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

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Development ; 148(5)2021 03 09.
Artículo en Inglés | MEDLINE | ID: mdl-33574039

RESUMEN

In mice, the entry of germ cells into meiosis crucially depends on the expression of stimulated by retinoic acid gene 8 (Stra8). Stra8 is expressed specifically in pre-meiotic germ cells of females and males, at fetal and postnatal stages, respectively, but the mechanistic details of its spatiotemporal regulation are yet to be defined. In particular, there has been considerable debate regarding whether retinoic acid is required, in vivo, to initiate Stra8 expression in the mouse fetal ovary. We show that the distinctive anterior-to-posterior pattern of Stra8 initiation, characteristic of germ cells in the fetal ovary, is faithfully recapitulated when 2.9 kb of the Stra8 promoter is used to drive eGFP expression. Using in vitro transfection assays of cutdown and mutant constructs, we identified two functional retinoic acid responsive elements (RAREs) within this 2.9 kb regulatory element. We also show that the transcription factor DMRT1 enhances Stra8 expression, but only in the presence of RA and the most proximal RARE. Finally, we used CRISPR/Cas9-mediated targeted mutation studies to demonstrate that both RAREs are required for optimal Stra8 expression levels in vivo.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Células Germinativas/metabolismo , Secuencias Reguladoras de Ácidos Nucleicos/genética , Proteínas Adaptadoras Transductoras de Señales/genética , Animales , Sitios de Unión , Sistemas CRISPR-Cas/genética , Femenino , Desarrollo Fetal/genética , Feto/citología , Feto/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Células Germinativas/citología , Meiosis , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Mutagénesis , Ovario/citología , Ovario/metabolismo , Regiones Promotoras Genéticas , Receptores X Retinoide/genética , Receptores X Retinoide/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Factores de Transcripción/farmacología , Tretinoina/farmacología
2.
Hum Mol Genet ; 25(21): 4674-4685, 2016 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-28173120

RESUMEN

TMEM70, a 21-kDa protein localized in the inner mitochondrial membrane, has been shown to facilitate the biogenesis of mammalian F1Fo ATP synthase. Mutations of the TMEM70 gene represent the most frequent cause of isolated ATP synthase deficiency resulting in a severe mitochondrial disease presenting as neonatal encephalo-cardiomyopathy (OMIM 604273). To better understand the biological role of this factor, we generated Tmem70-deficient mice and found that the homozygous Tmem70-/- knockouts exhibited profound growth retardation and embryonic lethality at ∼9.5 days post coitum. Blue-Native electrophoresis demonstrated an isolated deficiency in fully assembled ATP synthase in the Tmem70-/- embryos (80% decrease) and a marked accumulation of F1 complexes indicative of impairment in ATP synthase biogenesis that was stalled at the early stage, following the formation of F1 oligomer. Consequently, a decrease in ADP-stimulated State 3 respiration, respiratory control ratio and ATP/ADP ratios, indicated compromised mitochondrial ATP production. Tmem70-/- embryos exhibited delayed development of the cardiovascular system and a disturbed heart mitochondrial ultrastructure, with concentric or irregular cristae structures. Tmem70+/- heterozygous mice were fully viable and displayed normal postnatal growth and development of the mitochondrial oxidative phosphorylation system. Nevertheless, they presented with mild deterioration of heart function. Our results demonstrated that Tmem70 knockout in the mouse results in embryonic lethality due to the lack of ATP synthase and impairment of mitochondrial energy provision. This is analogous to TMEM70 dysfunction in humans and verifies the crucial role of this factor in the biosynthesis and assembly of mammalian ATP synthase.


Asunto(s)
Proteínas de la Membrana/genética , Proteínas Mitocondriales/genética , ATPasas de Translocación de Protón Mitocondriales/genética , Adenosina Trifosfato/metabolismo , Animales , Cardiomiopatías/metabolismo , Femenino , Homocigoto , Proteínas de la Membrana/deficiencia , Proteínas de la Membrana/metabolismo , Errores Innatos del Metabolismo/metabolismo , Ratones , Ratones Noqueados , Mitocondrias/metabolismo , Enfermedades Mitocondriales/genética , Enfermedades Mitocondriales/metabolismo , Membranas Mitocondriales/metabolismo , Proteínas Mitocondriales/deficiencia , Proteínas Mitocondriales/metabolismo , ATPasas de Translocación de Protón Mitocondriales/biosíntesis , ATPasas de Translocación de Protón Mitocondriales/metabolismo , Mutación , Fosforilación Oxidativa , Embarazo
3.
Development ; 138(1): 65-74, 2011 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-21098558

RESUMEN

An interplay of transcription factors interprets signalling pathways to define anteroposterior positions along the vertebrate axis. In the hindbrain, these transcription factors prompt the position-appropriate appearance of seven to eight segmental structures, known as rhombomeres (r1-r8). The evolutionarily conserved Cdx caudal-type homeodomain transcription factors help specify the vertebrate trunk and tail but have not been shown to directly regulate hindbrain patterning genes. Mafb (Kreisler, Krml1, valentino), a basic domain leucine zipper transcription factor, is required for development of r5 and r6 and is the first gene to show restricted expression within these two segments. The homeodomain protein vHnf1 (Hnf1b) directly activates Mafb expression. vHnf1 and Mafb share an anterior expression limit at the r4/r5 boundary but vHnf1 expression extends beyond the posterior limit of Mafb and, therefore, cannot establish the posterior Mafb expression boundary. Upon identifying regulatory sequences responsible for posterior Mafb repression, we have used in situ hybridization, immunofluorescence and chromatin immunoprecipitation (ChIP) analyses to determine that Cdx1 directly inhibits early Mafb expression in the neural tube posterior of the r6/r7 boundary, which is the anteriormost boundary of Cdx1 expression in the hindbrain. Cdx1 dependent repression of Mafb is transient. After the 10-somite stage, another mechanism acts to restrict Mafb expression in its normal r5 and r6 domain, even in the absence of Cdx1. Our findings identify Mafb as one of the earliest direct targets of Cdx1 and show that Cdx1 plays a direct role in early hindbrain patterning. Thus, just as Cdx2 and Cdx4 govern the trunk-to-tail transition, Cdx1 may regulate the hindbrain-to-spinal cord transition.


Asunto(s)
Elementos de Facilitación Genéticos/genética , Proteínas de Homeodominio/metabolismo , Factor de Transcripción MafB/metabolismo , Rombencéfalo/embriología , Rombencéfalo/metabolismo , Animales , Sitios de Unión , Inmunoprecipitación de Cromatina , Ensayo de Cambio de Movilidad Electroforética , Técnica del Anticuerpo Fluorescente , Proteínas de Homeodominio/genética , Hibridación in Situ , Factor de Transcripción MafB/genética , Ratones , Ratones Transgénicos , Unión Proteica/genética , Unión Proteica/fisiología
4.
Sci Adv ; 10(6): eadk3384, 2024 Feb 09.
Artículo en Inglés | MEDLINE | ID: mdl-38335290

RESUMEN

Disruption of cell division cycle associated 7 (CDCA7) has been linked to aberrant DNA hypomethylation, but the impact of DNA methylation loss on transcription has not been investigated. Here, we show that CDCA7 is critical for maintaining global DNA methylation levels across multiple tissues in vivo. A pathogenic Cdca7 missense variant leads to the formation of large, aberrantly hypomethylated domains overlapping with the B genomic compartment but without affecting the deposition of H3K9 trimethylation (H3K9me3). CDCA7-associated aberrant DNA hypomethylation translated to localized, tissue-specific transcriptional dysregulation that affected large gene clusters. In the brain, we identify CDCA7 as a transcriptional repressor and epigenetic regulator of clustered protocadherin isoform choice. Increased protocadherin isoform expression frequency is accompanied by DNA methylation loss, gain of H3K4 trimethylation (H3K4me3), and increased binding of the transcriptional regulator CCCTC-binding factor (CTCF). Overall, our in vivo work identifies a key role for CDCA7 in safeguarding tissue-specific expression of gene clusters via the DNA methylation pathway.


Asunto(s)
Proteínas de Ciclo Celular , Proteínas Nucleares , ADN , Metilación de ADN , Isoformas de Proteínas/genética , Proteínas Represoras/genética , Factores de Transcripción/genética , Animales , Ratones , Proteínas de Ciclo Celular/metabolismo , Proteínas Nucleares/metabolismo
5.
Biol Reprod ; 86(1): 1-12, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-21900680

RESUMEN

Disruptions in the regulatory pathways controlling sex determination and differentiation can cause disorders of sex development, often compromising reproductive function. Although extensive efforts have been channeled into elucidating the regulatory mechanisms controlling the many aspects of sexual differentiation, the majority of disorders of sex development phenotypes are still unexplained at the molecular level. In this study, we have analyzed the potential involvement of Wnt5a in sexual development and show in mice that Wnt5a is male-specifically upregulated within testicular interstitial cells at the onset of gonad differentiation. Homozygous deletion of Wnt5a affected sexual development in male mice, causing testicular hypoplasia and bilateral cryptorchidism despite the Leydig cells producing factors such as Hsd3b1 and Insl3. Additionally, Wnt5a-null embryos of both sexes showed a significant reduction in gonadal germ cell numbers, which was caused by aberrant primordial germ cell migration along the hindgut endoderm prior to gonadal colonization. Our results indicate multiple roles for Wnt5a during mammalian reproductive development and help to clarify further the etiology of Robinow syndrome (OMIM 268310), a disease previously linked to the WNT5A pathway.


Asunto(s)
Regulación del Desarrollo de la Expresión Génica/fisiología , Maduración Sexual/fisiología , Proteínas Wnt/metabolismo , Animales , Criptorquidismo , Feto/fisiología , Células Germinativas , Homocigoto , Masculino , Ratones , Procesos de Determinación del Sexo/fisiología , Transducción de Señal , Testículo/embriología , Proteínas Wnt/genética , Proteína Wnt-5a
6.
Nature ; 438(7070): 937-45, 2005 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-16355211

RESUMEN

The intricate patterning processes that establish the complex vascular system during development depend on a combination of intrinsic pre-patterning and extrinsic responses to environmental parameters. Mutational studies in mice and fish have shown that the vascular system is highly sensitive to genetic disruption and have identified potential targets for therapeutic interventions. New insights into non-vascular roles of vascular endothelial growth factor and the requirement for endothelial cells in adult organs and stem-cell niches highlight possible side effects of anti-angiogenic therapy and the need for new targets.


Asunto(s)
Células Endoteliales/metabolismo , Neovascularización Fisiológica/fisiología , Factor A de Crecimiento Endotelial Vascular/metabolismo , Animales , Células Endoteliales/citología , Organogénesis/fisiología , Transducción de Señal , Células Madre/citología , Células Madre/fisiología
7.
Proc Natl Acad Sci U S A ; 105(22): 7756-61, 2008 Jun 03.
Artículo en Inglés | MEDLINE | ID: mdl-18511567

RESUMEN

Cdx genes (Cdx1, Cdx2, and Cdx4) encode a family of caudal-related transcription factors that mediate anterior-posterior patterning during embryogenesis through Hox gene regulation. Homologues in the zebrafish have been shown to play key roles in blood formation. To define the role of Cdx genes during embryonic hematopoiesis in a mammalian system, we examined the hematopoietic potential of Cdx-deficient mouse embryonic stem cells (ESCs) in vitro and in vivo. Individual Cdx-deficient ESCs exhibited impaired embryonic hematopoietic progenitor formation and altered Hox gene expression, most notably for Cdx2 deficiency. A more severe hematopoietic defect was observed with compound Cdx deficiency than loss of function of any single Cdx gene. Reduced hematopoietic progenitor formation of ESCs deficient in multiple Cdx genes could be rescued by ectopic expression of Cdx4, concomitant with partially restored Hox gene expression. These results reveal an essential and partially redundant role for multiple Cdx genes during embryonic hematopoiesis in the mouse.


Asunto(s)
Embrión de Mamíferos/citología , Células Madre Embrionarias/citología , Hematopoyesis/genética , Células Madre Hematopoyéticas/citología , Proteínas de Homeodominio/fisiología , Factores de Transcripción/fisiología , Animales , Factor de Transcripción CDX2 , Embrión de Mamíferos/metabolismo , Células Madre Embrionarias/metabolismo , Células Madre Hematopoyéticas/metabolismo , Proteínas de Homeodominio/genética , Ratones , Ratones Noqueados , Factores de Transcripción/genética
8.
J Clin Invest ; 117(11): 3369-82, 2007 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-17948123

RESUMEN

The Notch family of cell surface receptors and its ligands are highly conserved proteins that regulate cell fate determination, including those involved in mammalian vascular development. We report that Notch induces VEGFR-3 expression in vitro in human endothelial cells and in vivo in mice. In vitro, Notch in complex with the DNA-binding protein CBF-1/suppressor of hairless/Lag1 (CSL) bound the VEGFR-3 promoter and transactivated VEGFR-3 specifically in endothelial cells. Through induction of VEGFR-3, Notch increased endothelial cell responsiveness to VEGF-C, promoting endothelial cell survival and morphological changes. In vivo, VEGFR-3 was upregulated in endothelial cells with active Notch signaling. Mice heterozygous for null alleles of both Notch1 and VEGFR-3 had significantly reduced viability and displayed midgestational vascular patterning defects analogous to Notch1 nullizygous embryos. We found that Notch1 and Notch4 were expressed in normal and tumor lymphatic endothelial cells and that Notch1 was activated in lymphatic endothelium of invasive mammary micropapillary carcinomas. These results demonstrate that Notch1 and VEGFR-3 interact genetically, that Notch directly induces VEGFR-3 in blood endothelial cells to regulate vascular development, and that Notch may function in tumor lymphangiogenesis.


Asunto(s)
Células Endoteliales/metabolismo , Receptores Notch/metabolismo , Factor A de Crecimiento Endotelial Vascular/metabolismo , Receptor 3 de Factores de Crecimiento Endotelial Vascular/metabolismo , Animales , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Forma de la Célula , Supervivencia Celular , Células Cultivadas , Embrión de Mamíferos/anatomía & histología , Embrión de Mamíferos/fisiología , Células Endoteliales/citología , Femenino , Regulación de la Expresión Génica , Humanos , Ratones , Receptores Notch/genética , Transducción de Señal/fisiología , Factor A de Crecimiento Endotelial Vascular/genética , Receptor 3 de Factores de Crecimiento Endotelial Vascular/genética
9.
Genes (Basel) ; 10(11)2019 11 14.
Artículo en Inglés | MEDLINE | ID: mdl-31739541

RESUMEN

The Cdx2 homeobox gene is important in assigning positional identity during the finely orchestrated process of embryogenesis. In adults, regenerative responses to tissues damage can require a replay of these same developmental pathways. Errors in reassigning positional identity during regeneration can cause metaplasias-normal tissue arising in an abnormal location-and this in turn, is a well-recognized cancer risk factor. In animal models, a gain of Cdx2 function can elicit a posterior shift in tissue identity, modeling intestinal-type metaplasias of the esophagus (Barrett's esophagus) and stomach. Conversely, loss of Cdx2 function can elicit an anterior shift in tissue identity, inducing serrated-type lesions expressing gastric markers in the colon. These metaplasias are major risk factors for the later development of esophageal, stomach and colon cancer. Leukemia, another cancer in which Cdx2 is ectopically expressed, may have mechanistic parallels with epithelial cancers in terms of stress-induced reprogramming. This review will address how animal models have refined our understanding of the role of Cdx2 in these common human cancers.


Asunto(s)
Factor de Transcripción CDX2/metabolismo , Neoplasias del Colon/genética , Neoplasias Esofágicas/genética , Regulación Neoplásica de la Expresión Génica , Genes Homeobox/genética , Neoplasias Gástricas/genética , Animales , Factor de Transcripción CDX2/genética , Colon/patología , Neoplasias del Colon/patología , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Neoplasias Esofágicas/patología , Esófago/patología , Humanos , Metaplasia/genética , Metaplasia/patología , Estómago/patología , Neoplasias Gástricas/patología
10.
Sci Rep ; 8(1): 5762, 2018 Apr 05.
Artículo en Inglés | MEDLINE | ID: mdl-29622773

RESUMEN

A correction to this article has been published and is linked from the HTML and PDF versions of this paper. The error has been fixed in the paper.

11.
Biotechniques ; 42(3): 317-8, 320, 322-5, 2007 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-17390538

RESUMEN

The trophoblast layers of the mammalian placenta carry out many complex functions required to pattern the developing embryo and maintain its growth and survival in the uterine environment. Genetic disruption of many gene pathways can result in embryonic lethality because of placental failure, potentially confusing the interpretation of mouse knockout phenotypes. Development of tools to specifically and efficiently manipulate gene expression in the trophoblast lineage would greatly aid understanding of the relative roles of different genetic pathways in the trophoblast versus embryonic lineages. We show that short-term lentivirus-mediated infection of mouse blastocysts can lead to rapid expression of a green fluorescent protein (GFP) transgene specifically in the outer trophoblast progenitors and their later placental derivatives. Efficient trophoblast-specific gene knockdown can also be produced by lentivirus-mediated pol III-driven short hairpin RNA (shRNA) and efficient trophoblast-specific gene knockout by pol II-driven Cre recombinase lentiviral vectors. This lentivirus lineage-specific infection system thus facilitates both gain and loss of function studies during placental development in the mouse and potentially other mammalian species.


Asunto(s)
Vectores Genéticos , Lentivirus/genética , Trofoblastos/metabolismo , Animales , Biotecnología/métodos , Blastocisto/metabolismo , Proteínas Fluorescentes Verdes/metabolismo , Integrasas/genética , Operón Lac , Ratones , Ratones Noqueados , Plásmidos/metabolismo , ARN/metabolismo , ARN Interferente Pequeño , Factores de Tiempo
12.
Sci Rep ; 7(1): 9322, 2017 08 24.
Artículo en Inglés | MEDLINE | ID: mdl-28839193

RESUMEN

Gastrulation initiates with the formation of the primitive streak, during which, cells of the epiblast delaminate to form the mesoderm and definitive endoderm. At this stage, the pluripotent cell population of the epiblast undergoes very rapid proliferation and extensive epigenetic programming. Here we show that Fam208a, a new epigenetic modifier, is essential for early post-implantation development. We show that Fam208a mutation leads to impaired primitive streak elongation and delayed epithelial-to-mesenchymal transition. Fam208a mutant epiblasts had increased expression of p53 pathway genes as well as several pluripotency-associated long non-coding RNAs. Fam208a mutants exhibited an increase in p53-driven apoptosis and complete removal of p53 could partially rescue their gastrulation block. This data demonstrates a new in vivo function of Fam208a in maintaining epiblast fitness, establishing it as an important factor at the onset of gastrulation when cells are exiting pluripotency.


Asunto(s)
Epigénesis Genética , Estratos Germinativos/embriología , Estratos Germinativos/fisiología , Proteínas Nucleares/metabolismo , Animales , Apoptosis , Transición Epitelial-Mesenquimal , Ratones , Mutación , Proteínas Nucleares/genética , Línea Primitiva/embriología
13.
PLoS One ; 9(4): e94813, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24743337

RESUMEN

The Y-chromosomal gene SRY acts as the primary trigger for male sex determination in mammalian embryos. Correct regulation of SRY is critical: aberrant timing or level of Sry expression is known to disrupt testis development in mice and we hypothesize that mutations that affect regulation of human SRY may account for some of the many cases of XY gonadal dysgenesis that currently remain unexplained. However, the cis-sequences involved in regulation of Sry have not been identified, precluding a test of this hypothesis. Here, we used a transgenic mouse approach aimed at identifying mouse Sry 5' flanking regulatory sequences within 8 kb of the Sry transcription start site (TSS). To avoid problems associated with conventional pronuclear injection of transgenes, we used a published strategy designed to yield single-copy transgene integration at a defined, transcriptionally open, autosomal locus, Col1a1. None of the Sry transgenes tested was expressed at levels compatible with activation of Sox9 or XX sex reversal. Our findings indicate either that the Col1a1 locus does not provide an appropriate context for the correct expression of Sry transgenes, or that the cis-sequences required for Sry expression in the developing gonads lie beyond 8 kb 5' of the TSS.


Asunto(s)
Técnicas de Transferencia de Gen , Genes sry/genética , Secuencias Reguladoras de Ácidos Nucleicos/genética , Testículo/metabolismo , Animales , Línea Celular , Colágeno Tipo I/genética , Cadena alfa 1 del Colágeno Tipo I , Sitios Genéticos/genética , Técnicas de Genotipaje , Masculino , Ratones , Sitio de Iniciación de la Transcripción , Transgenes/genética
14.
PLoS One ; 6(9): e25228, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21980401

RESUMEN

The Tmem26 gene encodes a novel protein that we have previously shown to be regulated by hedgehog signalling in the mouse limb. We now report that Tmem26 expression is spatially and temporally restricted in other regions of the mouse embryo, most notably the facial primordia. In particular, Tmem26 expression in the mesenchyme of the maxillary and nasal prominences is coincident with fusion of the primary palate. In the secondary palate, Tmem26 is expressed in the palatal shelves during their growth and fusion but is downregulated once fusion is complete. Expression was also detected at the midline of the expanding mandible and at the tips of the eyelids as they migrate across the cornea. Given the spatio-temporally restricted expression of Tmem26, we sought to uncover a functional role in embryonic development through targeted gene inactivation in the mouse. However, ubiquitous inactivation of Tmem26 led to no overt phenotype in the resulting embryos or adult mice, suggesting that TMEM26 function is dispensable for embryonic survival.


Asunto(s)
Extremidades/embriología , Glicoproteínas de Membrana/metabolismo , Hueso Paladar/embriología , Animales , Huesos Faciales/embriología , Regulación del Desarrollo de la Expresión Génica/genética , Regulación del Desarrollo de la Expresión Génica/fisiología , Glicoproteínas de Membrana/genética , Ratones , Ratones Endogámicos C57BL , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
15.
Proc Natl Acad Sci U S A ; 104(3): 846-51, 2007 Jan 16.
Artículo en Inglés | MEDLINE | ID: mdl-17210915

RESUMEN

Notch-mediated induction of Nodal at the vertebrate node is a critical step in initiating left-right (LR) asymmetry. In mice and zebrafish we show that Baf60c, a subunit of the Swi/Snf-like BAF chromatin remodeling complex, is essential for establishment of LR asymmetry. Baf60c knockdown mouse embryos fail to activate Nodal at the node and also have abnormal node morphology with mixing of crown and pit cells. In cell culture, Baf60c is required for Notch-dependent transcriptional activation and functions to stabilize interactions between activated Notch and its DNA-binding partner, RBP-J. Brg1 is also required for these processes, suggesting that BAF complexes are key components of nuclear Notch signaling. We propose a critical role for Baf60c in Notch-dependent transcription and LR asymmetry.


Asunto(s)
Tipificación del Cuerpo , Núcleo Celular/metabolismo , Proteínas Cromosómicas no Histona/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Proteínas Musculares/metabolismo , Receptores Notch/metabolismo , Transducción de Señal , Proteínas de Pez Cebra/metabolismo , Animales , Animales Modificados Genéticamente , Proteínas Cromosómicas no Histona/deficiencia , Proteínas Cromosómicas no Histona/genética , Embrión de Mamíferos/embriología , Embrión de Mamíferos/metabolismo , Embrión no Mamífero , Regulación del Desarrollo de la Expresión Génica , Péptidos y Proteínas de Señalización Intracelular/genética , Ratones , Proteínas Musculares/deficiencia , Proteínas Musculares/genética , Proteína Nodal , Transcripción Genética/genética , Factor de Crecimiento Transformador beta/genética , Factor de Crecimiento Transformador beta/metabolismo , Pez Cebra/embriología , Pez Cebra/genética , Pez Cebra/metabolismo , Proteínas de Pez Cebra/genética
16.
Science ; 312(5773): 596-600, 2006 Apr 28.
Artículo en Inglés | MEDLINE | ID: mdl-16574820

RESUMEN

Germ cells in the mouse embryo can develop as oocytes or spermatogonia, depending on molecular cues that have not been identified. We found that retinoic acid, produced by mesonephroi of both sexes, causes germ cells in the ovary to enter meiosis and initiate oogenesis. Meiosis is retarded in the fetal testis by the action of the retinoid-degrading enzyme CYP26B1, ultimately leading to spermatogenesis. In testes of Cyp26b1-knockout mouse embryos, germ cells enter meiosis precociously, as if in a normal ovary. Thus, precise regulation of retinoid levels during fetal gonad development provides the molecular control mechanism that specifies germ cell fate.


Asunto(s)
Sistema Enzimático del Citocromo P-450/metabolismo , Células Germinativas/fisiología , Meiosis , Oogénesis , Transducción de Señal , Espermatogénesis , Tretinoina/metabolismo , Animales , Sistema Enzimático del Citocromo P-450/genética , Femenino , Regulación del Desarrollo de la Expresión Génica , Genes Reporteros , Hibridación in Situ , Operón Lac , Masculino , Mesonefro/metabolismo , Ratones , Ratones Noqueados , Naftalenos/farmacología , Ovario/embriología , Ovario/metabolismo , Receptores de Ácido Retinoico/antagonistas & inhibidores , Ácido Retinoico 4-Hidroxilasa , Células de Sertoli/enzimología , Caracteres Sexuales , Testículo/embriología , Testículo/metabolismo , Técnicas de Cultivo de Tejidos , Tretinoina/farmacología
17.
Development ; 132(9): 2093-102, 2005 May.
Artículo en Inglés | MEDLINE | ID: mdl-15788452

RESUMEN

Blastocyst formation marks the segregation of the first two cell lineages in the mammalian preimplantation embryo: the inner cell mass (ICM) that will form the embryo proper and the trophectoderm (TE) that gives rise to the trophoblast lineage. Commitment to ICM lineage is attributed to the function of the two transcription factors, Oct4 (encoded by Pou5f1) and Nanog. However, a positive regulator of TE cell fate has not been described. The T-box protein eomesodermin (Eomes) and the caudal-type homeodomain protein Cdx2 are expressed in the TE, and both Eomes and Cdx2 homozygous mutant embryos die around the time of implantation. A block in early TE differentiation occurs in Eomes mutant blastocysts. However, Eomes mutant blastocysts implant, and Cdx2 and Oct4 expression are correctly restricted to the ICM TE. Blastocoel formation initiates in Cdx2 mutants but epithelial integrity is not maintained and embryos fail to implant. Loss of Cdx2 results in failure to downregulate Oct4 and Nanog in outer cells of the blastocyst and subsequent death of those cells. Thus, Cdx2 is essential for segregation of the ICM and TE lineages at the blastocyst stage by ensuring repression of Oct4 and Nanog in the TE.


Asunto(s)
Blastocisto/fisiología , Diferenciación Celular/fisiología , Ectodermo/fisiología , Proteínas de Homeodominio/fisiología , Factores de Transcripción/fisiología , Animales , Biomarcadores , Blastocisto/citología , Factor de Transcripción CDX2 , Proteínas de Unión al ADN/metabolismo , Ectodermo/citología , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Ratones , Proteína Homeótica Nanog , Factor 3 de Transcripción de Unión a Octámeros , Proteínas de Dominio T Box/metabolismo , Factores de Transcripción/deficiencia , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
18.
Proc Natl Acad Sci U S A ; 101(20): 7641-5, 2004 May 18.
Artículo en Inglés | MEDLINE | ID: mdl-15136723

RESUMEN

Inactivation of Cdx2 leads to preimplantation embryonic lethality. Rescue of the implantation defect by tetraploid fusion established that Cdx2 is necessary for trophoblastic development, vasculogenesis in the yolk sac mesoderm, allantoic growth, and chorioallantoic fusion. "Rescued" Cdx2 mutants die at late gastrulation stages because of failure of placental development. Cdx2 is also needed for the completion of the normal process of gastrulation and tail bud elongation. Presegmental paraxial mesoderm is severely restricted in amount and somites posterior to somite 5 are abnormal. The Cdx2 mutation, like mutations impairing Wnt and Fgf signaling, causes posterior truncations and disturbs axial patterning of the embryonic structures, indicated by changes in the Hox expression domains. The gene appears to be important in the integration of the pathways controlling embryonic axial elongation, and anterior-posterior patterning.


Asunto(s)
Blastocisto/metabolismo , Tipificación del Cuerpo/fisiología , Proteínas de Homeodominio/metabolismo , Ratones/embriología , Animales , Factor de Transcripción CDX2 , Ectodermo/metabolismo , Endodermo/metabolismo , Gástrula/metabolismo , Regulación del Desarrollo de la Expresión Génica/fisiología , Genes Homeobox/fisiología , Proteínas de Homeodominio/genética , Mesodermo/metabolismo , Fenotipo , Poliploidía , Células Madre/metabolismo , Factores de Transcripción , Trofoblastos/metabolismo
19.
Development ; 129(9): 2181-93, 2002 May.
Artículo en Inglés | MEDLINE | ID: mdl-11959827

RESUMEN

Mouse Cdx and Hox genes presumably evolved from genes on a common ancestor cluster involved in anteroposterior patterning. Drosophila caudal (cad) is involved in specifying the posterior end of the early embryo, and is essential for patterning tissues derived from the most caudal segment, the analia. Two of the three mouse Cdx paralogues, Cdx 1 and Cdx2, are expressed early in a Hox-like manner in the three germ layers. In the nascent paraxial mesoderm, both genes are expressed in cells contributing first to the most rostral, and then to progressively more caudal parts of the vertebral column. Later, expression regresses from the anterior sclerotomes, and is only maintained for Cdx1 in the dorsal part of the somites, and for both genes in the tail bud. Cdx1 null mutants show anterior homeosis of upper cervical and thoracic vertebrae. Cdx2-null embryos die before gastrulation, and Cdx2 heterozygotes display anterior transformations of lower cervical and thoracic vertebrae. We have analysed the genetic interactions between Cdx1 and Cdx2 in compound mutants. Combining mutant alleles for both genes gives rise to anterior homeotic transformations along a more extensive length of the vertebral column than do single mutations. The most severely affected Cdx1 null/Cdx2 heterozygous mice display a posterior shift of their cranio-cervical, cervico-thoracic, thoraco-lumbar, lumbo-sacral and sacro-caudal transitions. The effects of the mutations in Cdx1 and Cdx2 were co-operative in severity, and a more extensive posterior shift of the expression of three Hox genes was observed in double mutants. The alteration in Hox expression boundaries occurred early. We conclude that both Cdx genes cooperate at early stages in instructing the vertebral progenitors all along the axis, at least in part by setting the rostral expression boundaries of Hox genes. In addition, Cdx mutants transiently exhibit alterations in the extent of Hox expression domains in the spinal cord, reminding of the strong effects of overexpressing Cdx genes on Hox gene expression in the neurectoderm. Phenotypical alterations in the peripheral nervous system were observed at mid-gestation stages. Strikingly, the altered phenotype at caudal levels included a posterior truncation of the tail, mildly affecting Cdx2 heterozygotes, but more severely affecting Cdx1/Cdx2 double heterozygotes and Cdx1 null/Cdx2 heterozygotes. Mutations in Cdx1 and Cdx2 therefore also interfere with axis elongation in a cooperative way. The function of Cdx genes in morphogenetic processes during gastrulation and tail bud extension, and their relationship with the Hox genes are discussed in the light of available data in Amphioxus, C. elegans, Drosophila and mice.


Asunto(s)
Tipificación del Cuerpo/genética , Genes Homeobox , Proteínas de Homeodominio/genética , Columna Vertebral/embriología , Animales , Animales Recién Nacidos , Evolución Biológica , Factor de Transcripción CDX2 , Sistema Digestivo/embriología , Extremidades/embriología , Ganglios Espinales/anomalías , Ganglios Espinales/embriología , Regulación del Desarrollo de la Expresión Génica , Heterocigoto , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos CBA , Ratones Noqueados , Ratones Transgénicos , Familia de Multigenes , Mutación , Sistema Nervioso/embriología , Fenotipo , Columna Vertebral/anomalías , Transactivadores
20.
Dev Biol ; 255(2): 399-406, 2003 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-12648499

RESUMEN

Inactivation of Cdx2 by homologous recombination results in the development of forestomach epithelium at ectopic sites in pericaecal areas of the midgut of heterozygote mice. Local factors subsequently result in the secondary induction of tissues exhibiting an orderly sequence of tissue types between the ectopic forestomach tissue and the surrounding colon. Clonal analysis of this secondarily generated tissue using Y chromosome painting in chimaeric mice indicates that once differentiated to express Cdx2, host colonic epithelium can only form small intestinal-type epithelium, while Cdx2 mutant cells give rise to a succession of gastric-type tissue but never to a small intestine morphology. Our results indicate a difference in potency between forestomach and midgut precursor endodermal cells.


Asunto(s)
Sistema Digestivo/crecimiento & desarrollo , Genes Homeobox , Proteínas de Homeodominio/genética , Animales , Factor de Transcripción CDX2 , Quimera/genética , Pintura Cromosómica , Colon/crecimiento & desarrollo , Colon/metabolismo , Sistema Digestivo/metabolismo , Endodermo/citología , Endodermo/metabolismo , Femenino , Mucosa Gástrica/metabolismo , Proteínas de Homeodominio/metabolismo , Inmunohistoquímica , Hibridación in Situ , Pólipos Intestinales/genética , Pólipos Intestinales/metabolismo , Pólipos Intestinales/patología , Masculino , Ratones , Ratones Noqueados , Fenotipo , Embarazo , Estómago/crecimiento & desarrollo , Distribución Tisular , Transactivadores , Cromosoma Y/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA