Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 92
Filtrar
1.
Sci Rep ; 8(1): 9542, 2018 06 22.
Artigo em Inglês | MEDLINE | ID: mdl-29934521

RESUMO

Smad ubiquitin regulatory factor 1 (SMURF1) is a HECT-type E3 ubiquitin ligase that plays a critical role in vertebrate development by regulating planar cell polarity (PCP) signaling and convergent extension (CE). Here we show that SMURF1 is involved in mammalian heart development. We find that SMURF1 is highly expressed in outflow tract cushion mesenchyme and Smurf1-/- mouse embryos show delayed outflow tract septation. SMURF1 is expressed in smooth muscle cells of the coronary arteries and great vessels. Thickness of the aortic smooth muscle cell layer is reduced in Smurf1-/- mouse embryos. We show that SMURF1 is a negative regulator of cardiomyogenesis and a positive regulator of smooth muscle cell and cardiac fibroblast differentiation, indicating that SMURF1 is important for cell-type specification during heart development. Finally, we provide evidence that SMURF1 localizes at the primary cilium where it may regulate bone morphogenetic protein (BMP) signaling, which controls the initial phase of cardiomyocyte differentiation. In summary, our results demonstrate that SMURF1 is a critical regulator of outflow tract septation and cell-type specification during heart development, and that these effects may in part be mediated via control of cilium-associated BMP signaling.


Assuntos
Coração/crescimento & desenvolvimento , Miócitos Cardíacos/citologia , Ubiquitina-Proteína Ligases/metabolismo , Animais , Aorta/citologia , Diferenciação Celular , Linhagem Celular , Regulação da Expressão Gênica no Desenvolvimento , Técnicas de Silenciamento de Genes , Coração/fisiologia , Humanos , Camundongos , Miócitos de Músculo Liso/metabolismo , Transdução de Sinais , Ubiquitina-Proteína Ligases/deficiência , Ubiquitina-Proteína Ligases/genética
2.
Oncogene ; 35(32): 4256-68, 2016 08 11.
Artigo em Inglês | MEDLINE | ID: mdl-26725322

RESUMO

Postnatal proliferation of cerebellar granule neuron precursors (CGNPs), proposed cells of origin for the SHH-associated subgroup of medulloblastoma, is driven by Sonic hedgehog (Shh) and insulin-like growth factor (IGF) in the developing cerebellum. Shh induces the oncogene Yes-associated protein (YAP), which drives IGF2 expression in CGNPs and mouse Shh-associated medulloblastomas. To determine how IGF2 expression is regulated downstream of YAP, we carried out an unbiased screen for transcriptional regulators bound to IGF2 promoters. We report that Y-box binding protein-1 (YB-1), an onco-protein regulating transcription and translation, binds to IGF2 promoter P3. We observed that YB-1 is upregulated across human medulloblastoma subclasses as well as in other varieties of pediatric brain tumors. Utilizing the cerebellar progenitor model for the Shh subgroup of medulloblastoma in mice, we show for the first time that YB-1 is induced by Shh in CGNPs. Its expression is YAP-dependent and it is required for IGF2 expression in CGNPs. Finally, both gain-of function and loss-of-function experiments reveal that YB-1 activity is required for sustaining CGNP and medulloblastoma cell (MBC) proliferation. Collectively, our findings describe a novel role for YB-1 in driving proliferation in the developing cerebellum and MBCs and they identify the SHH:YAP:YB1:IGF2 axis as a powerful target for therapeutic intervention in medulloblastomas.


Assuntos
Neoplasias Cerebelares/patologia , Cerebelo/patologia , Proteínas Hedgehog/metabolismo , Meduloblastoma/patologia , Células-Tronco Neurais/patologia , Proteína 1 de Ligação a Y-Box/metabolismo , Animais , Linhagem Celular Tumoral , Proliferação de Células , Neoplasias Cerebelares/genética , Neoplasias Cerebelares/metabolismo , Regulação Neoplásica da Expressão Gênica , Fator de Crescimento Insulin-Like II/genética , Meduloblastoma/genética , Meduloblastoma/metabolismo , Camundongos , Transdução de Sinais
3.
Oncogene ; 31(48): 5019-28, 2012 Nov 29.
Artigo em Inglês | MEDLINE | ID: mdl-22286767

RESUMO

RB(+/-) individuals develop retinoblastoma and, subsequently, many other tumors. The Rb relatives p107 and p130 protect the tumor-resistant Rb(-/-) mouse retina. Determining the mechanism underlying this tumor suppressor function may expose novel strategies to block Rb pathway cancers. p107/p130 are best known as E2f inhibitors, but here we implicate E2f-independent Cdk2 inhibition as the critical p107 tumor suppressor function in vivo. Like p107 loss, deleting p27 or inactivating its Cdk inhibitor (CKI) function (p27(CK-)) cooperated with Rb loss to induce retinoblastoma. Genetically, p107 behaved like a CKI because inactivating Rb and one allele each of p27 and p107 was tumorigenic. Although Rb loss induced canonical E2f targets, unexpectedly p107 loss did not further induce these genes, but instead caused post-transcriptional Skp2 induction and Cdk2 activation. Strikingly, Cdk2 activity correlated with tumor penetrance across all the retinoblastoma models. Therefore, Rb restrains E2f, but p107 inhibits cross talk to Cdk. While removing either E2f2 or E2f3 genes had little effect, removing only one E2f1 allele blocked tumorigenesis. More importantly, exposing retinoblastoma-prone fetuses to small molecule inhibitors of E2f (HLM006474) or Cdk (R547) for merely 1 week dramatically inhibited subsequent tumorigenesis in adult mice. Protection was achieved without disrupting normal proliferation. Thus, exquisite sensitivity of the cell-of-origin to E2f and Cdk activity can be exploited to prevent Rb pathway-induced cancer in vivo without perturbing normal cell division. These data suggest that E2f inhibitors, never before tested in vivo, or CKIs, largely disappointing as therapeutics, may be effective preventive agents.


Assuntos
Quinase 2 Dependente de Ciclina/fisiologia , Fator de Transcrição E2F1/fisiologia , Retinoblastoma/fisiopatologia , Animais , Modelos Animais de Doenças , Camundongos , Camundongos Knockout , Retinoblastoma/patologia , Proteína do Retinoblastoma/genética , Proteína do Retinoblastoma/fisiologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa
4.
Oncogene ; 29(16): 2368-80, 2010 Apr 22.
Artigo em Inglês | MEDLINE | ID: mdl-20101206

RESUMO

Matrix remodeling, degradation, inflammation and invasion liberate peptide fragments that can subsequently interact with cells in an attachment-independent manner. Such 'soluble' matrix components, including collagens, fibronectin and laminin, induced Smad activation (termed crosstalk signaling), which follows a similar chronological sequence and R-Smad specificity as induced by transforming growth factor (TGF)-beta1. Smad4 nuclear translocation occurred in response to collagen binding, indicating downstream signal propagation. TGF-beta scavenging antibody affected only TGF-beta1, but not crosstalk-induced responses. TGF-beta type II receptor mutation (DR26Delta25), which is deficient in TGF-beta type I receptor recruitment to the ligand, induced a heterotetramer signaling complex, and propagated Smad2 activation only through collagen induction and not TGF-beta signaling. Consequentially, TGF-beta ligand participation is not required for crosstalk signaling. This signaling requires a functional integrin beta1 receptor as showed by RNA interference. Co-immunoprecipitation (co-IP) and fluorescent microscopy indicate the involvement of focal adhesion kinase (FAK) and Src activity in collagen-induced signal propagation, and suggest a membrane signaling complex formation that includes both TGF-beta receptors and integrins. The related gene expressional responses are distinct from that evoked by TGF-beta1, supporting its separate function. This signaling mechanism expands and partially explains TGF-beta receptor dynamics and consequential signaling diversity-related gene expressional plasticity.


Assuntos
Matriz Extracelular/fisiologia , Receptores de Fatores de Crescimento Transformadores beta/fisiologia , Transdução de Sinais/fisiologia , Linhagem Celular Tumoral , Colágeno/farmacologia , Humanos , Integrina beta1/fisiologia , Modelos Biológicos , Fosforilação , Receptor Cross-Talk/fisiologia , Receptores de Colágeno/fisiologia , Proteínas Smad/metabolismo
5.
Genet Mol Res ; 8(4): 1331-43, 2009 Nov 03.
Artigo em Inglês | MEDLINE | ID: mdl-19937589

RESUMO

Chondroitin-4-sulfotransferase-1(C4ST-1)/carbohydrate sulfotransferase 11 (CHST11) is a Golgi-bound enzyme involved in the biosynthesis of the glycosaminoglycan chondroitin sulfate. The sulfation pattern of chondroitin is tightly regulated during development, injury and disease, with the temporal and spatial expression of chondroitin sulfotransferase genes believed to be a crucial determinant of the fine balance of chondroitin sulfation. We have previously identified mouse C4st-1 as a target gene of ligands of the TGFbeta superfamily of growth factors, which could positively regulate C4st-1 expression in a number of cell types. We have also shown that a gene trap loss-of-function mutation in C4st-1 leads to severe skeletal abnormalities during mouse embryogenesis. In addition, we described a highly specific temporal and spatial expression pattern of C4st-1 during mouse embryogenesis. However, the transcriptional regulatory mechanisms that control C4st-1 gene expression remain unexplored. In order to gain knowledge on the transcriptional regulation of C4ST-1, we used a bioinformatical approach to identify conserved putative long-range cis-regulatory modules in a region of 120 kb spanning the 5' end of the C4ST-1 gene. Luciferase reporter assays in human HEK293T and mouse NmuMG cells identified a functional C4ST-1 promoter, as well as a number of cis-regulatory modules able to positively and negatively regulate C4ST-1 expression. Moreover, we identified TGFbeta- responsive regulatory modules that can function in a cell type-specific fashion. Taken together, our results identify TGFbeta-dependent and -independent cis-regulatory modules of the C4ST-1 gene.


Assuntos
Sulfatos de Condroitina/metabolismo , Sequências Reguladoras de Ácido Nucleico , Sulfotransferases/metabolismo , Fator de Crescimento Transformador beta/fisiologia , Animais , Sequência de Bases , Linhagem Celular , Primers do DNA , Humanos , Camundongos
6.
J Biomed Mater Res A ; 68(4): 704-16, 2004 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-14986325

RESUMO

We sought to develop bioactive hydrogels to facilitate arterial healing, e.g., after balloon angioplasty. Toward this end, we developed a new class of proteolytically sensitive, biologically active polyethylene glycol (PEG)-peptide hydrogels that can be formed in situ to temporarily protect the arterial injury from blood contact. Furthermore, we incorporated endothelial cell-specific biological signals with the goal of enhancing arterial reendothelialization. Here we demonstrate efficient endothelial cell anchorage and activation on PEG hydrogel matrices modified by conjugation with both the cell adhesive peptide motif RGD and an engineered variant of vascular endothelial growth factor (VEGF). By crosslinking peptide sequences for cleavage by MMP-2 into the polymer backbone, the hydrogels became sensitive to proteolytic degradation by cell-derived matrix metalloproteinases (MMPs). Analysis of molecular hallmarks associated with endothelial cell activation by VEGF-RGD hydrogel matrices revealed a 70% increase in production of the latent MMP-2 zymogen compared with PEG-peptide hydrogels lacking VEGF. By additional provision of transforming growth factor beta1 (TGF-beta1) within the PEG-peptide hydrogel, conversion of the latent MMP zymogen into its active form was demonstrated. As a result of MMP-2 activation, strongly enhanced hydrogel degradation by activated endothelial cells was observed. Our data illustrate the critical importance of growth factor activities for remodeling of synthetic biomaterials into native tissue, as it is desired in many applications of regenerative medicine. Functionalized PEG-peptide hydrogels could help restore the native vessel wall and improve the performance of angioplasty procedures.


Assuntos
Artérias/lesões , Materiais Biocompatíveis/metabolismo , Hidrogéis/metabolismo , Metaloproteinase 2 da Matriz/metabolismo , Fator A de Crescimento do Endotélio Vascular/metabolismo , Adesão Celular/fisiologia , Células Endoteliais/fisiologia , Humanos , Fatores de Tempo
7.
Mol Cell Biol ; 23(20): 7230-42, 2003 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-14517293

RESUMO

Myostatin, a transforming growth factor beta (TGF-beta) family member, is a potent negative regulator of skeletal muscle growth. In this study we characterized the myostatin signal transduction pathway and examined its effect on bone morphogenetic protein (BMP)-induced adipogenesis. While both BMP7 and BMP2 activated transcription from the BMP-responsive I-BRE-Lux reporter and induced adipogenic differentiation, myostatin inhibited BMP7- but not BMP2-mediated responses. To dissect the molecular mechanism of this antagonism, we characterized the myostatin signal transduction pathway. We showed that myostatin binds the type II Ser/Thr kinase receptor. ActRIIB, and then partners with a type I receptor, either activin receptor-like kinase 4 (ALK4 or ActRIB) or ALK5 (TbetaRI), to induce phosphorylation of Smad2/Smad3 and activate a TGF-beta-like signaling pathway. We demonstrated that myostatin prevents BMP7 but not BMP2 binding to its receptors and that BMP7-induced heteromeric receptor complex formation is blocked by competition for the common type II receptor, ActRIIB. Thus, our results reveal a strikingly specific antagonism of BMP7-mediated processes by myostatin and suggest that myostatin is an important regulator of adipogenesis.


Assuntos
Adipócitos/metabolismo , Transdução de Sinais , Fator de Crescimento Transformador beta/metabolismo , Animais , Proteína Morfogenética Óssea 2 , Proteína Morfogenética Óssea 7 , Proteínas Morfogenéticas Ósseas/metabolismo , Células COS , Diferenciação Celular , Linhagem Celular , Proteínas de Ligação a DNA/metabolismo , Genes Reporter , Homeostase , Humanos , Camundongos , Camundongos Endogâmicos C3H , Modelos Biológicos , Miostatina , Células NIH 3T3 , Fosforilação , Reação em Cadeia da Polimerase , Ligação Proteica , RNA/metabolismo , Interferência de RNA , Proteína Smad2 , Fatores de Tempo , Transativadores/metabolismo , Transcrição Gênica , Ativação Transcricional , Transfecção
8.
Genes Dev ; 15(21): 2822-36, 2001 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-11691834

RESUMO

Smad proteins mediate transforming growth factor-beta (TGF-beta) signaling to regulate cell growth and differentiation. SnoN is an important negative regulator of TGF-beta signaling that functions to maintain the repressed state of TGF-beta target genes in the absence of ligand. On TGF-beta stimulation, Smad3 and Smad2 translocate into the nucleus and induce a rapid degradation of SnoN, allowing activation of TGF-beta target genes. We show that Smad2- or Smad3-induced degradation of SnoN requires the ubiquitin-dependent proteasome and can be mediated by the anaphase-promoting complex (APC) and the UbcH5 family of ubiquitin-conjugating enzymes. Smad3 and to a lesser extent, Smad2, interact with both the APC and SnoN, resulting in the recruitment of the APC to SnoN and subsequent ubiquitination of SnoN in a destruction box (D box)-dependent manner. In addition to the D box, efficient ubiquitination and degradation of SnoN also requires the Smad3 binding site in SnoN as well as key lysine residues necessary for ubiquitin attachment. Mutation of either the Smad3 binding site or lysine residues results in stabilization of SnoN and in enhanced antagonism of TGF-beta signaling. Our studies elucidate an important mechanism and pathway for the degradation of SnoN and more importantly, reveal a novel role of the APC in the regulation of TGF-beta signaling.


Assuntos
Proteínas de Ligação a DNA/química , Proteínas de Ligação a DNA/metabolismo , Proteínas Proto-Oncogênicas/química , Proteínas Proto-Oncogênicas/metabolismo , Transativadores/química , Transativadores/metabolismo , Ubiquitina/metabolismo , Sequência de Aminoácidos , Sítios de Ligação , Western Blotting , Divisão Celular , Linhagem Celular , Relação Dose-Resposta a Droga , Deleção de Genes , Humanos , Peptídeos e Proteínas de Sinalização Intracelular , Ligantes , Lisina/química , Modelos Biológicos , Modelos Genéticos , Dados de Sequência Molecular , Testes de Precipitina , Ligação Proteica , Estrutura Terciária de Proteína , Retroviridae/genética , Transdução de Sinais , Proteína Smad2 , Proteína Smad3 , Fatores de Tempo , Transfecção , Fator de Crescimento Transformador beta/metabolismo , Fator de Crescimento Transformador beta1
9.
Nat Cell Biol ; 3(6): 587-95, 2001 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-11389444

RESUMO

The receptor-regulated Smad proteins are essential intracellular mediators of signal transduction by the transforming growth factor-beta (TGF-beta) superfamily of growth factors and are also important as regulators of gene transcription. Here we describe a new role for TGF-beta-regulated Smad2 and Smad3 as components of a ubiquitin ligase complex. We show that in the presence of TGF-beta signalling, Smad2 interacts through its proline-rich PPXY motif with the tryptophan-rich WW domains of Smurf2, a recently identified E3 ubiquitin ligases. TGF-beta also induces the association of Smurf2 with the transcriptional co-repressor SnoN and we show that Smad2 can function to mediate this interaction. This allows Smurf2 HECT domain to target SnoN for ubiquitin-mediated degradation by the proteasome. Thus, stimulation by TGF-beta can induce the assembly of a Smad2-Smurf2 ubiquitin ligase complex that functions to target substrates for degradation.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Ligases/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Transativadores/metabolismo , Fator de Crescimento Transformador beta/metabolismo , Motivos de Aminoácidos , Animais , Células Cultivadas , Humanos , Peptídeos e Proteínas de Sinalização Intracelular , Vison , Fosforilação , Proteína Smad2 , Ubiquitina-Proteína Ligases , Ubiquitinas/metabolismo
10.
Genes Dev ; 15(10): 1257-71, 2001 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-11358869

RESUMO

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


Assuntos
Padronização Corporal , Proteínas de Ligação a DNA/genética , Gástrula/metabolismo , Transdução de Sinais , Fatores de Transcrição/genética , Ativinas , Animais , Proteínas de Ligação a DNA/metabolismo , Fatores de Transcrição Forkhead , Regulação da Expressão Gênica no Desenvolvimento , Hibridização In Situ , Inibinas/genética , Inibinas/metabolismo , Camundongos , Camundongos Endogâmicos ICR , Camundongos Knockout , Mutagênese , Proteína Nodal , Reação em Cadeia da Polimerase , Deleção de Sequência , Proteína Smad2 , Transativadores/genética , Transativadores/metabolismo , Fatores de Transcrição/metabolismo , Fator de Crescimento Transformador beta/genética , Fator de Crescimento Transformador beta/metabolismo
11.
Nucleic Acids Res ; 29(3): 732-42, 2001 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-11160896

RESUMO

An emerging theme in transforming growth factor-ss (TGF-ss) signalling is the association of the Smad proteins with diverse groups of transcriptional regulatory proteins. Several Smad cofactors have been identified to date but the diversity of TGF-ss effects on gene transcription suggests that interactions with other co-regulators must occur. In these studies we addressed the possible interaction of Smad proteins with the myocyte enhancer-binding factor 2 (MEF2) transcriptional regulators. Our studies indicate that Smad2 and 4 (Smad2/4) complexes cooperate with MEF2 regulatory proteins in a GAL4-based one-hybrid reporter gene assay. We have also observed in vivo interactions between Smad2 and MEF2A using co-immunoprecipitation assays. This interaction is confirmed by glutathione S:-transferase pull-down analysis. Immunofluorescence studies in C2C12 myotubes show that Smad2 and MEF2A co-localise in the nucleus of multinuclear myotubes during differentiation. Interestingly, phospho-acceptor site mutations of MEF2 that render it unresponsive to p38 MAP kinase signalling abrogate the cooperativity with the Smads suggesting that p38 MAP Kinase-catalysed phosphorylation of MEF2 is a prerequisite for the Smad-MEF2 interaction. Thus, the association between Smad2 and MEF2A may subserve a physical link between TGF-ss signalling and a diverse array of genes controlled by the MEF2 cis element.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Proteínas de Ligação a DNA/fisiologia , Transativadores/fisiologia , Fatores de Transcrição/metabolismo , Animais , Sítios de Ligação , Células COS , Linhagem Celular , Núcleo Celular/metabolismo , Citoplasma/metabolismo , DNA Recombinante , Proteínas de Ligação a DNA/genética , Fatores de Transcrição MEF2 , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Fatores de Regulação Miogênica/genética , Fatores de Regulação Miogênica/metabolismo , Fosforilação , Plasmídeos/genética , Ligação Proteica , Receptores de Fatores de Crescimento Transformadores beta/genética , Receptores de Fatores de Crescimento Transformadores beta/fisiologia , Proteína Smad2 , Transativadores/genética , Transativadores/metabolismo , Fatores de Transcrição/genética , Ativação Transcricional/efeitos dos fármacos , Transfecção , Fator de Crescimento Transformador beta/farmacologia , Proteínas Quinases p38 Ativadas por Mitógeno
12.
Mol Cell Biol ; 20(24): 9346-55, 2000 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-11094085

RESUMO

Smad proteins are effector molecules that transmit signals from the receptors for the transforming growth factor beta (TGF-beta) superfamily to the nucleus; of the Smad proteins, Smad2 and Smad4 are essential components for mouse early embryogenesis. We demonstrated that Hgs, a FYVE domain protein, binds to Smad2 in its C-terminal half and cooperates with another FYVE domain protein, the Smad anchor for receptor activation (SARA), to stimulate activin receptor-mediated signaling through efficient recruitment of Smad2 to the receptor. Furthermore, a LacZ knock-in allele of the C-terminal half-deletion mutant of mouse Hgs was created by gene targeting. The introduced mutation causes an embryonic lethality between embryonic days 8.5 and 10.5. Mutant cells showed significantly decreased responses to stimulation with activin and TGF-beta. These findings suggest that the two FYVE domain proteins, Hgs and SARA, are prerequisites for receptor-mediated activation of Smad2.


Assuntos
Proteínas de Transporte/metabolismo , Proteínas de Ligação a DNA/metabolismo , Fosfoproteínas/metabolismo , Receptores de Fatores de Crescimento/metabolismo , Transdução de Sinais , Transativadores/metabolismo , Receptores de Ativinas , Ativinas , Animais , Proteínas de Transporte/genética , Diferenciação Celular , Linhagem Celular , Quimera/genética , Quimera/imunologia , Quimera/metabolismo , Proteínas de Ligação a DNA/genética , Embrião de Mamíferos/anatomia & histologia , Embrião de Mamíferos/fisiologia , Complexos Endossomais de Distribuição Requeridos para Transporte , Marcação de Genes , Genes Reporter/efeitos dos fármacos , Inibinas/farmacologia , Substâncias Macromoleculares , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos/anatomia & histologia , Camundongos Transgênicos/genética , Camundongos Transgênicos/metabolismo , Fosfoproteínas/genética , Fosforilação , Testes de Precipitina , Proteína Smad2 , Proteína Smad3 , Transativadores/genética , Fator de Crescimento Transformador beta/farmacologia
13.
J Biol Chem ; 275(43): 33205-8, 2000 Oct 27.
Artigo em Inglês | MEDLINE | ID: mdl-10973944

RESUMO

Activation of transforming growth factor-beta (TGF-beta) receptors triggers phosphorylation of Smad2 and Smad3. After binding to Smad4, the complex enters the nucleus and interacts with other transcription factors to activate gene transcription. Unlike other Smads, Smad7 inhibits phosphorylation of Smad2 and Smad3, and its transcription is induced by TGF-beta, suggesting a negative feedback loop. Here, we show that TFE3 and Smad3 synergistically mediate TGF-beta-induced transcription from the Smad7 promoter by binding to an E-box and two adjacent Smad binding elements (SBEs), respectively. A precise 3-base pair spacer between one SBE and the E-box is essential. Previously, we showed that a similar arrangement between a SBE and an E-box of an element is essential for TGF-beta-dependent transcription of the plasminogen activator inhibitor-1 gene (PAI-1) and that TGF-beta-induced phosphorylation of Smad3 triggers its association with TFE3. Thus, TFE3-Smad3 response elements may represent a common target for TGF-beta-induced gene expression.


Assuntos
Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/farmacologia , Transativadores/genética , Transativadores/farmacologia , Fatores de Transcrição/farmacologia , Transcrição Gênica/efeitos dos fármacos , Fator de Crescimento Transformador beta/farmacologia , Fatores de Transcrição de Zíper de Leucina e Hélice-Alça-Hélix Básicos , Linhagem Celular , Proteínas de Ligação a DNA/metabolismo , Sinergismo Farmacológico , Humanos , Regiões Promotoras Genéticas , Proteína Smad3 , Proteína Smad7 , Transativadores/metabolismo , Fatores de Transcrição/metabolismo
15.
Cytokine Growth Factor Rev ; 11(1-2): 5-13, 2000.
Artigo em Inglês | MEDLINE | ID: mdl-10708948

RESUMO

Transforming growth factor-beta superfamily member signals are conveyed through cell-surface serine/threonine kinase receptors to the intracellular mediators known as Smads. Activation of Smads causes their translocation from the cytoplasm to the nucleus where they function to control gene expression. In this review we will focus on proteins that modulate Smad activity, including SARA, for Smad Anchor for Receptor Activation, which functions during the initiation of signalling and on components of the ubiquitin-proteasome pathway, such as Smurf1, which can negatively regulate Smad signalling. In addition, we will summarize recent findings on the role of Smads as transcriptional co-modulators.


Assuntos
Proteínas de Transporte/metabolismo , Cisteína Endopeptidases/metabolismo , Proteínas de Ligação a DNA/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular , Complexos Multienzimáticos/metabolismo , Serina Endopeptidases , Transativadores/metabolismo , Fator de Crescimento Transformador beta/metabolismo , Animais , Sítios de Ligação , Proteínas de Transporte/química , Núcleo Celular/metabolismo , Proteínas de Ligação a DNA/química , Humanos , Fosfoproteínas/metabolismo , Complexo de Endopeptidases do Proteassoma , Transdução de Sinais , Proteínas Smad , Proteína Smad2 , Proteína Smad4 , Proteína Smad5 , Transativadores/química , Transcrição Gênica , Ubiquitinas/metabolismo
16.
Curr Opin Cell Biol ; 12(2): 235-43, 2000 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-10712925

RESUMO

The Smad signalling pathway is critical for transmitting transforming growth factor-beta (TGF-beta) superfamily signals from the cell surface to the nucleus. In the nucleus, Smads regulate transcriptional responses by recruiting co-activators and co-repressors to a wide array of DNA-binding partners. Thus, Smads function as transcriptional co-modulators to regulate TGFbeta-dependent gene expression.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular , Serina Endopeptidases , Transdução de Sinais , Transativadores/metabolismo , Transcrição Gênica , Fator de Crescimento Transformador beta/metabolismo , Animais , Proteínas de Transporte/metabolismo , Núcleo Celular/fisiologia , Drosophila , Humanos , Proteína Smad4 , Ubiquitinas/metabolismo
18.
J Cell Sci ; 113 Pt 2: 269-78, 2000 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-10633078

RESUMO

Bone morphogenetic protein (BMP)-2 and hepatocyte growth factor (HGF) exert antagonistic effects on renal collecting duct formation during embryogenesis. A current model proposes HGF inhibits BMP-2 signaling at the level of Smad1 in a common target cell. Here, we show that BMP-2 and HGF control collecting duct formation via parallel pathways. We examined the interactions between BMP-2 and HGF in the mIMCD-3 model of collecting duct morphogenesis. During tubule formation, HGF rescued the inhibitory effects of BMP-2 and of a constitutive active form of the BMP-2 receptor, ALK3, stably expressed in mIMCD-3 cells. To determine whether the effect of HGF occurs through known mediators which act downstream of the BMP-2/ALK3 complex, we examined the effect of HGF on BMP-2-induced Smad1 phosphorylation, Smad1/Smad4 complex formation, and Smad1 nuclear translocation. Neither HGF nor other receptor tyrosine kinase ligands (EGF, FGF-4) induced phosphorylation of endogenous Smad1 in mIMCD-3 cells or in Mv1Lu, MC3T3-E1 or P19 cells. Furthermore, none of these ligands blocked induction of the BMP-responsive promoter, Tlx2. Thus, HGF overcomes the inhibitory effects of BMP-2 on collecting duct morphogenesis without interrupting any of the known signaling events in the BMP-2 dependent Smad1 signaling pathway. We conclude that BMP-2/ALK3 and HGF function to control parallel pathways downstream of their respective cell surface receptors. Integration of these signals likely occurs at the level of transcriptional or post-transcriptional events.


Assuntos
Proteínas Morfogenéticas Ósseas/metabolismo , Fator de Crescimento de Hepatócito/metabolismo , Túbulos Renais Coletores/embriologia , Túbulos Renais Coletores/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Receptores de Superfície Celular/metabolismo , Receptores de Fatores de Crescimento , Fator de Crescimento Transformador beta , Receptores de Ativinas , Animais , Proteína Morfogenética Óssea 2 , Receptores de Proteínas Morfogenéticas Ósseas , Proteínas Morfogenéticas Ósseas/farmacologia , Linhagem Celular , Proteínas de Ligação a DNA/metabolismo , Fator de Crescimento de Hepatócito/farmacologia , Túbulos Renais Coletores/citologia , Modelos Biológicos , Morfogênese/efeitos dos fármacos , Fosforilação , Proteínas Serina-Treonina Quinases/genética , Receptores de Superfície Celular/genética , Transdução de Sinais , Proteínas Smad , Transativadores/metabolismo , Transfecção
19.
J Biol Chem ; 275(3): 2063-70, 2000 Jan 21.
Artigo em Inglês | MEDLINE | ID: mdl-10636910

RESUMO

The tumor suppressor gene Smad4 has been proposed to be a common mediator of transforming growth factor beta (TGFbeta)-related signaling pathways. We investigated the role of Smad4 in TGFbeta-related pathways by targeted disruption of its locus in murine cell lines. TGFbeta responses, including growth arrest, induction of the endogenous PAI-1 gene, and other extracellular matrix components, were normal in Smad4-deficient fibroblasts. Assembly of a TGFbeta-induced DNA-binding complex on one of two regulatory regions in the human plasminogen activator inhibitor (PAI)-1 promoter did not require Smad4 but was, instead, dependent on a TFE-3 binding site. In contrast, Smad4 was required for activation of the Xenopus Mix.2 promoter in response to TGFbeta/activin. Smad4 was also involved in the regulation of the Msx homeobox protein family members in response to bone morphogenetic protein (BMP). Interestingly, the expression of the endogenous Msx-2 was reduced, whereas that of Msx-3 was activated in differentiating Smad4(-/-) ES cells relative to wild-type cells. Moreover, reporter assays of the Msx-2 promoter revealed an absolute requirement for Smad4 in fibroblasts and ES cells for activation. Our results indicate that Smad4 is dispensable for critical TGFbeta-induced responses but is required for others in murine fibroblasts. We have identified transcriptional targets for Smad4 in the BMP signaling pathway, which may contribute to the genetic defect observed in the Smad4-deficient embryos.


Assuntos
Proteínas de Ligação a DNA/fisiologia , Transativadores/fisiologia , Fator de Crescimento Transformador beta/metabolismo , Proteínas de Xenopus , Ativinas , Animais , Ligação Competitiva , Quimera/genética , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Relação Dose-Resposta a Droga , Matriz Extracelular/metabolismo , Fibroblastos , Regulação da Expressão Gênica , Glucose-6-Fosfato Isomerase/metabolismo , Proteínas de Homeodomínio/metabolismo , Humanos , Inibinas/farmacologia , Camundongos , Camundongos Endogâmicos C57BL , Fatores de Crescimento Neural , Regiões Promotoras Genéticas , Recombinação Genética , Sequências Reguladoras de Ácido Nucleico , Transdução de Sinais , Proteínas Smad , Proteína Smad4 , Fatores de Tempo , Transativadores/genética , Transfecção
20.
Mol Cell ; 6(6): 1365-75, 2000 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-11163210

RESUMO

Ubiquitin-mediated proteolysis regulates the activity of diverse receptor systems. Here, we identify Smurf2, a C2-WW-HECT domain ubiquitin ligase and show that Smurf2 associates constitutively with Smad7. Smurf2 is nuclear, but binding to Smad7 induces export and recruitment to the activated TGF beta receptor, where it causes degradation of receptors and Smad7 via proteasomal and lysosomal pathways. IFN gamma, which stimulates expression of Smad7, induces Smad7-Smurf2 complex formation and increases TGF beta receptor turnover, which is stabilized by blocking Smad7 or Smurf2 expression. Furthermore, Smad7 mutants that interfere with recruitment of Smurf2 to the receptors are compromised in their inhibitory activity. These studies thus define Smad7 as an adaptor in an E3 ubiquitin-ligase complex that targets the TGF beta receptor for degradation.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Ligases/metabolismo , Proteínas Nucleares/metabolismo , Receptores de Fatores de Crescimento Transformadores beta/metabolismo , Transativadores/metabolismo , Animais , Linhagem Celular , Cisteína Endopeptidases/metabolismo , Proteínas de Ligação a DNA/genética , Regulação para Baixo/efeitos dos fármacos , Regulação da Expressão Gênica/efeitos dos fármacos , Immunoblotting , Interferon gama/farmacologia , Ligases/química , Lisossomos/metabolismo , Substâncias Macromoleculares , Modelos Biológicos , Dados de Sequência Molecular , Complexos Multienzimáticos/metabolismo , Mutação , Complexo de Endopeptidases do Proteassoma , Ligação Proteica/efeitos dos fármacos , Estrutura Terciária de Proteína , Transporte Proteico , Proteínas Recombinantes de Fusão , Proteína Smad7 , Transativadores/genética , Transfecção , Ubiquitina-Proteína Ligases
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA