Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Stem Cell Reports ; 16(1): 56-74, 2021 01 12.
Artículo en Inglés | MEDLINE | ID: mdl-33382978

RESUMEN

After reprogramming to naive pluripotency, human pluripotent stem cells (PSCs) still exhibit very low ability to make interspecies chimeras. Whether this is because they are inherently devoid of the attributes of chimeric competency or because naive PSCs cannot colonize embryos from distant species remains to be elucidated. Here, we have used different types of mouse, human, and rhesus monkey naive PSCs and analyzed their ability to colonize rabbit and cynomolgus monkey embryos. Mouse embryonic stem cells (ESCs) remained mitotically active and efficiently colonized host embryos. In contrast, primate naive PSCs colonized host embryos with much lower efficiency. Unlike mouse ESCs, they slowed DNA replication after dissociation and, after injection into host embryos, they stalled in the G1 phase and differentiated prematurely, regardless of host species. We conclude that human and non-human primate naive PSCs do not efficiently make chimeras because they are inherently unfit to remain mitotically active during colonization.


Asunto(s)
Diferenciación Celular , Quimera/metabolismo , Puntos de Control de la Fase G1 del Ciclo Celular , Células Madre Pluripotentes/citología , Animales , Apoptosis , Reprogramación Celular , Transferencia de Embrión , Embrión de Mamíferos/citología , Embrión de Mamíferos/metabolismo , Humanos , Macaca mulatta , Ratones , Células Madre Embrionarias de Ratones/citología , Células Madre Embrionarias de Ratones/metabolismo , Células Madre Pluripotentes/metabolismo , Conejos , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
2.
Exp Cell Res ; 386(1): 111712, 2020 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-31697928

RESUMEN

The leukemia inhibitory factor (LIF)/glycoprotein (GP) 130/Janus kinase (JAK)/signal transducer and activator of transcription (STAT) 3 signaling pathway is a hallmark of naïve pluripotency in mice. In this review, we discuss the role of the LIF/GP130/JAK/STAT3 pathway in supporting the naïve-state pluripotency in human, monkey, and pig pluripotent stem cells (PSCs). We highlight the role of LIF/GP130/JAK/STAT3 signaling in reprogramming conventional human and monkey PSCs to naïve-like pluripotency. We analyze published single-cell RNA sequencing datasets of human and monkey embryos and note that the main components of the GP130/JAK/STAT3 pathway are expressed in pluripotent cells at preimplantation stages. We conclude that there is a growing body of evidence supporting the involvement of GP130/JAK/STAT3 in the regulation of naïve pluripotency in non-rodent species including humans, monkeys, and pigs.


Asunto(s)
Diferenciación Celular , Receptor gp130 de Citocinas/metabolismo , Células Madre Pluripotentes/metabolismo , Transducción de Señal , Animales , Haplorrinos , Humanos , Quinasas Janus/metabolismo , Células Madre Pluripotentes/citología , Factores de Transcripción STAT/metabolismo , Porcinos
3.
Cell Cycle ; 18(20): 2697-2712, 2019 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-31462142

RESUMEN

Continuous, non-cell cycle-dependent expression of cyclin E is a characteristic feature of mouse embryonic stem cells (mESCs). We studied the 5' regulatory region of Cyclin E, also known as Ccne1, and identified binding sites for transcription factors of the naïve pluripotency network, including Esrrb, Klf4, and Tfcp2l1 within 1 kilobase upstream of the transcription start site. Luciferase assay and chromatin immunoprecipitation-quantitative polymerase chain reaction (ChiP-qPCR) study highlighted one binding site for Esrrb that is essential to transcriptional activity of the promoter region, and three binding sites for Klf4 and Tfcp2l1. Knockdown of Esrrb, Klf4, and Tfcp2l1 reduced Cyclin E expression whereas overexpression of Esrrb and Klf4 increased it, indicating a strong correlation between the expression level of these factors and that of cyclin E. We observed that cyclin E overexpression delays differentiation induced by Esrrb depletion, suggesting that cyclin E is an important target of Esrrb for differentiation blockade. We observed that mESCs express a low level of miR-15a and that transfection of a miR-15a mimic decreases Cyclin E mRNA level. These results lead to the conclusion that the high expression level of Cyclin E in mESCs can be attributed to transcriptional activation by Esrrb as well as to the absence of its negative regulator, miR-15a.


Asunto(s)
Diferenciación Celular/genética , Ciclina E/metabolismo , Ciclinas/metabolismo , Células Madre Embrionarias de Ratones/metabolismo , Regiones Promotoras Genéticas , Animales , Sitios de Unión , Línea Celular , Inmunoprecipitación de Cromatina , Ciclina E/genética , Ciclinas/genética , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Técnicas de Silenciamiento del Gen , Factor 4 Similar a Kruppel , Factores de Transcripción de Tipo Kruppel/genética , Factores de Transcripción de Tipo Kruppel/metabolismo , Ratones , MicroARNs/genética , MicroARNs/metabolismo , ARN Interferente Pequeño , Receptores de Estrógenos/genética , Receptores de Estrógenos/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Regulación hacia Arriba
4.
Med Sci (Paris) ; 34(11): 944-953, 2018 Nov.
Artículo en Francés | MEDLINE | ID: mdl-30526839

RESUMEN

Since the derivation of the first pluripotent embryonic stem cell lines in mice in the early 1980s, a plethora of lines has been obtained from various mammalian species including rodents, lagomorphs and primates. These lines are distinguished by their molecular and functional characteristics and correspond to the different pluripotency states observed in the developing embryo between the "blastocyst" and "gastrula" stages. These cell lines are positioned along a gradient, or continuum of pluripotency, the ends of which are epitomized by the naïve and primed states, respectively. Conventional human pluripotent stem cells self-renew in the primed state of pluripotency (ie, at the bottom of the gradient), a position that is undoubtedly the cause of their natural instability. Recent studies aim to generate naive human pluripotent stem cells (at the top of the gradient). The importance of this research in the perspective of medical applications will be discussed.


Asunto(s)
Células Madre Pluripotentes/clasificación , Células Madre Pluripotentes/fisiología , Animales , Diferenciación Celular/fisiología , Desarrollo Embrionario/fisiología , Células Madre Embrionarias/fisiología , Regulación del Desarrollo de la Expresión Génica , Células Madre Embrionarias Humanas/fisiología , Humanos , Ratones
5.
Stem Cell Res ; 16(3): 557-67, 2016 05.
Artículo en Inglés | MEDLINE | ID: mdl-26999759

RESUMEN

The imprinted genes of primate embryonic stem cells (ESCs) often show altered DNA methylation. It is unknown whether these alterations emerge while deriving the ESCs. Here we studied the methylation patterns of two differentially methylated regions (DMRs), SNRPN and H19/IGF2 DMRs, during the derivation of monkey ESCs. We show that the SNRPN DMR is characteristically methylated at maternal alleles, whereas the H19/IGF2 DMR is globally highly methylated, with unusual methylation on the maternal alleles. These methylation patterns remain stable from the early stages of ESC derivation to late passages of monkey ESCs and following differentiation. Importantly, the methylation status of H19/IGF2 DMR and the expression levels of IGF2, H19, and DNMT3B mRNAs in early embryo-derived cells were correlated with their capacity to generate genuine ESC lines. Thus, we propose that these markers could be useful to predict the outcomes of establishing an ESC line in primates.


Asunto(s)
Células Madre Embrionarias/citología , Factor II del Crecimiento Similar a la Insulina/genética , ARN Largo no Codificante/genética , Proteínas Nucleares snRNP/genética , Animales , Secuencia de Bases , Línea Celular , Metilación de ADN , Células Madre Embrionarias/metabolismo , Epigenómica , Impresión Genómica , Macaca mulatta , Ratones , Ratones SCID , Teratoma/patología , Trasplante Heterólogo
6.
Nat Commun ; 6: 7095, 2015 May 13.
Artículo en Inglés | MEDLINE | ID: mdl-25968054

RESUMEN

Leukemia inhibitory factor (LIF)/STAT3 signalling is a hallmark of naive pluripotency in rodent pluripotent stem cells (PSCs), whereas fibroblast growth factor (FGF)-2 and activin/nodal signalling is required to sustain self-renewal of human PSCs in a condition referred to as the primed state. It is unknown why LIF/STAT3 signalling alone fails to sustain pluripotency in human PSCs. Here we show that the forced expression of the hormone-dependent STAT3-ER (ER, ligand-binding domain of the human oestrogen receptor) in combination with 2i/LIF and tamoxifen allows human PSCs to escape from the primed state and enter a state characterized by the activation of STAT3 target genes and long-term self-renewal in FGF2- and feeder-free conditions. These cells acquire growth properties, a gene expression profile and an epigenetic landscape closer to those described in mouse naive PSCs. Together, these results show that temporarily increasing STAT3 activity is sufficient to reprogramme human PSCs to naive-like pluripotent cells.


Asunto(s)
Células Madre Embrionarias/fisiología , Regulación de la Expresión Génica/fisiología , Células Madre Pluripotentes/fisiología , Factor de Transcripción STAT3/metabolismo , Animales , Células Madre Embrionarias/citología , Células Madre Embrionarias/efectos de los fármacos , Células Nutrientes , Factor 2 de Crecimiento de Fibroblastos/genética , Factor 2 de Crecimiento de Fibroblastos/metabolismo , Fibroblastos/citología , Fibroblastos/fisiología , Humanos , Factor Inhibidor de Leucemia/genética , Factor Inhibidor de Leucemia/metabolismo , Ratones , Análisis por Matrices de Proteínas , Factor de Transcripción STAT3/genética , Transducción de Señal , Tamoxifeno/farmacología
7.
Nat Commun ; 5: 3719, 2014 Apr 28.
Artículo en Inglés | MEDLINE | ID: mdl-24770696

RESUMEN

Krüppel-like factors (Klf) 4 and 5 are two closely related members of the Klf family, known to play key roles in cell cycle regulation, somatic cell reprogramming and pluripotency. Here we focus on the functional divergence between Klf4 and Klf5 in the inhibition of mouse embryonic stem (ES) cell differentiation. Using microarrays and chromatin immunoprecipitation coupled to ultra-high-throughput DNA sequencing, we show that Klf4 negatively regulates the expression of endodermal markers in the undifferentiated ES cells, including transcription factors involved in the commitment of pluripotent stem cells to endoderm differentiation. Knockdown of Klf4 enhances differentiation towards visceral and definitive endoderm. In contrast, Klf5 negatively regulates the expression of mesodermal markers, some of which control commitment to the mesoderm lineage, and knockdown of Klf5 specifically enhances differentiation towards mesoderm. We conclude that Klf4 and Klf5 differentially inhibit mesoderm and endoderm differentiation in murine ES cells.


Asunto(s)
Diferenciación Celular/fisiología , Células Madre Embrionarias/fisiología , Endodermo/embriología , Regulación del Desarrollo de la Expresión Génica/fisiología , Factores de Transcripción de Tipo Kruppel/metabolismo , Mesodermo/embriología , Animales , Western Blotting , Inmunoprecipitación de Cromatina , Citometría de Flujo , Técnicas de Silenciamiento del Gen , Secuenciación de Nucleótidos de Alto Rendimiento , Factor 4 Similar a Kruppel , Factores de Transcripción de Tipo Kruppel/genética , Ratones , Análisis por Micromatrices , Reacción en Cadena en Tiempo Real de la Polimerasa
8.
Stem Cell Res ; 10(1): 118-31, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23178806

RESUMEN

A short G1 phase is a characteristic feature of mouse embryonic stem cells (ESCs). To determine if there is a causal relationship between G1 phase restriction and pluripotency, we made use of the Fluorescence Ubiquitination Cell Cycle Indicator (FUCCI) reporter system to FACS-sort ESCs in the different cell cycle phases. Hence, the G1 phase cells appeared to be more susceptible to differentiation, particularly when ESCs self-renewed in the naïve state of pluripotency. Transitions from ground to naïve, then from naïve to primed states of pluripotency were associated with increased durations of the G1 phase, and cyclin E-mediated alteration of the G1/S transition altered the balance between self-renewal and differentiation. LIF withdrawal resulted in a lengthening of the G1 phase in naïve ESCs, which occurred prior to the appearance of early lineage-specific markers, and could be reversed upon LIF supplementation. We concluded that the short G1 phase observed in murine ESCs was a determinant of naïve pluripotency and was partially under the control of LIF signaling.


Asunto(s)
Células Madre Embrionarias/citología , Fase G1 , Animales , Diferenciación Celular , Ciclina E/antagonistas & inhibidores , Ciclina E/genética , Ciclina E/metabolismo , Fase G1/efectos de los fármacos , Humanos , Factor Inhibidor de Leucemia/farmacología , Ratones , Microscopía Confocal , Interferencia de ARN , ARN Interferente Pequeño/metabolismo , Ratas , Transducción de Señal , Imagen de Lapso de Tiempo , Ubiquitinación
9.
Differentiation ; 81(3): 142-52, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21296479

RESUMEN

Non-human primate (NHP) embryonic stem (ES) cells show unlimited proliferative capacities and a great potential to generate multiple cell lineages. These properties make them an ideal resource both for investigating early developmental processes and for assessing their therapeutic potential in numerous models of degenerative diseases. They share the same markers and the same properties with human ES cells, and thus provide an invaluable transitional model that can be used to address the safety issues related to the clinical use of human ES cells. Here, we review the available information on the derivation and the specific features of monkey ES cells. We comment on the capacity of primate ES cells to differentiate into neural lineages and the current protocols to generate self-renewing neural stem cells. We also highlight the signalling pathways involved in the maintenance of these neural cell types. Finally, we discuss the potential of monkey ES cells for neuronal differentiation.


Asunto(s)
Diferenciación Celular/fisiología , Células Madre Embrionarias/fisiología , Haplorrinos , Neuronas/fisiología , Animales , Biomarcadores/metabolismo , Ciclo Celular/fisiología , Células Madre Embrionarias/citología , Humanos , Neuronas/citología , Transducción de Señal/fisiología
10.
BMC Biol ; 8: 125, 2010 Sep 27.
Artículo en Inglés | MEDLINE | ID: mdl-20875146

RESUMEN

Recent papers have demonstrated a role for Krüppel-like transcription factors 2, 4 and 5 in the control of mouse embryonic stem cell pluripotency. However, it is not clear whether each factor has a unique role or whether they are functionally redundant. A paper by Parisi and colleagues in BMC Biology now sheds light on the mechanism by which Klf5 regulates pluripotency. See research article http://www.biomedcentral.com/1741-7007/8/128.


Asunto(s)
Regulación de la Expresión Génica/genética , Factores de Transcripción de Tipo Kruppel/metabolismo , Células Madre Pluripotentes/citología , Células Madre Pluripotentes/metabolismo , Animales , Diferenciación Celular/fisiología , Ratones , Regulón/genética
11.
PLoS One ; 4(9): e6804, 2009 Sep 03.
Artículo en Inglés | MEDLINE | ID: mdl-19727443

RESUMEN

Embryonic stem (ES) cells have high self-renewal capacity and the potential to differentiate into a large variety of cell types. To investigate gene networks operating in pluripotent ES cells and their derivatives, the "Functional Genomics in Embryonic Stem Cells" consortium (FunGenES) has analyzed the transcriptome of mouse ES cells in eleven diverse settings representing sixty-seven experimental conditions. To better illustrate gene expression profiles in mouse ES cells, we have organized the results in an interactive database with a number of features and tools. Specifically, we have generated clusters of transcripts that behave the same way under the entire spectrum of the sixty-seven experimental conditions; we have assembled genes in groups according to their time of expression during successive days of ES cell differentiation; we have included expression profiles of specific gene classes such as transcription regulatory factors and Expressed Sequence Tags; transcripts have been arranged in "Expression Waves" and juxtaposed to genes with opposite or complementary expression patterns; we have designed search engines to display the expression profile of any transcript during ES cell differentiation; gene expression data have been organized in animated graphs of KEGG signaling and metabolic pathways; and finally, we have incorporated advanced functional annotations for individual genes or gene clusters of interest and links to microarray and genomic resources. The FunGenES database provides a comprehensive resource for studies into the biology of ES cells.


Asunto(s)
Bases de Datos Genéticas , Genómica , Células Madre/citología , Animales , Diferenciación Celular , Línea Celular , Análisis por Conglomerados , Etiquetas de Secuencia Expresada , Perfilación de la Expresión Génica , Ratones , Familia de Multigenes , Análisis de Secuencia por Matrices de Oligonucleótidos , Transducción de Señal , Programas Informáticos
12.
Stem Cells ; 27(8): 1760-71, 2009 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-19544440

RESUMEN

Leukemia inhibitory factor (LIF) activates the transcription factor signal transducer and activator of transcription 3 (STAT3), which results in the maintenance of mouse embryonic stem cells in the pluripotent state by inhibiting both mesodermal and endodermal differentiation. How the LIF/STAT3 pathway inhibits commitment to both mesoderm and endoderm lineages is presently unknown. Using a hormone-dependent STAT3 and with microarray analysis, we identified 58 targets of STAT3 including 20 unknown genes. Functional analysis showed that 22 among the 23 STAT3 target genes analyzed contribute to the maintenance of the undifferentiated state, as evidenced by an increase in the frequency of differentiated colonies in a self-renewal assay and a concomitant elevation of early differentiation markers upon knockdown. Fourteen of them, including Dact1, Klf4, Klf5, Rgs16, Smad7, Ccrn4l, Cnnm1, Ocln, Ier3, Pim1, Cyr61, and Sgk, were also regulated by Nanog. Analysis of lineage-specific markers showed that the STAT3 target genes fell into three distinct categories, depending on their capacity to inhibit either mesoderm or endoderm differentiation or both. The identification of genes that harness self-renewal and are downstream targets of both STAT3 and Nanog shed light on the mechanisms underlying functional redundancy between STAT3 and Nanog in mouse embryonic stem cells.


Asunto(s)
Ectodermo/fisiología , Células Madre Embrionarias/fisiología , Proteínas de Homeodominio/genética , Mesodermo/fisiología , Factor de Transcripción STAT3/genética , Animales , Diferenciación Celular/genética , Procesos de Crecimiento Celular/genética , Línea Celular , Ectodermo/citología , Células Madre Embrionarias/citología , Células Madre Embrionarias/metabolismo , Regulación de la Expresión Génica , Proteínas de Homeodominio/metabolismo , Factor 4 Similar a Kruppel , Factor Inhibidor de Leucemia , Mesodermo/citología , Ratones , Ratones SCID , Proteína Homeótica Nanog , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factor de Transcripción STAT3/metabolismo , Transducción de Señal , Activación Transcripcional
13.
Stem Cells ; 25(12): 2996-3004, 2007 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-17717068

RESUMEN

pim-1 and pim-3 encode serine/threonine kinases involved in the regulation of cell proliferation and apoptosis in response to cytokine stimulation. We analyzed the regulation of pim-1 and pim-3 by the leukemia inhibitory factor (LIF)/gp130/signal transducer and activator of transcription-3 (STAT3) pathway and the role of Pim-1 and Pim-3 kinases in mouse embryonic stem (ES) cell self-renewal. Making use of ES cells expressing a granulocyte colony-stimulating factor:gp130 chimeric receptor and a hormone-dependent signal transducer and activator of transcription-3 estrogen receptor (STAT3-ER(T2)), we showed that expression of pim-1 and pim-3 was upregulated by LIF/gp130-dependent signaling and the STAT3 transcription factor. ES cells overexpressing pim-1 and pim-3 had a greater capacity to self-renew and displayed a greater resistance to LIF starvation based on a clonal assay. In contrast, knockdown of pim-1 and pim-3 increased the rate of spontaneous differentiation in a self-renewal assay. Knockdown of pim-1 and pim-3 was also detrimental to the growth of undifferentiated ES cell colonies and increased the rate of apoptosis. These findings provide a novel role of Pim-1 and Pim-3 kinases in the control of self-renewal of ES cells. Disclosure of potential conflicts of interest is found at the end of this article.


Asunto(s)
Diferenciación Celular/fisiología , Células Madre Embrionarias/citología , Células Madre Embrionarias/enzimología , Proteínas Serina-Treonina Quinasas/fisiología , Proteínas Proto-Oncogénicas c-pim-1/fisiología , Proteínas Proto-Oncogénicas/fisiología , Animales , Proliferación Celular , Células Cultivadas , Ratones , Proteínas Serina-Treonina Quinasas/genética , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas c-pim-1/genética
14.
J Biol Chem ; 282(9): 6265-73, 2007 Mar 02.
Artículo en Inglés | MEDLINE | ID: mdl-17204467

RESUMEN

Embryonic stem (ES) cell pluripotency is regulated by a combination of extrinsic and intrinsic factors. Previously we have demonstrated that phosphoinositide 3-kinase (PI3K)-dependent signaling is required for efficient self-renewal of murine ES cells. In the study presented here, we have investigated the downstream molecular mechanisms that contribute to the ability of PI3Ks to regulate pluripotency. We show that inhibition of PI3K activity with either pharmacological or genetic tools results in decreased expression of RNA for the homeodomain transcription factor Nanog and decreased Nanog protein levels. Inhibition of glycogen synthase kinase 3 (GSK-3) activity by PI3Ks plays a key role in regulation of Nanog expression, because blockade of GSK-3 activity effectively reversed the effects of PI3K inhibition on Nanog RNA, and protein expression and self-renewal under these circumstances were restored. Furthermore, GSK-3 mutants mimicked the effects of PI3K or GSK-3 inhibition on Nanog expression. Importantly, expression of an inducible form of Nanog prevented the loss of self-renewal observed upon inhibition of PI3Ks, supporting a functional relationship between PI3Ks and Nanog expression. In addition, expression of a number of putative Nanog target genes was sensitive to PI3K inhibition. Thus, the new evidence provided in this study shows that PI3K-dependent regulation of ES cell self-renewal is mediated, at least in part, by the ability of PI3K signaling to maintain Nanog expression. Regulation of GSK-3 activity by PI3Ks appears to play a key role in this process.


Asunto(s)
Proteínas de Unión al ADN/genética , Células Madre Embrionarias/metabolismo , Regulación de la Expresión Génica , Proteínas de Homeodominio/genética , Fosfatidilinositol 3-Quinasas/fisiología , Transducción de Señal , Animales , Células Madre Embrionarias/citología , Células Madre Embrionarias/enzimología , Inhibidores Enzimáticos/farmacología , Regulación de la Expresión Génica/efectos de los fármacos , Glucógeno Sintasa Quinasa 3/antagonistas & inhibidores , Glucógeno Sintasa Quinasa 3/fisiología , Ratones , Proteína Homeótica Nanog , Inhibidores de las Quinasa Fosfoinosítidos-3 , Células Madre Pluripotentes
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...