Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 80
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Mech Dev ; 121(6): 507-18, 2004 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-15172682

RESUMO

The Hedgehog signalling pathway is deployed repeatedly during normal animal development and its inappropriate activity is associated with various tumours in human. The serpentine protein Smoothened (Smo) is essential for cells to respond to the Hedeghog (Hh) signal; oncogenic forms of Smo have been isolated from human basal cell carcinomas. Despite similarities with ligand binding G-protein coupled receptors, the molecular basis of Smo activity and its regulation remains unclear. In non-responding cells, Smo is suppressed by the activity of another multipass membrane spanning protein Ptc, which acts as the Hh receptor. In Drosophila, binding of Hh to Ptc has been shown to cause an accumulation of phosphorylated Smo protein and a concomitant stabilisation of the activated form of the Ci transcription factor. Here, we identify domains essential for Smo activity and investigate the sub-cellular distribution of the wild type protein in vivo. We find that deletion of the amino terminus and the juxtamembrane region of the carboxy terminus of the protein result in the loss of normal Smo activity. Using Green Fluorescent Protein (GFP) and horseradish peroxidase fusion proteins we show that Smo accumulates in the plasma membrane of cells in which Ptc activity is abrogated by Hh but is targeted to the degradative pathway in cells where Ptc is active. We further demonstrate that Smo accumulation is likely to be a cause, rather than a consequence, of Hh signal transduction.


Assuntos
Proteínas de Drosophila/biossíntese , Proteínas de Drosophila/química , Proteínas de Drosophila/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Receptores Acoplados a Proteínas G/biossíntese , Receptores Acoplados a Proteínas G/química , Transdução de Sinais , Alelos , Animais , Proteínas de Bactérias/metabolismo , Células COS , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Proteínas de Ligação a DNA/metabolismo , Drosophila melanogaster , Deleção de Genes , Proteínas de Fluorescência Verde/metabolismo , Proteínas Hedgehog , Homozigoto , Peroxidase do Rábano Silvestre/metabolismo , Ligantes , Proteínas Luminescentes/metabolismo , Proteínas de Membrana/metabolismo , Microscopia , Microscopia Confocal , Microscopia Eletrônica de Transmissão , Mutagênese , Proteínas Mutantes Quiméricas , Mutação , Fenótipo , Fosforilação , Mutação Puntual , Ligação Proteica , Conformação Proteica , Estrutura Terciária de Proteína , Receptores de Superfície Celular , Proteínas Recombinantes de Fusão/metabolismo , Receptor Smoothened , Fatores de Transcrição
2.
Dev Biol ; 269(2): 381-92, 2004 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-15110707

RESUMO

Searches of zebrafish EST and whole genome shotgun sequence databases for sequences encoding the sterol-sensing domain (SSD) protein motif identified two sets of DNA sequences with significant homology to the Drosophila dispatched gene required for release of secreted Hedgehog protein. Using morpholino antisense oligonucleotides, we found that inhibition of one of these genes, designated Disp1, results in a phenotype similar to that of the "you-type" mutants, previously implicated in signalling by Hedgehog proteins in the zebrafish embryo. Injection of disp1 mRNA into embryos homozygous for one such mutation, chameleon (con) results in rescue of the mutant phenotype. Radiation hybrid mapping localised disp1 to the same region of LG20 to which the con mutation was mapped by meiotic recombination analysis. Sequence analysis of disp1 cDNA derived from homozygous con mutant embryos revealed that both mutant alleles are associated with premature termination codons in the disp1 coding sequence. By analysing the expression of markers of specific cell types in the neural tube, pancreas and myotome of con mutant and Disp1 morphant embryos, we conclude that Disp1 activity is essential for the secretion of lipid-modified Hh proteins from midline structures.


Assuntos
Proteínas de Membrana/fisiologia , Transdução de Sinais , Transativadores/fisiologia , Proteínas de Peixe-Zebra/fisiologia , Peixe-Zebra/embriologia , Peixe-Zebra/genética , Sequência de Aminoácidos , Animais , Mapeamento Cromossômico , Regulação da Expressão Gênica no Desenvolvimento , Proteínas Hedgehog , Dados de Sequência Molecular , Mutação
3.
Science ; 294(5548): 1879-81, 2001 Nov 30.
Artigo em Inglês | MEDLINE | ID: mdl-11729305

RESUMO

Hedgehog proteins constitute one of the major classes of intercellular signals that control inductive interactions during animal development. These proteins undergo unusual lipid modifications and signal through an unconventional transmembrane protein receptor that is characterized by a sequence motif implicated in sterol sensing. Recent studies suggest that the lipid adducts regulate the range and potency of the signals, whereas the sterol-sensing domain is essential for receptor activity.


Assuntos
Colesterol/metabolismo , Proteínas de Insetos/metabolismo , Metabolismo dos Lipídeos , Transdução de Sinais , Transativadores/metabolismo , Animais , Proteínas de Drosophila/química , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Proteínas Hedgehog , Humanos , Proteínas de Insetos/química , Proteínas de Insetos/genética , Ligação Proteica , Processamento de Proteína Pós-Traducional , Transporte Proteico , Transativadores/química , Transativadores/genética
5.
Development ; 128(16): 3081-94, 2001 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-11688558

RESUMO

We describe a new zebrafish mutation, neckless, and present evidence that it inactivates retinaldehyde dehydrogenase type 2, an enzyme involved in retinoic acid biosynthesis. neckless embryos are characterised by a truncation of the anteroposterior axis anterior to the somites, defects in midline mesendodermal tissues and absence of pectoral fins. At a similar anteroposterior level within the nervous system, expression of the retinoic acid receptor a and hoxb4 genes is delayed and significantly reduced. Consistent with a primary defect in retinoic acid signalling, some of these defects in neckless mutants can be rescued by application of exogenous retinoic acid. We use mosaic analysis to show that the reduction in hoxb4 expression in the nervous system is a non-cell autonomous effect, reflecting a requirement for retinoic acid signalling from adjacent paraxial mesoderm. Together, our results demonstrate a conserved role for retinaldehyde dehydrogenase type 2 in patterning the posterior cranial mesoderm of the vertebrate embryo and provide definitive evidence for an involvement of endogenous retinoic acid in signalling between the paraxial mesoderm and neural tube.


Assuntos
Aldeído Oxirredutases/genética , Mesoderma/metabolismo , Mutação , Rombencéfalo/metabolismo , Aldeído Oxirredutases/metabolismo , Sequência de Aminoácidos , Animais , Clonagem Molecular , Ectoderma/metabolismo , Ligação Genética , Proteínas de Homeodomínio/metabolismo , Hibridização In Situ , Marcação In Situ das Extremidades Cortadas , Modelos Biológicos , Modelos Genéticos , Dados de Sequência Molecular , Crista Neural/embriologia , Notocorda/embriologia , Fenótipo , RNA Mensageiro/metabolismo , Receptores do Ácido Retinoico/metabolismo , Retinal Desidrogenase , Receptor alfa de Ácido Retinoico , Rombencéfalo/embriologia , Homologia de Sequência de Aminoácidos , Transdução de Sinais , Fatores de Transcrição/metabolismo , Tretinoína/metabolismo , Tretinoína/farmacologia , Peixe-Zebra , Proteínas de Peixe-Zebra/agonistas
6.
Curr Biol ; 11(17): 1358-63, 2001 Sep 04.
Artigo em Inglês | MEDLINE | ID: mdl-11553330

RESUMO

Recent studies have implicated the signaling factor Sonic hedgehog (Shh) as a negative regulator of pancreatic development, but as a positive regulator of pancreas function in amniotes [1-4]. Here, using genetic analysis, we show that specification of the pancreas in the teleost embryo requires the activity of Hh proteins. Zebrafish embryos compromised in Hh signaling exhibit disruption in the expression of the pancreas-specifying homeobox gene pdx-1 and concomitantly show almost complete absence of the endocrine pancreas. Reciprocally, ubiquitous activation of the Hh pathway in wild-type embryos causes ectopic induction of endodermal pdx-1 expression and the differentiation of supernumerary endocrine cells. Our results suggest that Hh proteins influence pancreas specification via inductive interactions from the axial midline rather than through their localized expression in the endodermal cells themselves.


Assuntos
Proteínas de Homeodomínio , Pâncreas/embriologia , Receptores Acoplados a Proteínas G , Transdução de Sinais/fisiologia , Transativadores/metabolismo , Proteínas de Peixe-Zebra , Animais , Expressão Gênica , Proteínas Hedgehog , Receptores de Superfície Celular/genética , Receptores de Superfície Celular/metabolismo , Transdução de Sinais/genética , Receptor Smoothened , Transativadores/genética , Peixe-Zebra/embriologia
7.
Genes Dev ; 15(12): 1563-76, 2001 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-11410536

RESUMO

Developmental programs that govern the embryonic diversification of distinct kinds of muscles in vertebrates remain obscure. For instance, the most widely recognized attribute of early diversity among skeletal myoblasts is their ability to differentiate exclusively into fibers with slow or fast contractile properties. However, we know little about the developmental basis and genetic regulation of this seminal event in vertebrate myogenesis. Here we show that in the zebrafish, the u-boot gene acts as a myogenic switch that regulates the choice of myoblasts to adopt slow versus fast fiber developmental pathways. In u-boot mutant embryos, slow muscle precursors abort their developmental program, failing to activate expression of the homeobox gene prox1 and transfating into muscle cells with fast fiber properties. Using oligonucleotide-mediated translational inhibition, we have investigated the role of prox1 in this program. We find that it functions in the terminal step of the u-boot controlled slow fiber developmental pathway in the regulation of slow myofibril assembly. Our findings provide new insight into the genetic control of slow versus fast fiber specification and differentiation and indicate that dedicated developmental pathways exist in vertebrates for the elaboration of distinct elements of embryonic muscle pattern.


Assuntos
Proteínas de Homeodomínio/metabolismo , Fibras Musculares de Contração Rápida/fisiologia , Fibras Musculares de Contração Lenta/fisiologia , Proteínas/metabolismo , Transativadores , Animais , Diferenciação Celular , Expressão Gênica , Proteínas Hedgehog , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/fisiologia , Fatores de Transcrição Kruppel-Like , Mesoderma/citologia , Mesoderma/fisiologia , Fibras Musculares Esqueléticas/fisiologia , Fibras Musculares de Contração Lenta/citologia , Cadeias Pesadas de Miosina/genética , Proteínas/genética , Transdução de Sinais/fisiologia , Células-Tronco/citologia , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Proteínas Supressoras de Tumor , Peixe-Zebra/embriologia , Proteína Gli2 com Dedos de Zinco
8.
Curr Biol ; 11(8): 608-13, 2001 Apr 17.
Artigo em Inglês | MEDLINE | ID: mdl-11369206

RESUMO

The tumor suppressor gene patched (ptc) encodes an approximately 140 kDa polytopic transmembrane protein [1-3] [corrected] that binds members of the Hedgehog (Hh) family of signaling proteins [4-6] [corrected] and regulates the activity of Smoothened (Smo), a G protein-coupled receptor-like protein essential for Hh signal transduction [7-9] [corrected]. Ptc contains a sterol-sensing domain (SSD) [10, 11] [corrected], a motif found in proteins implicated in the intracellular trafficking of cholesterol [12] [corrected], and/or other cargoes [13-15] [corrected]. Cholesterol plays a critical role in Hedgehog (Hh) signaling by facilitating the regulated secretion and sequestration of the Hh protein [16] [corrected], to which it is covalently coupled. In addition, cholesterol synthesis inhibitors block the ability of cells to respond to Hh [18, 19] [corrected], and this finding points to an additional requirement for the lipid in regulating downstream components of the Hh signaling pathway. Although the SSD of Ptc has been linked to both the sequestration of, and the cellular response to Hh [16, 20, 21] [corrected], definitive evidence for its function has so far been lacking. Here we describe the identification and characterization of two missense mutations in the SSD of Drosophila Ptc; strikingly, while both mutations abolish Smo repression, neither affects the ability of Ptc to interact with Hh. We speculate that Ptc may control Smo activity by regulating an intracellular trafficking process dependent upon the integrity of the SSD.


Assuntos
Proteínas de Drosophila , Proteínas de Insetos/fisiologia , Proteínas de Membrana/fisiologia , Mutação de Sentido Incorreto , Receptores de Superfície Celular/metabolismo , Receptores Acoplados a Proteínas G , Sequência de Aminoácidos , Animais , Transporte Biológico , Drosophila/genética , Drosophila/crescimento & desenvolvimento , Drosophila/metabolismo , Feminino , Proteínas Hedgehog , Proteínas de Insetos/genética , Proteínas de Insetos/metabolismo , Masculino , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Dados de Sequência Molecular , Transdução de Sinais/fisiologia , Receptor Smoothened , Esteróis , Frações Subcelulares
9.
Dev Biol ; 231(1): 201-16, 2001 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-11180963

RESUMO

The anterior-posterior identities of cells in the hindbrain and cranial neural crest are thought to be determined by their Hox gene expression status, but how and when cells become committed to these identities remain unclear. Here we address this in zebrafish by cell transplantation, to test plasticity in hox expression in single cells. We transplanted cells alone, or in small groups, between hindbrain rhombomeres or between the neural crest primordia of pharyngeal arches. We found that transplanted cells regulated hox expression according to their new environments. The degree of plasticity, however, depended on both the timing and the size of the transplant. At later stages transplanted cells were more likely to be irreversibly committed and maintain their hox expression, demonstrating a progressive loss of responsiveness to the environmental signals that specify segmental identities. Individual transplanted cells also showed greater plasticity than those lying within the center of larger groups, suggesting that a community effect normally maintains hox expression within segments. We also raised experimental embryos to larval stages to analyze transplanted cells after differentiation and found that neural crest cells contributed to pharyngeal cartilages appropriate to the anterior-posterior level of the new cellular environment. Thus, consistent with models implicating hox expression in control of segmental identity, plasticity in hox expression correlates with plasticity in final cell fate.


Assuntos
Regulação da Expressão Gênica no Desenvolvimento , Proteínas de Homeodomínio/genética , Crista Neural/metabolismo , Rombencéfalo/metabolismo , Proteínas de Xenopus , Proteínas de Peixe-Zebra , Peixe-Zebra/embriologia , Animais , Osso e Ossos/embriologia , Ectoderma/fisiologia
10.
Curr Biol ; 10(20): 1315-8, 2000 Oct 19.
Artigo em Inglês | MEDLINE | ID: mdl-11069117

RESUMO

Hedgehog (Hh) signalling plays a central role in many developmental processes in both vertebrates and invertebrates [1]. The multipass membrane-spanning proteins Patched (Ptc) [2-4] and Smoothened (Smo) [5-7] have been proposed to act as subunits of a putative Hh receptor complex. According to this view, Smo functions as the transducing subunit, the activity of which is blocked by a direct interaction with the ligand-binding subunit, Ptc [8]. Activation of the intracellular signalling pathway occurs when Hh binds to Ptc [8-11], an event assumed to release Smo from Ptc-mediated inhibition. Evidence for a physical interaction between Smo and Ptc is so far limited to studies of the vertebrate versions of these proteins when overexpressed in tissue culture systems [8,12]. To test this model, we have overexpressed the Drosophila Smo protein in vivo and found that increasing the levels of Smo protein per se was not sufficient for activation of the pathway. Immunohistochemical staining of wild-type and transgenic embryos revealed distinct patterns of Smo distribution, depending on which region of the protein was detected by the antibody. Our findings suggest that Smo is modified to yield a non-functional form and this modification is promoted by Ptc in a non-stoichiometric manner.


Assuntos
Proteínas de Drosophila , Embrião não Mamífero/fisiologia , Proteínas de Insetos/fisiologia , Proteínas de Membrana/fisiologia , Receptores de Superfície Celular/metabolismo , Receptores Acoplados a Proteínas G , Animais , Drosophila/embriologia , Embrião não Mamífero/citologia , Proteínas Hedgehog , Morfogênese , Receptores de Superfície Celular/fisiologia , Proteínas Recombinantes/análise , Transdução de Sinais/fisiologia , Receptor Smoothened , Transfecção , beta-Galactosidase/análise
11.
J Cell Sci ; 113 ( Pt 15): 2695-703, 2000 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-10893185

RESUMO

Muscles are composed of several fibre types, the precise combination of which determines muscle function. Whereas neonatal and adult fibre type is influenced by a number of extrinsic factors, such as neural input and muscle load, there is little knowledge of how muscle cells are initially determined in the early embryo. In the zebrafish, fibres of the slow twitch class arise from precociously specified myoblasts that lie close to the midline whereas the remainder of the myotome differentiates as fast myosin expressing muscle. In vivo evidence has suggested the Sonic Hedgehog glycoprotein, secreted from the notochord, controls the formation of slow twitch and fast twitch muscle fates. Here we describe an in vitro culture system that we have developed to test directly the ability of zebrafish myoblasts to respond to exogenous Sonic Hedgehog peptide. We find that Sonic Hedgehog peptide can control the binary cell fate choice of embryonic zebrafish myoblasts in vitro. We have also used this culture system to assay the relative activities of different Hedgehog-family proteins and to investigate the possible involvement of heterotrimeric G-proteins in Hedgehog signal transduction.


Assuntos
Fibras Musculares de Contração Lenta/citologia , Fibras Musculares de Contração Lenta/fisiologia , Proteínas/fisiologia , Transdução de Sinais/fisiologia , Transativadores , Animais , Diferenciação Celular/fisiologia , Células Cultivadas , Indução Embrionária/fisiologia , Proteínas Hedgehog , Proteínas Heterotriméricas de Ligação ao GTP/fisiologia , Técnicas In Vitro , Músculo Esquelético/citologia , Músculo Esquelético/embriologia , Peixe-Zebra
13.
Dev Biol ; 219(2): 350-63, 2000 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-10694427

RESUMO

The floor plate is a morphologically distinct structure of epithelial cells situated along the midline of the ventral spinal cord in vertebrates. It is a source of guidance molecules directing the growth of axons along and across the midline of the neural tube. In the zebrafish, the floor plate is about three cells wide and composed of cuboidal cells. Two cell populations can be distinguished by the expression patterns of several marker genes, including sonic hedgehog (shh) and the fork head-domain gene fkd4: a single row of medial floor plate (MFP) cells, expressing both shh and fkd4, is flanked by rows of lateral floor plate (LFP) cells that express fkd4 but not shh. Systematic mutant searches in zebrafish embryos have identified a number of genes, mutations in which visibly reduce the floor plate. In these mutants either the MFP or the LFP cells are absent, as revealed by the analysis of the shh and fkd4 expression patterns. MFP cells are absent, but LFP cells are present, in mutants of cyclops, one-eyed pinhead, and schmalspur, whose development of midline structures is affected. LFP cells are absent, but MFP cells are present, in mutants of four genes, sonic you, you, you-too, and chameleon, collectively called the you-type genes. This group of mutants also shows defects in patterning of the paraxial mesoderm, causing U- instead of V-shaped somites. One of the you-type genes, sonic you, was recently shown to encode the zebrafish Shh protein, suggesting that the you-type genes encode components of the Shh signaling pathway. It has been shown previously that in the zebrafish shh is required for the induction of LFP cells, but not for the development of MFP cells. This conclusion is supported by the finding that injection of shh RNA causes an increase in the number of LFP, but not MFP cells. Embryos mutant for iguana, detour, and umleitung share the lack of LFP cells with you-type mutants while somite patterning is not severely affected. In mutants that fail to develop a notochord, MFP cells may be present, but are always surrounded by LFP cells. These data indicate that shh, expressed in the notochord and/or the MFP cells, induces the formation of LFP cells. In embryos doubly mutant for cyclops (cyc) and sonic you (syu) both LFP and MFP cells are deleted. The number of primary motor neurons is strongly reduced in cyc;syu double mutants, while almost normal in single mutants, suggesting that the two different pathways have overlapping functions in the induction of primary motor neurons.


Assuntos
Medula Espinal/citologia , Medula Espinal/embriologia , Transativadores , Peixe-Zebra/embriologia , Animais , Padronização Corporal/genética , Regulação da Expressão Gênica no Desenvolvimento , Proteínas Hedgehog , Hibridização In Situ , Camundongos , Neurônios Motores/citologia , Mutação , Notocorda/citologia , Notocorda/embriologia , Proteínas/genética , Especificidade da Espécie , Peixe-Zebra/genética
14.
Curr Biol ; 10(5): R180-3, 2000 Mar 09.
Artigo em Inglês | MEDLINE | ID: mdl-10712894

RESUMO

By coupling to cholesterol, Hedgehog can be anchored to the cells where it is made, yet to act as a morphogen, it must be able to move away from its source. Novel genes have now been identified that control the release and dispersal of Hedgehog, shedding new light on the part played by cholesterol in these processes.


Assuntos
Colesterol/fisiologia , Proteínas de Drosophila , Proteínas de Insetos/metabolismo , Transdução de Sinais , Animais , Ligação Competitiva , Drosophila/genética , Proteínas Hedgehog , Proteínas de Insetos/genética , Modelos Biológicos , Proteoglicanas/metabolismo , Asas de Animais/citologia
15.
Mech Dev ; 90(2): 237-52, 2000 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-10640707

RESUMO

Fli-1 is an ETS-domain transcription factor whose locus is disrupted in Ewing's Sarcoma and F-MuLV induced erythroleukaemia. To gain a better understanding of its normal function, we have isolated the zebrafish homologue. Similarities with other vertebrates, in the amino acid sequence and DNA binding properties of Fli-1 from zebrafish, suggest that its function has been conserved during vertebrate evolution. The initial expression of zebrafish fli-1 in the posterior lateral mesoderm overlaps with that of gata2 in a potential haemangioblast population which likely contains precursors of blood and endothelium. Subsequently, fli-1 and gata2 expression patterns diverge, with separate fli-1 and gata2 expression domains arising in the developing vasculature and in sites of blood formation respectively. Elsewhere in the embryo, fli-1 is expressed in sites of vasculogenesis. The expression of fli-1 was investigated in a number of zebrafish mutants, which affect the circulatory system. In cloche, endothelium is absent and blood is drastically reduced. In contrast to the blood and endothelial markers that have been studied previously, fli-1 expression was initiated normally in cloche embryos, indicating that induction of fli-1 is one of the earliest indicators of haemangioblast formation. Furthermore, although fli-1 expression in the trunk was not maintained, the normal expression pattern in the anterior half of the embryo was retained. These anterior cells did not, however, condense to form blood vessels. These data indicate that cloche has previously unsuspected roles at multiple stages in the formation of the vasculature. Analysis of fli-1 expression in midline patterning mutants floating head and squint, confirms a requirement for the notochord in the formation of the dorsal-aorta. The formation of endothelium in one-eyed pinhead, cyclops and squint embryos indicates a novel role for the endoderm in the formation of the axial vein. The phenotype of sonic-you mutants implies a likely role for Sonic Hedgehog in mediating these processes.


Assuntos
Proteínas de Ligação a DNA/genética , Neovascularização Fisiológica/fisiologia , Proteínas Proto-Oncogênicas , Transativadores/genética , Proteínas de Peixe-Zebra , Sequência de Aminoácidos , Animais , Sequência de Bases , Clonagem Molecular , DNA/metabolismo , Endotélio Vascular/citologia , Endotélio Vascular/metabolismo , Fator de Transcrição GATA2 , Expressão Gênica , Células-Tronco Hematopoéticas/metabolismo , Proteínas de Homeodomínio/genética , Humanos , Peptídeos e Proteínas de Sinalização Intracelular , Camundongos , Dados de Sequência Molecular , Mutação , Ligantes da Sinalização Nodal , Proteína Proto-Oncogênica c-fli-1 , Homologia de Sequência de Aminoácidos , Células-Tronco/citologia , Células-Tronco/metabolismo , Fatores de Transcrição/genética , Fator de Crescimento Transformador beta/genética , Peixe-Zebra
16.
Mech Dev ; 90(2): 299-304, 2000 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-10640716

RESUMO

T-box (tbx) genes constitute a large family of transcriptional regulators involved in developmental patterning processes. In tetrapods, tbx5 has been implicated in specifying forelimb type identity. Here, we report the cloning of the zebrafish tbx5.1 gene and characterise its expression during zebrafish embryogenesis and early larval development of wild type and mutant embryos that affect pectoral fin patterning. tbx5.1 is expressed during development of the heart, the pectoral fins and the eye. Notably, its expression in the lateral plate mesoderm defines a single and continuous region of heart and pectoral fin precursor cells, and constitutes the earliest specific marker for pectoral fin development in the zebrafish.


Assuntos
Regulação da Expressão Gênica no Desenvolvimento , Proteínas com Domínio T/genética , Sequência de Aminoácidos , Animais , Sequência de Bases , DNA Complementar , Humanos , Camundongos , Dados de Sequência Molecular , Homologia de Sequência de Aminoácidos , Distribuição Tecidual , Peixe-Zebra
19.
Dev Biol ; 210(2): 277-87, 1999 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-10357891

RESUMO

Left-right (LR) asymmetry of the heart in vertebrates is regulated by early asymmetric signals in the embryo, including the secreted signal Sonic hedgehog (Shh), but less is known about LR asymmetries of visceral organs. Here we show that Shh also specifies asymmetries in visceral precursors in the zebrafish and that cardiac and visceral sidedness are independent. The transcription factors fli-1 and Nkx-2.5 are expressed asymmetrically in the precardiac mesoderm and subsequently in the heart; an Eph receptor, rtk2, and an adhesion protein, DM-GRASP, mark early asymmetries in visceral endoderm. Misexpression of shh mRNA, or a dominant negative form of protein kinase A, on the right side reverses the expression of these asymmetries in precursors of both the heart and the viscera. Reversals in the heart and gut are uncoordinated, suggesting that each organ interprets the signal independently. Misexpression of Bone Morphogenetic Protein (BMP4) on the right side reverses the heart, but visceral organs are unaffected, consistent with a function for BMPs locally in the heart field. Zebrafish mutants with midline defects show independent reversals of cardiac and visceral laterality. Thus, hh signals influence the development of multiple organ asymmetries in zebrafish and different organs appear to respond to a central cascade of midline signaling independently, which in the heart involves BMP4.


Assuntos
Padronização Corporal , Proteínas Morfogenéticas Ósseas/fisiologia , Embrião não Mamífero/fisiologia , Indução Embrionária , Proteínas/fisiologia , Proteínas Proto-Oncogênicas , Fatores de Transcrição , Proteínas de Xenopus , Peixe-Zebra/embriologia , Animais , Proteína Morfogenética Óssea 4 , Proteínas Morfogenéticas Ósseas/genética , Proteínas de Ligação a DNA/genética , Embrião não Mamífero/citologia , Regulação da Expressão Gênica no Desenvolvimento , Coração/embriologia , Proteínas Hedgehog , Proteína Homeobox Nkx-2.5 , Proteínas de Homeodomínio/genética , Mesoderma/fisiologia , Proteínas/genética , Proteína Proto-Oncogênica c-fli-1 , Transdução de Sinais , Transativadores/genética , Vísceras/embriologia , Proteínas de Peixe-Zebra
20.
Development ; 126(11): 2397-407, 1999 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-10225999

RESUMO

talpid3 is an embryonic-lethal chicken mutation in a molecularly un-characterised autosomal gene. The recessive, pleiotropic phenotype includes polydactylous limbs with morphologically similar digits. Previous analysis established that hox-D and bmp genes, that are normally expressed posteriorly in the limb bud in response to a localised, posterior source of Sonic Hedgehog (Shh) are expressed symmetrically across the entire anteroposterior axis in talpid3 limb buds. In contrast, Shh expression itself is unaffected. Here we examine expression of patched (ptc), which encodes a component of the Shh receptor, and is probably itself a direct target of Shh signalling, to establish whether talpid3 acts in the Shh pathway. We find that ptc expression is significantly reduced in talpid3 embryos. We also demonstrate that talpid3 function is not required for Shh signal production but is required for normal response to Shh signals, implicating talpid3 in transduction of Shh signals in responding cells. Our analysis of expression of putative components of the Shh pathway, gli1, gli3 and coupTFII shows that genes regulated by Shh are either ectopically expressed or no longer responsive to Shh signals in talpid3 limbs, suggesting possible bifurcation in the Shh pathway. We also describe genetic mapping of gli1, ptc, shh and smoothened in chickens and confirm by co-segregation analysis that none of these genes correspond to talpid3.


Assuntos
Proteínas/genética , Receptores Acoplados a Proteínas G , Receptores de Esteroides , Transativadores , Fator de Crescimento Transformador beta , Animais , Proteína Morfogenética Óssea 2 , Proteínas Morfogenéticas Ósseas/genética , Fatores de Transcrição COUP , Embrião de Galinha , Mapeamento Cromossômico , Proteínas de Ligação a DNA/genética , Regulação da Expressão Gênica no Desenvolvimento , Genes Letais , Proteínas Hedgehog , Hibridização In Situ , Botões de Extremidades/embriologia , Proteínas de Membrana , Mutação , Proteínas Oncogênicas/genética , Receptores Patched , Fenótipo , Reação em Cadeia da Polimerase , Polimorfismo Conformacional de Fita Simples , Receptores de Superfície Celular/genética , Transdução de Sinais , Transplante de Tecidos , Fatores de Transcrição/genética , Proteína GLI1 em Dedos de Zinco
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA