Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 42
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Int J Dev Biol ; 61(6-7): 465-470, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28695967

RESUMO

Multiple members of the transforming growth factor beta (TGFß) family of secreted factors play central inductive and patterning roles during embryogenesis. During gastrulation in vertebrates, the bone morphogenetic protein (BMP) sub-family is linked to formation of the embryonic organizer, Spemann's organizer in Xenopus, and dorsal-ventral mesoderm patterning. Our knowledge regarding the BMP receptors mediating this signaling is still very incomplete. The BMPR1A (ALK3) and BMPR1B (ALK6) receptors are known to mediate the BMP4 signal. These receptors belong to the ALK1 subfamily of type I receptors that also includes ACVR1 (ALK2), and ACVRL1 (ALK1). We studied by qPCR and in situ hybridization the spatio-temporal expression patterns of ALK2 and ALK1 and compared them to ALK3 and ALK6, and to the main BMPs expressed during gastrulation, i.e., BMP4, BMP7, BMP2, and ADMP, in an attempt to establish a link between ligands and receptors. There is extensive overlap between BMP4, and ALk3 and Alk6 expression, supporting their functional interaction. Robust Alk6 expression was observed from mid-gastrula. Animal region expression of both receptors shows co-expression with BMP4 and BMP7. Alk2 transcripts were detected within the organizer, overlapping with its proposed ligand, ADMP, suggesting a probable function within the organizer. Alk1 is very weakly expressed during gastrula, but its transcripts were localized to the lateral marginal zone flanking the organizer domain. No receptor closely matched the maternal BMP2 expression, although Alk2, Alk3, and Alk6, have transcripts of maternal origin. Our analysis shows that the BMP ligands and their receptors exhibit dynamic expression patterns during gastrula stages.


Assuntos
Receptores de Proteínas Morfogenéticas Ósseas/metabolismo , Embrião não Mamífero/metabolismo , Gástrula/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Proteínas de Xenopus/metabolismo , Xenopus laevis/metabolismo , Receptores de Activinas Tipo II/metabolismo , Animais , Proteínas Morfogenéticas Ósseas/metabolismo , Embrião não Mamífero/citologia , Desenvolvimento Embrionário , Feminino , Gástrula/embriologia , Organizadores Embrionários , Filogenia , Transdução de Sinais , Xenopus laevis/embriologia
2.
Sci Rep ; 7: 43010, 2017 02 21.
Artigo em Inglês | MEDLINE | ID: mdl-28220837

RESUMO

Goosecoid (Gsc) expression marks the primary embryonic organizer in vertebrates and beyond. While functions have been assigned during later embryogenesis, the role of Gsc in the organizer has remained enigmatic. Using conditional gain-of-function approaches in Xenopus and mouse to maintain Gsc expression in the organizer and along the axial midline, neural tube closure defects (NTDs) arose and dorsal extension was compromised. Both phenotypes represent convergent extension (CE) defects, arising from impaired Wnt/planar cell polarity (PCP) signaling. Dvl2 recruitment to the cell membrane was inhibited by Gsc in Xenopus animal cap assays and key Wnt/PCP factors (RhoA, Vangl2, Prickle, Wnt11) rescued Gsc-mediated NTDs. Re-evaluation of endogenous Gsc functions in MO-mediated gene knockdown frog and knockout mouse embryos unearthed PCP/CE-related phenotypes as well, including cartilage defects in Xenopus and misalignment of inner ear hair cells in mouse. Our results assign a novel function to Gsc as an inhibitor of Wnt/PCP-mediated CE. We propose that in the organizer Gsc represses CE as well: Gsc-expressing prechordal cells, which leave the organizer first, migrate and do not undergo CE like the Gsc-negative notochordal cells, which subsequently emerge from the organizer. In this model, Gsc provides a switch between cell migration and CE, i.e. cell intercalation.


Assuntos
Proteína Goosecoid/metabolismo , Organizadores Embrionários/metabolismo , Proteínas Wnt/metabolismo , Proteínas de Xenopus/metabolismo , Xenopus laevis/metabolismo , Animais , Polaridade Celular , Proteínas Desgrenhadas/metabolismo , Embrião de Mamíferos/metabolismo , Embrião de Mamíferos/patologia , Embrião não Mamífero/metabolismo , Embrião não Mamífero/patologia , Desenvolvimento Embrionário , Genes Reporter , Proteína Goosecoid/deficiência , Proteína Goosecoid/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Microscopia Eletrônica de Varredura , Transdução de Sinais , Proteínas de Xenopus/genética
3.
Differentiation ; 93: 27-38, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-27875771

RESUMO

The Wnt/planar cell polarity (PCP) pathway directs cell migration during vertebrate gastrulation and is essential for proper embryonic development. Paraxial protocadherin (PAPC, Gene Symbol pcdh8.2) is an important activator of Wnt/PCP signaling during Xenopus gastrulation, but how PAPC activity is controlled is incompletely understood. Here we show that Nemo-like kinase 1 (Nlk1), an atypical mitogen-activated protein (MAP) kinase, physically associates with the C-terminus of PAPC. This interaction mutually stabilizes both proteins by inhibiting polyubiquitination. The Nlk1 mediated stabilization of PAPC is essential for Wnt/PCP signaling, tissue separation and gastrulation movements. We identified two conserved putative phosphorylation sites in the PAPC C-terminus that are critical for Nlk1 mediated PAPC stabilization and Wnt/PCP regulation. Intriguingly, the kinase activity of Nlk1 itself was not essential for its cooperation with PAPC, suggesting an indirect regulation for example by impeding a different kinase that promotes protein degradation. Overall these results outline a novel, kinase independent role of Nlk1, wherein Nlk1 regulates PAPC stabilization and thereby controls gastrulation movements and Wnt/PCP signaling during development.


Assuntos
Caderinas/genética , Desenvolvimento Embrionário/genética , Gastrulação/genética , Proteínas Quinases Ativadas por Mitógeno/genética , Via de Sinalização Wnt/genética , Proteínas de Xenopus/genética , Animais , Caderinas/metabolismo , Movimento Celular/genética , Polaridade Celular/genética , Embrião não Mamífero , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Fosforilação , Mapas de Interação de Proteínas/genética , Protocaderinas , Proteínas de Xenopus/metabolismo , Xenopus laevis/genética , Xenopus laevis/crescimento & desenvolvimento
4.
J Biol Chem ; 291(26): 13730-42, 2016 Jun 24.
Artigo em Inglês | MEDLINE | ID: mdl-27129770

RESUMO

Convergent extension movements during vertebrate gastrulation require a balanced activity of non-canonical Wnt signaling pathways, but the factors regulating this interplay on the molecular level are poorly characterized. Here we show that sFRP2, a member of the secreted frizzled-related protein (sFRP) family, is required for morphogenesis and papc expression during Xenopus gastrulation. We further provide evidence that sFRP2 redirects non-canonical Wnt signaling from Frizzled 7 (Fz7) to the receptor tyrosine kinase-like orphan receptor 2 (Ror2). During this process, sFRP2 promotes Ror2 signal transduction by stabilizing Wnt5a-Ror2 complexes at the membrane, whereas it inhibits Fz7 signaling, probably by blocking Fz7 receptor endocytosis. The cysteine-rich domain of sFRP2 is sufficient for Ror2 activation, and related sFRPs can substitute for this function. Notably, direct interaction of the two receptors via their cysteine-rich domains also promotes Ror2-mediated papc expression but inhibits Fz7 signaling. We propose that sFRPs can act as a molecular switch, channeling the signal input for different non-canonical Wnt pathways during vertebrate gastrulation.


Assuntos
Receptores Órfãos Semelhantes a Receptor Tirosina Quinase/metabolismo , Receptores de Superfície Celular/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Via de Sinalização Wnt/fisiologia , Proteína Wnt-5a/metabolismo , Proteínas de Xenopus/metabolismo , Proteínas de Peixe-Zebra/metabolismo , Peixe-Zebra/embriologia , Animais , Gástrula , Células HEK293 , Humanos , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Receptores Órfãos Semelhantes a Receptor Tirosina Quinase/genética , Receptores de Superfície Celular/genética , Receptores Acoplados a Proteínas G/genética , Proteína Wnt-5a/genética , Proteínas de Xenopus/genética , Xenopus laevis , Proteínas de Peixe-Zebra/genética
5.
PLoS One ; 10(8): e0135504, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26270962

RESUMO

Protein Kinase Domain Containing, Cytoplasmic (PKDCC) is a protein kinase which has been implicated in longitudinal bone growth through regulation of chondrocytes formation. Nevertheless, the mechanism by which this occurs remains unknown. Here, we identified two new members of the PKDCC family, Pkdcc1 and Pkdcc2 from Xenopus laevis. Interestingly, our knockdown experiments revealed that these two proteins are both involved on blastopore and neural tube closure during gastrula and neurula stages, respectively. In vertebrates, tissue polarity and cell movement observed during gastrulation and neural tube closure are controlled by Wnt/Planar Cell Polarity (PCP) molecular pathway. Our results showed that Pkdcc1 and Pkdcc2 promote the recruitment of Dvl to the plasma membrane. But surprisingly, they revealed different roles in the induction of a luciferase reporter under the control of Atf2 promoter. While Pkdcc1 induces Atf2 expression, Pkdcc2 does not, and furthermore inhibits its normal induction by Wnt11 and Wnt5a. Altogether our data show, for the first time, that members of the PKDCC family are involved in the regulation of JNK dependent Wnt/PCP signaling pathway.


Assuntos
Proteínas Tirosina Quinases/metabolismo , Proteínas de Xenopus/metabolismo , Xenopus laevis/embriologia , Xenopus laevis/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Animais , Movimento Celular , Polaridade Celular , Clonagem Molecular/métodos , Proteínas Desgrenhadas , Regulação da Expressão Gênica no Desenvolvimento , Sistema de Sinalização das MAP Quinases , Fosfoproteínas/metabolismo , Proteínas Tirosina Quinases/genética , Via de Sinalização Wnt , Proteínas de Xenopus/genética , Xenopus laevis/genética
6.
J Inherit Metab Dis ; 38(6): 1137-46, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26141167

RESUMO

Reduced phosphomannomutase 2 activity in man leads to hypoglycosylation of glycoconjugates causing PMM2-CDG, the most common type of congenital disorders of glycosylation. Here we show that an antisense morpholino-mediated knockdown of the Xenopus laevis phosphomannomutase 2 gene provoked a general underglycosylation in frog embryos, which led to an altered phenotype and reduced glycosylation of Wnt5a as member of the non-canonical Wnt signalling. Loss of function experiments in hemi-sectioned embryos proved that due to the phosphomannomutase 2 knockdown expression of the Wnt5a/Ror2 target gene paraxial protocadherin was significantly decreased. Regarding the expression of paraxial protocadherin, a gain of function could only be achieved by injections of wnt5a and ror2 in dorsal neighbouring blastomeres, while a parallel injection of phosphomannomutase 2 morpholino led to a significant reduced level of expression. Our data show for the first time that a knockdown of phosphomannomutase 2 influences in vivo the non-canonical Wnt signalling during early embryogenesis.


Assuntos
Morfogênese/genética , Fosfotransferases (Fosfomutases)/genética , Via de Sinalização Wnt/genética , Xenopus laevis , Animais , Técnicas de Silenciamento de Genes , Receptores Órfãos Semelhantes a Receptor Tirosina Quinase/genética , Proteínas Wnt/genética , Proteína Wnt-5a , Proteínas de Xenopus/genética
7.
Eur J Hum Genet ; 23(12): 1627-33, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25804400

RESUMO

Congenital cataract (CC) is one of the most important causes for blindness or visual impairment in infancy. A substantial proportion of isolated CCs has monogenic causes. The disease is genetically heterogeneous, and all Mendelian modes of inheritance have been reported. We mapped a locus for isolated CC on 19p13.1-q13.2 in a distantly consanguineous German family with two sisters affected by dense white cataracts. Whole-exome sequencing identified a homozygous nonsense variant c.4489C>T (p.(R1497*)) in SIPA1L3 (signal-induced proliferation-associated 1 like 3) in both affected children. SIPA1L3 encodes a GTPase-activating protein (GAP), which interacts with small GTPases of the Rap family via its Rap-GAP-domain. The suggested role of Rap GTPases in cell growth, differentiation and organization of the cytoskeleton in the human lens, and lens-enriched expression of the murine ortholog gene Sipa1l3 in embryonic mice indicates that this gene is crucial for early lens development. Our results provide evidence that sequence variants in human SIPA1L3 cause autosomal recessive isolated CC and give new insight into the molecular pathogenesis underlying human cataracts.


Assuntos
Catarata/congênito , Exoma , Proteínas Ativadoras de GTPase/genética , Ligação Genética , Catarata/diagnóstico , Catarata/genética , Pré-Escolar , Cromossomos Humanos Par 19/genética , Códon sem Sentido , Feminino , Homozigoto , Humanos , Linhagem
8.
J Cell Sci ; 128(6): 1139-49, 2015 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-25616895

RESUMO

Cranial neural crest (CNC) cells are a transient population of stem cells that originate at the border of the neural plate and the epidermis, and migrate ventrally to contribute to most of the facial structures including bones, cartilage, muscles and ganglia. ADAM13 is a cell surface metalloprotease that is essential for CNC cell migration. Here, we show in Xenopus laevis embryos that the Wnt receptor Fz4 binds to the cysteine-rich domain of ADAM13 and negatively regulates its proteolytic activity in vivo. Gain of Fz4 function inhibits CNC cell migration and can be rescued by gain of ADAM13 function. Loss of Fz4 function also inhibits CNC cell migration and induces a reduction of mature ADAM13, together with an increase in the ADAM13 cytoplasmic fragment that is known to translocate into the nucleus to regulate gene expression. We propose that Fz4 associates with ADAM13 during its transport to the plasma membrane to regulate its proteolytic activity.


Assuntos
Proteínas ADAM/metabolismo , Embrião não Mamífero/metabolismo , Receptores Frizzled/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Proteínas de Membrana/metabolismo , Crista Neural/metabolismo , Proteínas Wnt/metabolismo , Proteínas de Xenopus/metabolismo , Proteínas ADAM/genética , Animais , Células COS , Membrana Celular/metabolismo , Movimento Celular , Núcleo Celular/metabolismo , Proliferação de Células , Células Cultivadas , Chlorocebus aethiops , Embrião não Mamífero/citologia , Imunofluorescência , Receptores Frizzled/genética , Células HEK293 , Humanos , Imunoprecipitação , Hibridização In Situ , Proteínas de Membrana/genética , Crista Neural/citologia , RNA Mensageiro/genética , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Proteínas Wnt/genética , Proteínas de Xenopus/genética , Xenopus laevis/genética , Xenopus laevis/crescimento & desenvolvimento , Xenopus laevis/metabolismo
9.
Differentiation ; 87(5): 209-19, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25287945

RESUMO

Mouse F9 teratocarcinoma cells are an established model for the differentiation of extra-embryonic endoderm (ExEn). Primitive endoderm, parietal and visceral endoderm can be generated by stimulation of F9 cells with retinoic acid and dibutyryl cyclic adenosine monophosphate. Here we show that Wnt/ß-Catenin signaling is down-regulated during ExEn differentiation in F9 cells and that the inhibition of the Wnt pathway promotes differentiation of the three extra-embryonic endoderm lineages. Wnt inhibition is achieved through the IGF pathway, which is up-regulated during differentiation. IGF signaling antagonizes the Wnt pathway by stimulating transcription of axin2 and by stabilizing Axin1 protein. Both Axin1 and Axin2 are components of the ß-Catenin destruction complex and act as intra-cellular inhibitors of the Wnt/ß-Catenin pathway. The data presented reveal a mechanism which restricts pluripotency of undifferentiated cells and directs them toward extra-embryonic lineages.


Assuntos
Diferenciação Celular/genética , Endoderma/metabolismo , Fator de Crescimento Insulin-Like I/metabolismo , Via de Sinalização Wnt/genética , Animais , Proteína Axina/metabolismo , Diferenciação Celular/efeitos dos fármacos , Endoderma/crescimento & desenvolvimento , Camundongos , Transdução de Sinais/efeitos dos fármacos , Tretinoína/administração & dosagem , beta Catenina/antagonistas & inibidores , beta Catenina/genética
10.
BMC Biol ; 12: 44, 2014 May 30.
Artigo em Inglês | MEDLINE | ID: mdl-24885675

RESUMO

BACKGROUND: Wnt proteins are a family of secreted signaling molecules that regulate key developmental processes in metazoans. The molecular basis of Wnt binding to Frizzled and LRP5/6 co-receptors has long been unknown due to the lack of structural data on Wnt ligands. Only recently, the crystal structure of the Wnt8-Frizzled8-cysteine-rich-domain (CRD) complex was solved, but the significance of interaction sites that influence Wnt signaling has not been assessed. RESULTS: Here, we present an extensive structure-function analysis of mouse Wnt3a in vitro and in vivo. We provide evidence for the essential role of serine 209, glycine 210 (site 1) and tryptophan 333 (site 2) in Fz binding. Importantly, we discovered that valine 337 in the site 2 binding loop is critical for signaling without contributing to binding. Mutations in the presumptive second CRD binding site (site 3) partly abolished Wnt binding. Intriguingly, most site 3 mutations increased Wnt signaling, probably by inhibiting Wnt-CRD oligomerization. In accordance, increasing amounts of soluble Frizzled8-CRD protein modulated Wnt3a signaling in a biphasic manner. CONCLUSIONS: We propose a concentration-dependent switch in Wnt-CRD complex formation from an inactive aggregation state to an activated high mobility state as a possible modulatory mechanism in Wnt signaling gradients.


Assuntos
Receptores Acoplados a Proteínas G/química , Receptores Acoplados a Proteínas G/metabolismo , Via de Sinalização Wnt , Proteína Wnt3A/química , Proteína Wnt3A/metabolismo , Sequência de Aminoácidos , Animais , Embrião não Mamífero/metabolismo , Células HEK293 , Humanos , Camundongos , Modelos Biológicos , Modelos Moleculares , Dados de Sequência Molecular , Proteínas Mutantes/metabolismo , Mutação Puntual/genética , Ligação Proteica , Estrutura Terciária de Proteína , Solubilidade , Relação Estrutura-Atividade , Peixe-Zebra/embriologia
11.
Hum Mol Genet ; 23(13): 3618-28, 2014 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-24549042

RESUMO

17ß-Hydroxysteroid dehydrogenase type 10 (HSD10) is multifunctional protein coded by the X-chromosomal HSD17B10 gene. Mutations in this gene cause HSD10 disease characterized by progressive neurological abnormalities and cardiomyopathy. Disease progression and severity of symptoms is unrelated to the protein's dehydrogenase activity. Recently, it was shown that HSD10 is an essential component of mitochondrial Ribonuclease P (RNase P), an enzyme required for mitochondrial tRNA processing, but little is known about the role of HSD10 in RNase P function. RNase P consists of three different proteins MRPP1, MRPP2 (HSD10) and MRPP3, each of which is essential for RNase P function. Here, we show that HSD10 protein levels are significantly reduced in fibroblasts from patients carrying the HSD17B10 mutation p.R130C. A reduction in HSD10 levels was accompanied by a reduction in MRPP1 protein but not MRPP3 protein. In HSD10 knock-down cells, MRPP1 protein content was also reduced, indicating that HSD10 is important for the maintenance of normal MRPP1 protein levels. Ectopic expression of HSD10 partially restored RNA processing in HSD10 knock-down cells and fibroblasts, and also expression of MRPP1 protein was restored to values comparable to controls. In both, patient fibroblasts and HSD10 knock-down cells, there was evidence of impaired processing of precursor tRNA transcripts of the mitochondrial heavy strand but not the light strand compared with controls. Our findings indicate that HSD10 is important for the maintenance of the MRPP1-HSD10 subcomplex of RNase P and that loss of HSD10 causes impaired mitochondrial precursor transcript processing which may explain mitochondrial dysfunction observed in HSD10 disease.


Assuntos
3-Hidroxiacil-CoA Desidrogenases/metabolismo , Metiltransferases/metabolismo , 3-Hidroxiacil-CoA Desidrogenases/genética , Fibroblastos/citologia , Fibroblastos/metabolismo , Humanos , Metiltransferases/genética , Mutação , RNA de Transferência/genética , Ribonuclease P/genética , Ribonuclease P/metabolismo
12.
Cell Commun Signal ; 11: 89, 2013 Nov 19.
Artigo em Inglês | MEDLINE | ID: mdl-24252524

RESUMO

BACKGROUND: Activation of the Wnt signalling cascade is primarily based on the interplay between Wnt ligands, their receptors and extracellular modulators. One prominent family of extracellular modulators is represented by the SFRP (secreted Frizzled-related protein) family. These proteins have significant similarity to the extracellular domain of Frizzled receptors, suggesting that they bind Wnt ligands and inhibit signalling. The SFRP-type protein Fz4-v1, a splice variant of the Frizzled-4 receptor found in humans and Xenopus, was shown to augment Wnt/ß-catenin signalling, and also interacts with those Wnt ligands that act on ß-catenin-independent Wnt pathways. FINDINGS: Here we show that Xenopus Fz4-v1 can activate and inhibit the ß-catenin-dependent Wnt pathway. Gain-of-function experiments revealed that high Wnt/ß-catenin activity is inhibited by low and high concentrations of Fz4-v1. In contrast, signals generated by low amounts of Wnt ligands were enhanced by low concentrations of Fz4-v1 but were repressed by high concentrations. This biphasic activity of Fz4-v1 was not observed in non-canonical Wnt signalling. Fz4-v1 enhanced ß-catenin-independent Wnt signalling triggered by either low or high doses of Wnt11. Antisense morpholino-mediated knock-down experiments demonstrated that in early Xenopus embryos Fz4-v1 is required for the migration of cranial neural crest cells and for the development of the dorsal fin. CONCLUSIONS: For the first time, we show that a splice variant of the Frizzled-4 receptor modulates Wnt signalling in a dose-dependent, biphasic manner. These results also demonstrate that the cystein-rich domain (CRD), which is shared by Fz4-v1 and SFRPs, is sufficient for the biphasic activity of these secreted Wnt modulators.


Assuntos
Receptores Frizzled/fisiologia , Via de Sinalização Wnt/fisiologia , Proteínas de Xenopus/fisiologia , Animais , Embrião não Mamífero , Desenvolvimento Embrionário/fisiologia , Sistema de Sinalização das MAP Quinases/fisiologia , Isoformas de Proteínas , Xenopus laevis
13.
J Cell Biol ; 202(1): 81-95, 2013 Jul 08.
Artigo em Inglês | MEDLINE | ID: mdl-23816619

RESUMO

Meiotic maturation in vertebrate oocytes is an excellent model system for microtubule reorganization during M-phase spindle assembly. Here, we surveyed changes in the pattern of microtubule-interacting proteins upon Xenopus laevis oocyte maturation by quantitative proteomics. We identified the synovial sarcoma X breakpoint protein (SSX2IP) as a novel spindle protein. Using X. laevis egg extracts, we show that SSX2IP accumulated at spindle poles in a Dynein-dependent manner and interacted with the γ-tubulin ring complex (γ-TuRC) and the centriolar satellite protein PCM-1. Immunodepletion of SSX2IP impeded γ-TuRC loading onto centrosomes. This led to reduced microtubule nucleation and spindle assembly failure. In rapidly dividing blastomeres of medaka (Oryzias latipes) and in somatic cells, SSX2IP knockdown caused fragmentation of pericentriolar material and chromosome segregation errors. We characterize SSX2IP as a novel centrosome maturation and maintenance factor that is expressed at the onset of vertebrate development. It preserves centrosome integrity and faithful mitosis during the rapid cleavage division of blastomeres and in somatic cells.


Assuntos
Centríolos/metabolismo , Centrossomo/metabolismo , Proteínas de Neoplasias/metabolismo , Proteínas Repressoras/metabolismo , Proteínas de Xenopus/metabolismo , Xenopus laevis/metabolismo , Animais , Blastômeros/metabolismo , Blastômeros/patologia , Centríolos/genética , Segregação de Cromossomos , Cromossomos/genética , Cromossomos/metabolismo , Embrião não Mamífero/citologia , Embrião não Mamífero/metabolismo , Técnicas de Silenciamento de Genes , Mitose , Proteínas de Neoplasias/genética , Oócitos/metabolismo , Oryzias/embriologia , Oryzias/genética , Oryzias/metabolismo , Proteômica , Proteínas Repressoras/genética , Fuso Acromático/genética , Fuso Acromático/metabolismo , Imagem com Lapso de Tempo , Tubulina (Proteína)/genética , Tubulina (Proteína)/metabolismo , Xenopus laevis/genética
14.
Hypertension ; 61(6): 1177-83, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23509077

RESUMO

Heart failure has an increasing contribution to cardiovascular disease burden and is governed by the myocardial remodeling process. The contribution of Wnt signaling to cardiac remodeling has recently drawn significant attention. Here, we report that upregulation of Dapper-1 in a transgenic mouse model activates the canonical/ß-catenin-dependent Wnt pathway through dishevelled-2. These mice exhibited increased heart weight/tibia length ratio, myocyte cross-sectional area, and upregulation of hypertrophic marker genes compared with wild-type mice. Furthermore, impairment of left ventricular systolic and diastolic function was observed in all indicating features of myocardial remodeling. Depletion of Dapper-1 and dishevelled-2 in cardiomyocytes demonstrated that Dapper-1 functions upstream of dishevelled-2 and that activity of both Dapper-1 and dishevelled-2 is essential for activating canonical Wnt signaling. Moreover, Dapper-1 depletion alleviated Wnt3a- and phenylephrine-induced cardiomyocyte hypertrophy. These observations provide evidence that Dapper-1-mediated activation of canonical Wnt signaling is necessary and sufficient to induce cardiomyocyte hypertrophy. Inhibition of this pathway may thus serve as a novel therapeutic strategy for alleviating cardiac hypertrophy.


Assuntos
Cardiomegalia/genética , Peptídeos e Proteínas de Sinalização Intracelular/genética , Miócitos Cardíacos/efeitos dos fármacos , RNA Mensageiro/genética , Regulação para Cima , Remodelação Ventricular/genética , Via de Sinalização Wnt/genética , Animais , Cardiomegalia/metabolismo , Cardiomegalia/fisiopatologia , Modelos Animais de Doenças , Peptídeos e Proteínas de Sinalização Intracelular/biossíntese , Camundongos , Camundongos Transgênicos , Miocárdio/metabolismo , Miocárdio/patologia , Miócitos Cardíacos/metabolismo , Miócitos Cardíacos/patologia , Proteínas de Ligação a RNA , Ratos
15.
Basic Res Cardiol ; 108(2): 339, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23455426

RESUMO

The heart's rhythm is initiated and regulated by a group of specialized cells in the sinoatrial node (SAN), the primary pacemaker of the heart. Abnormalities in the development of the SAN can result in irregular heart rates (arrhythmias). Although several of the critical genes important for SAN formation have been identified, our understanding of the transcriptional network controlling SAN development remains at a relatively early stage. The homeodomain transcription factor Shox2 is involved in the specification and patterning of the SAN. While the Shox2 knockout in mice results in embryonic lethality due to severe cardiac defects including improper SAN development, Shox2 knockdown in zebrafish causes a reduced heart rate (bradycardia). In order to gain deeper insight into molecular pathways involving Shox2, we compared gene expression levels in right atria of wildtype and Shox2 (-/-) hearts using microarray experiments and identified the LIM homeodomain transcription factor Islet1 (Isl1) as one of its putative target genes. The downregulation of Isl1 expression in Shox2 (-/-) hearts was confirmed and the affected region narrowed down to the SAN by whole-mount in situ hybridization. Using luciferase reporter assays and EMSA studies, we identified two specific SHOX2 binding sites within intron 2 of the ISL1 locus. We also provide functional evidence for Isl1 as a transcriptional target of Shox2 by rescuing the Shox2-mediated bradycardia phenotype with Isl1 using zebrafish as a model system. Our findings demonstrate a novel epistatic relationship between Shox2 and Isl1 in the heart with important developmental consequences for SAN formation and heart beat.


Assuntos
Bradicardia/genética , Regulação da Expressão Gênica/fisiologia , Proteínas de Homeodomínio/genética , Proteínas com Homeodomínio LIM/metabolismo , Fatores de Transcrição/metabolismo , Animais , Bradicardia/metabolismo , Bradicardia/fisiopatologia , Células Cultivadas , Ensaio de Desvio de Mobilidade Eletroforética , Redes Reguladoras de Genes , Imuno-Histoquímica , Hibridização In Situ , Camundongos , Análise em Microsséries , Reação em Cadeia da Polimerase em Tempo Real , Nó Sinoatrial/fisiologia , Transcrição Gênica , Peixe-Zebra
16.
Chem Biol ; 19(11): 1423-36, 2012 Nov 21.
Artigo em Inglês | MEDLINE | ID: mdl-23177197

RESUMO

Regulatory Smads (R-Smads), Smad1/5/8 and Smad2/3, are the central mediators of TGFß and BMP signaling pathways. Here, we screened indirubin derivatives, known kinase inhibitors, and observed strong interference with BMP signaling. We found that indirubin derivative E738 inhibited both TGFß and BMP pathways through ubiquitin-proteasome-mediated depletion of total R-Smad pools, although phospho-R-Smad levels were initially stabilized by GSK3ß and cyclin-dependent kinase inhibition. E738 also enhanced p38 and JNK phosphorylation, involved in Smad-independent TGFß/BMP signaling. Additionally, using a small siRNA screen, we showed that depletion of ubiquitin proteases USP9x and USP34 significantly reduced total R-Smad levels, mimicking E738 treatment. In fact, both USP9x and USP34 levels were significantly reduced in E738-treated cells. Our findings not only describe the complex activity profile of the indirubin derivative E738, but also reveal a mechanism for controlling TGFß/BMP signaling, the control of R-Smad protein levels through deubiquitination.


Assuntos
Proteínas Morfogenéticas Ósseas/metabolismo , Inibidores de Proteínas Quinases/farmacologia , Proteínas Smad Reguladas por Receptor/metabolismo , Fator de Crescimento Transformador beta/metabolismo , Ubiquitina/metabolismo , Animais , Embrião não Mamífero , Fibroblastos/efeitos dos fármacos , Regulação da Expressão Gênica no Desenvolvimento/efeitos dos fármacos , Células HeLa/efeitos dos fármacos , Humanos , Indóis/química , Indóis/farmacologia , Células Jurkat/efeitos dos fármacos , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Fosforilação/efeitos dos fármacos , RNA Interferente Pequeno , Transdução de Sinais/efeitos dos fármacos , Ubiquitina Tiolesterase/metabolismo , Xenopus/embriologia , Xenopus/genética
17.
J Cell Biol ; 198(4): 695-709, 2012 Aug 20.
Artigo em Inglês | MEDLINE | ID: mdl-22908314

RESUMO

Wnt-11/planar cell polarity signaling polarizes mesodermal cells undergoing convergent extension during Xenopus laevis gastrulation. These shape changes associated with lateral intercalation behavior require a dynamic modulation of cell adhesion. In this paper, we report that Wnt-11/frizzled-7 (Fz7) controls cell adhesion by forming separate adhesion-modulating complexes (AMCs) with the paraxial protocadherin (PAPC; denoted as AMCP) and C-cadherin (denoted as AMCC) via distinct Fz7 interaction domains. When PAPC was part of a Wnt-11-Fz7 complex, its Dynamin1- and clathrin-dependent internalization was blocked. This membrane stabilization of AMCP (Fz7/PAPC) by Wnt-11 prevented C-cadherin clustering, resulting in reduced cell adhesion and modified cell sorting activity. Importantly, Wnt-11 did not influence C-cadherin internalization; instead, it promoted the formation of AMCC (Fz7/Cadherin), which competed with cis-dimerization of C-cadherin. Because PAPC and C-cadherin did not directly interact and did not form a joint complex with Fz7, we suggest that Wnt-11 triggers the formation of two distinct complexes, AMCC and AMCP, that act in parallel to reduce cell adhesion by hampering lateral clustering of C-cadherin.


Assuntos
Caderinas/metabolismo , Adesão Celular/fisiologia , Receptores Acoplados a Proteínas G/fisiologia , Proteínas Wnt/fisiologia , Proteínas de Xenopus/metabolismo , Proteínas de Xenopus/fisiologia , Animais , Caderinas/antagonistas & inibidores , Caderinas/genética , Regulação para Baixo/genética , Regulação para Baixo/fisiologia , Embrião não Mamífero/citologia , Embrião não Mamífero/fisiologia , Feminino , Gastrulação/fisiologia , Complexos Multiproteicos/metabolismo , Oócitos/citologia , Oócitos/fisiologia , Multimerização Proteica , Protocaderinas , Receptores Acoplados a Proteínas G/genética , Proteínas Wnt/genética , Proteínas de Xenopus/antagonistas & inibidores , Proteínas de Xenopus/genética , Xenopus laevis
18.
EMBO Rep ; 13(2): 129-34, 2012 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-22193776

RESUMO

Xenopus paraxial protocadherin (PAPC) regulates cadherin-mediated cell adhesion and promotes the planar cell polarity (PCP) pathway. Here we report that PAPC functions in the Xenopus gastrula as an inhibitor of the Wnt/ß-catenin pathway. The intracellular domain of PAPC interacts with casein kinase 2 beta (CK2ß), which is part of the CK2 holoenzyme. The CK2α/ß complex stimulates Wnt/ß-catenin signalling, and the physical interaction of CK2ß with PAPC antagonizes this activity. By this mechanism, PAPC restricts the expression of Wnt target genes during gastrulation. These experiments identify a novel function of protocadherins as regulators of the Wnt pathway.


Assuntos
Caderinas/metabolismo , Caseína Quinase II/metabolismo , Via de Sinalização Wnt , Proteínas de Xenopus/metabolismo , Xenopus/metabolismo , Animais , Caderinas/química , Modelos Biológicos , Ligação Proteica , Estrutura Terciária de Proteína , Protocaderinas , Regulação para Cima , Xenopus/embriologia , Proteínas de Xenopus/química
19.
Wiley Interdiscip Rev Dev Biol ; 1(2): 294-300, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23801443

RESUMO

Tissue border formation is an important process that prevents mixing of cells during embryonic development. The establishment of tissue borders is not a trivial problem, particularly in early embryos when cells and tissues are not fully differentiated. An example of an early tissue separation process is the formation of Brachet's cleft in Xenopus. During early gastrulation, this morphologically visible cleft separates mesendoderm and ectoderm. Over the last decade, it was recognized that morphogenetic processes, including tissue separation, can be experimentally uncoupled from embryonic patterning events. In this study, we summarize the data explaining the regulation of Brachet's cleft and introduce the experimental arsenal that was used for this analysis. The formation of Brachet's cleft involves the activity of transcription factors, cell adhesion molecules, and signaling modules, which act in a complex regulatory network. According to the current state of knowledge, Rho signaling seems to be the central player during this process. The mechanisms that regulate Rho during tissue separation and the experimental approaches to monitor Rho activity are discussed.


Assuntos
Gastrulação , Transdução de Sinais , Xenopus/embriologia , Animais , Xenopus/metabolismo , Proteínas de Xenopus/genética , Proteínas de Xenopus/metabolismo , Proteínas rho de Ligação ao GTP/genética , Proteínas rho de Ligação ao GTP/metabolismo
20.
Dev Genes Evol ; 221(1): 29-41, 2011 May.
Artigo em Inglês | MEDLINE | ID: mdl-21509535

RESUMO

Cerberus-related molecules are well-known Wnt, Nodal, and BMP inhibitors that have been implicated in different processes including anterior­posterior patterning and left­right asymmetry. In both mouse and frog, two Cerberus-related genes have been isolated, mCer-1 and mCer-2, and Xcer and Xcoco, respectively. Until now, little is known about the mechanisms involved in their transcriptional regulation. Here, we report a heterologous analysis of the mouse Cerberus-1 gene upstream regulatory regions, responsible for its expression in the visceral endodermal cells. Our analysis showed that the consensus sequences for a TATA, CAAT, or GC boxes were absent but a TGTGG sequence was present at position -172 to -168 bp, relative to the ATG. Using a series of deletion constructs and transient expression in Xenopus embryos, we found that a fragment of 1.4 kb of Cer-1 promoter sequence could reproduce the endogenous expression pattern of Xenopus cerberus. A 0.7-kb mcer-1 upstream region was able to drive reporter expression to the involuting mesendodermal cells, while further deletions abolished reporter gene expression. Our results suggest that although no sequence similarity was found between mouse and Xenopus cerberus cis-regulatory regions, the signaling cascades regulating cerberus expression, during gastrulation, is conserved.


Assuntos
Proteínas/genética , Sequências Reguladoras de Ácido Nucleico/genética , Proteínas de Xenopus/genética , Xenopus , Animais , Sequência de Bases , Proteínas Morfogenéticas Ósseas/genética , Clonagem Molecular/métodos , Citocinas , Gastrulação/fisiologia , Regulação da Expressão Gênica no Desenvolvimento , Camundongos , Proteína Nodal/genética , Proteínas Wnt/genética , Xenopus/embriologia , Xenopus/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA