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

Banco de datos
Tipo del documento
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
3.
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
4.
Nat Rev Mol Cell Biol ; 10(2): 91-103, 2009 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-19129791

RESUMEN

Genetic studies have identified the key signalling pathways and developmentally regulated transcription factors that govern cell lineage allocation and axis patterning in the early mammalian embryo. Recent advances have uncovered details of the molecular circuits that tightly control cell growth and differentiation in the mammalian embryo from the blastocyst stage, through the establishment of initial anterior-posterior polarity, to gastrulation, when the germ cells are set aside and the three primary germ layers are specified. Relevant studies in lower vertebrates indicate the conservation and divergence of regulatory mechanisms for cell lineage allocation and axis patterning.


Asunto(s)
Tipificación del Cuerpo/fisiología , Linaje de la Célula , Embrión de Mamíferos , Animales , Proteínas Morfogenéticas Óseas/genética , Proteínas Morfogenéticas Óseas/metabolismo , Proteínas de Unión al ADN , Embrión de Mamíferos/anatomía & histología , Embrión de Mamíferos/fisiología , Gastrulación , Ratones , Morfogénesis , Factor 1 de Unión al Dominio 1 de Regulación Positiva , Proteínas de Unión al ARN , Transducción de Señal/fisiología , Proteínas Smad/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
5.
Genes Dev ; 27(9): 997-1002, 2013 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-23651855

RESUMEN

Reciprocal inductive interactions between the embryonic and extraembryonic tissues establish the anterior-posterior (AP) axis of the early mouse embryo. The anterior visceral endoderm (AVE) signaling center emerges at the distal tip of the embryo at embryonic day 5.5 and translocates to the prospective anterior side of the embryo. The process of AVE induction and migration are poorly understood. Here we demonstrate that the T-box gene Eomesodermin (Eomes) plays an essential role in AVE recruitment, in part by directly activating the homeobox transcription factor Lhx1. Thus, Eomes function in the visceral endoderm (VE) initiates an instructive transcriptional program controlling AP identity.


Asunto(s)
Endodermo/metabolismo , Regulación del Desarrollo de la Expresión Génica , Proteínas de Dominio T Box/metabolismo , Animales , Tipificación del Cuerpo/genética , Línea Celular , Embrión de Mamíferos , Proteínas con Homeodominio LIM/genética , Proteínas con Homeodominio LIM/metabolismo , Ratones , Mutación , Proteínas de Dominio T Box/genética , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
6.
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
7.
Genes Dev ; 26(18): 2063-74, 2012 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-22987638

RESUMEN

Developmental arrest of Blimp1/Prdm1 mutant embryos at around embryonic day 10.5 (E10.5) has been attributed to placental disturbances. Here we investigate Blimp1/Prdm1 requirements in the trophoblast cell lineage. Loss of function disrupts specification of the invasive spiral artery-associated trophoblast giant cells (SpA-TGCs) surrounding maternal blood vessels and severely compromises the ability of the spongiotrophoblast layer to expand appropriately, secondarily causing collapse of the underlying labyrinth layer. Additionally, we identify a population of proliferating Blimp1(+) diploid cells present within the spongiotrophoblast layer. Lineage tracing experiments exploiting a novel Prdm1.Cre-LacZ allele demonstrate that these Blimp1(+) cells give rise to the mature SpA-TGCs, canal TGCs, and glycogen trophoblasts. In sum, the transcriptional repressor Blimp1/Prdm1 is required for terminal differentiation of SpA-TGCs and defines a lineage-restricted progenitor cell population contributing to placental growth and morphogenesis.


Asunto(s)
Diferenciación Celular , Células Gigantes/citología , Células Madre/citología , Factores de Transcripción/metabolismo , Trofoblastos/citología , Animales , Femenino , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Ratones , Placenta/irrigación sanguínea , Placenta/citología , Factor 1 de Unión al Dominio 1 de Regulación Positiva , Embarazo , Células Madre/metabolismo , Factores de Transcripción/genética
8.
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
9.
Genes Dev ; 25(3): 238-50, 2011 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-21245162

RESUMEN

Understanding the molecular mechanisms controlling early cell fate decisions in mammals is a major objective toward the development of robust methods for the differentiation of human pluripotent stem cells into clinically relevant cell types. Here, we used human embryonic stem cells and mouse epiblast stem cells to study specification of definitive endoderm in vitro. Using a combination of whole-genome expression and chromatin immunoprecipitation (ChIP) deep sequencing (ChIP-seq) analyses, we established an hierarchy of transcription factors regulating endoderm specification. Importantly, the pluripotency factors NANOG, OCT4, and SOX2 have an essential function in this network by actively directing differentiation. Indeed, these transcription factors control the expression of EOMESODERMIN (EOMES), which marks the onset of endoderm specification. In turn, EOMES interacts with SMAD2/3 to initiate the transcriptional network governing endoderm formation. Together, these results provide for the first time a comprehensive molecular model connecting the transition from pluripotency to endoderm specification during mammalian development.


Asunto(s)
Diferenciación Celular , Endodermo , Regulación del Desarrollo de la Expresión Génica , Células Madre Pluripotentes , Proteínas de Dominio T Box/metabolismo , Activinas/metabolismo , Animales , Biomarcadores/metabolismo , Línea Celular , Endodermo/citología , Endodermo/metabolismo , Redes Reguladoras de Genes/genética , Proteínas de Homeodominio/metabolismo , Humanos , Ratones , Proteína Homeótica Nanog , Proteína Nodal/metabolismo , Factor 3 de Transcripción de Unión a Octámeros/metabolismo , Células Madre Pluripotentes/citología , Células Madre Pluripotentes/metabolismo , Factores de Transcripción SOXB1/metabolismo , Proteínas de Dominio T Box/genética
10.
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
11.
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
12.
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
13.
Proc Natl Acad Sci U S A ; 108(26): 10585-90, 2011 Jun 28.
Artículo en Inglés | MEDLINE | ID: mdl-21670299

RESUMEN

Female mammals produce milk to feed their newborn offspring before teeth develop and permit the consumption of solid food. Intestinal enterocytes dramatically alter their biochemical signature during the suckling-to-weaning transition. The transcriptional repressor Blimp1 is strongly expressed in immature enterocytes in utero, but these are gradually replaced by Blimp1(-) crypt-derived adult enterocytes. Here we used a conditional inactivation strategy to eliminate Blimp1 function in the developing intestinal epithelium. There was no noticeable effect on gross morphology or formation of mature cell types before birth. However, survival of mutant neonates was severely compromised. Transcriptional profiling experiments reveal global changes in gene expression patterns. Key components of the adult enterocyte biochemical signature were substantially and prematurely activated. In contrast, those required for processing maternal milk were markedly reduced. Thus, we conclude Blimp1 governs the developmental switch responsible for postnatal intestinal maturation.


Asunto(s)
Enterocitos/citología , Intestinos/crecimiento & desarrollo , Proteínas Represoras/fisiología , Factores de Transcripción/fisiología , Transcripción Genética , Animales , Femenino , Perfilación de la Expresión Génica , Intestinos/citología , Masculino , Ratones , Factor 1 de Unión al Dominio 1 de Regulación Positiva , Factores de Transcripción/genética
14.
Proc Natl Acad Sci U S A ; 108(10): 4006-11, 2011 Mar 08.
Artículo en Inglés | MEDLINE | ID: mdl-21330551

RESUMEN

We report that the dominant human missense mutations G303E and G296S in GATA4, a cardiac-specific transcription factor gene, cause atrioventricular septal defects and valve abnormalities by disrupting a signaling cascade involved in endocardial cushion development. These GATA4 missense mutations, but not a mutation causing secundum atrial septal defects (S52F), demonstrated impaired protein interactions with SMAD4, a transcription factor required for canonical bone morphogenetic protein/transforming growth factor-ß (BMP/TGF-ß) signaling. Gata4 and Smad4 genetically interact in vivo: atrioventricular septal defects result from endothelial-specific Gata4 and Smad4 compound haploinsufficiency. Endothelial-specific knockout of Smad4 caused an absence of valve-forming activity: Smad4-deficient endocardium was associated with acellular endocardial cushions, absent epithelial-to-mesenchymal transformation, reduced endocardial proliferation, and loss of Id2 expression in valve-forming regions. We show that Gata4 and Smad4 cooperatively activated the Id2 promoter, that human GATA4 mutations abrogated this activity, and that Id2 deficiency in mice could cause atrioventricular septal defects. We suggest that one determinant of the phenotypic spectrum caused by human GATA4 mutations is differential effects on GATA4/SMAD4 interactions required for endocardial cushion development.


Asunto(s)
Factor de Transcripción GATA4/genética , Válvulas Cardíacas/embriología , Proteína Smad4/genética , Animales , Proteínas Morfogenéticas Óseas/metabolismo , Transición Epitelial-Mesenquimal , Femenino , Humanos , Masculino , Ratones , Morfogénesis , Mutación , Linaje , Regiones Promotoras Genéticas , Factor de Crecimiento Transformador beta/metabolismo
15.
Am J Pathol ; 178(3): 1069-79, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21356359

RESUMEN

Bone morphogenetic protein-6 (BMP-6) suppresses inflammatory genes in renal proximal tubular cells and regulates iron metabolism by inducing hepcidin. In diabetic patients, an increase of myofibroblast progenitor cells (MFPCs), also known as fibrocytes, was found to be associated with decreased BMP-6 expression. We hypothesized that loss of endogenous BMP-6 would aggravate renal injury and fibrosis. Wild type (WT) and BMP-6 null mice underwent unilateral ureteral obstruction. In WT mice, ureteral obstruction down-regulated BMP-6. Obstructed kidneys of BMP-6 null mice showed more casts (1.5-fold), epithelial necrosis (1.4-fold), and brush border loss (1.3-fold). This was associated with more inflammation (1.8-fold more CD45(+) cells) and more pronounced overexpression of profibrotic genes for αSMA (2.0-fold), collagen I (6.8-fold), fibronectin (4.3-fold), CTGF (1.8-fold), and PAI-1 (3.8-fold), despite similar BMP-7 expression. Also, 1.3-fold more MFPCs were obtained from BMP-6 null than from WT mononuclear cell cultures, but in vivo only very few MFPCs were observed in obstructed kidneys, irrespective of BMP-6 genotype. The obstructed kidneys of BMP-6 null mice showed 2.2-fold more iron deposition, in association with 3.3-fold higher expression of the oxidative stress marker HO-1. Thus, ureteral obstruction leads to down-regulation of BMP-6 expression, and BMP-6 deficiency aggravates tubulointerstitial damage and fibrosis independent of BMP-7. This process appears to involve loss of both direct anti-inflammatory and antifibrotic action and indirect suppressive effects on renal iron deposition, oxidative stress, and MFPCs.


Asunto(s)
Proteína Morfogenética Ósea 6/deficiencia , Riñón/metabolismo , Riñón/patología , Actinas/metabolismo , Animales , Peso Corporal , Proteína Morfogenética Ósea 6/metabolismo , Cadherinas/genética , Cadherinas/metabolismo , Factor de Crecimiento del Tejido Conjuntivo/genética , Factor de Crecimiento del Tejido Conjuntivo/metabolismo , Matriz Extracelular/genética , Fibroblastos/metabolismo , Fibroblastos/patología , Fibrosis , Regulación de la Expresión Génica , Hemo-Oxigenasa 1/metabolismo , Inflamación/metabolismo , Inflamación/patología , Hierro/metabolismo , Túbulos Renales/enzimología , Túbulos Renales/patología , Ratones , Inhibidor 1 de Activador Plasminogénico/genética , Inhibidor 1 de Activador Plasminogénico/metabolismo , Transducción de Señal , Células Madre/metabolismo , Células Madre/patología , Factor de Crecimiento Transformador beta/genética , Factor de Crecimiento Transformador beta/metabolismo
16.
Dev Cell ; 11(3): 313-23, 2006 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-16950123

RESUMEN

During early mouse development, the subtilisin-like proprotein convertases (SPC) Furin and PACE4 pattern the primitive ectoderm and visceral endoderm, presumably by activating the TGFss-related Nodal precursor. Here, mutation of the SPC motif provides direct evidence that Nodal processing is essential to specify anterior visceral endoderm and mesendoderm. Surprisingly, however, the Nodal precursor binds and activates activin receptors to maintain expression of Furin, PACE4, and Bmp4 in extraembryonic ectoderm at a distance from the Nodal source. In return, Bmp4 induces Wnt3, which amplifies Nodal expression in the epiblast and mediates induction of mesoderm. We conclude that uncleaved Nodal sustains the extraembryonic source of proprotein convertases and Bmp4 to amplify Nodal signaling in two nonredundant feedback loops with dual timescales and to localize primitive streak formation at the posterior pole. Based on mathematical modeling, we discuss how these sequential loops control cell fate.


Asunto(s)
Receptores de Activinas/metabolismo , Tipificación del Cuerpo , Proteínas Morfogenéticas Óseas/metabolismo , Mesodermo/fisiología , Proproteína Convertasas/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Animales , Secuencia de Bases , Elementos de Facilitación Genéticos , Retroalimentación Fisiológica , Regulación del Desarrollo de la Expresión Génica , Ratones , Ratones Endogámicos C57BL , Modelos Biológicos , Datos de Secuencia Molecular , Proteína Nodal , Precursores de Proteínas/metabolismo , Homología de Secuencia de Ácido Nucleico , Transducción de Señal , Factor de Crecimiento Transformador beta/genética , Factor de Crecimiento Transformador beta/fisiología , Proteínas Wnt/fisiología , Proteína Wnt3
17.
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
18.
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
19.
Genesis ; 47(11): 775-81, 2009 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-19830823

RESUMEN

During mouse embryonic development, the T-box transcription factor Eomes/Tbr2 is expressed in highly dynamic patterns in various progenitor cell types. Those include the undifferentiated cells of the trophectoderm, ingressing nascent mesoderm at the primitive streak, and intermediate progenitor cells of the developing cerebral cortex. We generated an Eomes(GFP)- targeted allele to follow the highly dynamic patterns of Eomes expression and to allow for the identification of novel expression domains. We show that our novel allele recapitulates endogenous gene expression at known sites of expression and confirm our results by anti-Eomes immunofluorescent staining. Using this novel allele we were able to identify previously undocumented domains of Eomes expression within the visceral endoderm and at various locations in the developing and adult mouse brain.


Asunto(s)
Proteínas Fluorescentes Verdes/genética , Proteínas de Dominio T Box/genética , Alelos , Animales , Secuencia de Bases , Cartilla de ADN , Genes Reporteros , Hibridación in Situ , Ratones
20.
Dev Biol ; 318(1): 184-93, 2008 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-18448090

RESUMEN

The three fibronectin leucine-rich repeat transmembrane (FLRT) proteins contain 10 leucine-rich repeats (LRR), a type III fibronectin (FN) domain, followed by the transmembrane region, and a short cytoplasmic tail. XFLRT3, a Nodal/TGFbeta target, regulates cell adhesion and modulates FGF signalling during Xenopus gastrulation. The present study describes the onset and pattern of FLRT1-3 expression in the early mouse embryo. FLRT3 expression is activated in the anterior visceral endoderm (AVE), and during gastrulation appears in anterior streak derivatives namely the node, notochord and the emerging definitive endoderm. To explore FLRT3 function we generated a null allele via gene targeting. Early Nodal activities required for anterior-posterior (A-P) patterning, primitive streak formation and left-right (L-R) axis determination were unperturbed. However, FLRT3 mutant embryos display defects in headfold fusion, definitive endoderm migration and a failure of the lateral edges of the ventral body wall to fuse, leading to cardia bifida. Surprisingly, the mutation has no effect on FGF signalling. Collectively these experiments demonstrate that FLRT3 plays a key role in controlling cell adhesion and tissue morphogenesis in the developing mouse embryo.


Asunto(s)
Movimiento Celular/fisiología , Embrión de Mamíferos/fisiología , Endodermo/fisiología , Glicoproteínas de Membrana/metabolismo , Morfogénesis , Animales , Adhesión Celular/fisiología , Embrión de Mamíferos/anatomía & histología , Endodermo/citología , Factores de Crecimiento de Fibroblastos/metabolismo , Regulación del Desarrollo de la Expresión Génica , Marcación de Gen , Genes Reporteros , Genotipo , Humanos , Hibridación in Situ , Glicoproteínas de Membrana/genética , Ratones , Ratones Transgénicos , Transducción de Señal/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA