Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
1.
Development ; 149(17)2022 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-35976266

RESUMEN

Mouse embryonic stem cells have an inherent propensity to explore gene regulatory states associated with either self-renewal or differentiation. This property depends on ERK, which downregulates pluripotency genes such as Nanog. Here, we aimed at identifying repressive histone modifications that would mark Nanog for inactivation in response to ERK activity. We found that the transcription factor ZFP57, which binds methylated DNA to nucleate heterochromatin, is recruited upstream of Nanog, within a region enriched for histone H3 lysine 9 tri-methylation (H3K9me3). Whereas before differentiation H3K9me3 at Nanog depends on ERK, in somatic cells it becomes independent of ERK. Moreover, the loss of H3K9me3 at Nanog, induced by deleting the region or by knocking out DNA methyltransferases or Zfp57, is associated with reduced heterogeneity of NANOG, delayed commitment into differentiation and impaired ability to acquire a primitive endoderm fate. Hence, a network axis centred on DNA methylation, ZFP57 and H3K9me3 links Nanog regulation to ERK activity for the timely establishment of new cell identities. We suggest that establishment of irreversible H3K9me3 at specific master regulators allows the acquisition of particular cell fates during differentiation.


Asunto(s)
Células Madre Embrionarias , Endodermo , Código de Histonas , Proteína Homeótica Nanog/genética , Animales , Diferenciación Celular , Endodermo/metabolismo , Genes Homeobox , Ratones , Proteína Homeótica Nanog/metabolismo
2.
Nucleic Acids Res ; 50(13): 7367-7379, 2022 07 22.
Artículo en Inglés | MEDLINE | ID: mdl-35762231

RESUMEN

Histone H3 Lysine 9 (H3K9) methylation, a characteristic mark of heterochromatin, is progressively implemented during development to contribute to cell fate restriction as differentiation proceeds. Accordingly, in undifferentiated and pluripotent mouse Embryonic Stem (ES) cells the global levels of H3K9 methylation are rather low and increase only upon differentiation. How global H3K9 methylation levels are coupled with the loss of pluripotency remains largely unknown. Here, we identify SUV39H1, a major H3K9 di- and tri-methylase, as an indirect target of the pluripotency network of Transcription Factors (TFs). We find that pluripotency TFs, principally OCT4, activate the expression of Suv39h1as, an antisense long non-coding RNA to Suv39h1. In turn, Suv39h1as downregulates Suv39h1 transcription in cis via a mechanism involving the modulation of the chromatin status of the locus. The targeted deletion of the Suv39h1as promoter region triggers increased SUV39H1 expression and H3K9me2 and H3K9me3 levels, affecting all heterochromatic regions, particularly peri-centromeric major satellites and retrotransposons. This increase in heterochromatinization efficiency leads to accelerated and more efficient commitment into differentiation. We report, therefore, a simple genetic circuitry coupling the genetic control of pluripotency with the global efficiency of H3K9 methylation associated with a major cell fate restriction, the irreversible loss of pluripotency.


Asunto(s)
Histonas , Metiltransferasas/metabolismo , Factor 3 de Transcripción de Unión a Octámeros/metabolismo , ARN Largo no Codificante , Proteínas Represoras/metabolismo , Animales , Cromatina , Código de Histonas , Histonas/genética , Histonas/metabolismo , Metilación , Metiltransferasas/genética , Ratones , ARN Largo no Codificante/genética , ARN Largo no Codificante/metabolismo , Proteínas Represoras/genética
3.
RNA ; 24(12): 1803-1812, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30242063

RESUMEN

The contribution of basal cellular processes to the regulation of tissue homeostasis has just started to be appreciated. However, our knowledge of the modulation of ribosome biogenesis activity in situ within specific lineages remains very limited. This is largely due to the lack of assays that enable quantitation of ribosome biogenesis in small numbers of cells in vivo. We used a technique, named Flow-FISH, combining cell surface antibody staining and flow cytometry with intracellular ribosomal RNA (rRNA) FISH, to measure the levels of pre-rRNAs of hematopoietic cells in vivo. Here, we show that Flow-FISH reports and quantifies ribosome biogenesis activity in hematopoietic cell populations, thereby providing original data on this fundamental process notably in rare populations such as hematopoietic stem and progenitor cells. We unravel variations in pre-rRNA levels between different hematopoietic progenitor compartments and during erythroid differentiation. In particular, our data indicate that, contrary to what may be anticipated from their quiescent state, hematopoietic stem cells have significant ribosome biogenesis activity. Moreover, variations in pre-rRNA levels do not correlate with proliferation rates, suggesting that cell type-specific mechanisms might regulate ribosome biogenesis in hematopoietic stem cells and progenitors. Our study contributes to a better understanding of the cellular physiology of the hematopoietic system in vivo in unperturbed situations.


Asunto(s)
Células Madre Hematopoyéticas/metabolismo , Procesamiento Postranscripcional del ARN/genética , ARN Ribosómico/biosíntesis , Ribosomas/genética , Animales , Diferenciación Celular/genética , Ratones , Precursores del ARN/genética , Proteínas Ribosómicas , Ribosomas/metabolismo
4.
Stem Cells ; 37(7): 888-898, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-30913328

RESUMEN

Receptor tyrosine kinase signaling pathways are key regulators for the formation of the primitive endoderm (PrE) and the epiblast (Epi) from the inner cell mass (ICM) of the mouse preimplantation embryo. Among them, FGF signaling is critical for PrE cell specification, whereas PDGF signaling is critical for the survival of committed PrE cells. Here, we investigated possible functional redundancies among FGF, PDGF, and KIT signaling and showed that only PDGF signaling is involved in PrE cell survival. In addition, we analyzed the effectors downstream of PDGFRα. Our results suggest that the role of PDGF signaling in PrE cell survival is mediated through PI3K-mTOR and independently from p53. Lastly, we uncovered a role for PI3K-mTOR signaling in the survival of Epi cells. Taken together, we propose that survival of ICM cell lineages relies on the regulation of PI3K-mTOR signaling through the regulation of multiple signaling pathways. Stem Cells 2019;37:888-898.


Asunto(s)
Masa Celular Interna del Blastocisto/metabolismo , Linaje de la Célula/genética , Endodermo/metabolismo , Regulación del Desarrollo de la Expresión Génica , Fosfatidilinositol 3-Quinasas/genética , Factor de Crecimiento Derivado de Plaquetas/genética , Serina-Treonina Quinasas TOR/genética , Animales , Blastocisto , Masa Celular Interna del Blastocisto/citología , Supervivencia Celular , Endodermo/citología , Endodermo/crecimiento & desarrollo , Femenino , Factores de Crecimiento de Fibroblastos/genética , Factores de Crecimiento de Fibroblastos/metabolismo , Masculino , Ratones , Fosfatidilinositol 3-Quinasas/metabolismo , Factor de Crecimiento Derivado de Plaquetas/metabolismo , Proteínas Proto-Oncogénicas c-kit/genética , Proteínas Proto-Oncogénicas c-kit/metabolismo , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/genética , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/metabolismo , Transducción de Señal , Serina-Treonina Quinasas TOR/metabolismo , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo
5.
Development ; 142(21): 3649-60, 2015 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-26534985

RESUMEN

NOTCH signalling is an evolutionarily conserved pathway involved in intercellular communication essential for cell fate choices during development. Although dispensable for early aspects of mouse development, canonical RBPJ-dependent NOTCH signalling has been shown to influence lineage commitment during embryonic stem cell (ESC) differentiation. NOTCH activation in ESCs promotes the acquisition of a neural fate, whereas its suppression favours their differentiation into cardiomyocytes. This suggests that NOTCH signalling is implicated in the acquisition of distinct embryonic fates at early stages of mammalian development. In order to investigate in vivo such a role for NOTCH signalling in shaping cell fate specification, we use genetic approaches to constitutively activate the NOTCH pathway in the mouse embryo. Early embryonic development, including the establishment of anterior-posterior polarity, is not perturbed by forced NOTCH activation. By contrast, widespread NOTCH activity in the epiblast triggers dramatic gastrulation defects. These are fully rescued in a RBPJ-deficient background. Epiblast-specific NOTCH activation induces acquisition of neurectoderm identity and disrupts the formation of specific mesodermal precursors including the derivatives of the anterior primitive streak, the mouse organiser. In addition, we show that forced NOTCH activation results in misregulation of NODAL signalling, a major determinant of early embryonic patterning. Our study reveals a previously unidentified role for canonical NOTCH signalling during mammalian gastrulation. It also exemplifies how in vivo studies can shed light on the mechanisms underlying cell fate specification during in vitro directed differentiation.


Asunto(s)
Embrión de Mamíferos/metabolismo , Desarrollo Embrionario , Gastrulación , Receptores Notch/metabolismo , Transducción de Señal , Animales , Ectodermo/metabolismo , Implantación del Embrión , Estratos Germinativos/metabolismo , Ratones , Proteína Nodal/metabolismo
6.
RNA ; 18(2): 253-64, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22201644

RESUMEN

Over the last years, the microRNA (miRNA) pathway has emerged as a key component of the regulatory network of pluripotency. Although clearly distinct states of pluripotency have been described in vivo and ex vivo, differences in miRNA expression profiles associated with the developmental modulation of pluripotency have not been extensively studied so far. Here, we performed deep sequencing to profile miRNA expression in naive (embryonic stem cell [ESC]) and primed (epiblast stem cell [EpiSC]) pluripotent stem cells derived from mouse embryos of identical genetic background. We developed a graphical representation method allowing the rapid identification of miRNAs with an atypical profile including mirtrons, a small nucleolar RNA (snoRNA)-derived miRNA, and miRNAs whose biogenesis may differ between ESC and EpiSC. Comparison of mature miRNA profiles revealed that ESCs and EpiSCs exhibit very different miRNA signatures with one third of miRNAs being differentially expressed between the two cell types. Notably, differential expression of several clusters, including miR290-295, miR17-92, miR302/367, and a large repetitive cluster on chromosome 2, was observed. Our analysis also showed that differentiation priming of EpiSC compared to ESC is evidenced by changes in miRNA expression. These dynamic changes in miRNAs signature are likely to reflect both redundant and specific roles of miRNAs in the fine-tuning of pluripotency during development.


Asunto(s)
Células Madre Embrionarias/metabolismo , MicroARNs/biosíntesis , MicroARNs/genética , Células Madre Pluripotentes/metabolismo , Animales , Diferenciación Celular/genética , Línea Celular , Bases de Datos de Ácidos Nucleicos , Células Madre Embrionarias/citología , Perfilación de la Expresión Génica/métodos , Ratones , Células Madre Pluripotentes/citología
7.
Methods Mol Biol ; 2214: 11-30, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-32944900

RESUMEN

The mouse preimplantation embryo is an excellent system for studying how mammalian cells organize dynamically into increasingly complex structures. Accessible to experimental and genetic manipulations, its normal or perturbed development can be scrutinized ex vivo by real-time imaging from fertilization to late blastocyst stage. High-resolution imaging of multiple embryos at the same time can be compromised by embryos displacement during imaging. We have developed an inexpensive and easy-to-produce imaging device that facilitates greatly the imaging of preimplantation embryo. In this chapter, we describe the different steps of production and storage of the imaging device as well as its use for live imaging of mouse preimplantation embryos expressing fluorescent reporters from genetically modified alleles or after in vitro transcribed mRNA transfer by microinjection or electroporation.


Asunto(s)
Blastocisto/ultraestructura , Microscopía Confocal/métodos , Animales , Electroporación/métodos , Técnicas de Cultivo de Embriones/métodos , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , Microinyecciones/métodos
8.
Cell Death Differ ; 27(10): 2872-2887, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32355182

RESUMEN

Ribosome biogenesis inhibition causes cell cycle arrest and apoptosis through the activation of tumor suppressor-dependent surveillance pathways. These responses are exacerbated in cancer cells, suggesting that targeting ribosome synthesis may be beneficial to patients. Here, we characterize the effect of the loss-of-function of Notchless (Nle), an essential actor of ribosome biogenesis, on the intestinal epithelium undergoing tumor initiation due to acute Apc loss-of-function. We show that ribosome biogenesis dysfunction strongly alleviates Wnt-driven tumor initiation by restoring cell cycle exit and differentiation in Apc-deficient progenitors. Conversely Wnt hyperactivation attenuates the cellular responses to surveillance pathways activation induced by ribosome biogenesis dysfunction, as proliferation was maintained at control-like levels in the stem cells and progenitors of double mutants. Thus, our data indicate that, while ribosome biogenesis inhibition efficiently reduces cancer cell proliferation in the intestinal epithelium, enhanced resistance of Apc-deficient stem and progenitor cells to ribosome biogenesis defects may be an important concern when using a therapeutic strategy targeting ribosome production for the treatment of Wnt-dependent tumorigenesis.


Asunto(s)
Proteína de la Poliposis Adenomatosa del Colon/fisiología , Transformación Celular Neoplásica , Mucosa Intestinal , Proteínas de la Membrana/fisiología , Ribosomas/metabolismo , Vía de Señalización Wnt , Animales , Mucosa Intestinal/citología , Mucosa Intestinal/metabolismo , Mucosa Intestinal/patología , Ratones , Ratones Endogámicos C57BL , Biogénesis de Organelos
9.
Neuron ; 107(4): 617-630.e6, 2020 08 19.
Artículo en Inglés | MEDLINE | ID: mdl-32559415

RESUMEN

Stable genomic integration of exogenous transgenes is essential in neurodevelopmental and stem cell studies. Despite tools driving increasingly efficient genomic insertion with DNA vectors, transgenesis remains fundamentally hindered by the impossibility of distinguishing integrated from episomal transgenes. Here, we introduce an integration-coupled On genetic switch, iOn, which triggers gene expression upon incorporation into the host genome through transposition, thus enabling rapid and accurate identification of integration events following transfection with naked plasmids. In vitro, iOn permits rapid drug-free stable transgenesis of mouse and human pluripotent stem cells with multiple vectors. In vivo, we demonstrate faithful cell lineage tracing, assessment of regulatory elements, and mosaic analysis of gene function in somatic transgenesis experiments that reveal neural progenitor potentialities and interaction. These results establish iOn as a universally applicable strategy to accelerate and simplify genetic engineering in cultured systems and model organisms by conditioning transgene activation to genomic integration.


Asunto(s)
Expresión Génica , Técnicas de Transferencia de Gen , Células-Madre Neurales , Transgenes , Animales , Linaje de la Célula , Vectores Genéticos , Humanos , Ratones
10.
Mol Cell Biol ; 26(9): 3541-9, 2006 May.
Artículo en Inglés | MEDLINE | ID: mdl-16611995

RESUMEN

Notch signaling is an evolutionarily conserved pathway involved in intercellular communication and is essential for proper cell fate choices. Numerous genes participate in the modulation of the Notch signaling pathway activity. Among them, Notchless (Nle) is a direct regulator of the Notch activity identified in Drosophila melanogaster. Here, we characterized the murine ortholog of Nle and demonstrated that it has conserved the ability to modulate Notch signaling. We also generated mice deficient for mouse Nle (mNle) and showed that its disruption resulted in embryonic lethality shortly after implantation. In late mNle(-/-) blastocysts, inner cell mass (ICM) cells died through a caspase 3-dependent apoptotic process. Most deficient embryos exhibited a delay in the temporal down-regulation of Oct4 expression in the trophectoderm (TE). However, mNle-deficient TE was able to induce decidual swelling in vivo and properly differentiated in vitro. Hence, our results indicate that mNle is mainly required in ICM cells, being instrumental for their survival, and raise the possibility that the death of mNle-deficient embryos might result from abnormal Notch signaling during the first steps of development.


Asunto(s)
Linaje de la Célula/genética , Implantación del Embrión/genética , Embrión de Mamíferos/citología , Genes Letales , Proteínas de la Membrana/fisiología , Receptores Notch/fisiología , Secuencia de Aminoácidos , Animales , Apoptosis , Drosophila melanogaster/genética , Drosophila melanogaster/fisiología , Embrión no Mamífero/citología , Femenino , Masculino , Proteínas de la Membrana/genética , Ratones , Ratones Mutantes , Datos de Secuencia Molecular , Xenopus
11.
Elife ; 82019 10 10.
Artículo en Inglés | MEDLINE | ID: mdl-31599722

RESUMEN

The access of Transcription Factors (TFs) to their cognate DNA binding motifs requires a precise control over nucleosome positioning. This is especially important following DNA replication and during mitosis, both resulting in profound changes in nucleosome organization over TF binding regions. Using mouse Embryonic Stem (ES) cells, we show that the TF CTCF displaces nucleosomes from its binding site and locally organizes large and phased nucleosomal arrays, not only in interphase steady-state but also immediately after replication and during mitosis. Correlative analyses suggest this is associated with fast gene reactivation following replication and mitosis. While regions bound by other TFs (Oct4/Sox2), display major rearrangement, the post-replication and mitotic nucleosome positioning activity of CTCF is not unique: Esrrb binding regions are also characterized by persistent nucleosome positioning. Therefore, selected TFs such as CTCF and Esrrb act as resilient TFs governing the inheritance of nucleosome positioning at regulatory regions throughout the cell-cycle.


Asunto(s)
Factor de Unión a CCCTC/metabolismo , Replicación del ADN , Células Madre Embrionarias/fisiología , Mitosis , Nucleosomas/metabolismo , Animales , Células Cultivadas , Regulación de la Expresión Génica , Ratones , Activación Transcripcional
12.
Mol Cell Biol ; 25(14): 6289-302, 2005 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-15988037

RESUMEN

While highly conserved through evolution, the cell cycle has been extensively modified to adapt to new developmental programs. Recently, analyses of mouse mutants revealed that several important cell cycle regulators are either dispensable for development or have a tissue- or cell-type-specific function, indicating that many aspects of cell cycle regulation during mammalian embryo development remain to be elucidated. Here, we report on the characterization of a new gene, Omcg1, which codes for a nuclear zinc finger protein. Embryos lacking Omcg1 die by the end of preimplantation development. In vitro cultured Omcg1-null blastocysts exhibit a dramatic reduction in the total cell number, a high mitotic index, and the presence of abnormal mitotic figures. Importantly, we found that Omcg1 disruption results in the lengthening of M phase rather than in a mitotic block. We show that the mitotic delay in Omcg1-/- embryos is associated with neither a dysfunction of the spindle checkpoint nor abnormal global histone modifications. Taken together, these results suggest that Omcg1 is an important regulator of the cell cycle in the preimplantation embryo.


Asunto(s)
Blastocisto/citología , Proteínas de Ciclo Celular/fisiología , Desarrollo Embrionario , Genes Letales/genética , Mitosis/genética , Proteínas Nucleares/fisiología , Secuencia de Aminoácidos , Animales , Evolución Biológica , Blastocisto/metabolismo , Proteínas de Ciclo Celular/genética , Secuencia Conservada , Femenino , Expresión Génica , Histonas/metabolismo , Ratones , Datos de Secuencia Molecular , Mutación , Proteínas Nucleares/genética , Embarazo , Procesamiento Proteico-Postraduccional , Huso Acromático/genética , Huso Acromático/metabolismo , Dedos de Zinc
13.
Stem Cell Reports ; 10(2): 461-476, 2018 02 13.
Artículo en Inglés | MEDLINE | ID: mdl-29396181

RESUMEN

In mouse, although four Argonaute (AGO) proteins with partly overlapping functions in small-RNA pathways exist, only Ago2 deficiency causes embryonic lethality. To investigate the role of AGO2 during mouse early development, we generated Ago2-deficient mouse embryonic stem cells (mESCs) and performed a detailed characterization of their differentiation potential. Ago2 disruption caused a global reduction of microRNAs, which resulted in the misregulation of only a limited number of transcripts. We demonstrated, both in vivo and in vitro, that AGO2 is dispensable for the embryonic germ-layer formation. However, Ago2-deficient mESCs showed a specific defect during conversion into extra-embryonic endoderm cells. We proved that this defect is cell autonomous and can be rescued by both a catalytically active and an inactive Ago2, but not by Ago2 deprived of its RNA binding capacity or by Ago1 overexpression. Overall, our results suggest a role for AGO2 in stem cell differentiation.


Asunto(s)
Proteínas Argonautas/genética , Diferenciación Celular/genética , Desarrollo Embrionario/genética , Células Madre Embrionarias de Ratones/citología , Animales , Línea Celular , Endodermo/citología , Endodermo/crecimiento & desarrollo , Gastrulación/genética , Regulación del Desarrollo de la Expresión Génica/genética , Estratos Germinativos/citología , Estratos Germinativos/crecimiento & desarrollo , Ratones , MicroARNs/genética
14.
Mol Biol Cell ; 15(10): 4444-56, 2004 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-15229288

RESUMEN

Endocytosed membrane components are recycled to the cell surface either directly from early/sorting endosomes or after going through the endocytic recycling compartment (ERC). Studying recycling mechanisms is difficult, in part due to the fact that specific tools to inhibit this process are scarce. In this study, we have characterized a novel widely expressed protein, named Rififylin (Rffl) for RING Finger and FYVE-like domain-containing protein, that, when overexpressed in HeLa cells, induced the condensation of transferrin receptor-, Rab5-, and Rab11-positive recycling tubulovesicular membranes in the perinuclear region. Internalized transferrin was able to access these condensed endosomes but its exit from this compartment was delayed. Using deletion mutants, we show that the carboxy-terminal RING finger of Rffl is dispensable for its action. In contrast, the amino-terminal domain of Rffl, which shows similarities with the phosphatidylinositol-3-phosphate-binding FYVE finger, is critical for the recruitment of Rffl to recycling endocytic membranes and for the inhibition of recycling, albeit in a manner that is independent of PtdIns(3)-kinase activity. Rffl overexpression represents a novel means to inhibit recycling that will help to understand the mechanisms involved in recycling from the ERC to the plasma membrane.


Asunto(s)
Endocitosis/fisiología , Endosomas/metabolismo , Membranas Intracelulares/metabolismo , Proteínas/metabolismo , Secuencia de Aminoácidos , Animales , Proteínas Reguladoras de la Apoptosis , Secuencia de Bases , Endosomas/ultraestructura , Células HeLa , Humanos , Membranas Intracelulares/ultraestructura , Ratones , Datos de Secuencia Molecular , Mutación , Estructura Terciaria de Proteína , Proteínas/genética , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Alineación de Secuencia , Transferrina/metabolismo , Ubiquitina-Proteína Ligasas , Dedos de Zinc
15.
Sci Rep ; 7: 42661, 2017 02 17.
Artículo en Inglés | MEDLINE | ID: mdl-28209967

RESUMEN

Microinjection of the CRISPR/Cas9 system in zygotes is an efficient and comparatively fast method to generate genetically modified mice. So far, only few knock-in mice have been generated using this approach, and because no systematic study has been performed, parameters controlling the efficacy of CRISPR/Cas9-mediated targeted insertion are not fully established. Here, we evaluated the effect of several parameters on knock-in efficiency changing only one variable at a time. We found that knock-in efficiency was dependent on injected Cas9 mRNA and single-guide RNA concentrations and that cytoplasmic injection resulted in more genotypic complexity compared to pronuclear injection. Our results also indicated that injection into the pronucleus compared to the cytoplasm is preferable to generate knock-in alleles with an oligonucleotide or a circular plasmid. Finally, we showed that Cas9D10A nickase variant was less efficient than wild-type Cas9 for generating knock-in alleles and caused a higher rate of mosaicism. Thus, our study provides valuable information that will help to improve the future production of precise genetic modifications in mice.


Asunto(s)
Sistemas CRISPR-Cas , Edición Génica/métodos , Técnicas de Sustitución del Gen/métodos , Proteínas Fluorescentes Verdes/genética , Proteínas de la Membrana/genética , Plásmidos/metabolismo , Animales , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Proteína 9 Asociada a CRISPR , Núcleo Celular/genética , Núcleo Celular/metabolismo , Repeticiones Palindrómicas Cortas Agrupadas y Regularmente Espaciadas , Citosol/metabolismo , Embrión de Mamíferos , Endonucleasas/genética , Endonucleasas/metabolismo , Exones , Femenino , Proteínas Fluorescentes Verdes/metabolismo , Masculino , Proteínas de la Membrana/deficiencia , Ratones , Ratones Transgénicos , Microinyecciones , Mutación , Oligodesoxirribonucleótidos/genética , Oligodesoxirribonucleótidos/metabolismo , Plásmidos/administración & dosificación , Plásmidos/química , ARN Guía de Kinetoplastida/genética , ARN Guía de Kinetoplastida/metabolismo , Cigoto/citología , Cigoto/metabolismo
16.
Sci Rep ; 7(1): 12285, 2017 09 25.
Artículo en Inglés | MEDLINE | ID: mdl-28947813

RESUMEN

Inner cell Mass (ICM) specification into epiblast (Epi) and primitive endoderm (PrE) is an asynchronous and progressive process taking place between E3.0 to E3.75 under the control of the Fibroblast Growth Factor (FGF)/Extracellular signal-Regulated Kinase (ERK) signaling pathway. Here, we have analyzed in details the kinetics of specification and found that ICM cell responsiveness to the up and down regulation of FGF signaling activity are temporally distinct. We also showed that PrE progenitors are generated later than Epi progenitors. We further demonstrated that, during this late phase of specification, a 4 hours period of FGF/ERK inhibition prior E3.75 is sufficient to convert ICM cells into Epi. Finally, we showed that ICM conversion into Epi in response to inhibition during this short time window requires both transcription and proteasome degradation. Collectively, our data give new insights into the timing and mechanisms involved in the process of ICM specification.


Asunto(s)
Masa Celular Interna del Blastocisto/fisiología , Diferenciación Celular/fisiología , Factores de Crecimiento de Fibroblastos/metabolismo , Estratos Germinativos/fisiología , Sistema de Señalización de MAP Quinasas/fisiología , Animales , Plasticidad de la Célula/fisiología , Ratones , Proteolisis , Factores de Tiempo , Transcripción Genética/fisiología
17.
Nat Cell Biol ; 18(11): 1139-1148, 2016 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-27723719

RESUMEN

Pluripotent mouse embryonic stem cells maintain their identity throughout virtually infinite cell divisions. This phenomenon, referred to as self-renewal, depends on a network of sequence-specific transcription factors (TFs) and requires daughter cells to accurately reproduce the gene expression pattern of the mother. However, dramatic chromosomal changes take place in mitosis, generally leading to the eviction of TFs from chromatin. Here, we report that Esrrb, a major pluripotency TF, remains bound to key regulatory regions during mitosis. We show that mitotic Esrrb binding is highly dynamic, driven by specific recognition of its DNA-binding motif and is associated with early transcriptional activation of target genes after completion of mitosis. These results indicate that Esrrb may act as a mitotic bookmarking factor, opening another perspective to molecularly understand the role of sequence-specific TFs in the epigenetic control of self-renewal, pluripotency and genome reprogramming.


Asunto(s)
Autorrenovación de las Células/genética , Reprogramación Celular/genética , Regulación del Desarrollo de la Expresión Génica/genética , Mitosis/genética , Células Madre Pluripotentes/citología , Receptores de Estrógenos/metabolismo , Animales , Inmunoprecipitación de Cromatina/métodos , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Células Madre Embrionarias de Ratones/metabolismo , Unión Proteica/genética , Receptores de Estrógenos/genética , Secuencias Reguladoras de Ácidos Nucleicos/genética , Activación Transcripcional/genética
18.
Gene ; 296(1-2): 75-86, 2002 Aug 21.
Artículo en Inglés | MEDLINE | ID: mdl-12383505

RESUMEN

The DDK syndrome is defined as the embryonic lethality of F1 mouse embryos from crosses between DDK females and males from other strains (named hereafter as non-DDK strains). Genetically controlled by the Ovum mutant (Om) locus, it is due to a deleterious interaction between a maternal factor present in DDK oocytes and the non-DDK paternal pronucleus. Therefore, the DDK syndrome constitutes a unique genetic tool to study the crucial interactions that take place between the parental genomes and the egg cytoplasm during mammalian development. In this paper, we present an extensive analysis performed by exon trapping on the Om region. Twenty-seven trapped sequences were from genes in the databases: beta-adaptin, CCT zeta2, DNA LigaseIII, Notchless, Rad51l3 and Scya1. Twenty-eight other sequences presented similarities with expressed sequence tags and genomic sequences whereas 57 did not. The pattern of expression of 37 of these markers was established. Importantly, five of them are expressed in DDK oocytes and are candidate genes for the maternal factor, and 20 are candidate genes for the paternal factor since they are expressed in testis. This data is an important step towards identifying the genes responsible for the DDK syndrome.


Asunto(s)
Infertilidad Femenina/genética , Mapeo Físico de Cromosoma/métodos , Animales , Células COS , Cromosomas Artificiales Bacterianos/genética , Clonación Molecular , Embrión de Mamíferos/metabolismo , Exones/genética , Etiquetas de Secuencia Expresada , Femenino , Expresión Génica , Predisposición Genética a la Enfermedad/genética , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos , Datos de Secuencia Molecular , Mutación , Oocitos/metabolismo , ARN/genética , ARN/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Síndrome , Transcripción Genética
19.
Gene Expr Patterns ; 4(6): 713-7, 2004 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-15465494

RESUMEN

Notch signaling is an evolutionary conserved pathway involved in intercellular signaling and essential for proper cell fate choices during development. Thus, it could be involved in mouse preimplantation development where intercellular signaling plays a crucial role, particularly between the inner cell mass and the trophectoderm of the blastocyst. At their face value, the phenotypes observed when disrupting each of the four Notch genes known in the mouse do not support this view as none of them involves perturbation of preimplantation development. However this could be due to functional redundancy and/or maternal expression. As a first step to address this issue, we decided to examine the expression in early development of various genes known to participate in Notch signaling. Here, we report on the expression pattern of Notch1-4, Jagged1 (Jag1), Jag2, Delta-like1 (Dll-1), Dll-3, Dll-4, Rbpsuh, Deltex1(Dtx1)and Dtx2 genes during preimplantation development from unfertilized eggs until late blastocyst stage using a RT-PCR strategy. We show that Notch1, 2, Jag1-2, Dll-3, Rbpsuh and Dtx2 transcripts are expressed at all stages. Notch4 and Dll-4 mRNAs are synthesized from the 2-cell through to the hatched blastocyst stage. Notch3, Dll-1 and Dtx1exhibit a stage dependent expression as their mRNAs are detected in 2-cell embryos and in hatched blastocysts, but are absent or weakly detected at the morula stage. Finally, we show that all the above genes are expressed both in Embryonic and Trophoblast Stem cells (ES and TS cells, respectively). Our results suggest that the Notch pathway may be active during mouse preimplantation development.


Asunto(s)
Blastocisto , Regulación del Desarrollo de la Expresión Génica , Proteínas de la Membrana/biosíntesis , Animales , Proteínas de Unión al Calcio , Linaje de la Célula , Cartilla de ADN/química , Proteínas de Unión al ADN/biosíntesis , Proteína de Unión a la Señal Recombinante J de las Inmunoglobulinas , Péptidos y Proteínas de Señalización Intercelular , Péptidos y Proteínas de Señalización Intracelular , Proteína Jagged-1 , Proteínas de la Membrana/metabolismo , Ratones , Proteínas Nucleares/biosíntesis , Fenotipo , Reacción en Cadena de la Polimerasa , ARN Mensajero/metabolismo , Receptores Notch , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Proteínas Serrate-Jagged , Transducción de Señal , Células Madre/metabolismo , Factores de Tiempo , Trofoblastos/citología , Ubiquitina-Proteína Ligasas
20.
PLoS One ; 9(5): e98507, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24875805

RESUMEN

Maintenance of cell survival is essential for proper embryonic development. In the mouse, Notchless homolog 1 (Drosophila) (Nle1) is instrumental for survival of cells of the inner cell mass upon implantation. Here, we analyze the function of Nle1 after implantation using the Meox2(tm1(cre)Sor) mouse that expresses the Cre recombinase specifically in the epiblast at E5.5. First, we find that NLE1 function is required in epiblast cells, as Nle1-deficient cells are rapidly eliminated. In this report, we also show that the Meox2(Cre) transgene is active in specific tissues during organogenesis. In particular, we detect high Cre expression in the vertebral column, ribs, limbs and tailbud. We took advantage of this dynamic expression profile to analyze the effects of inducing mosaic deletion of Nle1 in the embryo. We show that Nle1 deletion in this context, results in severe developmental anomalies leading to lethality at birth. Mutant embryos display multiple developmental defects in particular during axial skeletal formation. We also provide evidence that axial defects are due to an increase in apoptotic cell death in the somite at E9.5. These data demonstrate an essential role for Nle1 during organogenesis and in particular during axial development.


Asunto(s)
Proteínas de Microfilamentos/genética , Columna Vertebral/embriología , Columna Vertebral/metabolismo , Animales , Apoptosis/genética , Caspasa 3/metabolismo , Implantación del Embrión , Embrión de Mamíferos , Femenino , Regulación del Desarrollo de la Expresión Génica , Estratos Germinativos/embriología , Estratos Germinativos/metabolismo , Proteínas de Homeodominio/genética , Ratones , Ratones Transgénicos , Mutación , Tubo Neural/embriología , Tubo Neural/metabolismo , Organogénesis/genética , Somitos/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA