Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 186
Filtrar
1.
Placenta ; 35(12): 1079-88, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25443433

RESUMO

INTRODUCTION: Fibroblast growth factor (FGF) signaling is essential for early trophoblast expansion and maintenance in the mouse, but is not required for trophectoderm specification during blastocyst formation. This signaling pathway is stably activated to expand the trophoblast stem cell compartment in vivo, while in vitro, FGFs are used for the derivation of trophoblast stem (TS) cells from blastocysts and early post-implantation mouse embryos. However, the function of FGFs during human trophoblast development is not known. METHODS: We sought to derive TS cells from human blastocysts in a number of culture conditions, including in the presence of FGFs and stem cell factor (SCF). We also investigated the expression of FGF receptors (FGFRs) in blastocysts, and the expression of FGFR2 and activated ERK1/2 in first trimester human placentae. RESULTS: We found that SCF, but not FGF2/4, improved the quality of blastocyst outgrowths, but we were unable to establish stable human TS cell lines. We observed CDX2 expression in the trophectoderm of fully blastocysts, but rarely observed transcription of FGFRs. FGFR2 protein was not detected in human blastocysts, but was strongly expressed in mouse blastocysts. However, we found robust FGFR2 expression and activated ERK1/2 in the cytotrophoblast layer of early human placenta. DISCUSSION: Our data suggests that initiation of FGF-dependent trophoblast expansion may occur later in human development, and is unlikely to drive maintenance of a TS cell compartment during the peri-implantation period. These findings suggest that cytotrophoblast preparations from early placentae may be a potential source of FGF-dependent human TS cells.


Assuntos
Blastocisto/metabolismo , Desenvolvimento Embrionário/fisiologia , Regulação da Expressão Gênica no Desenvolvimento , Receptores de Fatores de Crescimento de Fibroblastos/metabolismo , Trofoblastos/metabolismo , Animais , Blastocisto/citologia , Diferenciação Celular/genética , Feminino , Humanos , Camundongos , Gravidez , Receptores de Fatores de Crescimento de Fibroblastos/genética , Especificidade da Espécie , Trofoblastos/citologia
2.
Dev Dyn ; 239(6): 1888-900, 2010 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-20503384

RESUMO

Missense mutations in the Fibroblast Growth Factor Receptor 2 (FGFR2) have been identified in human craniosynostotic syndromes such as Crouzon (CS) and Pfeiffer (PS). FGFR2 has two major isoforms, IIIb and IIIc, generated through alternative splicing with their own temporal, spatial, and ligand-binding specificities. In this study, we report the identification and characterization of a missense mutation in codon 290 of murine Fgfr2 (W290R). The defects in W290R mutants are suggestive of disruption of signalling in both IIIb and IIIc isoforms of the Fgfr2 gene. Heterozygous mutants presented with features resembling those found in patients with CS. Fgfr2(W290R) homozygotes displayed constitutive FGFR2 activation with increased, but correct tissue-specific, expression of the IIIb and IIIc isoforms in many of the defective organs. Our Fgfr2(W290R) mouse model thus represents an excellent mouse model of CS to probe the many questions around the pathogenesis of craniosynostotic birth defects consequent to defects in FGF signaling.


Assuntos
Mutação de Sentido Incorreto , Receptor Tipo 2 de Fator de Crescimento de Fibroblastos/genética , Receptor Tipo 2 de Fator de Crescimento de Fibroblastos/metabolismo , Transdução de Sinais/genética , Processamento Alternativo , Animais , Disostose Craniofacial/genética , Craniossinostoses/genética , Cruzamentos Genéticos , Embrião de Mamíferos , Feminino , Heterozigoto , Homozigoto , Masculino , Camundongos , Camundongos Endogâmicos C3H , Camundongos Endogâmicos C57BL , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo
3.
Genes Brain Behav ; 6(8): 717-27, 2007 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-17376154

RESUMO

Dominant mutations of the P/Q-type Ca(2+) channel (CACNA1A) underlie several human neurological disorders, including episodic ataxia type 2, familial hemiplegic migraine 1 (FHM1) and spinocerebellar ataxia 6, but have not been found previously in the mouse. Here we report the first dominant ataxic mouse model of Cacna1a mutation. This Wobbly mutant allele of Cacna1a was identified in an ethylnitrosourea (ENU) mutagenesis dominant behavioral screen. Heterozygotes exhibit ataxia from 3 weeks of age and have a normal life span. Homozygotes have a righting reflex defect from postnatal day 8 and later develop severe ataxia and die prematurely. Both heterozygotes and homozygotes exhibit cerebellar atrophy with focal reduction of the molecular layer. No obvious loss of Purkinje cells or decrease in size of the granule cell layer was observed. Real-time polymerase chain reaction revealed altered expression levels of Cacna1g, Calb2 and Th in Wobbly cerebella, but Cacna1a messenger RNA and protein levels were unchanged. Positional cloning revealed that Wobbly mice have a missense mutation leading to an arginine to leucine (R1255L) substitution, resulting in neutralization of a positively charged amino acid in repeat III of voltage sensor segment S4. The dominance of the Wobbly mutation more closely resembles patterns of CACNA1A mutation in humans than previously described mouse recessive mutants (tottering, leaner, rolling Nagoya and rocker). Positive-charge neutralization in S4 has also been shown to underlie several cases of human dominant FHM1 with ataxia. The Wobbly mutant thus highlights the importance of the voltage sensor and provides a starting point to unravel the neuropathological mechanisms of this disease.


Assuntos
Ataxia/genética , Canais de Cálcio Tipo P/genética , Canais de Cálcio Tipo Q/genética , Cerebelo/patologia , Distonia/genética , Mutação de Sentido Incorreto/genética , Substituição de Aminoácidos/genética , Animais , Atrofia/genética , Canais de Cálcio Tipo N , Canais de Cálcio Tipo P/metabolismo , Canais de Cálcio Tipo Q/metabolismo , Cerebelo/metabolismo , Feminino , Marcha/genética , Genes Dominantes/genética , Masculino , Camundongos , Camundongos Endogâmicos C3H , Camundongos Mutantes , Polimorfismo de Nucleotídeo Único/genética
4.
Clin Genet ; 68(2): 106-12, 2005 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-15996204

RESUMO

'Stem cell' has practically become a household term, but what is a stem cell and where does it come from? Insight into these questions has come from the early mouse embryo, or blastocyst, from which three kinds of stem cells have been derived: embryonic stem (ES) cells, trophoblast stem (TS) cells, and extraembryonic endoderm (XEN) cells. These stem cells appear to derive from three distinct tissue lineages within the blastocyst: the epiblast, the trophectoderm, and the extraembryonic endoderm. Understanding how these lineages arise during development will illuminate efforts to understand the establishment and maintenance of the stem cell state and the mechanisms that restrict stem cell potency. Genetic analysis has enabled the identification of several genes important for lineage decisions in the mouse blastocyst. Among these, Oct4, Nanog, Cdx2, and Gata6 encode transcription factors required for the three lineages of the blastocyst and for the maintenance their respective stem cell types. Interestingly, genetic manipulation of several of these factors can cause lineage switching among these stem cells, suggesting that knowledge of key lineage-determining genes could help control differentiation of stem cells more generally. Pluripotent stem cells have also been isolated from the human blastocyst, but the relationship between these cells and stem cells of the mouse blastocyst remains to be explored. This review describes the genetic regulation of lineage allocation during blastocyst formation and discusses similarities and differences between mouse and human ES cells.


Assuntos
Linhagem da Célula/genética , Regulação da Expressão Gênica no Desenvolvimento , Camundongos/embriologia , Células-Tronco/fisiologia , Animais , Blastocisto/citologia , Humanos
5.
Placenta ; 25(7): 595-607, 2004 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-15193866

RESUMO

Little is known about the role of gap junctional intercellular communication (GJIC) in human trophoblast differentiation, particularly during the formation of extravillous trophoblast (EVT) cell columns and their subsequent differentiation into invasive cells. We have identified transcripts for five connexin gap junction proteins in the early human placenta (Cx32, Cx37, Cx40, Cx43 and Cx45). Of these, Cx40 and Cx45 proteins immunolocalize to EVT in anchoring cell columns. Cx40 expression is prominent in the anchoring column throughout the first trimester of pregnancy (6-14 weeks gestation). We used first trimester placental villous explant cultures to determine the functional significance of the inhibition of GJIC in EVT cell proliferation and differentiation using two known GJIC inhibitors, carbenoxolone (CBX) and heptanol. The morphology of EVT outgrowths changed dramatically upon GJIC-blockade, from compact and organized outgrowths into a scattered group of rounded individual trophoblast cells, reminiscent of an early invasive phenotype. Furthermore, the inhibition of GJIC in placental explants by CBX or heptanol induced a switch away from the proliferative and towards an invasive EVT phenotype, as evident from (a) the loss of the proliferation marker Ki67 and (b) an increase in the invasive marker alpha1 integrin. We also utilized antisense oligonucleotides to inhibit Cx40 protein expression in placental explants. Cx40 antisense treatment also resulted in the abolishment of outgrowth EVT cell proliferation (as determined by Ki67 immunostaining). Together, these results suggest that gap junctions composed particularly of Cx40 channels are required for the proliferation of EVT cells in anchoring cell columns, and that a loss of GJIC contributes to differentiation to the invasive EVT phenotype.


Assuntos
Divisão Celular , Junções Comunicantes/fisiologia , Placentação , Trofoblastos/ultraestrutura , Carbenoxolona/farmacologia , Comunicação Celular/efeitos dos fármacos , Diferenciação Celular , Células Cultivadas , Conexina 43/análise , Conexina 43/genética , Conexinas/análise , Conexinas/genética , Feminino , Imunofluorescência , Junções Comunicantes/efeitos dos fármacos , Humanos , Imuno-Histoquímica , Hibridização In Situ , Gravidez , Primeiro Trimestre da Gravidez , RNA Mensageiro/análise , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Técnicas de Cultura de Tecidos , Proteína beta-1 de Junções Comunicantes , Proteína alfa-5 de Junções Comunicantes , Proteína alfa-4 de Junções Comunicantes
6.
Placenta ; 25 Suppl A: S32-8, 2004 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-15033304

RESUMO

The first priority of a mammalian embryo is to establish an intimate relationship with its mother. This is accomplished by precocious differentiation of the trophoblast lineage, which mediates uterine implantation and initiates the process of placentation. Surprisingly little is known about the molecular mechanisms that drive trophectoderm differentiation from the equipotent blastomeres of the morula. Somewhat more is known about the maintenance of trophoblast stem cells, once this lineage has been established. The first half of this review will focus on determination of the mouse trophoblast lineage and the second half will discuss the maintenance of trophoblast stem cells.


Assuntos
Camundongos/embriologia , Células-Tronco Pluripotentes/fisiologia , Prenhez/fisiologia , Trofoblastos/citologia , Trofoblastos/fisiologia , Animais , Feminino , Gravidez
7.
Dev Cell ; 1(1): 37-49, 2001 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-11703922

RESUMO

Although FGF signaling plays an integral role in the migration and patterning of mesoderm at gastrulation, the mechanism and downstream targets of FGF activity have remained elusive. Here, we demonstrate that FGFR1 orchestrates the epithelial to mesenchymal transition and morphogenesis of mesoderm at the primitive streak by controlling Snail and E-cadherin expression. Furthermore, we show that FGFR1 functions in mesoderm cell fate specification by positively regulating Brachyury and Tbx6 expression. Finally, we provide evidence that the attenuation of Wnt3a signaling observed in Fgfr1 -/- embryos can be rescued by lowering E-cadherin levels. We propose that modulation of cytoplasmic beta-catenin levels, associated with FGF-induced downregulation of E-cadherin, provides a molecular link between FGF and Wnt signaling pathways at the streak.


Assuntos
Movimento Celular/fisiologia , Proteínas Fetais , Gástrula/fisiologia , Receptores Proteína Tirosina Quinases/genética , Receptores Proteína Tirosina Quinases/metabolismo , Receptores de Fatores de Crescimento de Fibroblastos/genética , Receptores de Fatores de Crescimento de Fibroblastos/metabolismo , Transativadores , Proteínas de Peixe-Zebra , Animais , Caderinas/genética , Células Cultivadas , Proteínas do Citoesqueleto/metabolismo , Proteínas de Ligação a DNA/genética , Feminino , Gástrula/citologia , Regulação da Expressão Gênica no Desenvolvimento , Masculino , Mesoderma/citologia , Mesoderma/fisiologia , Camundongos , Camundongos Endogâmicos ICR , Camundongos Mutantes , Mutação/fisiologia , Proteínas Proto-Oncogênicas/fisiologia , Receptor Tipo 1 de Fator de Crescimento de Fibroblastos , Transdução de Sinais/fisiologia , Fatores de Transcrição da Família Snail , Proteínas com Domínio T/genética , Fatores de Transcrição/genética , Proteínas Wnt , beta Catenina
8.
Trends Mol Med ; 7(11): 502-7, 2001 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-11689335

RESUMO

The powerful and wide-ranging genetic tools available in the laboratory mouse make it the major experimental model for studying mammalian gene function in vivo and modelling human disease traits. Large-scale random mutagenesis approaches, either gene-driven or phenotype-driven, promise to identify new clinically relevant phenotypes and their associated genes. Development of appropriate tools for assessing clinical phenotypes in mice is a crucial component of these endeavours, as is the establishment of the infrastructure for archiving and distribution of the growing mutant resource to the community. Integrated, multidisciplinary programs will be needed to fully exploit the power of the mouse in molecular medicine.


Assuntos
Genômica , Laboratórios , Camundongos/genética , Academias e Institutos , Animais , Modelos Animais de Doenças , Genômica/métodos , Genômica/organização & administração , Humanos , Mutagênese
9.
Stem Cells ; 19(6): 477-82, 2001.
Artigo em Inglês | MEDLINE | ID: mdl-11713338

RESUMO

Early differentiation of the mammalian embryo leads to the development of two distinct lineages-the inner cell mass (ICM) and the trophectoderm. Cells of the ICM are pluripotent and give rise to all tissues of the fetus, while trophectoderm cells are restricted in their potential to the trophoblast cell layers of the placenta. In the mouse, apparently immortal stem cell lines can be obtained from both cell types. These cell lines, embryonic stem (ES) cells and trophoblast stem (TS) cells, are morphologically and molecularly distinct and depend on different signaling pathways for their maintenance. They also show different cell fates when introduced into early embryos to generate chimeras. However, a change in the levels of expression of a key regulator of pluripotency, Oct4, can push ES cells towards the TS phenotype, when grown in TS cell conditions. Stem cell potential in the early embryo thus appears to depend on a combination of the levels of expression of key intrinsic regulators and the appropriate extrinsic environmental factors. Manipulation of both intrinsic and extrinsic regulators may be needed to reveal the full potential of stem cells from other stages of development and the adult.


Assuntos
Blastocisto/citologia , Células-Tronco/citologia , Animais , Humanos
10.
Science ; 294(5542): 559-63, 2001 Oct 19.
Artigo em Inglês | MEDLINE | ID: mdl-11577199

RESUMO

The embryonic role of endothelial cells and nascent vessels in promoting organogenesis, prior to vascular function, is unclear. We find that early endothelial cells in mouse embryos surround newly specified hepatic endoderm and delimit the mesenchymal domain into which the liver bud grows. In flk-1 mutant embryos, which lack endothelial cells, hepatic specification occurs, but liver morphogenesis fails prior to mesenchyme invasion. We developed an embryo tissue explant system that permits liver bud vasculogenesis and show that in the absence of endothelial cells, or when the latter are inhibited, there is a selective defect in hepatic outgrowth. We conclude that vasculogenic endothelial cells and nascent vessels are critical for the earliest stages of organogenesis, prior to blood vessel function.


Assuntos
Indução Embrionária , Endoderma/fisiologia , Endotélio Vascular/fisiologia , Fígado/embriologia , Mitógenos , Animais , Vasos Sanguíneos/citologia , Vasos Sanguíneos/embriologia , Vasos Sanguíneos/fisiologia , Técnicas de Cultura , Endotélio Vascular/citologia , Endotélio Vascular/embriologia , Feminino , Fator de Crescimento de Hepatócito/antagonistas & inibidores , Fator de Crescimento de Hepatócito/metabolismo , Fator de Crescimento de Hepatócito/farmacologia , Hepatócitos/fisiologia , Fígado/irrigação sanguínea , Fígado/citologia , Fígado/efeitos dos fármacos , Masculino , Mesoderma/fisiologia , Camundongos , Camundongos Endogâmicos C3H , Morfogênese , Mutação , Neovascularização Fisiológica , Receptores Proteína Tirosina Quinases/genética , Receptores Proteína Tirosina Quinases/fisiologia , Receptores de Fatores de Crescimento/genética , Receptores de Fatores de Crescimento/fisiologia , Receptores de Fatores de Crescimento do Endotélio Vascular , Transdução de Sinais/efeitos dos fármacos
11.
Genome Biol ; 2(7): REVIEWS1019, 2001.
Artigo em Inglês | MEDLINE | ID: mdl-11516341

RESUMO

Microarrays of mouse genes are now available from several sources, and they have so far given new insights into gene expression in embryonic development, regions of the brain and during apoptosis. Microarray data posted on the internet can be reanalyzed to study a range of questions.


Assuntos
Bases de Dados de Ácidos Nucleicos , Perfilação da Expressão Gênica/métodos , Análise de Sequência com Séries de Oligonucleotídeos , Animais , Encéfalo/metabolismo , Etiquetas de Sequências Expressas , Internet , Pulmão/metabolismo , Camundongos
12.
Nat Rev Genet ; 2(7): 538-48, 2001 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-11433360

RESUMO

The placenta is the first organ to form during mammalian embryogenesis. Problems in its formation and function underlie many aspects of early pregnancy loss and pregnancy complications in humans. Because the placenta is critical for survival, it is very sensitive to genetic disruption, as reflected by the ever-increasing list of targeted mouse mutations that cause placental defects. Recent studies of mouse mutants with disrupted placental development indicate that signalling interactions between the placental trophoblast and embryonic cells have a key role in placental morphogenesis. Furthering our understanding of mouse trophoblast development should provide novel insights into human placental function.


Assuntos
Camundongos/embriologia , Placentação , Trofoblastos/fisiologia , Animais , Diferenciação Celular , Feminino , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Camundongos Knockout , Gravidez , Especificidade da Espécie
13.
Int J Dev Biol ; 45(3): 577-82, 2001.
Artigo em Inglês | MEDLINE | ID: mdl-11417901

RESUMO

Back at the first half of the 1980s, there was no mammalian experimental embryology in Hungary. One of us, AN, took up the challenge of establishing a small group in the field. In the absence of local information, AN and his former colleague, Andras Paldi (AP), used their tourist passport to visit several laboratories in Western Europe and collect information and advice. This is how AN and AP ended up one day sitting in Anne McLaren's office in the MRC Mammalian Development Unit at University College, London. They never forgot her endless enthusiasm and the way she clearly explained the important points of preimplantation embryo manipulation, chimaera making and embryo transfer. As well as the extremely useful suggestions, which were crucial to starting the lab in Hungary, they also took back her deep love for embryo development. They remember her telling them, 'never waste an embryo--there is always another unanswered question it can solve'. Many who have been lucky and experienced Anne's spirit and advice later realized how useful it was to generate 'new' ideas by following the 'not wasting' principle. Our views on chimaeras presented below definitely contain elements which grew out from this principle.


Assuntos
Quimera , Mosaicismo , Animais , Diploide , Embriologia/história , Feminino , História do Século XX , Masculino , Modelos Genéticos , Mutação , Fenótipo , Poliploidia , Células-Tronco/citologia
14.
Proc Natl Acad Sci U S A ; 98(10): 5643-8, 2001 May 08.
Artigo em Inglês | MEDLINE | ID: mdl-11344305

RESUMO

Notch proteins function as receptors for membrane-bound ligands (Jagged and Delta-like) to regulate cell-fate determination. We have investigated the role of Notch signaling in embryonic endothelium of the mouse by expressing an activated form of the Notch4 protein in vasculature under the regulation of the Flk1 (VEGFR) locus. Expression of activated Notch4 results in a growth and developmental delay and embryonic lethality at about 10 days postcoitum. The extent of the developing vasculature in mutant embryos was restricted, fewer small vessels were seen, and vascular networks were disorganized. The brain periphery of mutant embryos contained large dilated vessels with evidence of compromised vessel-wall integrity and large areas of necrosis; yolk-sac vasculature was abnormal. Expression of an activated form of Notch4 in embryonic vasculature leads to abnormal vessel structure and patterning, implicating the Notch pathway in phases of vascular development associated with vessel patterning and remodeling.


Assuntos
Vasos Sanguíneos/embriologia , Padronização Corporal/genética , Embrião de Mamíferos/metabolismo , Endotélio Vascular/metabolismo , Expressão Gênica , Proteínas Proto-Oncogênicas/genética , Receptores de Superfície Celular , Animais , Camundongos , Receptor Notch4 , Receptores Notch
15.
Neuron ; 30(1): 65-78, 2001 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-11343645

RESUMO

Little is known about how neural stem cells are formed initially during development. We investigated whether a default mechanism of neural specification could regulate acquisition of neural stem cell identity directly from embryonic stem (ES) cells. ES cells cultured in defined, low-density conditions readily acquire a neural identity. We characterize a novel primitive neural stem cell as a component of neural lineage specification that is negatively regulated by TGFbeta-related signaling. Primitive neural stem cells have distinct growth factor requirements, express neural precursor markers, generate neurons and glia in vitro, and have neural and non-neural lineage potential in vivo. These results are consistent with a default mechanism for neural fate specification and support a model whereby definitive neural stem cell formation is preceded by a primitive neural stem cell stage during neural lineage commitment.


Assuntos
Padronização Corporal/fisiologia , Diferenciação Celular/fisiologia , Linhagem da Célula/fisiologia , Interleucina-6 , Mamíferos/embriologia , Proteínas do Tecido Nervoso , Sistema Nervoso/embriologia , Sistema Nervoso/crescimento & desenvolvimento , Neurônios/citologia , Células-Tronco/citologia , Animais , Padronização Corporal/efeitos dos fármacos , Diferenciação Celular/efeitos dos fármacos , Linhagem da Célula/efeitos dos fármacos , Tamanho Celular/genética , Células Cultivadas/citologia , Células Cultivadas/efeitos dos fármacos , Células Cultivadas/metabolismo , Quimera/embriologia , Quimera/genética , Quimera/metabolismo , Meios de Cultura Livres de Soro/farmacologia , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Inibidores do Crescimento/farmacologia , Substâncias de Crescimento/deficiência , Proteínas de Filamentos Intermediários/efeitos dos fármacos , Proteínas de Filamentos Intermediários/metabolismo , Fator Inibidor de Leucemia , Linfocinas/farmacologia , Mamíferos/metabolismo , Camundongos , Sistema Nervoso/citologia , Nestina , Neurônios/metabolismo , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia , Proteína Smad4 , Células-Tronco/efeitos dos fármacos , Células-Tronco/metabolismo , Transativadores/genética , Transativadores/metabolismo , Fator de Crescimento Transformador beta/efeitos dos fármacos , Fator de Crescimento Transformador beta/metabolismo
16.
Genes Dev ; 15(10): 1257-71, 2001 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-11358869

RESUMO

The node and the anterior visceral endoderm (AVE) are important organizing centers that pattern the mouse embryo by establishing the anterior-posterior (A-P), dorsal-ventral (D-V), and left-right (L-R) axes. Activin/nodal signaling through the Smad2 pathway has been implicated in AVE formation and in morphogenesis of the primitive streak, the anterior end of which gives rise to the node. The forkhead DNA-binding protein, FoxH1 (or Fast), functions as a Smad DNA-binding partner to regulate transcription in response to activin signaling. Here, we show that deletion of FoxH1 in mice results in failure to pattern the anterior primitive streak (APS) and form node, prechordal mesoderm, notochord, and definitive endoderm. In contrast, formation of the AVE can occur in the absence of FoxH1. The FoxH1 mutant phenotype is remarkably similar to that of mice deficient in the forkhead protein Foxa2 (HNF3beta), and we show that Foxa2 expression is dependent on FoxH1 function. These results show that FoxH1 functions in an activin/nodal-Smad signaling pathway that acts upstream of Foxa2 and is required specifically for patterning the APS and node in the mouse.


Assuntos
Padronização Corporal , Proteínas de Ligação a DNA/genética , Gástrula/metabolismo , Transdução de Sinais , Fatores de Transcrição/genética , Ativinas , Animais , Proteínas de Ligação a DNA/metabolismo , Fatores de Transcrição Forkhead , Regulação da Expressão Gênica no Desenvolvimento , Hibridização In Situ , Inibinas/genética , Inibinas/metabolismo , Camundongos , Camundongos Endogâmicos ICR , Camundongos Knockout , Mutagênese , Proteína Nodal , Reação em Cadeia da Polimerase , Deleção de Sequência , Proteína Smad2 , Transativadores/genética , Transativadores/metabolismo , Fatores de Transcrição/metabolismo , Fator de Crescimento Transformador beta/genética , Fator de Crescimento Transformador beta/metabolismo
17.
Int J Dev Biol ; 45(1): 357-65, 2001.
Artigo em Inglês | MEDLINE | ID: mdl-11291866

RESUMO

Patterning the developing nervous system in the mouse has been proposed to depend on two separate sources of signals, the anterior visceral endoderm (AVE) and the node or organizer. Mutation of the winged-helix gene HNF3beta leads to loss of the node and its derivatives, while mutation of the homeobox gene Otx2 results in loss of head structures, apparently at least partially because of defects in the AVE. To investigate the potential genetic interactions between the two signaling centers, we crossed Otx2+/- and HNF3beta+/- mice and found that very few Otx2+/-;HNF3beta+/- double heterozygous mutants survived to weaning. Normal Mendelian ratios of genotypes were observed during gestation, but more than half the double heterozygotes displayed a severe anterior patterning phenotype that would be incompatible with postnatal survival. The phenotype was characterized by varying degrees of holoprosencephaly, cyclopia with proboscis-like structures, and anterior forebrain truncations. Regional marker analysis revealed that ventral forebrain structures of Otx2+/-;HNF3beta+/- mutant embryos were most severely affected. Shh expression was completely absent in the anterior region of Otx2+/-;HNF3beta+/- embryos, suggesting that Otx2 and HNF3beta genetically interact, directly or indirectly, to regulate Shh expression in the anterior midline. In addition, the forebrain truncations suggest an involvement of both genes in anterior patterning, through their overlapping expression domains in either the AVE and/or the prechordal mesoderm.


Assuntos
Padronização Corporal/genética , Proteínas de Ligação a DNA/genética , Proteínas de Homeodomínio , Proteínas do Tecido Nervoso/genética , Proteínas Nucleares/genética , Transativadores/genética , Fatores de Transcrição , Animais , Padronização Corporal/fisiologia , Proteínas de Ligação a DNA/fisiologia , Endoderma/citologia , Regulação da Expressão Gênica no Desenvolvimento , Fator 3-beta Nuclear de Hepatócito , Heterozigoto , Hibridização In Situ , Mesoderma/citologia , Camundongos , Camundongos Mutantes , Proteínas do Tecido Nervoso/fisiologia , Sistema Nervoso/embriologia , Proteínas Nucleares/fisiologia , Organizadores Embrionários , Fatores de Transcrição Otx , Fenótipo , Transdução de Sinais , Transativadores/fisiologia
18.
Genes Dev ; 15(7): 833-8, 2001 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-11297507

RESUMO

The orphan nuclear receptor ERR beta is expressed in undifferentiated trophoblast stem cell lines and extraembryonic ectoderm, and genetic ablation of ERR beta results in abnormal trophoblast proliferation and precocious differentiation toward the giant cell lineage. Here, we show that the synthetic estrogen diethylstilbestrol (DES) promotes coactivator release from ERR beta and inhibits its transcriptional activity. Strikingly, treatment of trophoblast stem cells with DES led to their differentiation toward the polyploid giant cell lineage. In addition, DES-treated pregnant mice exhibited abnormal early placenta development associated with an overabundance of trophoblast giant cells and an absence of diploid trophoblast. These results define a novel pathway for DES action and provide evidence for steroidlike control of trophoblast development.


Assuntos
Dietilestilbestrol/farmacologia , Estrogênios não Esteroides/farmacologia , Etiocolanolona/análogos & derivados , Receptores de Estrogênio/antagonistas & inibidores , Células-Tronco/efeitos dos fármacos , Trofoblastos/efeitos dos fármacos , Animais , Células COS , Diferenciação Celular/efeitos dos fármacos , Chlorocebus aethiops , Estradiol/farmacologia , Etiocolanolona/farmacologia , Feminino , Fatores de Crescimento de Fibroblastos/farmacologia , Fluorometria , Genes Reporter , Idade Gestacional , Células Gigantes/patologia , Ligantes , Luciferases/biossíntese , Luciferases/genética , Camundongos , Estrutura Molecular , Coativador 2 de Receptor Nuclear , Placenta/efeitos dos fármacos , Placenta/patologia , Gravidez , Pregnanolona/farmacologia , Receptores Citoplasmáticos e Nucleares/efeitos dos fármacos , Receptores Citoplasmáticos e Nucleares/genética , Receptores de Estrogênio/efeitos dos fármacos , Receptores de Estrogênio/genética , Receptores de Estrogênio/fisiologia , Proteínas Recombinantes de Fusão/biossíntese , Células-Tronco/patologia , Fatores de Transcrição/metabolismo , Ativação Transcricional/efeitos dos fármacos , Transfecção , Trofoblastos/patologia , Receptor ERRalfa Relacionado ao Estrogênio
20.
Genes Dev ; 15(2): 213-25, 2001 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-11157777

RESUMO

Retinoic acid (RA), a derivative of vitamin A, plays a pivotal role in vertebrate development. The level of RA may be determined by the balance between its synthesis and degradation. We have examined the role of CYP26, a P450 enzyme that may degrade RA, by generating mutant mice that lack CYP26. CYP26(-/-) mice exhibited anomalies, including caudal agenesis, similar to those induced by administration of excess RA. The concentration of endogenous RA, as revealed by marker gene activity, was markedly increased in the tailbud of the mutant animals, in which CYP26 is normally expressed. Expression of T (Brachyury) and Wnt3a in the tailbud was down-regulated in CYP26(-/-) mice, which may underlie the caudal truncation. The lack of CYP26 also resulted in homeotic transformation of vertebrae as well as in misspecification of the rostral hindbrain associated with anterior expansion of RA-positive domains. These results suggest that local degradation of RA by CYP26 is required for establishing an uneven distribution of RA along the anterio-posterior axis, which is essential for patterning the hindbrain, vertebrae, and tailbud.


Assuntos
Sistema Enzimático do Citocromo P-450/metabolismo , Embrião de Mamíferos/metabolismo , Proteínas Fetais , Tretinoína/metabolismo , Animais , Sequência de Bases , Padronização Corporal , Sistema Enzimático do Citocromo P-450/deficiência , Sistema Enzimático do Citocromo P-450/genética , Primers do DNA/genética , Regulação para Baixo , Membro Anterior/embriologia , Regulação da Expressão Gênica no Desenvolvimento , Camundongos , Camundongos Knockout , Camundongos Mutantes , Camundongos Transgênicos , Proteínas/genética , Proteínas/metabolismo , Ácido Retinoico 4 Hidroxilase , Rombencéfalo/embriologia , Somitos/metabolismo , Proteínas com Domínio T/genética , Proteínas com Domínio T/metabolismo , Distribuição Tecidual , Tretinoína/antagonistas & inibidores , Proteínas Wnt , Proteína Wnt3 , Proteína Wnt3A
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA