Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 78
Filtrar
1.
Proc Natl Acad Sci U S A ; 112(40): 12426-31, 2015 Oct 06.
Artigo em Inglês | MEDLINE | ID: mdl-26392552

RESUMO

The gene encoding the E3 ubiquitin ligase Ligand of Numb protein-X (Lnx)2a is expressed in the ventral-anterior pancreatic bud of zebrafish embryos in addition to its expression in the brain. Knockdown of Lnx2a by using an exon 2/intron 2 splice morpholino resulted in specific inhibition of the differentiation of ventral bud derived exocrine cell types, with little effect on endocrine cell types. A frame shifting null mutation in lnx2a did not mimic this phenotype, but a mutation that removed the exon 2 splice donor site did. We found that Lnx2b functions in a redundant manner with its paralog Lnx2a. Inhibition of lnx2a exon 2/3 splicing causes exon 2 skipping and leads to the production of an N-truncated protein that acts as an interfering molecule. Thus, the phenotype characterized by inhibition of exocrine cell differentiation requires inactivation of both Lnx2a and Lnx2b. Human LNX1 is known to destabilize Numb, and we show that inhibition of Numb expression rescues the Lnx2a/b-deficient phenotype. Further, Lnx2a/b inhibition leads to a reduction in the number of Notch active cells in the pancreas. We suggest that Lnx2a/b function to fine tune the regulation of Notch through Numb in the differentiation of cell types in the early zebrafish pancreas. Further, the complex relationships among genotype, phenotype, and morpholino effect in this case may be instructive in the ongoing consideration of morpholino use.


Assuntos
Proteínas de Transporte/genética , Diferenciação Celular/genética , Pâncreas Exócrino/metabolismo , Pâncreas/metabolismo , Ubiquitina-Proteína Ligases/genética , Proteínas de Peixe-Zebra/genética , Processamento Alternativo , Sequência de Aminoácidos , Animais , Sequência de Bases , Proteínas de Transporte/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Técnicas de Silenciamento de Genes , Células HEK293 , Humanos , Immunoblotting , Hibridização In Situ , Microscopia Confocal , Morfolinos/genética , Mutação , Pâncreas/citologia , Pâncreas/embriologia , Pâncreas Exócrino/citologia , Pâncreas Exócrino/embriologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Ubiquitina-Proteína Ligases/metabolismo , Peixe-Zebra/embriologia , Peixe-Zebra/genética , Peixe-Zebra/metabolismo , Proteínas de Peixe-Zebra/metabolismo
2.
Proc Natl Acad Sci U S A ; 111(36): 13099-104, 2014 Sep 09.
Artigo em Inglês | MEDLINE | ID: mdl-25157132

RESUMO

Precise control of the canonical Wnt pathway is crucial in embryogenesis and all stages of life, and dysregulation of this pathway is implicated in many human diseases including cancers and birth defect disorders. A key aspect of canonical Wnt signaling is the cytoplasmic to nuclear translocation of ß-catenin, a process that remains incompletely understood. Here we report the identification of a previously undescribed component of the canonical Wnt signaling pathway termed Custos, originally isolated as a Dishevelled-interacting protein. Custos contains casein kinase phosphorylation sites and nuclear localization sequences. In Xenopus, custos mRNA is expressed maternally and then widely throughout embryogenesis. Depletion or overexpression of Custos produced defective anterior head structures by inhibiting the formation of the Spemann-Mangold organizer. In addition, Custos expression blocked secondary axis induction by positive signaling components of the canonical Wnt pathway and inhibited ß-catenin/TCF-dependent transcription. Custos binds to ß-catenin in a Wnt responsive manner without affecting its stability, but rather modulates the cytoplasmic to nuclear translocation of ß-catenin. This effect on nuclear import appears to be the mechanism by which Custos inhibits canonical Wnt signaling. The function of Custos is conserved as loss-of-function and gain-of-function studies in zebrafish also demonstrate a role for Custos in anterior head development. Our studies suggest a role for Custos in fine-tuning canonical Wnt signal transduction during embryogenesis, adding an additional layer of regulatory control in the Wnt-ß-catenin signal transduction cascade.


Assuntos
Desenvolvimento Embrionário , Cabeça/embriologia , Proteínas de Homeodomínio/metabolismo , Vertebrados/embriologia , Vertebrados/metabolismo , Proteínas de Xenopus/metabolismo , Proteínas de Peixe-Zebra/metabolismo , beta Catenina/metabolismo , Animais , Padronização Corporal , Núcleo Celular/metabolismo , Células HEK293 , Humanos , Dados de Sequência Molecular , Ligação Proteica , Transporte Proteico , Via de Sinalização Wnt , Xenopus laevis/embriologia , Peixe-Zebra/embriologia
3.
Proc Natl Acad Sci U S A ; 110(8): 2870-5, 2013 Feb 19.
Artigo em Inglês | MEDLINE | ID: mdl-23382213

RESUMO

The neural crest develops in vertebrate embryos within a discrete domain at the neural plate boundary and eventually gives rise to a migrating population of cells that differentiate into a multitude of derivatives. We have shown that the broad-complex, tramtrack and bric a brac (BTB) domain-containing factor potassium channel tetramerization domain containing 15 (Kctd15) inhibits neural crest formation, and we proposed that its function is to delimit the neural crest domain. Here we report that Kctd15 is a highly effective inhibitor of transcription factor activating enhancer binding protein 2 (AP-2) in zebrafish embryos and in human cells; AP-2 is known to be critical for several steps of neural crest development. Kctd15 interacts with AP-2α but does not interfere with its nuclear localization or binding to cognate sites in the genome. Kctd15 binds specifically to the activation domain of AP-2α and efficiently inhibits transcriptional activation by a hybrid protein composed of the regulatory protein Gal4 DNA binding and AP-2α activation domains. Mutation of one proline residue in the activation domain to an alanine (P59A) yields a protein that is highly active but largely insensitive to Kctd15. These results indicate that Kctd15 acts in the embryo at least in part by specifically binding to the activation domain of AP-2α, thereby blocking the function of this critical factor in the neural crest induction hierarchy.


Assuntos
Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Crista Neural/crescimento & desenvolvimento , Canais de Potássio/fisiologia , Fator de Transcrição AP-2/fisiologia , Imunoprecipitação da Cromatina , DNA/metabolismo , Células HEK293 , Humanos , Hibridização In Situ , Ligação Proteica , Fator de Transcrição AP-2/metabolismo
4.
EMBO J ; 30(14): 2894-907, 2011 Jun 10.
Artigo em Inglês | MEDLINE | ID: mdl-21666599

RESUMO

The caudal homeobox (cdx) gene family is critical for specification of caudal body formation and erythropoiesis. In zebrafish, cdx4 expression is controlled by the Wnt pathway, but the molecular mechanism of this regulation is not fully understood. Here, we provide evidence that Tcf3 suppresses cdx4 expression through direct binding to multiple sites in the cdx4 gene regulatory region. Tcf3 requires corepressor molecules such as Groucho (Gro)/TLE and HDAC1 for activity. Using zebrafish embryos and cultured mammalian cells, we show that the transcription factor E4f1 derepresses cdx4 by dissociating corepressor proteins from Tcf3 without inhibiting its binding to cis-regulatory sites in the DNA. Further, the E3 ubiquitin ligase Lnx2b, acting as a scaffold protein irrespective of its enzymatic activity, counteracts the effects of E4f1. We propose that the modulation of Tcf3 repressor function by E4f1 assures precise and robust regulation of cdx4 expression in the caudal domain of the embryo.


Assuntos
Regulação da Expressão Gênica no Desenvolvimento , Proteínas de Homeodomínio/genética , Proteínas Repressoras/metabolismo , Proteínas de Peixe-Zebra/metabolismo , Peixe-Zebra/embriologia , Animais , Western Blotting , Padronização Corporal , Células Cultivadas , Imunoprecipitação da Cromatina , Embrião não Mamífero/citologia , Embrião não Mamífero/metabolismo , Histona Desacetilase 1/genética , Histona Desacetilase 1/metabolismo , Proteínas de Homeodomínio/metabolismo , Imunoprecipitação , Luciferases/metabolismo , Regiões Promotoras Genéticas , Ligação Proteica , RNA Mensageiro/genética , Sequências Reguladoras de Ácido Nucleico , Proteínas Repressoras/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fatores de Transcrição , Ubiquitina-Proteína Ligases/genética , Ubiquitina-Proteína Ligases/metabolismo , Proteínas Wnt/genética , Proteínas Wnt/metabolismo , Peixe-Zebra/genética , Peixe-Zebra/metabolismo , Proteínas de Peixe-Zebra/genética
5.
Proc Natl Acad Sci U S A ; 109(22): 8594-9, 2012 May 29.
Artigo em Inglês | MEDLINE | ID: mdl-22592794

RESUMO

Liver and ventral pancreas develop from neighboring territories within the endoderm of gastrulae. ventral pancreatic precursor 1 (vpp1) is a marker gene that is differentially expressed in a cell population within the dorsal endoderm in a pattern partially overlapping with that of hematopoietically expressed homeobox (hhex) during gastrulation. In tail bud embryos, vpp1 expression specifically demarcates two ventral pancreatic buds, whereas hhex expression is mainly restricted to the liver diverticulum. Ectopic expression of a critical dose of hhex led to a greatly enlarged vpp1-positive domain and, subsequently, to the formation of giant ventral pancreata, putatively by conversion of intestinal to ventral pancreatic precursor cells. Conversely, antisense morpholino oligonucleotide-mediated knockdown of hhex resulted in a down-regulation of vpp1 expression and a specific loss of the ventral pancreas. Furthermore, titration of hhex with a dexamethasone-inducible hhex-VP16GR fusion construct suggested that endogenous hhex activity during gastrulation is essential for the formation of ventral pancreatic progenitor cells. These observations suggest that, beyond its role in liver development, hhex controls specification of a vpp1-positive endodermal cell population during gastrulation that is required for the formation of the ventral pancreas.


Assuntos
Embrião não Mamífero/metabolismo , Proteínas de Homeodomínio/genética , Mucosa Intestinal/metabolismo , Pâncreas/metabolismo , Proteínas de Xenopus/genética , Xenopus laevis/genética , Processamento Alternativo , Sequência de Aminoácidos , Animais , Embrião não Mamífero/embriologia , Endoderma/citologia , Endoderma/embriologia , Endoderma/metabolismo , Gástrula/embriologia , Gástrula/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Técnicas de Inativação de Genes , Hibridização In Situ , Intestinos/embriologia , Dados de Sequência Molecular , Pâncreas/embriologia , Isoformas de Proteínas/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Homologia de Sequência de Aminoácidos , Fatores de Tempo , Transfecção , Xenopus laevis/embriologia
6.
Proc Natl Acad Sci U S A ; 109(9): 3371-6, 2012 Feb 28.
Artigo em Inglês | MEDLINE | ID: mdl-22331904

RESUMO

The yolk syncytial layer (YSL) in the zebrafish embryo is a multinucleated syncytium essential for embryo development, but the molecular mechanisms underlying YSL formation remain largely unknown. Here we show that zebrafish solute carrier family 3 member 2 (Slc3a2) is expressed specifically in the YSL and that slc3a2 knockdown causes severe YSL defects including clustering of the yolk syncytial nuclei and enhanced cell fusion, accompanied by disruption of microtubule networks. Expression of a constitutively active RhoA mimics the YSL phenotypes caused by slc3a2 knockdown, whereas attenuation of RhoA or ROCK activity rescues the slc3a2-knockdown phenotypes. Furthermore, slc3a2 knockdown significantly reduces tyrosine phosphorylation of c-Src, and overexpression of a constitutively active Src restores the slc3a2-knockdown phenotypes. Our data demonstrate a signaling pathway regulating YSL formation in which Slc3a2 inhibits the RhoA/ROCK pathway via phosphorylation of c-Src to modulate YSL microtubule dynamics. This work illuminates processes at a very early stage of zebrafish embryogenesis and more generally informs the mechanism of cell dynamics during syncytium formation.


Assuntos
Proteínas do Ovo/fisiologia , Gema de Ovo/citologia , Células Gigantes/citologia , Microtúbulos/ultraestrutura , Proteínas de Peixe-Zebra/fisiologia , Peixe-Zebra/embriologia , Animais , Blastoderma/metabolismo , Proteína Tirosina Quinase CSK , Gema de Ovo/enzimologia , Embrião não Mamífero/citologia , Gástrula/metabolismo , Técnicas de Silenciamento de Genes , Genes src , Proteínas Monoméricas de Ligação ao GTP/fisiologia , Fosforilação , Processamento de Proteína Pós-Traducional , Proteínas Tirosina Quinases/fisiologia , Quinases Associadas a rho/fisiologia , Quinases da Família src
7.
Proc Natl Acad Sci U S A ; 109(43): 17484-9, 2012 Oct 23.
Artigo em Inglês | MEDLINE | ID: mdl-23045671

RESUMO

Transcription activator-like effector nucleases (TALENs) are an approach for directed gene disruption and have been proved to be effective in various animal models. Here, we report that TALENs can induce somatic mutations in Xenopus embryos with reliably high efficiency and that such mutations are heritable through germ-line transmission. We modified the Golden Gate method for TALEN assembly to make the product suitable for RNA transcription and microinjection into Xenopus embryos. Eight pairs of TALENs were constructed to target eight Xenopus genes, and all resulted in indel mutations with high efficiencies of up to 95.7% at the targeted loci. Furthermore, mutations induced by TALENs were highly efficiently passed through the germ line to F(1) frogs. Together with simple and reliable PCR-based approaches for detecting TALEN-induced mutations, our results indicate that TALENs are an effective tool for targeted gene editing/knockout in Xenopus.


Assuntos
Endonucleases/metabolismo , Marcação de Genes , Transativadores/metabolismo , Xenopus/embriologia , Animais , Sequência de Bases , DNA , Embrião não Mamífero , Dados de Sequência Molecular , Xenopus/genética
8.
Dev Dyn ; 242(9): 1033-42, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-23749482

RESUMO

BACKGROUND: The zebrafish pineal gland (epiphysis) is a site of melatonin production, contains photoreceptor cells, and functions as a circadian clock pacemaker. Since it is located on the surface of the forebrain, it is accessible for manipulation and, therefore, is a useful model system to analyze pineal gland function and development. We previously analyzed the pineal transcriptome during development and showed that many genes exhibit a highly dynamic expression pattern in the pineal gland. RESULTS: Among genes preferentially expressed in the zebrafish pineal gland, we identified a tissue-specific form of the unc119 gene family, unc119c, which is highly preferentially expressed in the pineal gland during day and night at all stages examined from embryo to adult. When expression of unc119c was inhibited, the formation of the habenular commissure (HC) was specifically compromised. The Unc119c interacting factors Arl3l1 and Arl3l2 as well as Wnt4a also proved indispensible for HC formation. CONCLUSIONS: We suggest that Unc119c, together with Arl3l1/2, plays an important role in modulating Wnt4a production and secretion during HC formation in the forebrain of the zebrafish embryo.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/biossíntese , Estruturas Animais/embriologia , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Glândula Pineal/embriologia , Proteínas de Peixe-Zebra/biossíntese , Peixe-Zebra/embriologia , Proteínas Adaptadoras de Transdução de Sinal/genética , Animais , Ritmo Circadiano/fisiologia , Especificidade de Órgãos/fisiologia , Peixe-Zebra/genética , Proteínas de Peixe-Zebra/genética
9.
Development ; 137(18): 3013-8, 2010 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-20685732

RESUMO

Neural crest (NC) precursors are stem cells that are capable of forming many cell types after migration to different locations in the embryo. NC and placodes form at the neural plate border (NPB). The Wnt pathway is essential for specifying NC versus placodal identity in this cell population. Here we describe the BTB domain-containing protein Potassium channel tetramerization domain containing 15 (Kctd15) as a factor expressed in the NPB that efficiently inhibits NC induction in zebrafish and frog embryos. Whereas overexpression of Kctd15 inhibited NC formation, knockdown of Kctd15 led to expansion of the NC domain. Likewise, NC induction by Wnt3a plus Chordin in Xenopus animal explants was suppressed by Kctd15, but constitutively active beta-catenin reversed Kctd15-mediated suppression of NC induction. Suppression of NC induction by inhibition of Wnt8.1 was rescued by reduction of Kctd15 expression, linking Kctd15 action to the Wnt pathway. We propose that Kctd15 inhibits NC formation by attenuating the output of the canonical Wnt pathway, thereby restricting expansion of the NC domain beyond its normal range.


Assuntos
Crista Neural/embriologia , Crista Neural/metabolismo , Canais de Potássio de Abertura Dependente da Tensão da Membrana/metabolismo , Transdução de Sinais , Proteínas de Xenopus/metabolismo , Xenopus laevis/metabolismo , Proteínas de Peixe-Zebra/metabolismo , Peixe-Zebra/metabolismo , Animais , Diferenciação Celular , Regulação da Expressão Gênica no Desenvolvimento , Canais de Potássio de Abertura Dependente da Tensão da Membrana/genética , Proteínas Wnt/genética , Proteínas Wnt/metabolismo , Proteínas de Xenopus/genética , Xenopus laevis/embriologia , Xenopus laevis/genética , Peixe-Zebra/embriologia , Peixe-Zebra/genética , Proteínas de Peixe-Zebra/genética , beta Catenina/genética , beta Catenina/metabolismo
10.
Proc Natl Acad Sci U S A ; 107(8): 3564-9, 2010 Feb 23.
Artigo em Inglês | MEDLINE | ID: mdl-20142486

RESUMO

In vertebrates, Evx homeodomain transcription factor-encoding genes are expressed in the posterior region during embryonic development, and overexpression experiments have revealed roles in tail development in fish and frogs. We analyzed the molecular mechanisms of posterior neural development and axis formation regulated by eve1. We show that eve1 is involved in establishing trunk and tail neural ectoderm by two independent mechanisms: First, eve1 posteriorizes neural ectoderm via induction of aldh1a2, which encodes an enzyme that synthesizes retinoic acid; second, eve1 is involved in neural induction in the posterior ectoderm by attenuating BMP expression. Further, eve1 can restore trunk neural tube formation in the organizer-deficient ichabod(-/-) mutant. We conclude that eve1 is crucial for the organization of the antero-posterior and dorso-ventral axis in the gastrula ectoderm and also has trunk- and tail-promoting activity.


Assuntos
Padronização Corporal/genética , Ectoderma/embriologia , Regulação da Expressão Gênica no Desenvolvimento , Tubo Neural/embriologia , Proteínas Repressoras/metabolismo , Cauda/embriologia , Peixe-Zebra/embriologia , Animais , Proteínas Morfogenéticas Ósseas/biossíntese , Ectoderma/metabolismo , Embrião não Mamífero/metabolismo , Gástrula/embriologia , Gástrula/metabolismo , Genes Homeobox , Proteínas de Homeodomínio/biossíntese , Tubo Neural/metabolismo , Proteínas Repressoras/genética , Tretinoína/metabolismo , Peixe-Zebra/genética , Peixe-Zebra/metabolismo
11.
EMBO J ; 27(4): 606-17, 2008 Feb 20.
Artigo em Inglês | MEDLINE | ID: mdl-18256687

RESUMO

The Wnt-PCP (planar cell polarity, PCP) pathway regulates cell polarity and convergent extension movements during axis formation in vertebrates by activation of Rho and Rac, leading to the re-organization of the actin cytoskeleton. Rho and Rac activation require guanine nucleotide-exchange factors (GEFs), but the identity of the GEF involved in Wnt-PCP-mediated convergent extension is unknown. Here we report the identification of the weak-similarity GEF (WGEF) gene by a microarray-based screen for notochord enriched genes, and show that WGEF is involved in Wnt-regulated convergent extension. Overexpression of WGEF activated RhoA and rescued the suppression of convergent extension by dominant-negative Wnt-11, whereas depletion of WGEF led to suppression of convergent extension that could be rescued by RhoA or Rho-associated kinase activation. WGEF protein preferentially localized at the plasma membrane, and Frizzled-7 induced colocalization of Dishevelled and WGEF. WGEF protein can bind to Dishevelled and Daam-1, and deletion of the Dishevelled-binding domain generates a hyperactive from of WGEF. These results indicate that WGEF is a component of the Wnt-PCP pathway that connects Dishevelled to Rho activation.


Assuntos
Gastrulação , Fatores de Troca do Nucleotídeo Guanina/metabolismo , Proteínas Wnt/metabolismo , Proteínas rho de Ligação ao GTP/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Animais , Linhagem Celular , Polaridade Celular , Proteínas Desgrenhadas , Fatores de Troca do Nucleotídeo Guanina/genética , Humanos , Notocorda/metabolismo , Fosfoproteínas/metabolismo , Xenopus laevis/fisiologia
12.
Biochem Biophys Res Commun ; 421(2): 367-74, 2012 May 04.
Artigo em Inglês | MEDLINE | ID: mdl-22510405

RESUMO

Both endothelial and erythroid cells are generated in the intermediate cell mass (ICM) during zebrafish embryogenesis, but the nature of the genes that contribute to the processes of erythrocyte maturation and blood vessel network formation is not fully understood. From our in situ-based screening, we have identified a novel factor, Vap (Vascular Associated Protein) that is predominantly expressed in the ICM, and subsequently enriched in endothelial cells. Vap expression in the ICM was drastically suppressed in the cloche mutant that has defects in both vasculogenesis and hematopoiesis, whereas Vap expression was not affected in the vlad tepes/gata1 mutant. Knockdown of Vap using anti-sense morpholinos (VAP-MO) not only resulted in decreased numbers of erythrocytes but also in the strong suppression of hemoglobin production. Further, we found that Vap knockdown caused the disorganization of the intersegmental vessels (ISVs), which show irregular branching. We propose that Vap plays an important role in the maturation of endothelial and erythroid cells in zebrafish.


Assuntos
Eritropoese/fisiologia , Proteínas de Membrana/fisiologia , Neovascularização Fisiológica/fisiologia , Proteínas de Peixe-Zebra/fisiologia , Peixe-Zebra/fisiologia , Sequência de Aminoácidos , Animais , Técnicas de Silenciamento de Genes , Proteínas de Membrana/química , Proteínas de Membrana/genética , Dados de Sequência Molecular , Neovascularização Fisiológica/genética , Peixe-Zebra/embriologia , Peixe-Zebra/genética , Proteínas de Peixe-Zebra/química , Proteínas de Peixe-Zebra/genética
13.
Development ; 136(21): 3543-8, 2009 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-19793890

RESUMO

We compared the transcriptome in the developing notochord of Xenopus laevis embryos with that of other embryonic regions. A coordinated and intense activation of a large set of secretory pathway genes was observed in the notochord, but not in notochord precursors in the axial mesoderm at early gastrula stage. The genes encoding Xbp1 and Creb3l2 were also activated in the notochord. These two transcription factors are implicated in the activation of secretory pathway genes during the unfolded protein response, where cells react to the stress of a build-up of unfolded proteins in their endoplasmic reticulum. Xbp1 and Creb3l2 are differentially expressed but not differentially activated in the notochord. Reduction of expression of Xbp1 or Creb3l2 by injection of antisense morpholinos led to strong deficits in notochord but not somitic muscle development. In addition, the expression of some, but not all, genes encoding secretory proteins was inhibited by injection of xbp1 morpholinos. Furthermore, expression of activated forms of Xbp1 or Creb3l2 in animal explants could activate a similar subset of secretory pathway genes. We conclude that coordinated activation of a battery of secretory pathway genes mediated by Xbp1 and Creb/ATF factors is a characteristic and necessary feature of notochord formation.


Assuntos
Via Secretória , Xenopus/embriologia , Animais , Fatores de Transcrição de Zíper de Leucina Básica/metabolismo , Embrião não Mamífero/metabolismo , Retículo Endoplasmático/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Notocorda/embriologia , Dobramento de Proteína , Xenopus/metabolismo , Proteínas de Xenopus/metabolismo
14.
Proc Natl Acad Sci U S A ; 106(7): 2230-5, 2009 Feb 17.
Artigo em Inglês | MEDLINE | ID: mdl-19164561

RESUMO

Establishment of left-right asymmetry in vertebrates requires nodal, Wnt-PCP and FGF signaling and involves ciliogenesis in a laterality organ. Effector genes through which FGF signaling affects laterality have not been described. We isolated the zebrafish ier2 and fibp1 genes as FGF target genes and show that their protein products interact. Knock down of these factors interferes with establishment of organ laterality and causes defective cilia formation in Kupffer's Vesicle, the zebrafish laterality organ. Cilia are also lost after suppression of FGF8, but can be rescued by injection of ier2 and fibp1 mRNA. We conclude that Ier2 and Fibp1 mediate FGF signaling in ciliogenesis in Kupffer's Vesicle and in the establishment of laterality in the zebrafish embryo.


Assuntos
Padronização Corporal , Proteínas de Transporte/fisiologia , Fatores de Crescimento de Fibroblastos/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Proteínas Imediatamente Precoces/fisiologia , Proteínas de Peixe-Zebra/metabolismo , Animais , Proteínas de Transporte/genética , Linhagem Celular , Biologia do Desenvolvimento , Humanos , Proteínas Imediatamente Precoces/genética , Hibridização In Situ , Modelos Biológicos , Fenótipo , RNA Mensageiro/metabolismo , Técnicas do Sistema de Duplo-Híbrido , Peixe-Zebra , Proteínas de Peixe-Zebra/genética , Proteínas de Peixe-Zebra/fisiologia
15.
Proc Natl Acad Sci U S A ; 106(5): 1462-7, 2009 Feb 03.
Artigo em Inglês | MEDLINE | ID: mdl-19171890

RESUMO

The gene networks underlying closure of the optic fissure during vertebrate eye development are poorly understood. Here, we profile global gene expression during optic fissure closure using laser capture microdissected (LCM) tissue from the margins of the fissure. From these data, we identify a unique role for the C(2)H(2) zinc finger proteins Nlz1 and Nlz2 in normal fissure closure. Gene knockdown of nlz1 and/or nlz2 in zebrafish leads to a failure of the optic fissure to close, a phenotype which closely resembles that seen in human uveal coloboma. We also identify misregulation of pax2 in the developing eye of morphant fish, suggesting that Nlz1 and Nlz2 act upstream of the Pax2 pathway in directing proper closure of the optic fissure.


Assuntos
Proteínas de Ligação a DNA/genética , Olho/embriologia , Perfilação da Expressão Gênica , Proteínas Repressoras/genética , Proteínas de Peixe-Zebra/genética , Animais , Sequência de Bases , Coloboma/genética , Primers do DNA , Regulação da Expressão Gênica no Desenvolvimento , Hibridização In Situ , Fator de Transcrição PAX2/genética , Reação em Cadeia da Polimerase , Regiões Promotoras Genéticas , RNA Mensageiro/genética , Peixe-Zebra , Dedos de Zinco
16.
Dev Biol ; 342(1): 11-25, 2010 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-20346941

RESUMO

Fibroblast growth factors (FGFs) are secreted molecules that activate the RAS/mitogen-activated protein kinase (MAPK) signaling pathway. In zebrafish development, FGF signaling is responsible for establishing dorsal polarity, maintaining the isthmic organizer, and cardiac ventricle formation. Because several ETS factors are known transcriptional mediators of MAPK signaling, we hypothesized that these factors function to mediate FGF signaling processes. In zebrafish, the simultaneous knock-down of three Pea3 ETS proteins, Etv5, Erm, and Pea3, produced phenotypes reminiscent of embryos deficient in FGF signaling. Morphant embryos displayed both cardiac and left/right patterning defects as well as disruption of the isthmic organizer. Furthermore, the expression of FGF target genes was abolished in Pea3 ETS depleted embryos. To understand how FGF signaling and ETS factors control gene expression, transcriptional regulation of dusp6 was studied in mouse and zebrafish. Conserved Pea3 ETS binding sites were identified within the Dusp6 promoter, and reporter assays showed that one of these sites is required for dusp6 induction by FGFs. We further demonstrated the interaction of Pea3 ETS factors with the Dusp6 promoter both in vitro and in vivo. These results revealed the requirement of ETS factors in transducing FGF signals in developmental processes.


Assuntos
Fatores de Crescimento de Fibroblastos/metabolismo , Proteínas Proto-Oncogênicas c-ets/metabolismo , Fatores de Transcrição/metabolismo , Peixe-Zebra/metabolismo , Animais , Sítios de Ligação/genética , Diferenciação Celular/genética , Fatores de Crescimento de Fibroblastos/genética , Expressão Gênica , Regulação da Expressão Gênica , Camundongos , Proteínas Quinases Ativadas por Mitógeno/genética , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Oncogenes , Proteínas Proto-Oncogênicas c-ets/genética , Transdução de Sinais/genética , Transdução de Sinais/fisiologia , Fatores de Transcrição/genética , Peixe-Zebra/genética
17.
Nat Cell Biol ; 4(2): 165-9, 2002 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-11802164

RESUMO

Fibroblast growth factors (FGFs) are members of a family of some 30 secreted proteins important in the regulation of cellular proliferation, migration, differentiation and survival. Here we report the identification of a novel modulator of FGF signal transduction, sef, isolated from a zebrafish embryo library through an in situ hybridization screen. The sef gene encodes a transmembrane protein, and belongs to the synexpression group that includes some of the fgf genes. Sef expression is positively regulated by FGF, and ectopic expression of sef in zebrafish or Xenopus laevis embryos specifically inhibits FGF signalling. In co-immunoprecipitation assays, the intracellular domain of Sef interacts with FGF receptors, FGFR1 and FGFR2. Injection of antisense sef morpholino oligos mimicked the phenotypes observed by ectopic fgf8 expression, suggesting that Sef is required to limit FGF signalling during development.


Assuntos
Fatores de Crescimento de Fibroblastos/metabolismo , Proteínas de Membrana/metabolismo , Transdução de Sinais/fisiologia , Animais , Embrião não Mamífero/metabolismo , Fatores de Crescimento de Fibroblastos/genética , Hibridização In Situ , Proteínas de Membrana/genética , Dados de Sequência Molecular , Oligonucleotídeos Antissenso/genética , Oligonucleotídeos Antissenso/metabolismo , Receptores de Fatores de Crescimento de Fibroblastos/metabolismo , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Transplante de Tecidos , Xenopus laevis/fisiologia , Peixe-Zebra/fisiologia
18.
Dev Dyn ; 239(11): 2980-8, 2010 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-20925124

RESUMO

Kupffer's vesicle (KV), a ciliated fluid-filled sphere in the zebrafish embryo with a critical role in laterality determination, is derived from a group of superficial cells in the organizer region of the gastrula named the dorsal forerunner cells (DFC). We have examined the role of the expression of sox17 and chordin (chd) in the DFC in KV formation and laterality determination. Whereas sox17 was known to be expressed in DFC, its function in these cells was not studied before. Further, expression of chd in these cells has not been reported previously. Targeted knockdown of Sox17 and Chd in DFC led to aberrant Left-Right (L-R) asymmetry establishment, as visualized by the expression of southpaw and lefty, and heart and pancreas placement in the embryo. These defects correlated with the formation of small KVs with apparently diminished cilia, consistent with the known requirement for ciliary function in the laterality organ for the establishment of L-R asymmetry.


Assuntos
Líquidos Corporais/metabolismo , Padronização Corporal/fisiologia , Embrião não Mamífero/citologia , Embrião não Mamífero/metabolismo , Glicoproteínas/metabolismo , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Fatores de Transcrição SOXF/metabolismo , Proteínas de Peixe-Zebra/metabolismo , Animais , Padronização Corporal/genética , Regulação da Expressão Gênica no Desenvolvimento/genética , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Glicoproteínas/genética , Imuno-Histoquímica , Hibridização In Situ , Peptídeos e Proteínas de Sinalização Intercelular/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fatores de Transcrição SOXF/genética , Peixe-Zebra , Proteínas de Peixe-Zebra/genética
19.
Biochem Biophys Res Commun ; 402(4): 626-30, 2010 Nov 26.
Artigo em Inglês | MEDLINE | ID: mdl-20971071

RESUMO

Coordinated Nodal-related signals and Bozozok (Boz) activity are critical for the initial specification of dorsal mesoderm and anterior neuroectoderm during zebrafish embryogenesis. Overexpression of Boz expands gsc expression into the ventro-lateral marginal blastomeres where Nodal signaling is active, but is insufficient to induce ectopic gsc expression in the animal region. We found that overexpression of Boz together with depletion of Lnx-2b (previously named Lnx-like, Lnx-l), but not each manipulation alone, causes robust gsc expression in all blastomeres. Furthermore, nodal-related signals are required for gsc expression in embryos with elevated Boz activity. Through targeted injection into single cells at the 128-cell stage we illustrate the role of maternally deposited Lnx-2b to restrict the expansion of gsc expression into the presumptive ectodermal region. This report provides a novel mechanism for limiting dorsal organizer specification to a defined region of the early zebrafish embryo.


Assuntos
Proteína Goosecoid/biossíntese , Proteínas de Homeodomínio/metabolismo , Mesoderma/metabolismo , Ligantes da Sinalização Nodal/metabolismo , Ubiquitina-Proteína Ligases/metabolismo , Proteínas de Peixe-Zebra/biossíntese , Proteínas de Peixe-Zebra/metabolismo , Peixe-Zebra/embriologia , Animais , Ectoderma/metabolismo , Ubiquitina-Proteína Ligases/genética , Peixe-Zebra/metabolismo , Proteínas de Peixe-Zebra/genética
20.
Dev Biol ; 318(2): 335-46, 2008 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-18468594

RESUMO

Protocadherin-18a (Pcdh18a) belongs to the delta 2-protocadherins, which constitute the largest subgroup within the cadherin superfamily. Here we present isolation of a full-length zebrafish cDNA that encodes a protein highly similar to human and mouse Pcdh18. Zebrafish pcdh18a is expressed in a complex and dynamic pattern in the nervous system from gastrula stages onward, with lesser expression in mesodermal derivatives. Pcdh18a-eGFP fusion protein is expressed in a punctate manner on the membranes between cells. Overexpression of pcdh18a in embryos caused cyclopia, mislocalization of hatching gland tissue, and duplication or splitting of the neural tube. Most neural markers tested were expressed in an approximately correct A-P pattern. By cell transplantation we showed that overexpression of pcdh18a causes diminished cell migration and reduced cell protrusions, resulting in a tendency of cells to stay more firmly aggregated, probably due to increased cell adhesion. In contrast, knockdown of pcdh18a by a morpholino oligonucleotide caused defects in epiboly, and led to reduced cell adhesion as shown by cell dissociation, sorting and transplantation experiments. These results suggest a role for Pcdh18a in cell adhesion, migration and behavior but not cell specification during gastrula and segmentation stages of development.


Assuntos
Caderinas/metabolismo , Comunicação Celular , Movimento Celular , Proteínas de Peixe-Zebra/metabolismo , Peixe-Zebra/embriologia , Animais , Caderinas/genética , Adesão Celular , Embrião não Mamífero/metabolismo , Gastrulação , Regulação da Expressão Gênica no Desenvolvimento , Dados de Sequência Molecular , Protocaderinas , Peixe-Zebra/metabolismo , Proteínas de Peixe-Zebra/genética
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa