génica , Análisis de la Célula Individual , Activinas/farmacología , Activinas/fisiología , Antígenos de Diferenciación/genética , Antígenos de Diferenciación/metabolismo , Forma de la Célula , Células Cultivadas , Células Madre Embrionarias/fisiología , Endodermo/citología , Humanos , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factores de Transcripción SOXF/genética , Factores de Transcripción SOXF/metabolismo , Transcriptoma
17.
PLoS One ; 6(10): e26026, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-22043302

RESUMEN

BACKGROUND: Diabetic retinopathy and retinopathy of prematurity are diseases caused by pathological angiogenesis in the retina as a consequence of local hypoxia. The underlying mechanism for epiretinal neovascularization (tuft formation), which contributes to blindness, has yet to be identified. Neural cell adhesion molecule (N-CAM) is expressed by Müller cells and astrocytes, which are in close contact with the retinal vasculature, during normal developmental angiogenesis. METHODOLOGY/PRINCIPAL FINDINGS: Notably, during oxygen induced retinopathy (OIR) N-CAM accumulated on astrocytes surrounding the epiretinal tufts. Here, we show that N-CAM ablation results in reduced vascular tuft formation due to reduced endothelial cell proliferation despite an elevation in VEGFA mRNA expression, whereas retinal developmental angiogenesis was unaffected. CONCLUSION/SIGNIFICANCE: We conclude that N-CAM exhibits a regulatory function in pathological angiogenesis in OIR. This is a novel finding that can be of clinical relevance in diseases associated with proliferative vasculopathy.


Asunto(s)
Retinopatía Diabética/etiología , Hipoxia/complicaciones , Neovascularización Patológica , Moléculas de Adhesión de Célula Nerviosa/fisiología , Neovascularización Retiniana , Animales , Proliferación Celular , Células Endoteliales/patología , Ratones , Moléculas de Adhesión de Célula Nerviosa/análisis , Vasos Retinianos/crecimiento & desarrollo , Vasos Retinianos/patología , Factor A de Crecimiento Endotelial Vascular/genética
18.
Dev Biol ; 352(2): 267-77, 2011 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-21281624

RESUMEN

Endoderm development is dependent on inductive signals from different structures in close vicinity, including the notochord, lateral plate mesoderm and endothelial cells. Recently, we demonstrated that a functional vascular system is necessary for proper pancreas development, and that sphingosine-1-phosphate (S1P) exhibits the traits of a blood vessel-derived molecule involved in early pancreas morphogenesis. To examine whether S1P(1)-signaling plays a more general role in endoderm development, S1P(1)-deficient mice were analyzed. S1P(1) ablation results in compromised growth of several foregut-derived organs, including the stomach, dorsal and ventral pancreas and liver. Within the developing pancreas the reduction in organ size was due to deficient proliferation of Pdx1(+) pancreatic progenitors, whereas endocrine cell differentiation was unaffected. Ablation of endothelial cells in vitro did not mimic the S1P(1) phenotype, instead, increased organ size and hyperbranching were observed. Consistent with a negative role for endothelial cells in endoderm organ expansion, excessive vasculature was discovered in S1P(1)-deficient embryos. Altogether, our results show that endothelial cell hyperplasia negatively influences organ development in several foregut-derived organs.


Asunto(s)
Endodermo/embriología , Células Endoteliales/citología , Células Endoteliales/metabolismo , Animales , Diferenciación Celular , Proliferación Celular , Desarrollo Embrionario , Endodermo/citología , Endodermo/metabolismo , Regulación del Desarrollo de la Expresión Génica , Edad Gestacional , Proteínas de Homeodominio/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Morfogénesis/genética , Morfogénesis/fisiología , Páncreas/irrigación sanguínea , Páncreas/citología , Páncreas/embriología , Páncreas/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Receptores de Lisoesfingolípidos/deficiencia , Receptores de Lisoesfingolípidos/genética , Receptores de Esfingosina-1-Fosfato , Transactivadores/metabolismo
19.
PLoS One ; 5(9)2010 Sep 02.
Artículo en Inglés | MEDLINE | ID: mdl-20824089

RESUMEN

BACKGROUND: Pluripotency and self-renewal of human embryonic stem cells (hESCs) is mediated by a complex interplay between extra- and intracellular signaling pathways, which regulate the expression of pluripotency-specific transcription factors. The homeodomain transcription factor NANOG plays a central role in maintaining hESC pluripotency, but the precise role and regulation of NANOG are not well defined. METHODOLOGY/PRINCIPAL FINDINGS: To facilitate the study of NANOG expression and regulation in viable hESC cultures, we generated fluorescent NANOG reporter cell lines by gene targeting in hESCs. In these reporter lines, the fluorescent reporter gene was co-expressed with endogenous NANOG and responded to experimental induction or repression of the NANOG promoter with appropriate changes in expression levels. Furthermore, NANOG reporter lines facilitated the separation of hESC populations based on NANOG expression levels and their subsequent characterization. Gene expression arrays on isolated hESC subpopulations revealed genes with differential expression in NANOG(high) and NANOG(low) hESCs, providing candidates for NANOG downstream targets hESCs. CONCLUSION/SIGNIFICANCE: The newly derived NANOG reporter hESC lines present novel tools to visualize NANOG expression in viable hESCs. In future applications, these reporter lines can be used to elucidate the function and regulation of NANOG in pluripotent hESCs.


Asunto(s)
Línea Celular/metabolismo , Células Madre Embrionarias/metabolismo , Marcación de Gen , Genes Reporteros , Proteínas de Homeodominio/genética , Regulación de la Expresión Génica , Proteínas de Homeodominio/metabolismo , Humanos , Proteína Homeótica Nanog
20.
PLoS One ; 5(8): e12413, 2010 Aug 26.
Artículo en Inglés | MEDLINE | ID: mdl-20865032

RESUMEN

BACKGROUND: Constitutive promoters that ensure sustained and high level gene expression are basic research tools that have a wide range of applications, including studies of human embryology and drug discovery in human embryonic stem cells (hESCs). Numerous cellular/viral promoters that ensure sustained gene expression in various cell types have been identified but systematic comparison of their activities in hESCs is still lacking. METHODOLOGY/PRINCIPAL FINDINGS: We have quantitatively compared promoter activities of five commonly used constitutive promoters, including the human ß-actin promoter (ACTB), cytomegalovirus (CMV), elongation factor-1α, (EF1α), phosphoglycerate kinase (PGK) and ubiquitinC (UbC) in hESCs. Lentiviral gene transfer was used to ensure stable integration of promoter-eGFP constructs into the hESCs genome. Promoter activities were quantitatively compared in long term culture of undifferentiated hESCs and in their differentiated progenies. CONCLUSION/SIGNIFICANCE: The ACTB, EF1α and PGK promoters showed stable activities during long term culture of undifferentiated hESCs. The ACTB promoter was superior by maintaining expression in 75-80% of the cells after 50 days in culture. During embryoid body (EB) differentiation, promoter activities of all five promoters decreased. Although the EF1α promoter was downregulated in approximately 50% of the cells, it was the most stable promoter during differentiation. Gene expression analysis of differentiated eGFP+ and eGFP- cells indicate that promoter activities might be restricted to specific cell lineages, suggesting the need to carefully select optimal promoters for constitutive gene expression in differentiated hESCs.


Asunto(s)
Diferenciación Celular , Células Madre Embrionarias/citología , Regiones Promotoras Genéticas , Actinas/genética , Línea Celular , Regulación de la Expresión Génica , Humanos , Factor 1 de Elongación Peptídica/genética , Fosfoglicerato Quinasa/genética , Ubiquitina C/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...