Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
1.
MicroPubl Biol ; 20242024.
Artigo em Inglês | MEDLINE | ID: mdl-38601902

RESUMO

TGF-ß signals are important for proliferation, differentiation, and cell fate determination during embryonic development and tissue homeostasis in adults. Drosophila Activin/TGF-ß signals are transduced intracellularly when its transcription factor dSmad2 (also called Smad on X or Smox) is C-terminally phosphorylated by pathway receptors. Recently, it has been shown that receptor-activated dSmad2 undergoes bulk degradation, however, the mechanism of how this occurs is unknown. Here we investigated if two putative linker phosphorylation sites are involved in dSmad2 degradation. We demonstrate that degradation of activated-dSmad2 occurs independently of threonine phosphorylation at linker sites 252 and 277. We also show that dSmad2 degradation is not carried out by cellular proteasomes.

2.
Sci Rep ; 7: 46923, 2017 12 22.
Artigo em Inglês | MEDLINE | ID: mdl-29271405

RESUMO

This corrects the article DOI: 10.1038/srep32269.

3.
PLoS One ; 12(11): e0187721, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-29107946

RESUMO

The regulatory networks governing morphogenesis of a pleomorphic fungus, Candida albicans are extremely complex and remain to be completely elucidated. This study investigated the function of C. albicans yeast casein kinase 2 (CaYck2p). The yck2Δ/yck2Δ strain displayed constitutive pseudohyphae in both yeast and hyphal growth conditions, and formed enhanced biofilm under non-biofilm inducing condition. This finding was further supported by gene expression analysis of the yck2Δ/yck2Δ strain which showed significant upregulation of UME6, a key transcriptional regulator of hyphal transition and biofilm formation, and cell wall protein genes ALS3, HWP1, and SUN41, all of which are associated with morphogenesis and biofilm architecture. The yck2Δ/yck2Δ strain was hypersensitive to cell wall damaging agents and had increased compensatory chitin deposition in the cell wall accompanied by an upregulation of the expression of the chitin synthase genes, CHS2, CHS3, and CHS8. Absence of CaYck2p also affected fungal-host interaction; the yck2Δ/yck2Δ strain had significantly reduced ability to damage host cells. However, the yck2Δ/yck2Δ strain had wild-type susceptibility to cyclosporine and FK506, suggesting that CaYck2p functions independently from the Ca+/calcineurin pathway. Thus, in C. albicans, Yck2p is a multifunctional kinase that governs morphogenesis, biofilm formation, cell wall integrity, and host cell interactions.


Assuntos
Biofilmes/crescimento & desenvolvimento , Candida albicans/enzimologia , Caseína Quinase II/metabolismo , Parede Celular , Candida albicans/crescimento & desenvolvimento , Quitina/biossíntese , Reação em Cadeia da Polimerase em Tempo Real
4.
Sci Rep ; 6: 32269, 2016 08 31.
Artigo em Inglês | MEDLINE | ID: mdl-27578171

RESUMO

Bone morphogenetic proteins (BMPs) are growth factors that provide essential signals for normal embryonic development and adult tissue homeostasis. A key step in initiating BMP signaling is ligand induced phosphorylation of receptor Smads (R-Smads) by type I receptor kinases, while linker phosphorylation of R-Smads has been shown to cause BMP signal termination. Here we present data demonstrating that the phosphatase Dullard is involved in dephosphorylating the Drosophila R-Smad, Mad, and is integral in controlling BMP signal duration. We show that a hypomorphic Dullard allele or Dullard knockdown leads to increased Mad phosphorylation levels, while Dullard overexpression resulted in reduced Mad phosphorylations. Co-immunoprecipitation binding assays demonstrate phosphorylated Mad and Dullard physically interact, while mutation of Dullard's phosphatase domain still allowed Mad-Dullard interactions but abolished its ability to regulate Mad phosphorylations. Finally, we demonstrate that linker and C-terminally phosphorylated Mad can be regulated by one of two terminating mechanisms, degradation by proteasomes or dephosphorylation by the phosphatase Dullard.


Assuntos
Proteínas Morfogenéticas Ósseas/fisiologia , Proteínas de Ligação a DNA/metabolismo , Proteínas de Drosophila/metabolismo , Fosfoproteínas Fosfatases/metabolismo , Processamento de Proteína Pós-Traducional , Fatores de Transcrição/metabolismo , Animais , Linhagem Celular , Drosophila melanogaster , Discos Imaginais/enzimologia , Masculino , Fosforilação , Proteólise , Transdução de Sinais
5.
Sci Rep ; 4: 6927, 2014 Nov 07.
Artigo em Inglês | MEDLINE | ID: mdl-25377173

RESUMO

The BMP ligand Dpp, operates as a long range morphogen to control many important functions during Drosophila development from tissue patterning to growth. The BMP signal is transduced intracellularly via C-terminal phosphorylation of the BMP transcription factor Mad, which forms an activity gradient in developing embryonic tissues. Here we show that Cyclin dependent kinase 8 and Shaggy phosphorylate three Mad linker serines. We demonstrate that linker phosphorylations control the peak intensity and range of the BMP signal across rapidly developing embryonic tissues. Shaggy knockdown broadened the range of the BMP-activity gradient and increased high threshold target gene expression in the early embryo, while expression of a Mad linker mutant in the wing disc resulted in enhanced levels of C-terminally phosphorylated Mad, a 30% increase in wing tissue, and elevated BMP target genes. In conclusion, our results describe how Mad linker phosphorylations work to control the peak intensity and range of the BMP signal in rapidly developing Drosophila tissues.


Assuntos
Proteínas Morfogenéticas Ósseas/genética , Proteínas de Ligação a DNA/genética , Proteínas de Drosophila/genética , Drosophila melanogaster/genética , Regulação da Expressão Gênica no Desenvolvimento , Morfogênese/genética , Fatores de Transcrição/genética , Asas de Animais/metabolismo , Animais , Proteínas Morfogenéticas Ósseas/metabolismo , Quinase 8 Dependente de Ciclina/genética , Quinase 8 Dependente de Ciclina/metabolismo , Proteínas de Ligação a DNA/metabolismo , Proteínas de Drosophila/antagonistas & inibidores , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/embriologia , Drosophila melanogaster/metabolismo , Embrião não Mamífero , Quinase 3 da Glicogênio Sintase/antagonistas & inibidores , Quinase 3 da Glicogênio Sintase/genética , Quinase 3 da Glicogênio Sintase/metabolismo , Fosforilação , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Transdução de Sinais , Fatores de Transcrição/metabolismo , Asas de Animais/citologia , Asas de Animais/embriologia
6.
Int Forum Allergy Rhinol ; 4(11): 893-900, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25196914

RESUMO

BACKGROUND: Airway secretions contain endogenous antimicrobial factors (AMFs) that contribute to the innate host defense of the respiratory tract. Antibacterial peptides as well as host-derived lipids including cholesteryl esters have been detected in maxillary lavage fluid. Sterol O-acyltransferase 1 (SOAT1) is a key enzyme in cholesteryl ester production. The purpose of this study is to determine if such intrinsic microbicidal molecules are acutely expressed within sinus tissue and to compare levels of expression between patients with and without chronic rhinosinusitis (CRS). METHODS: Sinus tissue was obtained from subjects with (24) and without (9) a history of CRS. Six CRS patients had nasal polyposis (CRSwNP). Immunofluorescence staining for human neutrophil peptide (HNP) was done as a marker for inflammation. Real-time polymerase chain reaction (RT-PCR) following RNA extraction was used to quantify the expression of SOAT-1, the epithelial beta-defensins (HBD2 and HBD3), and the cathelicidin LL37 with ribosomal protein, large, P0 (RPLP0) as the housekeeping gene. RESULTS: Immunofluorescence showed significant increase in HNP staining in CRS patients without nasal polyposis (CRSsNP) vs non-CRS specimens (p = 0.010), in agreement with clinical inflammation status. SOAT1 messenger RNA (mRNA) expression was also upregulated in CRSsNP compared to non-CRS (p = 0.041) and CRSwNP (p = 0.005) patients, whereas increases for HBD2 and HBD3 were less prominent. LL37 was either absent or expressed at very low levels in all samples. CONCLUSION: Increased biosynthesis of SOAT1, a key enzyme for antimicrobial cholesteryl ester production, was observed in the sinus tissue of CRSsNP patients but not in CRSwNP patients. This further supports the novel concept of lipid-mediated innate mucosal defense and delineates CRS with and without nasal polyposis as distinct subtypes.


Assuntos
Peptídeos Catiônicos Antimicrobianos/metabolismo , Imunidade Inata/fisiologia , Rinite/imunologia , Sinusite/imunologia , Doença Crônica , Expressão Gênica , Humanos , Mucosa/imunologia , Mucosa/metabolismo , Mucosa Nasal/imunologia , Mucosa Nasal/metabolismo , Neutrófilos/imunologia , Seios Paranasais/imunologia , Seios Paranasais/metabolismo , Proteínas Ribossômicas/metabolismo , Esterol O-Aciltransferase/metabolismo , Regulação para Cima , alfa-Defensinas/metabolismo
7.
Sci Signal ; 4(194): ra68, 2011 Oct 11.
Artigo em Inglês | MEDLINE | ID: mdl-21990430

RESUMO

Bone morphogenetic proteins (BMPs) and Wnts are growth factors that provide essential patterning signals for cell proliferation and differentiation. Here, we describe a molecular mechanism by which the phosphorylation state of the Drosophila transcription factor Mad determines its ability to transduce either BMP or Wingless (Wg) signals. Previously, Mad was thought to function in gene transcription only when phosphorylated by BMP receptors. We found that the unphosphorylated form of Mad was required for canonical Wg signaling by interacting with the Pangolin-Armadillo transcriptional complex. Phosphorylation of the carboxyl terminus of Mad by BMP receptor directed Mad toward BMP signaling, thereby preventing Mad from functioning in the Wg pathway. The results show that Mad has distinct signal transduction roles in the BMP and Wnt pathways depending on its phosphorylation state.


Assuntos
Proteínas Morfogenéticas Ósseas/metabolismo , Proteínas de Ligação a DNA/metabolismo , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/metabolismo , Modelos Biológicos , Transdução de Sinais/fisiologia , Fatores de Transcrição/metabolismo , Proteína Wnt1/metabolismo , Animais , Linhagem Celular , Clonagem Molecular , Drosophila melanogaster/anatomia & histologia , Inativação Gênica , Quinase 3 da Glicogênio Sintase/metabolismo , Humanos , Imunoprecipitação , Larva/anatomia & histologia , Larva/metabolismo , Fosforilação , Poliubiquitina/metabolismo , Proteínas Repressoras/metabolismo , Asas de Animais/anatomia & histologia
8.
PLoS One ; 5(4): e10383, 2010 Apr 28.
Artigo em Inglês | MEDLINE | ID: mdl-20442782

RESUMO

In the vertebrates, the BMP/Smad1 and TGF-beta/Smad2 signaling pathways execute antagonistic functions in different contexts of development. The differentiation of specific structures results from the balance between these two pathways. For example, the gastrula organizer/node of the vertebrates requires a region of low Smad1 and high Smad2 signaling. In Drosophila, Mad regulates tissue determination and growth in the wing, but the function of dSmad2 in wing patterning is largely unknown. In this study, we used an RNAi loss-of-function approach to investigate dSmad2 signaling during wing development. RNAi-mediated knockdown of dSmad2 caused formation of extra vein tissue, with phenotypes similar to those seen in Dpp/Mad gain-of-function. Clonal analyses revealed that the normal function of dSmad2 is to inhibit the response of wing intervein cells to the extracellular Dpp morphogen gradient that specifies vein formation, as measured by expression of the activated phospho-Mad protein. The effect of dSmad2 depletion in promoting vein differentiation was dependent on Medea, the co-factor shared by Mad and dSmad2. Furthermore, double RNAi experiments showed that Mad is epistatic to dSmad2. In other words, depletion of Smad2 had no effect in Mad-deficient wings. Our results demonstrate a novel role for dSmad2 in opposing Mad-mediated vein formation in the wing. We propose that the main function of dActivin/dSmad2 in Drosophila wing development is to antagonize Dpp/Mad signaling. Possible molecular mechanisms for the opposition between dSmad2 and Mad signaling are discussed.


Assuntos
Proteínas de Ligação a DNA/antagonistas & inibidores , Proteínas de Drosophila/antagonistas & inibidores , Proteínas de Drosophila/fisiologia , Transdução de Sinais/fisiologia , Proteína Smad2/fisiologia , Fatores de Transcrição/antagonistas & inibidores , Veias/crescimento & desenvolvimento , Asas de Animais/crescimento & desenvolvimento , Ativinas/metabolismo , Animais , Drosophila/crescimento & desenvolvimento , Proteínas de Drosophila/metabolismo , Feminino , Proteínas Smad Reguladas por Receptor , Asas de Animais/irrigação sanguínea
9.
Cytokine Growth Factor Rev ; 20(5-6): 357-65, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19896409

RESUMO

BMPs pattern the dorsal-ventral axis of vertebrate embryos. Smad1/5/8 transduces the BMP signal, and receives phosphorylation inputs from both MAPK and GSK3. Phosphorylation of Smad1 by MAPK and GSK3 result in its polyubiquitination and transport to the centrosome where it is degraded by the proteasome. These linker phosphorylations inhibit BMP/Smad1signaling by shortening its duration. Wnt, which negatively regulates GSK3 activity, prolongs the BMP/Smad1 signal. Remarkably, linker-phosphorylated Smad1 has been shown to be inherited asymmetrically during cell division. Drosophila contains a single Smad1/5/8 homologue, Mad, and is stabilized by phosphorylation-resistant mutations at GSK3 sites, causing Wingless-like effects. We summarize here the significance of linker-phosphorylated Smad1/Mad in relation to signal intensity and duration, and how this integrates the Wnt and BMP pathways during cell differentiation.


Assuntos
Proteínas Morfogenéticas Ósseas/fisiologia , Proteínas de Ligação a DNA/fisiologia , Proteínas de Drosophila/fisiologia , Proteínas Smad/fisiologia , Fatores de Transcrição/fisiologia , Proteínas Wnt/fisiologia , Animais , Proteínas Morfogenéticas Ósseas/genética , Proteínas de Ligação a DNA/genética , Drosophila/genética , Proteínas de Drosophila/genética , Humanos , Modelos Biológicos , Transdução de Sinais/genética , Transdução de Sinais/fisiologia , Proteínas Smad/genética , Proteína Smad1/genética , Proteína Smad1/fisiologia , Proteína Smad5/genética , Proteína Smad5/fisiologia , Proteína Smad8/genética , Proteína Smad8/fisiologia , Fatores de Transcrição/genética , Vertebrados/genética , Proteínas Wnt/genética
10.
PLoS One ; 4(8): e6543, 2009 Aug 06.
Artigo em Inglês | MEDLINE | ID: mdl-19657393

RESUMO

A key question in developmental biology is how growth factor signals are integrated to generate pattern. In this study we investigated the integration of the Drosophila BMP and Wingless/GSK3 signaling pathways via phosphorylations of the transcription factor Mad. Wingless was found to regulate the phosphorylation of Mad by GSK3 in vivo. In epistatic experiments, the effects of Wingless on wing disc molecular markers (senseless, distalless and vestigial) were suppressed by depletion of Mad with RNAi. Wingless overexpression phenotypes, such as formation of ectopic wing margins, were induced by Mad GSK3 phosphorylation-resistant mutant protein. Unexpectedly, we found that Mad phosphorylation by GSK3 and MAPK occurred in segmental patterns. Mad depletion or overexpression produced Wingless-like embryonic segmentation phenotypes. In Xenopus embryos, segmental border formation was disrupted by Smad8 depletion. The results show that Mad is required for Wingless signaling and for the integration of gradients of positional information.


Assuntos
Padronização Corporal , Proteínas de Ligação a DNA/fisiologia , Proteínas de Drosophila/metabolismo , Proteínas de Drosophila/fisiologia , Drosophila/embriologia , Transdução de Sinais/fisiologia , Fatores de Transcrição/fisiologia , Asas de Animais/embriologia , Proteína Wnt1/metabolismo , Animais , Proteínas de Ligação a DNA/genética , Proteínas de Drosophila/genética , Mutação , Fenótipo , Interferência de RNA , Fatores de Transcrição/genética
11.
Curr Opin Genet Dev ; 18(4): 304-10, 2008 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-18590818

RESUMO

The intensity of the BMP signal is determined by cell surface receptors that phosphorylate Smad1/5/8 at the C-terminus. In addition to this BMP-activated phosphorylation, recent studies have shown that sequential phosphorylations by MAPK and GSK3 kinases can negatively regulate the activity of the pSmad1Cter signal. These phosphorylations in the linker region cause Smad1 to be transported to the centrosomal region, polyubiquitinylated and degraded by the proteasomal machinery. In Xenopus embryos, Wnt signals, which regulate GSK3, induce ectoderm to adopt an epidermal fate, and this Wnt effect requires an active BMP-Smad1/5/8 signaling pathway. These findings have profound implications for understanding how dorsal-ventral and anterior-posterior patterning are seamlessly integrated in the early embryonic morphogenetic field.


Assuntos
Padronização Corporal/fisiologia , Proteína Smad1/fisiologia , Proteína Smad5/fisiologia , Proteína Smad8/fisiologia , Sequência de Aminoácidos , Animais , Padronização Corporal/genética , Embrião não Mamífero , Humanos , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Proteínas Quinases Ativadas por Mitógeno/fisiologia , Modelos Biológicos , Dados de Sequência Molecular , Fosforilação , Transdução de Sinais , Proteína Smad1/antagonistas & inibidores , Proteína Smad1/genética , Proteína Smad1/metabolismo , Proteína Smad5/genética , Proteína Smad8/genética , Xenopus/embriologia , Xenopus/genética
12.
Proc Natl Acad Sci U S A ; 105(22): 7732-7, 2008 Jun 03.
Artigo em Inglês | MEDLINE | ID: mdl-18511557

RESUMO

Mitotic cell division ensures that two daughter somatic cells inherit identical genetic material. Previous work has shown that signaling by the Smad1 transcription factor is terminated by polyubiquitinylation and proteasomal degradation after essential phosphorylations by MAPK and glycogen synthase kinase 3 (GSK3). Here, we show that, unexpectedly, proteins specifically targeted for proteasomal degradation are inherited preferentially by one mitotic daughter during somatic cell division. Experiments with dividing human embryonic stem cells and other mammalian cultured cell lines demonstrated that in many supposedly equal mitoses the segregation of proteins destined for degradation (Smad1 phosphorylated by MAPK and GSK3, phospho-beta-catenin, and total polyubiquitinylated proteins) was asymmetric. Transport of pSmad1 targeted for degradation to the centrosome required functional microtubules. In vivo, an antibody specific for Mad phosphorylated by MAPK showed that this antigen was associated preferentially with one of the two centrosomes in Drosophila embryos at cellular blastoderm stage. We propose that this remarkable cellular property may be explained by the asymmetric inheritance of peripheral centrosomal proteins when centrioles separate and migrate to opposite poles of the cell, so that one mitotic daughter remains pristine. We conclude that many mitotic divisions are unequal, unlike what was previously thought.


Assuntos
Mitose , Poliubiquitina/metabolismo , Complexo de Endopeptidases do Proteassoma/metabolismo , Proteínas/metabolismo , Ubiquitinação , Animais , Blastoderma/citologia , Blastoderma/metabolismo , Proteínas Morfogenéticas Ósseas/metabolismo , Células COS/metabolismo , Linhagem Celular , Centrossomo/metabolismo , Chlorocebus aethiops , Drosophila/citologia , Drosophila/embriologia , Drosophila/metabolismo , Células-Tronco Embrionárias/citologia , Células-Tronco Embrionárias/metabolismo , Quinase 3 da Glicogênio Sintase/metabolismo , Humanos , Microtúbulos/metabolismo , Quinases de Proteína Quinase Ativadas por Mitógeno/metabolismo , Modelos Biológicos , Fosforilação , Transporte Proteico , Proteína Smad1/metabolismo , beta Catenina/metabolismo
13.
Cell ; 131(5): 980-93, 2007 Nov 30.
Artigo em Inglês | MEDLINE | ID: mdl-18045539

RESUMO

BMP receptors determine the intensity of BMP signals via Smad1 C-terminal phosphorylations. Here we show that a finely controlled cell biological pathway terminates this activity. The duration of the activated pSmad1(Cter) signal was regulated by sequential Smad1 linker region phosphorylations at conserved MAPK and GSK3 sites required for its polyubiquitinylation and transport to the centrosome. Proteasomal degradation of activated Smad1 and total polyubiquitinated proteins took place in the centrosome. Inhibitors of the Erk, p38, and JNK MAPKs, as well as GSK3 inhibitors, prolonged the duration of a pulse of BMP7. Wnt signaling decreased pSmad1(GSK3) antigen levels and redistributed it from the centrosome to cytoplasmic LRP6 signalosomes. In Xenopus embryos, it was found that Wnts induce epidermis and that this required an active BMP-Smad pathway. Epistatic experiments suggested that the dorsoventral (BMP) and anteroposterior (Wnt/GSK3) patterning gradients are integrated at the level of Smad1 phosphorylations during embryonic pattern formation.


Assuntos
Proteínas Morfogenéticas Ósseas/metabolismo , Quinase 3 da Glicogênio Sintase/fisiologia , Transdução de Sinais/fisiologia , Proteína Smad1/metabolismo , Proteínas Wnt/fisiologia , Sequência de Aminoácidos , Animais , Proteínas Morfogenéticas Ósseas/fisiologia , Células COS , Células Cultivadas , Centrossomo/metabolismo , Chlorocebus aethiops , Embrião não Mamífero , Quinase 3 da Glicogênio Sintase/metabolismo , Humanos , Camundongos , Quinases de Proteína Quinase Ativadas por Mitógeno/fisiologia , Modelos Biológicos , Dados de Sequência Molecular , Fosforilação , Processamento de Proteína Pós-Traducional , Proteína Smad1/fisiologia , Fatores de Tempo , Ubiquitinação/fisiologia , Xenopus/embriologia
14.
Dev Genes Evol ; 216(3): 144-51, 2006 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-16411117

RESUMO

The endosome/lysosome system plays key roles in embryonic development, but difficulties posed by inaccessible mammalian embryos have hampered detailed studies. The accessible, transparent embryos of Danio rerio, together with the genetic and experimental approaches possible with this organism, provide many advantages over rodents. In mammals, mannose 6-phosphate receptors (MPRs) target acid hydrolases to endosomes and lysosomes, but nothing is known of acid hydrolase targeting in zebrafish. Here, we describe the sequence of the zebrafish cation-dependent MPR (CD-MPR) and cation-independent MPR (CI-MPR), and compare them with their mammalian orthologs. We show that all residues critical for mannose 6-phosphate (M6P) recognition are present in the extracellular domains of the zebrafish receptors, and that trafficking signals in the cytoplasmic tails are also conserved. This suggests that the teleost receptors possess M6P binding sites with properties similar to those of mammalian MPRs, and that targeting of lysosomal enzymes by MPRs represents an ancient pathway in vertebrate cell biology. We also determined the expression patterns of the CD-MPR and CI-MPR during embryonic development in zebrafish. Both genes are expressed from the one-cell stage through to the hatching period. In early embryos, expression is ubiquitous, but in later stages, expression of both receptors is restricted to the anterior region of the embryo, covering the forebrain, midbrain and hindbrain. The expression patterns suggest time- and tissue-specific functions for the receptors, with particular evidence for roles in neural development. Our study establishes zebrafish as a novel, genetically tractable model for in vivo studies of MPR function and lysosome biogenesis.


Assuntos
Receptor IGF Tipo 2/genética , Peixe-Zebra/genética , Sequência de Aminoácidos , Animais , Sítios de Ligação , Metabolismo dos Carboidratos , Cátions/metabolismo , Bovinos , Galinhas/genética , Clonagem Molecular , Sequência Conservada , Estruturas Citoplasmáticas/metabolismo , Embrião não Mamífero , Desenvolvimento Embrionário , Expressão Gênica , Humanos , Fator de Crescimento Insulin-Like II/metabolismo , Camundongos , Dados de Sequência Molecular , Neurônios/metabolismo , Gambás/genética , Filogenia , Ornitorrinco/genética , Homologia de Sequência de Aminoácidos , Transdução de Sinais , Distribuição Tecidual
15.
Int J Dev Biol ; 48(10): 1131-40, 2004 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-15602699

RESUMO

The insulin-like growth factor (IGF) signalling pathway has been highly conserved in animal evolution and, in mammals and Xenopus, plays a key role in embryonic growth and development, with the IGF-1 receptor (IGF-1R) being a crucial regulator of the signalling cascade. Here we report the first functional role for the IGF pathway in zebrafish. Expression of mRNA coding for a dominant negative IGF-1R resulted in embryos that were small in size compared to controls and had disrupted head and CNS development. At its most extreme, this phenotype was characterized by a complete loss of head and eye structures, an absence of notochord and the presence of abnormal somites. In contrast, up-regulation of IGF signalling following injection of IGF-1 mRNA, resulted in a greatly expanded development of anterior structures at the expense of trunk and tail. IGF-1R knockdown caused a significant decrease in the expression of Otx2, Rx3, FGF8, Pax6.2 and Ntl, while excess IGF signalling expanded Otx2 expression in presumptive forebrain tissue and widened the Ntl expression domain in the developing notochord. The observation that IGF-1R knockdown reduced expression of two key organizer genes (chordin and goosecoid) suggests that IGF signalling plays a role in regulating zebrafish organizer activity. This is supported by the expression of IGF-1, IGF-2 and IGF-1R in shield-stage zebrafish embryos and the demonstration that IGF signalling influences expression of BMP2b, a gene that plays an important role in zebrafish pattern formation. Our data is consistent with a common pathway for integration of IGF, FGF8 and anti-BMPs in early vertebrate development.


Assuntos
Embrião de Mamíferos/metabolismo , Embrião não Mamífero , Regulação da Expressão Gênica no Desenvolvimento , Somatomedinas/metabolismo , Animais , Western Blotting , Padronização Corporal , Sistema Nervoso Central/metabolismo , Técnicas de Cultura , Desenvolvimento Embrionário , Genes Dominantes , Glicoproteínas/metabolismo , Proteína Goosecoid , Proteínas de Homeodomínio/metabolismo , Hibridização In Situ , Fator de Crescimento Insulin-Like I/metabolismo , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Camundongos , Notocorda/metabolismo , Fatores de Transcrição Otx , Fenótipo , Fosforilação , Plasmídeos/metabolismo , Prosencéfalo/metabolismo , RNA Mensageiro/metabolismo , Receptor IGF Tipo 1/metabolismo , Proteínas Repressoras/metabolismo , Transdução de Sinais , Fatores de Transcrição/metabolismo , Regulação para Cima , Peixe-Zebra , Proteínas de Peixe-Zebra
16.
Genes Dev ; 17(24): 3023-8, 2003 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-14701872

RESUMO

How do very diverse signaling pathways induce neural differentiation in Xenopus? Anti-BMP (Chordin), FGF8, and IGF2 signals are integrated in the embryo via the regulation of Smad1 phosphorylation. Neural induction results from the combined inhibition of BMP receptor serine/threonine kinases and activation of receptor tyrosine kinases that signal through MAPK and phosphorylate Smad1 in the linker region, further inhibiting Smad1 transcriptional activity. This hard-wired molecular mechanism at the level of the Smad1 transcription factor may help explain the opposing activities of IGF, FGF, and BMP signals not only in neural induction, but also in other aspects of vertebrate development.


Assuntos
Proteínas Morfogenéticas Ósseas/metabolismo , Proteínas de Ligação a DNA/metabolismo , Indução Embrionária , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Sistema Nervoso/embriologia , Transdução de Sinais , Transativadores/metabolismo , Animais , Receptores de Proteínas Morfogenéticas Ósseas , Embrião não Mamífero , Fator 8 de Crescimento de Fibroblasto , Fatores de Crescimento de Fibroblastos/farmacologia , Fator de Crescimento Insulin-Like II/farmacologia , Camundongos , Células NIH 3T3 , Proteínas de Neoplasias/farmacologia , Fosforilação , Receptores de Fatores de Crescimento/antagonistas & inibidores , Proteínas Smad , Proteína Smad1 , Ativação Transcricional , Proteínas de Xenopus , Xenopus laevis
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA