Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 68
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
Proc Natl Acad Sci U S A ; 120(44): e2311946120, 2023 Oct 31.
Artículo en Inglés | MEDLINE | ID: mdl-37871215

RESUMEN

The T-box transcription factor Eomesodermin (Eomes), also known as Tbr2, plays essential roles in the early mouse embryo. Loss-of-function mutant embryos arrest at implantation due to Eomes requirements in the trophectoderm cell lineage. Slightly later, expression in the visceral endoderm promotes anterior visceral endoderm formation and anterior-posterior axis specification. Early induction in the epiblast beginning at day 6 is necessary for nascent mesoderm to undergo epithelial to mesenchymal transition (EMT). Eomes acts in a temporally and spatially restricted manner to sequentially specify the yolk sac haemogenic endothelium, cardiac mesoderm, definitive endoderm, and axial mesoderm progenitors during gastrulation. Little is known about the underlying molecular mechanisms governing Eomes actions during the formation of these distinct progenitor cell populations. Here, we introduced a degron-tag and mCherry reporter sequence into the Eomes locus. Our experiments analyzing homozygously tagged embryonic stem cells and embryos demonstrate that the degron-tagged Eomes protein is fully functional. dTAG (degradation fusion tag) treatment in vitro results in rapid protein degradation and recapitulates the Eomes-null phenotype. However in utero administration of dTAG resulted in variable and lineage-specific degradation, likely reflecting diverse cell type-specific Eomes expression dynamics. Finally, we demonstrate that Eomes protein rapidly recovers following dTAG wash-out in vitro. The ability to temporally manipulate Eomes protein expression in combination with cell marking by the mCherry-reporter offers a powerful tool for dissecting Eomes-dependent functional roles in these diverse cell types in the early embryo.


Asunto(s)
Transición Epitelial-Mesenquimal , Proteínas de Dominio T Box , Ratones , Animales , Proteínas de Dominio T Box/genética , Proteínas de Dominio T Box/metabolismo , Estratos Germinativos/metabolismo , Embrión de Mamíferos/metabolismo , Mesodermo/metabolismo , Endodermo/metabolismo , Regulación del Desarrollo de la Expresión Génica
2.
Differentiation ; 130: 28-31, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36543010

RESUMEN

The International Society of Differentiation was born from the First International Conference on Cell Differentiation conceived by D.V. and held in Nice, France in 1971. The conference also resulted in the creation of the journal of the Society named Differentiation. The Society advocates for the field of differentiation through the journal Differentiation, organizing and supporting international scientific conferences, honoring scientific achievements, and supporting trainees.


Asunto(s)
Diferenciación Celular , Sociedades Científicas , Sociedades Científicas/historia
4.
Nat Cell Biol ; 23(1): 61-74, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33420489

RESUMEN

Extra-embryonic mesoderm (ExM)-composed of the earliest cells that traverse the primitive streak-gives rise to the endothelium as well as haematopoietic progenitors in the developing yolk sac. How a specific subset of ExM becomes committed to a haematopoietic fate remains unclear. Here we demonstrate using an embryonic stem cell model that transient expression of the T-box transcription factor Eomesodermin (Eomes) governs haemogenic competency of ExM. Eomes regulates the accessibility of enhancers that the transcription factor stem cell leukaemia (SCL) normally utilizes to specify primitive erythrocytes and is essential for the normal development of Runx1+ haemogenic endothelium. Single-cell RNA sequencing suggests that Eomes loss of function profoundly blocks the formation of blood progenitors but not specification of Flk-1+ haematoendothelial progenitors. Our findings place Eomes at the top of the transcriptional hierarchy regulating early blood formation and suggest that haemogenic competence is endowed earlier during embryonic development than was previously appreciated.


Asunto(s)
Células Madre Embrionarias/citología , Hemangioblastos/citología , Mesodermo/citología , Proteínas de Dominio T Box/fisiología , Saco Vitelino/citología , Animales , Subunidad alfa 2 del Factor de Unión al Sitio Principal/genética , Subunidad alfa 2 del Factor de Unión al Sitio Principal/metabolismo , Células Madre Embrionarias/metabolismo , Femenino , Hemangioblastos/metabolismo , Masculino , Mesodermo/metabolismo , Ratones Noqueados , Embarazo , RNA-Seq , Análisis de la Célula Individual , Proteína 1 de la Leucemia Linfocítica T Aguda/genética , Proteína 1 de la Leucemia Linfocítica T Aguda/metabolismo , Receptor 2 de Factores de Crecimiento Endotelial Vascular/genética , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo , Saco Vitelino/metabolismo
5.
Nat Commun ; 11(1): 2782, 2020 06 03.
Artículo en Inglés | MEDLINE | ID: mdl-32493987

RESUMEN

The transcriptional repressor Blimp1 controls cell fate decisions in the developing embryo and adult tissues. Here we describe Blimp1 expression and functional requirements within maternal uterine tissues during pregnancy. Expression is robustly up-regulated at early post-implantation stages in the primary decidual zone (PDZ) surrounding the embryo. Conditional inactivation results in defective formation of the PDZ barrier and abnormal trophectoderm invasion. RNA-Seq analysis demonstrates down-regulated expression of genes involved in cell adhesion and markers of decidualisation. In contrast, genes controlling immune responses including IFNγ are up-regulated. ChIP-Seq experiments identify candidate targets unique to the decidua as well as those shared across diverse cell types including a highly conserved peak at the Csf-1 gene promoter. Interestingly Blimp1 inactivation results in up-regulated Csf1 expression and macrophage recruitment into maternal decidual tissues. These results identify Blimp1 as a critical regulator of tissue remodelling and maternal tolerance during early stages of pregnancy.


Asunto(s)
Decidua/metabolismo , Factor 1 de Unión al Dominio 1 de Regulación Positiva/metabolismo , Transcripción Genética , Animales , Decidua/ultraestructura , Ectodermo/metabolismo , Ectodermo/ultraestructura , Implantación del Embrión/genética , Femenino , Regulación del Desarrollo de la Expresión Génica , Factor Estimulante de Colonias de Macrófagos/genética , Factor Estimulante de Colonias de Macrófagos/metabolismo , Macrófagos/metabolismo , Masculino , Ratones Endogámicos C57BL , Mutación/genética , Embarazo , Regiones Promotoras Genéticas , Trofoblastos/metabolismo , Trofoblastos/ultraestructura , Regulación hacia Arriba/genética
6.
Nat Commun ; 10(1): 1089, 2019 03 06.
Artículo en Inglés | MEDLINE | ID: mdl-30842446

RESUMEN

The essential roles played by Nodal and Bmp signalling during early mouse development have been extensively documented. Here we use conditional deletion strategies to investigate functional contributions made by Nodal, Bmp and Smad downstream effectors during primordial germ cell (PGC) development. We demonstrate that Nodal and its target gene Eomes provide early instructions during formation of the PGC lineage. We discover that Smad2 inactivation in the visceral endoderm results in increased numbers of PGCs due to an expansion of the PGC niche. Smad1 is required for specification, whereas in contrast Smad4 controls the maintenance and migration of PGCs. Additionally we find that beside Blimp1, down-regulated phospho-Smad159 levels also distinguishes PGCs from their somatic neighbours so that emerging PGCs become refractory to Bmp signalling that otherwise promotes mesodermal development in the posterior epiblast. Thus balanced Nodal/Bmp signalling cues regulate germ cell versus somatic cell fate decisions in the early posterior epiblast.


Asunto(s)
Proteínas Morfogenéticas Óseas/metabolismo , Diferenciación Celular/fisiología , Regulación del Desarrollo de la Expresión Génica/fisiología , Células Germinativas/fisiología , Proteína Nodal/metabolismo , Animales , Proteínas Morfogenéticas Óseas/genética , Línea Celular , Movimiento Celular/fisiología , Embrión de Mamíferos , Endodermo/citología , Endodermo/fisiología , Femenino , Técnicas de Inactivación de Genes , Masculino , Ratones , Ratones Noqueados , Células Madre Embrionarias de Ratones , Proteína Nodal/genética , Transducción de Señal/genética , Proteínas Smad/genética , Proteínas Smad/metabolismo
8.
Cell Rep ; 24(8): 1977-1985.e7, 2018 08 21.
Artículo en Inglés | MEDLINE | ID: mdl-30134160

RESUMEN

Epiblast cells in the early post-implantation stage mammalian embryo undergo a transition described as lineage priming before cell fate allocation, but signaling pathways acting upstream remain ill defined. Genetic studies demonstrate that Smad2/3 double-mutant mouse embryos die shortly after implantation. To learn more about the molecular disturbances underlying this abrupt failure, here we characterized Smad2/3-deficient embryonic stem cells (ESCs). We found that Smad2/3 double-knockout ESCs induced to form epiblast-like cells (EpiLCs) display changes in naive and primed pluripotency marker gene expression, associated with the disruption of Oct4-bound distal regulatory elements. In the absence of Smad2/3, we observed enhanced Bmp target gene expression and de-repression of extra-embryonic gene expression. Cell fate allocation into all three embryonic germ layers is disrupted. Collectively, these experiments demonstrate that combinatorial Smad2/3 functional activities are required to maintain distinct embryonic and/or extra-embryonic cell identity during lineage priming in the epiblast before gastrulation.


Asunto(s)
Células Madre Embrionarias/metabolismo , Proteína Nodal/metabolismo , Animales , Diferenciación Celular , Humanos , Ratones , Transducción de Señal , Proteína Smad2
9.
Sci Rep ; 8(1): 237, 2018 01 10.
Artículo en Inglés | MEDLINE | ID: mdl-29321612

RESUMEN

The transcriptional repressor Blimp-1 originally cloned as a silencer of type I interferon (IFN)-ß gene expression controls cell fate decisions in multiple tissue contexts. Conditional inactivation in the mammary gland was recently shown to disrupt epithelial cell architecture. Here we report that Blimp-1 regulates expression of viral defense, IFN signaling and MHC class I pathways, and directly targets the transcriptional activator Stat1. Blimp-1 functional loss in 3D cultures of mammary epithelial cells (MECs) results in accumulation of dsRNA and expression of type III IFN-λ. Cultures treated with IFN lambda similarly display defective lumen formation. These results demonstrate that type III IFN-λ profoundly influences the behavior of MECs and identify Blimp-1 as a critical regulator of IFN signaling cascades.


Asunto(s)
Células Epiteliales/metabolismo , Interferones/metabolismo , Factor 1 de Unión al Dominio 1 de Regulación Positiva/metabolismo , Animales , Células Epiteliales/efectos de los fármacos , Femenino , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Silenciador del Gen , Interferones/farmacología , Ratones , Ratones Noqueados , Factor 1 de Unión al Dominio 1 de Regulación Positiva/genética , Unión Proteica , ARN Bicatenario/genética , ARN Bicatenario/metabolismo , Factor de Transcripción STAT1/metabolismo , Transducción de Señal
10.
Nat Commun ; 8(1): 1714, 2017 11 20.
Artículo en Inglés | MEDLINE | ID: mdl-29158490

RESUMEN

The hierarchical relationships between various stem and progenitor cell subpopulations driving mammary gland morphogenesis and homoeostasis are poorly understood. Conditional inactivation experiments previously demonstrated that expression of the zinc finger transcriptional repressor Blimp1/PRDM1 is essential for the establishment of epithelial cell polarity and functional maturation of alveolar cells. Here we exploit a Prdm1.CreERT2-LacZ reporter allele for lineage tracing experiments. Blimp1 expression marks a rare subpopulation of unipotent luminal stem cells that initially appear in the embryonic mammary gland at around E17.5 coincident with the segregation of the luminal and basal compartments. Fate mapping at multiple time points in combination with whole-mount confocal imaging revealed these long-lived unipotent luminal stem cells survive consecutive involutions and retain their identity throughout adult life. Blimp1+ luminal stem cells give rise to Blimp1- progeny that are invariably Elf5+ERα-PR-. Thus, Blimp1 expression defines a mammary stem cell subpopulation with unique functional characteristics.


Asunto(s)
Glándulas Mamarias Animales/metabolismo , Organogénesis/genética , Factor 1 de Unión al Dominio 1 de Regulación Positiva/genética , Células Madre/metabolismo , Animales , Linaje de la Célula/genética , Femenino , Regulación del Desarrollo de la Expresión Génica , Humanos , Masculino , Glándulas Mamarias Animales/embriología , Glándulas Mamarias Animales/crecimiento & desarrollo , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Microscopía Confocal , Factor 1 de Unión al Dominio 1 de Regulación Positiva/metabolismo
11.
Sci Rep ; 7(1): 6793, 2017 07 28.
Artículo en Inglés | MEDLINE | ID: mdl-28754907

RESUMEN

Trophoblast stem cells (TSCs) give rise to specialized cell types within the placenta. However, the regulatory mechanisms that guide trophoblast cell fate decisions during placenta development remain ill defined. Here we exploited ATAC-seq and transcriptional profiling strategies to describe dynamic changes in gene expression and chromatin accessibility during TSC differentiation. We detect significantly increased chromatin accessibility at key genes upregulated as TSCs exit from the stem cell state. However, downregulated gene expression is not simply due to the loss of chromatin accessibility in proximal regions. Additionally, transcriptional targets recognized by the zinc finger transcriptional repressor Prdm1/Blimp1, an essential regulator of placenta development, were identified in ChIP-seq experiments. Comparisons with previously reported ChIP-seq datasets for primordial germ cell-like cells and E18.5 small intestine, combined with functional annotation analysis revealed that Blimp1 has broadly shared as well as cell type-specific functional activities unique to the trophoblast lineage. Importantly, Blimp1 not only silences TSC gene expression but also prevents aberrant activation of divergent developmental programmes. Overall the present study provides new insights into the chromatin landscape and Blimp1-dependent regulatory networks governing trophoblast gene expression.


Asunto(s)
Diferenciación Celular , Cromatina/genética , Factor 1 de Unión al Dominio 1 de Regulación Positiva/genética , Trofoblastos/metabolismo , Animales , Células Cultivadas , Cromatina/metabolismo , Ensamble y Desensamble de Cromatina , Células Madre Embrionarias/citología , Células Madre Embrionarias/metabolismo , Regulación del Desarrollo de la Expresión Génica , Ratones , Factor 1 de Unión al Dominio 1 de Regulación Positiva/metabolismo , Trofoblastos/citología
12.
Development ; 144(7): 1249-1260, 2017 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-28174238

RESUMEN

The T-box transcription factor (TF) Eomes is a key regulator of cell fate decisions during early mouse development. The cis-acting regulatory elements that direct expression in the anterior visceral endoderm (AVE), primitive streak (PS) and definitive endoderm (DE) have yet to be defined. Here, we identified three gene-proximal enhancer-like sequences (PSE_a, PSE_b and VPE) that faithfully activate tissue-specific expression in transgenic embryos. However, targeted deletion experiments demonstrate that PSE_a and PSE_b are dispensable, and only VPE is required for optimal Eomes expression in vivo Embryos lacking this enhancer display variably penetrant defects in anterior-posterior axis orientation and DE formation. Chromosome conformation capture experiments reveal VPE-promoter interactions in embryonic stem cells (ESCs), prior to gene activation. The locus resides in a large (500 kb) pre-formed compartment in ESCs and activation during DE differentiation occurs in the absence of 3D structural changes. ATAC-seq analysis reveals that VPE, PSE_a and four additional putative enhancers display increased chromatin accessibility in DE that is associated with Smad2/3 binding coincident with transcriptional activation. By contrast, activation of the Eomes target genes Foxa2 and Lhx1 is associated with higher order chromatin reorganisation. Thus, diverse regulatory mechanisms govern activation of lineage specifying TFs during early development.


Asunto(s)
Embrión de Mamíferos/metabolismo , Regulación del Desarrollo de la Expresión Génica , Secuencias Reguladoras de Ácidos Nucleicos/genética , Proteínas de Dominio T Box/genética , Animales , Diferenciación Celular/genética , Cromatina/metabolismo , Endodermo/metabolismo , Elementos de Facilitación Genéticos , Femenino , Factores de Transcripción Forkhead/metabolismo , Gastrulación/genética , Eliminación de Gen , Marcación de Gen , Genes Reporteros , Genotipo , Ratones Endogámicos C57BL , Modelos Biológicos , Proteínas del Grupo Polycomb/metabolismo , Transducción de Señal/genética , Proteína Smad2/metabolismo , Proteínas de Dominio T Box/metabolismo
13.
Development ; 143(10): 1663-73, 2016 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-27190036

RESUMEN

Mammary gland morphogenesis depends on a tight balance between cell proliferation, differentiation and apoptosis, to create a defined functional hierarchy within the epithelia. The limited availability of stem cell/progenitor markers has made it challenging to decipher lineage relationships. Here, we identify a rare subset of luminal progenitors that express the zinc finger transcriptional repressor Blimp1, and demonstrate that this subset of highly clonogenic luminal progenitors is required for mammary gland development. Conditional inactivation experiments using K14-Cre and WAPi-Cre deleter strains revealed essential functions at multiple developmental stages. Thus, Blimp1 regulates proliferation, apoptosis and alveolar cell maturation during puberty and pregnancy. Loss of Blimp1 disrupts epithelial architecture and lumen formation both in vivo and in three-dimensional (3D) primary cell cultures. Collectively, these results demonstrate that Blimp1 is required to maintain a highly proliferative luminal subset necessary for mammary gland development and homeostasis.


Asunto(s)
Glándulas Mamarias Animales/embriología , Glándulas Mamarias Animales/metabolismo , Proteínas Represoras/metabolismo , Células Madre/citología , Células Madre/metabolismo , Factores de Transcripción/metabolismo , Animales , Compartimento Celular/efectos de los fármacos , Diferenciación Celular/efectos de los fármacos , Polaridad Celular/efectos de los fármacos , Células Cultivadas , Células Clonales , Células Epiteliales/citología , Células Epiteliales/efectos de los fármacos , Células Epiteliales/metabolismo , Células Epiteliales/ultraestructura , Femenino , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Hormonas/farmacología , Lactancia/efectos de los fármacos , Glándulas Mamarias Animales/citología , Ratones Endogámicos C57BL , Morfogénesis/efectos de los fármacos , Factor 1 de Unión al Dominio 1 de Regulación Positiva , Embarazo , Células Madre/efectos de los fármacos , Esteroides/farmacología , Regulación hacia Arriba/efectos de los fármacos
14.
Nat Commun ; 7: 11414, 2016 04 25.
Artículo en Inglés | MEDLINE | ID: mdl-27108815

RESUMEN

Growth and survival of the mammalian embryo within the uterine environment depends on the placenta, a highly complex vascularized organ comprised of both maternal and foetal tissues. Recent experiments demonstrate that the zinc finger transcriptional repressor Prdm1/Blimp1 is essential for specification of spiral artery trophoblast giant cells (SpA-TGCs) that invade and remodel maternal blood vessels. To learn more about functional contributions made by Blimp1+ cell lineages here we perform the first single-cell RNA-seq analysis of the placenta. Cell types of both foetal and maternal origin are profiled. Comparisons with microarray datasets from mutant placenta and in vitro differentiated trophoblast stem cells allow us to identify Blimp1-dependent transcripts enriched in SpA-TGCs. Our experiments provide new insights into the functionally distinct cell types present at the maternal-foetal interface and advance our knowledge of dynamic gene expression patterns controlling placental morphogenesis and vascular mimicry.


Asunto(s)
Células Gigantes/metabolismo , Ratones/embriología , Factor 1 de Unión al Dominio 1 de Regulación Positiva/metabolismo , ARN/genética , Trofoblastos/metabolismo , Animales , Diferenciación Celular , Femenino , Regulación del Desarrollo de la Expresión Génica , Células Gigantes/citología , Intercambio Materno-Fetal , Ratones/genética , Ratones/metabolismo , Placenta/citología , Placenta/metabolismo , Factor 1 de Unión al Dominio 1 de Regulación Positiva/genética , Embarazo , ARN/metabolismo , Análisis de Secuencia de ARN , Especificidad de la Especie , Transcripción Genética , Trofoblastos/citología
15.
Open Biol ; 6(1): 150200, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26791244

RESUMEN

Nodal is an evolutionarily conserved member of the transforming growth factor-ß (TGF-ß) superfamily of secreted signalling factors. Nodal factors are known to play key roles in embryonic development and asymmetry in a variety of organisms ranging from hydra and sea urchins to fish, mice and humans. In addition to embryonic patterning, Nodal signalling is required for maintenance of human embryonic stem cell pluripotency and mis-regulated Nodal signalling has been found associated with tumour metastases. Therefore, precise and timely regulation of this pathway is essential. Here, we discuss recent evidence from sea urchins, frogs, fish, mice and humans that show a role for transcriptional and translational repression of Nodal signalling during early development.


Asunto(s)
Proteína Nodal/metabolismo , Biosíntesis de Proteínas , Proteínas Represoras/metabolismo , Transducción de Señal , Transcripción Genética , Animales , Secuencia de Bases , Humanos , Modelos Biológicos , Datos de Secuencia Molecular
16.
Genes Dev ; 29(20): 2108-22, 2015 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-26494787

RESUMEN

Gene regulatory networks controlling functional activities of spatially and temporally distinct endodermal cell populations in the early mouse embryo remain ill defined. The T-box transcription factor Eomes, acting downstream from Nodal/Smad signals, directly activates the LIM domain homeobox transcription factor Lhx1 in the visceral endoderm. Here we demonstrate Smad4/Eomes-dependent Lhx1 expression in the epiblast marks the entire definitive endoderm lineage, the anterior mesendoderm, and midline progenitors. Conditional inactivation of Lhx1 disrupts anterior definitive endoderm development and impedes node and midline morphogenesis in part due to severe disturbances in visceral endoderm displacement. Transcriptional profiling and ChIP-seq (chromatin immunoprecipitation [ChIP] followed by high-throughput sequencing) experiments identified Lhx1 target genes, including numerous anterior definitive endoderm markers and components of the Wnt signaling pathway. Interestingly, Lhx1-binding sites were enriched at enhancers, including the Nodal-proximal epiblast enhancer element and enhancer regions controlling Otx2 and Foxa2 expression. Moreover, in proteomic experiments, we characterized a complex comprised of Lhx1, Otx2, and Foxa2 as well as the chromatin-looping protein Ldb1. These partnerships cooperatively regulate development of the anterior mesendoderm, node, and midline cell populations responsible for establishment of the left-right body axis and head formation.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Desarrollo Embrionario/genética , Regulación del Desarrollo de la Expresión Génica , Estratos Germinativos/embriología , Proteínas de Unión al ADN/genética , Embrión de Mamíferos , Elementos de Facilitación Genéticos/fisiología , Eliminación de Gen , Perfilación de la Expresión Génica , Estratos Germinativos/metabolismo , Factor Nuclear 3-beta del Hepatocito/metabolismo , Proteínas con Dominio LIM/metabolismo , Proteínas con Homeodominio LIM/genética , Proteínas con Homeodominio LIM/metabolismo , Complejos Multiproteicos/genética , Complejos Multiproteicos/metabolismo , Factores de Transcripción Otx/metabolismo , Unión Proteica , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Vía de Señalización Wnt
17.
PLoS Genet ; 11(7): e1005375, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-26158850

RESUMEN

The neonatal intestine is a very complex and dynamic organ that must rapidly adapt and remodel in response to a barrage of environmental stimuli during the first few postnatal weeks. Recent studies demonstrate that the zinc finger transcriptional repressor Blimp1/Prdm1 plays an essential role governing postnatal reprogramming of intestinal enterocytes during this period. Functional loss results in global changes in gene expression patterns, particularly in genes associated with metabolic function. Here we engineered a knock-in allele expressing an eGFP-tagged fusion protein under control of the endogenous regulatory elements and performed genome wide ChIP-seq analysis to identify direct Blimp1 targets and further elucidate the function of Blimp1 in intestinal development. Comparison with published human and mouse datasets revealed a highly conserved core set of genes including interferon-inducible promoters. Here we show that the interferon-inducible transcriptional activator Irf1 is constitutively expressed throughout fetal and postnatal intestinal epithelium development. ChIP-seq demonstrates closely overlapping Blimp1 and Irf1 peaks at key components of the MHC class I pathway in fetal enterocytes. The onset of MHC class I expression coincides with down-regulated Blimp1 expression during the suckling to weaning transition. Collectively, these experiments strongly suggest that in addition to regulating the enterocyte metabolic switch, Blimp1 functions as a gatekeeper in opposition to Irf1 to prevent premature activation of the MHC class I pathway in villus epithelium to maintain tolerance in the neonatal intestine.


Asunto(s)
Antígenos de Histocompatibilidad Clase I/inmunología , Factor 1 Regulador del Interferón/metabolismo , Mucosa Intestinal/metabolismo , Placenta/metabolismo , Factores de Transcripción/metabolismo , Animales , Diferenciación Celular/genética , Línea Celular Tumoral , Enterocitos/citología , Femenino , Regulación del Desarrollo de la Expresión Génica , Técnicas de Sustitución del Gen , Proteínas Fluorescentes Verdes/genética , Humanos , Factor 1 Regulador del Interferón/genética , Mucosa Intestinal/crecimiento & desarrollo , Ratones , Placenta/citología , Factor 1 de Unión al Dominio 1 de Regulación Positiva , Embarazo , Regiones Promotoras Genéticas/genética , Elementos Reguladores de la Transcripción/genética , Factores de Transcripción/genética
18.
Cereb Cortex ; 25(10): 3290-302, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-24927931

RESUMEN

The individual contribution of different progenitor subtypes towards the mature rodent cerebral cortex is not fully understood. Intermediate progenitor cells (IPCs) are key to understanding the regulation of neuronal number during cortical development and evolution, yet their exact contribution is much debated. Intermediate progenitors in the cortical subventricular zone are defined by expression of T-box brain-2 (Tbr2). In this study we demonstrate by using the Tbr2(Cre) mouse line and state-of-the-art cell lineage labeling techniques, that IPC derived cells contribute substantial proportions 67.5% of glutamatergic but not GABAergic or astrocytic cells to all cortical layers including the earliest generated subplate zone. We also describe the laminar dispersion of clonally derived cells from IPCs using a recently described clonal analysis tool (CLoNe) and show that pair-generated cells in different layers cluster closer (142.1 ± 76.8 µm) than unrelated cells (294.9 ± 105.4 µm). The clonal dispersion from individual Tbr2 positive intermediate progenitors contributes to increasing the cortical surface. Our study also describes extracortical contributions from Tbr2+ progenitors to the lateral olfactory tract and ventromedial hypothalamic nucleus.


Asunto(s)
Corteza Cerebral/embriología , Células-Madre Neurales/fisiología , Células Madre/fisiología , Proteínas de Dominio T Box/metabolismo , Animales , Astrocitos/metabolismo , Astrocitos/fisiología , Encéfalo/embriología , Encéfalo/metabolismo , Corteza Cerebral/metabolismo , Neuronas GABAérgicas/metabolismo , Neuronas GABAérgicas/fisiología , Ventrículos Laterales/metabolismo , Ventrículos Laterales/fisiología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Células-Madre Neurales/metabolismo , Células Madre/metabolismo , Proteínas de Dominio T Box/genética
19.
Semin Cell Dev Biol ; 32: 73-9, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24704361

RESUMEN

Nodal signals in the early post-implantation stage embryo are essential to establish initial proximal-distal (P-D) polarity and generate the final anterior-posterior (A-P) body axis. Nodal signaling in the epiblast results in the phosphorylation of Smad2 in the overlying visceral endoderm necessary to induce the AVE, in part via Smad2-dependent activation of the T-box gene Eomesodermin. Slightly later following mesoderm induction a continuum of dose-dependent Nodal signaling during the process of gastrulation underlies specification of mesodermal and definitive endoderm progenitors. Dynamic Nodal expression during the critical 72 h time window immediately following implantation, accomplished by a series of feed-back and feed-forward mechanisms serves to provide key positional cues required for establishment of the body plan and controls cell fate decisions in the early mammalian embryo.


Asunto(s)
Embrión de Mamíferos/metabolismo , Proteína Nodal/genética , Transducción de Señal/genética , Proteína Smad2/genética , Animales , Tipificación del Cuerpo/genética , Embrión de Mamíferos/citología , Embrión de Mamíferos/embriología , Regulación del Desarrollo de la Expresión Génica , Estratos Germinativos/citología , Estratos Germinativos/embriología , Estratos Germinativos/metabolismo , Ratones , Modelos Genéticos , Proteína Nodal/metabolismo , Proteína Smad2/metabolismo
20.
Mol Cell Biol ; 33(19): 3936-50, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23918801

RESUMEN

Prdm4 is a highly conserved member of the Prdm family of PR/SET domain zinc finger proteins. Many well-studied Prdm family members play critical roles in development and display striking loss-of-function phenotypes. Prdm4 functional contributions have yet to be characterized. Here, we describe its widespread expression in the early embryo and adult tissues. We demonstrate that DNA binding is exclusively mediated by the Prdm4 zinc finger domain, and we characterize its tripartite consensus sequence via SELEX (systematic evolution of ligands by exponential enrichment) and ChIP-seq (chromatin immunoprecipitation-sequencing) experiments. In embryonic stem cells (ESCs), Prdm4 regulates key pluripotency and differentiation pathways. Two independent strategies, namely, targeted deletion of the zinc finger domain and generation of a EUCOMM LacZ reporter allele, resulted in functional null alleles. However, homozygous mutant embryos develop normally and adults are healthy and fertile. Collectively, these results strongly suggest that Prdm4 functions redundantly with other transcriptional partners to cooperatively regulate gene expression in the embryo and adult animal.


Asunto(s)
Proteínas de Unión al ADN/genética , Embrión de Mamíferos/metabolismo , Células Madre Embrionarias/metabolismo , Regulación del Desarrollo de la Expresión Génica , Factores de Transcripción/genética , Animales , Secuencia de Bases , Sitios de Unión/genética , Northern Blotting , Western Blotting , Células Cultivadas , Inmunoprecipitación de Cromatina , Proteínas de Unión al ADN/metabolismo , Embrión de Mamíferos/citología , Embrión de Mamíferos/embriología , Femenino , Perfilación de la Expresión Génica , Hibridación in Situ , Factores de Transcripción de Tipo Kruppel/genética , Factores de Transcripción de Tipo Kruppel/metabolismo , Masculino , Ratones , Proteína Nodal/genética , Proteína Nodal/metabolismo , Análisis de Secuencia por Matrices de Oligonucleótidos , Unión Proteica , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Técnica SELEX de Producción de Aptámeros , Análisis de Secuencia de ADN , Factores de Transcripción/metabolismo , Dedos de Zinc/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA