Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 39
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Proc Natl Acad Sci U S A ; 120(23): e2216799120, 2023 06 06.
Artículo en Inglés | MEDLINE | ID: mdl-37252988

RESUMEN

ZC3H11A (zinc finger CCCH domain-containing protein 11A) is a stress-induced mRNA-binding protein required for efficient growth of nuclear-replicating viruses. The cellular functions of ZC3H11A during embryonic development are unknown. Here, we report the generation and phenotypic characterization of Zc3h11a knockout (KO) mice. Heterozygous null Zc3h11a mice were born at the expected frequency without distinguishable phenotypic differences compared with wild-type mice. In contrast, homozygous null Zc3h11a mice were missing, indicating that Zc3h11a is crucial for embryonic viability and survival. Zc3h11a -/- embryos were detected at the expected Mendelian ratios up to late preimplantation stage (E4.5). However, phenotypic characterization at E6.5 revealed degeneration of Zc3h11a -/- embryos, indicating developmental defects around the time of implantation. Transcriptomic analyses documented a dysregulation of glycolysis and fatty acid metabolic pathways in Zc3h11a-/- embryos at E4.5. Proteomic analysis indicated a tight interaction between ZC3H11A and mRNA-export proteins in embryonic stem cells. CLIP-seq analysis demonstrated that ZC3H11A binds a subset of mRNA transcripts that are critical for metabolic regulation of embryonic cells. Furthermore, embryonic stem cells with an induced deletion of Zc3h11a display an impaired differentiation toward epiblast-like cells and impaired mitochondrial membrane potential. Altogether, the results show that ZC3H11A is participating in export and posttranscriptional regulation of selected mRNA transcripts required to maintain metabolic processes in embryonic cells. While ZC3H11A is essential for the viability of the early mouse embryo, inactivation of Zc3h11a expression in adult tissues using a conditional KO did not lead to obvious phenotypic defects.


Asunto(s)
Implantación del Embrión , Proteínas Nucleares , Proteómica , Proteínas de Unión al ARN , Animales , Femenino , Ratones , Embarazo , Implantación del Embrión/genética , Embrión de Mamíferos/metabolismo , Desarrollo Embrionario/genética , Regulación del Desarrollo de la Expresión Génica , Ratones Noqueados , ARN Mensajero/metabolismo , Proteínas de Unión al ARN/genética , Proteínas Nucleares/genética
2.
Sci Rep ; 12(1): 13908, 2022 08 16.
Artículo en Inglés | MEDLINE | ID: mdl-35974030

RESUMEN

Early mouse development is characterized by structural and epigenetic changes while cells progress towards differentiation. At blastocyst stage, the segregation of the three primordial lineages is accompanied by establishment of differential patterns of DNA methylation and post-translational modifications of histones, such as H3K27me3. Here, we analysed the dynamics of H3K27me3 at pericentromeric heterochromatin (PCH) during early development. We also followed the localization of EZH2 and BEND3, previously shown in ESCs to drive PRC2 to hypomethylated PCH. We show that the location of H3K27me3 at PCH, in addition to H3K9me3, is a defining feature of embryonic cells in vivo. Moreover, it may play an important role in structuring PCH and preserving genomic integrity at a time of globally relaxed chromatin. At peri-implantation stages, while DNA methylation is still low, EZH2 and then H3K27me3, leave PCH in epiblast progenitors at the time of their spatial segregation from primitive endoderm cells, while BEND3 remains there up to implantation. The comparison with stem cells (ESCs and TSCs) reveals that the epigenetic marks (i.e. H3K9me3 and H3K27me3) of PCH are reset during in vitro derivation and only partially restored thereafter. This highlights possible divergences between in vitro and "in embryo" epigenetic regulation regarding constitutive heterochromatin.


Asunto(s)
Heterocromatina , Histonas , Animales , Blastocisto/metabolismo , Metilación de ADN , Epigénesis Genética , Heterocromatina/metabolismo , Histonas/metabolismo , Ratones
3.
Nat Commun ; 13(1): 3861, 2022 07 05.
Artículo en Inglés | MEDLINE | ID: mdl-35790717

RESUMEN

Mammalian pre-implantation embryos accumulate substantial lipids, which are stored in lipid droplets (LDs). Despite the fundamental roles of lipids in many cellular functions, the significance of building-up LDs for the developing embryo remains unclear. Here we report that the accumulation and mobilization of LDs upon implantation are causal in the morphogenesis of the pluripotent epiblast and generation of the pro-amniotic cavity in mouse embryos, a critical step for all subsequent development. We show that the CIDEA protein, found abundantly in adipocytes, enhances lipid storage in blastocysts and pluripotent stem cells by promoting LD enlargement through fusion. The LD-stored lipids are mobilized into lysosomes at the onset of lumenogenesis, but without CIDEA are prematurely degraded by cytosolic lipases. Loss of lipid storage or inactivation of lipophagy leads to the aberrant formation of multiple cavities within disorganised epithelial structures. Thus, our study reveals an unexpected role for LDs in orchestrating tissue remodelling and uncovers underappreciated facets of lipid metabolism in peri-implantation development.


Asunto(s)
Gotas Lipídicas , Metabolismo de los Lípidos , Adipocitos/metabolismo , Animales , Gotas Lipídicas/metabolismo , Metabolismo de los Lípidos/fisiología , Lípidos , Mamíferos , Ratones , Morfogénesis
4.
J Cell Sci ; 135(6)2022 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-35048992

RESUMEN

During the first cell cycles of early development, the chromatin of the embryo is highly reprogrammed while the embryonic genome starts its own transcription. The spatial organization of the genome is an important process that contributes to regulating gene transcription in time and space. It has, however, been poorly studied in the context of early embryos. To study the cause-and-effect link between transcription and spatial organization in embryos, we focused on ribosomal genes, which are silent initially but start to be transcribed in 2-cell mouse embryos. We demonstrated that ribosomal sequences and early unprocessed rRNAs are spatially organized in a very particular manner between 2-cell and 16-cell stage. By using drugs that interfere with ribosomal DNA transcription, we showed that this organization - which is totally different in somatic cells - depends on an active transcription of ribosomal genes and induces a unique chromatin environment that favors transcription of major satellite sequences once the 4-cell stage has been reached.


Asunto(s)
Cromatina , ARN Ribosómico , Animales , Cromatina/genética , Cromatina/metabolismo , ADN Ribosómico/genética , Embrión de Mamíferos/metabolismo , Ratones , ARN Ribosómico/genética , ARN Ribosómico/metabolismo , Ribosomas/metabolismo , Transcripción Genética
5.
Vet Res ; 52(1): 42, 2021 Mar 10.
Artículo en Inglés | MEDLINE | ID: mdl-33691745

RESUMEN

Within the past decades, major progress has been accomplished in isolating germ/stem/pluripotent cells, in refining culture medium and conditions and in establishing 3-dimensional culture systems, towards developing organoids for organs involved in reproduction in mice and to some extent in humans. Haploid male germ cells were generated in vitro from primordial germ cells. So were oocytes, with additional support from ovarian cells and subsequent follicle culture. Going on with the female reproductive tract, spherical oviduct organoids were obtained from adult stem/progenitor cells. Multicellular endometrial structures mimicking functional uterine glands were derived from endometrial cells. Trophoblastic stem cells were induced to form 3-dimensional syncytial-like structures and exhibited invasive properties, a crucial point for placentation. Finally, considering the embryo itself, pluripotent embryonic cells together with additional extra-embryonic cells, could self-organize into a blastoid, and eventually into a post-implantation-like embryo. Most of these accomplishments have yet to be reached in farm animals, but much effort is devoted towards this goal. Here, we review the progress and discuss the specific challenges of developing organoids for the study of reproductive biology in these species. We consider the use of such organoids in basic research to delineate the physiological mechanisms involved at each step of the reproductive process, or to understand how they are altered by environmental factors relevant to animal breeding. We evaluate their potential in reproduction of animals with a high genetic value, from a breeding point of view or in the context of preserving local breeds with limited headcounts.


Asunto(s)
Animales Domésticos/anatomía & histología , Técnicas de Cultivo de Célula/veterinaria , Organoides/citología , Reproducción , Técnicas Reproductivas/veterinaria , Animales , Técnicas de Cultivo de Célula/métodos
6.
Nat Commun ; 11(1): 1112, 2020 02 28.
Artículo en Inglés | MEDLINE | ID: mdl-32111830

RESUMEN

Clusters of enhancers, referred as to super-enhancers (SEs), control the expression of cell identity genes. The organisation of these clusters, and how they are remodelled upon developmental transitions remain poorly understood. Here, we report the existence of two types of enhancer units within SEs typified by distinctive CpG methylation dynamics in embryonic stem cells (ESCs). We find that these units are either prone for decommissioning or remain constitutively active in epiblast stem cells (EpiSCs), as further established in the peri-implantation epiblast in vivo. Mechanistically, we show a pivotal role for ESRRB in regulating the activity of ESC-specific enhancer units and propose that the developmentally regulated silencing of ESRRB triggers the selective inactivation of these units within SEs. Our study provides insights into the molecular events that follow the loss of ESRRB binding, and offers a mechanism by which the naive pluripotency transcriptional programme can be partially reset upon embryo implantation.


Asunto(s)
Islas de CpG , Metilación de ADN , Elementos de Facilitación Genéticos/genética , Células Madre Pluripotentes/metabolismo , Receptores de Estrógenos/metabolismo , Animales , Regulación del Desarrollo de la Expresión Génica , Estratos Germinativos/citología , Complejo Mediador/metabolismo , Ratones , Células Madre Embrionarias de Ratones/metabolismo , Unión Proteica , ARN Polimerasa II/metabolismo , Transcripción Genética
7.
Biochim Biophys Acta Gen Subj ; 1863(10): 1608-1618, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31278960

RESUMEN

BACKGROUND: The placenta controls exchanges between the mother and the fetus and therefore fetal development and growth. The maternal environment can lead to disturbance of placental functions, with consequences on the health of the offspring. Since the rabbit placenta is very close to that of humans, rabbit models can provide biomedical data to study human placental function. Yet, to limit the use of animal experiments and to investigate the mechanistic aspects of placental function, we developed a new cell culture model in which rabbit trophoblast cells are differentiated from rabbit trophoblast stem cells. METHODS: Rabbit trophoblast stems cells were derived from blastocysts and differentiated onto a collagen gel and in the presence of a flow of culture medium to mimic maternal blood flow. Transcriptome analysis was performed on the stem and differentiated cells. RESULTS: Our culture model allows the differentiation of trophoblast stem cells. In particular, the fluid shear stress enhances microvilli formation on the differentiated cell surface, lipid droplets formation and fusion of cytotrophoblasts into syncytiotrophoblasts. In addition, the transcriptome analysis confirms the early trophoblast identity of the derived stem cells and reveals upregulation of signaling pathways involved in trophoblast differentiation. CONCLUSION: Thereby, the culture model allows mimicking the in vivo conditions in which maternal blood flow exerts a shear stress on trophoblast cells that influences their phenotype. GENERAL SIGNIFICANCE: Our culture model can be used to study the differentiation of trophoblast stem cells into cytotrophoblasts and syncytiotrophoblasts, as well as the trophoblast function in physiological and pathological conditions.


Asunto(s)
Diferenciación Celular , Células Madre/citología , Estrés Mecánico , Trofoblastos/citología , Animales , Línea Celular , Femenino , Humanos , Conejos , Células Madre/metabolismo , Transcriptoma , Trofoblastos/metabolismo
8.
Adv Exp Med Biol ; 1123: 5-17, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31016592

RESUMEN

Epiblast stem cells (EpiSCs) are pluripotent cells that are derived from mouse embryos at gastrulation stages. They represent the primed state of pluripotency, in which cells are on the verge of differentiation and already express markers of the three primary lineages (mesoderm, endoderm, neurectoderm). EpiSCs display some heterogeneity intra- and inter-cell lines in the expression of some of these lineage markers. We relate this heterogeneity to signalling pathways that are active in EpiSCs, either due to addition of growth factors (FGF2 and activin) in the culture medium, or endogenously active (FGF, Nodal, and Wnt). By modulating Wnt or activin/nodal pathways, cell lines close to EpiSCs but with different properties can be obtained. These signalling pathways are all at work in vivo to pattern the pluripotent epiblast and specify cellular fates.


Asunto(s)
Estratos Germinativos/citología , Células Madre Pluripotentes/citología , Transducción de Señal , Animales , Diferenciación Celular , Embrión de Mamíferos/citología , Endodermo , Mesodermo , Ratones , Placa Neural
9.
Epigenetics Chromatin ; 12(1): 14, 2019 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-30767785

RESUMEN

BACKGROUND: Genomic imprinting, resulting in parent-of-origin specific gene expression, plays a critical role in mammalian development. Here, we apply allele-specific RNA-seq on isogenic B6D2F1 mice to assay imprinted genes in tissues from early embryonic tissues between E3.5 and E7.25 and in pluripotent cell lines to evaluate maintenance of imprinted gene expression. For the cell lines, we include embryonic stem cells (ESCs) and epiblast stem cells (EpiSCs) derived from fertilized embryos and from embryos obtained after nuclear transfer (NT) or parthenogenetic activation (PGA). RESULTS: As homozygous genomic regions of PGA-derived cells are not compatible with allele-specific RNA-seq, we developed an RNA-seq-based genotyping strategy allowing identification of informative heterozygous regions. Global analysis shows that proper imprinted gene expression as observed in embryonic tissues is largely lost in the ESC lines included in this study, which mainly consisted of female ESCs. Differentiation of ESC lines to embryoid bodies or NPCs does not restore monoallelic expression of imprinted genes, neither did reprogramming of the serum-cultured ESCs to the pluripotent ground state by the use of 2 kinase inhibitors. Fertilized EpiSC and EpiSC-NT lines largely maintain imprinted gene expression, as did EpiSC-PGA lines that show known paternally expressed genes being silent and known maternally expressed genes consistently showing doubled expression. Notably, two EpiSC-NT lines show aberrant silencing of Rian and Meg3, two critically imprinted genes in mouse iPSCs. With respect to female EpiSC, most of the lines displayed completely skewed X inactivation suggesting a (near) clonal origin. CONCLUSIONS: Altogether, our analysis provides a comprehensive overview of imprinted gene expression in pluripotency and provides a benchmark to allow identification of cell lines that faithfully maintain imprinted gene expression and therefore retain full developmental potential.


Asunto(s)
Alelos , Impresión Genómica , Células Madre Embrionarias de Ratones/metabolismo , ARN Mensajero/genética , Animales , Diferenciación Celular , Línea Celular , Células Cultivadas , Femenino , Perfilación de la Expresión Génica , Regulación del Desarrollo de la Expresión Génica , Silenciador del Gen , Estratos Germinativos/citología , Estratos Germinativos/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos DBA , Células Madre Embrionarias de Ratones/citología
10.
Biol Open ; 7(8)2018 Aug 17.
Artículo en Inglés | MEDLINE | ID: mdl-30026265

RESUMEN

During early mammalian development, transient pools of pluripotent cells emerge that can be immortalised upon stem cell derivation. The pluripotent state, 'naïve' or 'primed', depends on the embryonic stage and derivation conditions used. Here we analyse the temporal gene expression patterns of mouse, cattle and porcine embryos at stages that harbour different types of pluripotent cells. We document conserved and divergent traits in gene expression, and identify predictor genes shared across the species that are associated with pluripotent states in vivo and in vitro Amongst these are the pluripotency-linked genes Klf4 and Lin28b The novel genes discovered include naïve- (Spic, Scpep1 and Gjb5) and primed-associated (Sema6a and Jakmip2) genes as well as naïve to primed transition genes (Dusp6 and Trip6). Both Gjb5 and Dusp6 play a role in pluripotency since their knockdown results in differentiation and downregulation of key pluripotency genes. Our interspecies comparison revealed new insights of pluripotency, pluripotent stem cell identity and a new molecular criterion for distinguishing between pluripotent states in various species, including human.

11.
Sci Rep ; 8(1): 5776, 2018 04 10.
Artículo en Inglés | MEDLINE | ID: mdl-29636490

RESUMEN

Mouse embryonic stem cells (ESCs) and epiblast stem cells (EpiSCs) represent naive and primed pluripotency states, respectively, and are maintained in vitro by specific signalling pathways. Furthermore, ESCs cultured in serum-free medium with two kinase inhibitors (2i-ESCs) are thought to be the ground naïve pluripotent state. Here, we present a comparative study of the epigenetic and transcriptional states of pericentromeric heterochromatin satellite sequences found in these pluripotent states. We show that 2i-ESCs are distinguished from other pluripotent cells by a prominent enrichment in H3K27me3 and low levels of DNA methylation at pericentromeric heterochromatin. In contrast, serum-containing ESCs exhibit higher levels of major satellite repeat transcription, which is lower in 2i-ESCs and even more repressed in primed EpiSCs. Removal of either DNA methylation or H3K9me3 at PCH in 2i-ESCs leads to enhanced deposition of H3K27me3 with few changes in satellite transcript levels. In contrast, their removal in EpiSCs does not lead to deposition of H3K27me3 but rather removes transcriptional repression. Altogether, our data show that the epigenetic state of PCH is modified during transition from naive to primed pluripotency states towards a more repressive state, which tightly represses the transcription of satellite repeats.


Asunto(s)
ADN Satélite/metabolismo , Epigénesis Genética , Estratos Germinativos/metabolismo , Heterocromatina/metabolismo , Histonas/metabolismo , Células Madre Embrionarias de Ratones/metabolismo , Animales , Línea Celular , Metilación de ADN , Heterocromatina/genética , Metilación , Ratones , Procesamiento Proteico-Postraduccional
12.
Biol Reprod ; 96(3): 531-541, 2017 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-28339853

RESUMEN

Apoptotic activity is a common physiological process which culminates at the blastocyst stage in the preimplantation embryo of many mammals. The degree of embryonic cell death can be influenced by the oocyte microenvironment. However, the prognostic significance of the incidence of apoptosis remains undefined. Prostaglandin E2 (PGE2) derived from prostaglandin G/H synthase-2 (PTGS2) activity is a well-known prosurvival factor that is mainly studied in oncology. PGE2 is the predominant PTGS2-derived prostaglandin present in the oocyte microenvironment during the periconceptional period. Using an in vitro model of bovine embryo production followed by transfer and collection procedures, we investigated the impact of periconceptional PGE2 on the occurrence of spontaneous apoptosis in embryos and on subsequent in vivo posthatching development. Different periconceptional PGE2 environments were obtained using NS-398, a specific inhibitor of PTGS2 activity, and exogenous PGE2. We assessed the level of embryonic cell death in blastocysts at day 8 postfertilization by counting total cell numbers, by the immunohistochemical staining of active caspase-3, and by quantifying terminal deoxynucleotidyl transferase-mediated dUTP nick-end labeling signals and apoptosis regulator (BCL-2/BAX) mRNA expression. Morphometric parameters were used to estimate the developmental stage of the embryonic disk and the extent of trophoblast elongation on day 15 conceptuses. Our findings indicate that periconceptional PGE2 signaling durably impacts oocytes, conferring increased resistance to spontaneous apoptosis in blastocysts and promoting embryonic disk development and the elongation process during preimplantation development.


Asunto(s)
Apoptosis , Blastocisto/fisiología , Dinoprostona/fisiología , Desarrollo Embrionario , Animales , Blastocisto/citología , Bovinos , Femenino , Fertilización In Vitro , Técnicas de Maduración In Vitro de los Oocitos , Embarazo , Prostaglandina-Endoperóxido Sintasas/metabolismo
13.
PLoS One ; 11(10): e0165898, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27798681

RESUMEN

BACKGROUND: We previously showed that the homeodomain transcription factor HOXB9 is expressed in mammalian oocytes and early embryos. However, a systematic and exhaustive study of the localization of the HOXB9 protein, and HOX proteins in general, during mammalian early embryonic development has so far never been performed. RESULTS: The distribution of HOXB9 proteins in oocytes and the early embryo was characterized by immunofluorescence from the immature oocyte stage to the peri-gastrulation period in both the mouse and the bovine. HOXB9 was detected at all studied stages with a dynamic expression pattern. Its distribution was well conserved between the two species until the blastocyst stage and was mainly nuclear. From that stage on, trophoblastic cells always showed a strong nuclear staining, while the inner cell mass and the derived cell lines showed important dynamic variations both in staining intensity and in intra-cellular localization. Indeed, HOXB9 appeared to be progressively downregulated in epiblast cells and only reappeared after gastrulation had well progressed. The protein was also detected in the primitive endoderm and its derivatives with a distinctive presence in apical vacuoles of mouse visceral endoderm cells. CONCLUSIONS: Together, these results could suggest the existence of unsuspected functions for HOXB9 during early embryonic development in mammals.


Asunto(s)
Desarrollo Embrionario , Proteínas de Homeodominio/metabolismo , Mamíferos , Oocitos/metabolismo , Oogénesis , Animales , Blastocisto/metabolismo , Bovinos , Linaje de la Célula/genética , Desarrollo Embrionario/genética , Endodermo/metabolismo , Feto , Técnica del Anticuerpo Fluorescente , Gastrulación/genética , Expresión Génica , Técnicas de Inactivación de Genes , Proteínas de Homeodominio/genética , Humanos , Inmunohistoquímica , Ratones , Oogénesis/genética , Transporte de Proteínas , Cigoto/metabolismo
15.
Methods Mol Biol ; 1341: 41-8, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-25720369

RESUMEN

Mouse embryonic stem cells (ESCs) derive from the inner cell mass (ICM) of a blastocyst. These cells are pluripotent and thus able to generate both somatic and germinal lineages. It is possible to maintain ESCs in different pluripotent states depending on the in vitro culture conditions. Classically, ESCs are cultured in the presence of serum and LIF, which sustain the naive state of pluripotency but in this metastable state cells exhibit a large degree of heterogeneity. In the last few years, it has been discovered that when ESCs are cultured in a chemically defined medium (without serum), in the presence of LIF and with the addition of two small molecules (in particular the inhibitors of MAPK and Gsk-3 pathways), they reach a ground state of pluripotency where cells are more homogeneous and more "ICM-like." In this protocol, we describe how we culture mouse ESCs and the way we switch them from naive to ground state.


Asunto(s)
Técnicas de Cultivo de Célula/métodos , Medios de Cultivo/farmacología , Factor Inhibidor de Leucemia/farmacología , Células Madre Embrionarias de Ratones/citología , Células Madre Embrionarias de Ratones/efectos de los fármacos , Inhibidores de Proteínas Quinasas/farmacología , Suero/metabolismo , Animales , Criopreservación/métodos , Medios de Cultivo/química , Glucógeno Sintasa Quinasa 3/antagonistas & inhibidores , Glucógeno Sintasa Quinasa 3/metabolismo , Factor Inhibidor de Leucemia/química , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Ratones , Células Madre Embrionarias de Ratones/metabolismo , Inhibidores de Proteínas Quinasas/química , Suero/química
16.
Methods Mol Biol ; 1341: 209-16, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-25720370

RESUMEN

Mouse embryonic stem cells (ESCs) derive from the inner cell mass (ICM) of a blastocyst at E3.5 while mouse epiblast stem cells (EpiSCs) derive from the late epiblast of a post-implantation embryo at E5.5-E7.5. Both cells are able to differentiate into derivatives of the three germs layers but only ESCs are able to produce chimeras when they are introduced into a blastocyst. To support the naive state of pluripotency, ESC culture requires Leukemia inhibitory factor (Lif) and either serum or inhibitors of Erk and Gsk3 pathways (2i) while the primed pluripotency of EpiSCs is maintained using Activin A and Fibroblast Growth Factor 2 (FGF2). It is possible to obtain EpiSCs in vitro starting from ESCs but also to induce ESCs starting from EpiSCs even if this second process is very difficult and inefficient. In this protocol we describe how we perform the process of conversion from ESCs to EpiSCs.


Asunto(s)
Técnicas de Cultivo de Célula/métodos , Estratos Germinativos/citología , Células Madre Embrionarias de Ratones/citología , Células Madre Pluripotentes/citología , Animales , Clostridium histolyticum/enzimología , Medios de Cultivo/metabolismo , Estratos Germinativos/metabolismo , Metaloproteinasa 8 de la Matriz/metabolismo , Ratones , Células Madre Embrionarias de Ratones/metabolismo , Células Madre Pluripotentes/metabolismo
17.
Genome Biol ; 16: 149, 2015 Aug 03.
Artículo en Inglés | MEDLINE | ID: mdl-26235224

RESUMEN

BACKGROUND: During early embryonic development, one of the two X chromosomes in mammalian female cells is inactivated to compensate for a potential imbalance in transcript levels with male cells, which contain a single X chromosome. Here, we use mouse female embryonic stem cells (ESCs) with non-random X chromosome inactivation (XCI) and polymorphic X chromosomes to study the dynamics of gene silencing over the inactive X chromosome by high-resolution allele-specific RNA-seq. RESULTS: Induction of XCI by differentiation of female ESCs shows that genes proximal to the X-inactivation center are silenced earlier than distal genes, while lowly expressed genes show faster XCI dynamics than highly expressed genes. The active X chromosome shows a minor but significant increase in gene activity during differentiation, resulting in complete dosage compensation in differentiated cell types. Genes escaping XCI show little or no silencing during early propagation of XCI. Allele-specific RNA-seq of neural progenitor cells generated from the female ESCs identifies three regions distal to the X-inactivation center that escape XCI. These regions, which stably escape during propagation and maintenance of XCI, coincide with topologically associating domains (TADs) as present in the female ESCs. Also, the previously characterized gene clusters escaping XCI in human fibroblasts correlate with TADs. CONCLUSIONS: The gene silencing observed during XCI provides further insight in the establishment of the repressive complex formed by the inactive X chromosome. The association of escape regions with TADs, in mouse and human, suggests that TADs are the primary targets during propagation of XCI over the X chromosome.


Asunto(s)
Silenciador del Gen , Inactivación del Cromosoma X , Alelos , Animales , Cromatina/química , Cuerpos Embrioides/metabolismo , Células Madre Embrionarias/metabolismo , Femenino , Humanos , Ratones , Células-Madre Neurales/metabolismo , Análisis de Secuencia de ARN
18.
Methods Mol Biol ; 1222: 255-65, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25287352

RESUMEN

Whole-mount in situ hybridization (WISH) is widely used to visualize the site and dynamics of gene expression during embryonic development. Various methods of probe labeling and hybridization detection are available nowadays. Meanwhile the technique was adapted to be used on many different species and has evolved from a manual to a larger scale and automated procedure. Standardized automated protocols improve the chance to compare different experimental settings reliably. The high resolution of this method is ideally suited for examination of manipulated (e.g., cloned) embryos often displaying subtle changes only. Embedding and sectioning of in situ hybridized specimen further enhance the detailed examination of their gene expression and morphology.


Asunto(s)
Embrión de Mamíferos , Hibridación in Situ/métodos , Animales , Femenino , Perfilación de la Expresión Génica/métodos , Regulación del Desarrollo de la Expresión Génica , Hibridación in Situ/instrumentación , Microtomía/instrumentación , Embarazo , Sondas ARN , Adhesión del Tejido/instrumentación , Adhesión del Tejido/métodos , Fijación del Tejido/instrumentación , Fijación del Tejido/métodos
19.
Anim Reprod Sci ; 149(1-2): 67-79, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-25059199

RESUMEN

Pluripotency refers to the ability for a single cell to differentiate into the three embryonic germ layers. In mice, two types of pluripotent stem cells with different features have been obtained in vitro. Naive pluripotent stem cells are derived from the inner cell mass (ICM) of early blastocyst (ESCs) or reprogrammed from somatic cells (iPSCs), while primed pluripotent stem cells are derived from late epiblast (EpiSCs). Cells in a primed pluripotency state are more prone to differentiation and only naive pluripotent stem cells form germline chimera after injection into a blastocyst. Despite numerous attempts, capturing pluripotency in domestic mammalian species has been largely unsuccessful and only primed pluripotent stem cells have been obtained even starting from early blastocyst or reprogramming somatic cells. This raises two questions: whether inner cell mass and epiblast are in naive or primed pluripotency state and what are the transcriptome features of ESCs and iPSCs in these species. To address these questions we compared rabbit ICM, epiblast, ESCs and iPSCs transcriptomes. Our results show that: (i) molecular signature of naïve and primed pluripotency may differ between mice and rabbit embryos; (ii) Genes involved in G1/S transition of the cell-cycle, actin cytoskeleton signaling, development and differentiation pathways are upregulated in ESCs and iPSCs; (iii) ICM and epiblast upregulate pluripotency associated genes and display specific metabolic features. These results denote an advanced primed state of pluripotency for rabbit ESCs and iPSCs and evidence specific functions for ICM and epiblast that are not shared by ESCs and iPSCs.


Asunto(s)
Regulación del Desarrollo de la Expresión Génica/fisiología , Células Madre Pluripotentes/metabolismo , Conejos/embriología , Transcriptoma/fisiología , Animales , Biomarcadores , Blastocisto , Células Cultivadas , Análisis por Conglomerados , Estratos Germinativos , Ratones , Regulación hacia Arriba
20.
PLoS Biol ; 12(6): e1001890, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24960041

RESUMEN

During early development, modulations in the expression of Nodal, a TGFß family member, determine the specification of embryonic and extra-embryonic cell identities. Nodal has been extensively studied in the mouse, but aspects of its early expression remain unaccounted for. We identified a conserved hotspot for the binding of pluripotency factors at the Nodal locus and called this sequence "highly bound element" (HBE). Luciferase-based assays, the analysis of fluorescent HBE reporter transgenes, and a conditional mutation of HBE allowed us to establish that HBE behaves as an enhancer, is activated ahead of other Nodal enhancers in the epiblast, and is essential to Nodal expression in embryonic stem cells (ESCs) and in the mouse embryo. We also showed that HBE enhancer activity is critically dependent on its interaction with the pluripotency factor Oct4 and on Activin/Nodal signaling. Use of an in vitro model of epiblast maturation, relying on the differentiation of ESCs into epiblast stem cells (EpiSCs), revealed that this process entails a shift in the regulation of Nodal expression from an HBE-driven phase to an ASE-driven phase, ASE being another autoregulatory Nodal enhancer. Deletion of HBE in ESCs or in EpiSCs allowed us to show that HBE, although not necessary for Nodal expression in EpiSCs, is required in differentiating ESCs to activate the differentiation-promoting ASE and therefore controls this regulatory shift. Our findings clarify how early Nodal expression is regulated and suggest how this regulation can promote the specification of extra-embryonic precusors without inducing premature differentiation of epiblast cells. More generally, they open new perspectives on how pluripotency factors achieve their function.


Asunto(s)
Células Madre Embrionarias/metabolismo , Elementos de Facilitación Genéticos , Regulación del Desarrollo de la Expresión Génica , Estratos Germinativos/fisiología , Proteína Nodal/metabolismo , Animales , Diferenciación Celular , Línea Celular , Estratos Germinativos/citología , Proteínas de Homeodominio/metabolismo , Subunidades beta de Inhibinas/metabolismo , Ratones , Ratones Transgénicos , Proteína Homeótica Nanog , Factor 3 de Transcripción de Unión a Octámeros/metabolismo , Células Madre Pluripotentes/metabolismo , Proteína Smad2/metabolismo , Proteína smad3/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...