Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 92
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
Sci Rep ; 8(1): 9542, 2018 06 22.
Artículo en Inglés | MEDLINE | ID: mdl-29934521

RESUMEN

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.


Asunto(s)
Corazón/crecimiento & desarrollo , Miocitos Cardíacos/citología , Ubiquitina-Proteína Ligasas/metabolismo , Animales , Aorta/citología , Diferenciación Celular , Línea Celular , Regulación del Desarrollo de la Expresión Génica , Técnicas de Silenciamiento del Gen , Corazón/fisiología , Humanos , Ratones , Miocitos del Músculo Liso/metabolismo , Transducción de Señal , Ubiquitina-Proteína Ligasas/deficiencia , Ubiquitina-Proteína Ligasas/genética
2.
Oncogene ; 35(32): 4256-68, 2016 08 11.
Artículo en Inglés | MEDLINE | ID: mdl-26725322

RESUMEN

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.


Asunto(s)
Neoplasias Cerebelosas/patología , Cerebelo/patología , Proteínas Hedgehog/metabolismo , Meduloblastoma/patología , Células-Madre Neurales/patología , Proteína 1 de Unión a la Caja Y/metabolismo , Animales , Línea Celular Tumoral , Proliferación Celular , Neoplasias Cerebelosas/genética , Neoplasias Cerebelosas/metabolismo , Regulación Neoplásica de la Expresión Génica , Factor II del Crecimiento Similar a la Insulina/genética , Meduloblastoma/genética , Meduloblastoma/metabolismo , Ratones , Transducción de Señal
3.
Oncogene ; 31(48): 5019-28, 2012 Nov 29.
Artículo en Inglés | MEDLINE | ID: mdl-22286767

RESUMEN

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.


Asunto(s)
Quinasa 2 Dependiente de la Ciclina/fisiología , Factor de Transcripción E2F1/fisiología , Retinoblastoma/fisiopatología , Animales , Modelos Animales de Enfermedad , Ratones , Ratones Noqueados , Retinoblastoma/patología , Proteína de Retinoblastoma/genética , Proteína de Retinoblastoma/fisiología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
4.
Oncogene ; 29(16): 2368-80, 2010 Apr 22.
Artículo en Inglés | MEDLINE | ID: mdl-20101206

RESUMEN

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.


Asunto(s)
Matriz Extracelular/fisiología , Receptores de Factores de Crecimiento Transformadores beta/fisiología , Transducción de Señal/fisiología , Línea Celular Tumoral , Colágeno/farmacología , Humanos , Integrina beta1/fisiología , Modelos Biológicos , Fosforilación , Receptor Cross-Talk/fisiología , Receptores de Colágeno/fisiología , Proteínas Smad/metabolismo
5.
Genet Mol Res ; 8(4): 1331-43, 2009 Nov 03.
Artículo en Inglés | MEDLINE | ID: mdl-19937589

RESUMEN

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.


Asunto(s)
Sulfatos de Condroitina/metabolismo , Secuencias Reguladoras de Ácidos Nucleicos , Sulfotransferasas/metabolismo , Factor de Crecimiento Transformador beta/fisiología , Animales , Secuencia de Bases , Línea Celular , Cartilla de ADN , Humanos , Ratones
6.
J Biomed Mater Res A ; 68(4): 704-16, 2004 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-14986325

RESUMEN

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.


Asunto(s)
Arterias/lesiones , Materiales Biocompatibles/metabolismo , Hidrogeles/metabolismo , Metaloproteinasa 2 de la Matriz/metabolismo , Factor A de Crecimiento Endotelial Vascular/metabolismo , Adhesión Celular/fisiología , Células Endoteliales/fisiología , Humanos , Factores de Tiempo
7.
Mol Cell Biol ; 23(20): 7230-42, 2003 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-14517293

RESUMEN

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.


Asunto(s)
Adipocitos/metabolismo , Transducción de Señal , Factor de Crecimiento Transformador beta/metabolismo , Animales , Proteína Morfogenética Ósea 2 , Proteína Morfogenética Ósea 7 , Proteínas Morfogenéticas Óseas/metabolismo , Células COS , Diferenciación Celular , Línea Celular , Proteínas de Unión al ADN/metabolismo , Genes Reporteros , Homeostasis , Humanos , Ratones , Ratones Endogámicos C3H , Modelos Biológicos , Miostatina , Células 3T3 NIH , Fosforilación , Reacción en Cadena de la Polimerasa , Unión Proteica , ARN/metabolismo , Interferencia de ARN , Proteína Smad2 , Factores de Tiempo , Transactivadores/metabolismo , Transcripción Genética , Activación Transcripcional , Transfección
8.
Genes Dev ; 15(21): 2822-36, 2001 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-11691834

RESUMEN

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.


Asunto(s)
Proteínas de Unión al ADN/química , Proteínas de Unión al ADN/metabolismo , Proteínas Proto-Oncogénicas/química , Proteínas Proto-Oncogénicas/metabolismo , Transactivadores/química , Transactivadores/metabolismo , Ubiquitina/metabolismo , Secuencia de Aminoácidos , Sitios de Unión , Western Blotting , División Celular , Línea Celular , Relación Dosis-Respuesta a Droga , Eliminación de Gen , Humanos , Péptidos y Proteínas de Señalización Intracelular , Ligandos , Lisina/química , Modelos Biológicos , Modelos Genéticos , Datos de Secuencia Molecular , Pruebas de Precipitina , Unión Proteica , Estructura Terciaria de Proteína , Retroviridae/genética , Transducción de Señal , Proteína Smad2 , Proteína smad3 , Factores de Tiempo , Transfección , Factor de Crecimiento Transformador beta/metabolismo , Factor de Crecimiento Transformador beta1
9.
Nat Cell Biol ; 3(6): 587-95, 2001 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-11389444

RESUMEN

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.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Ligasas/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Transactivadores/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Secuencias de Aminoácidos , Animales , Células Cultivadas , Humanos , Péptidos y Proteínas de Señalización Intracelular , Visón , Fosforilación , Proteína Smad2 , Ubiquitina-Proteína Ligasas , Ubiquitinas/metabolismo
10.
Genes Dev ; 15(10): 1257-71, 2001 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-11358869

RESUMEN

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.


Asunto(s)
Tipificación del Cuerpo , Proteínas de Unión al ADN/genética , Gástrula/metabolismo , Transducción de Señal , Factores de Transcripción/genética , Activinas , Animales , Proteínas de Unión al ADN/metabolismo , Factores de Transcripción Forkhead , Regulación del Desarrollo de la Expresión Génica , Hibridación in Situ , Inhibinas/genética , Inhibinas/metabolismo , Ratones , Ratones Endogámicos ICR , Ratones Noqueados , Mutagénesis , Proteína Nodal , Reacción en Cadena de la Polimerasa , Eliminación de Secuencia , Proteína Smad2 , Transactivadores/genética , Transactivadores/metabolismo , Factores de Transcripción/metabolismo , Factor de Crecimiento Transformador beta/genética , Factor de Crecimiento Transformador beta/metabolismo
11.
Nucleic Acids Res ; 29(3): 732-42, 2001 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-11160896

RESUMEN

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.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Proteínas de Unión al ADN/fisiología , Transactivadores/fisiología , Factores de Transcripción/metabolismo , Animales , Sitios de Unión , Células COS , Línea Celular , Núcleo Celular/metabolismo , Citoplasma/metabolismo , ADN Recombinante , Proteínas de Unión al ADN/genética , Factores de Transcripción MEF2 , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Factores Reguladores Miogénicos/genética , Factores Reguladores Miogénicos/metabolismo , Fosforilación , Plásmidos/genética , Unión Proteica , Receptores de Factores de Crecimiento Transformadores beta/genética , Receptores de Factores de Crecimiento Transformadores beta/fisiología , Proteína Smad2 , Transactivadores/genética , Transactivadores/metabolismo , Factores de Transcripción/genética , Activación Transcripcional/efectos de los fármacos , Transfección , Factor de Crecimiento Transformador beta/farmacología , Proteínas Quinasas p38 Activadas por Mitógenos
12.
Mol Cell Biol ; 20(24): 9346-55, 2000 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-11094085

RESUMEN

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.


Asunto(s)
Proteínas Portadoras/metabolismo , Proteínas de Unión al ADN/metabolismo , Fosfoproteínas/metabolismo , Receptores de Factores de Crecimiento/metabolismo , Transducción de Señal , Transactivadores/metabolismo , Receptores de Activinas , Activinas , Animales , Proteínas Portadoras/genética , Diferenciación Celular , Línea Celular , Quimera/genética , Quimera/inmunología , Quimera/metabolismo , Proteínas de Unión al ADN/genética , Embrión de Mamíferos/anatomía & histología , Embrión de Mamíferos/fisiología , Complejos de Clasificación Endosomal Requeridos para el Transporte , Marcación de Gen , Genes Reporteros/efectos de los fármacos , Inhibinas/farmacología , Sustancias Macromoleculares , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos/anatomía & histología , Ratones Transgénicos/genética , Ratones Transgénicos/metabolismo , Fosfoproteínas/genética , Fosforilación , Pruebas de Precipitina , Proteína Smad2 , Proteína smad3 , Transactivadores/genética , Factor de Crecimiento Transformador beta/farmacología
13.
J Biol Chem ; 275(43): 33205-8, 2000 Oct 27.
Artículo en Inglés | MEDLINE | ID: mdl-10973944

RESUMEN

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.


Asunto(s)
Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/farmacología , Transactivadores/genética , Transactivadores/farmacología , Factores de Transcripción/farmacología , Transcripción Genética/efectos de los fármacos , Factor de Crecimiento Transformador beta/farmacología , Factores de Transcripción Básicos con Cremalleras de Leucinas y Motivos Hélice-Asa-Hélice , Línea Celular , Proteínas de Unión al ADN/metabolismo , Sinergismo Farmacológico , Humanos , Regiones Promotoras Genéticas , Proteína smad3 , Proteína smad7 , Transactivadores/metabolismo , Factores de Transcripción/metabolismo
15.
Cytokine Growth Factor Rev ; 11(1-2): 5-13, 2000.
Artículo en Inglés | MEDLINE | ID: mdl-10708948

RESUMEN

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.


Asunto(s)
Proteínas Portadoras/metabolismo , Cisteína Endopeptidasas/metabolismo , Proteínas de Unión al ADN/metabolismo , Péptidos y Proteínas de Señalización Intracelular , Complejos Multienzimáticos/metabolismo , Serina Endopeptidasas , Transactivadores/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Animales , Sitios de Unión , Proteínas Portadoras/química , Núcleo Celular/metabolismo , Proteínas de Unión al ADN/química , Humanos , Fosfoproteínas/metabolismo , Complejo de la Endopetidasa Proteasomal , Transducción de Señal , Proteínas Smad , Proteína Smad2 , Proteína Smad4 , Proteína Smad5 , Transactivadores/química , Transcripción Genética , Ubiquitinas/metabolismo
16.
Curr Opin Cell Biol ; 12(2): 235-43, 2000 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-10712925

RESUMEN

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.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Péptidos y Proteínas de Señalización Intracelular , Serina Endopeptidasas , Transducción de Señal , Transactivadores/metabolismo , Transcripción Genética , Factor de Crecimiento Transformador beta/metabolismo , Animales , Proteínas Portadoras/metabolismo , Núcleo Celular/fisiología , Drosophila , Humanos , Proteína Smad4 , Ubiquitinas/metabolismo
18.
J Biol Chem ; 275(3): 2063-70, 2000 Jan 21.
Artículo en Inglés | MEDLINE | ID: mdl-10636910

RESUMEN

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.


Asunto(s)
Proteínas de Unión al ADN/fisiología , Transactivadores/fisiología , Factor de Crecimiento Transformador beta/metabolismo , Proteínas de Xenopus , Activinas , Animales , Unión Competitiva , Quimera/genética , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Relación Dosis-Respuesta a Droga , Matriz Extracelular/metabolismo , Fibroblastos , Regulación de la Expresión Génica , Glucosa-6-Fosfato Isomerasa/metabolismo , Proteínas de Homeodominio/metabolismo , Humanos , Inhibinas/farmacología , Ratones , Ratones Endogámicos C57BL , Factores de Crecimiento Nervioso , Regiones Promotoras Genéticas , Recombinación Genética , Secuencias Reguladoras de Ácidos Nucleicos , Transducción de Señal , Proteínas Smad , Proteína Smad4 , Factores de Tiempo , Transactivadores/genética , Transfección
19.
J Cell Sci ; 113 Pt 2: 269-78, 2000 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-10633078

RESUMEN

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.


Asunto(s)
Proteínas Morfogenéticas Óseas/metabolismo , Factor de Crecimiento de Hepatocito/metabolismo , Túbulos Renales Colectores/embriología , Túbulos Renales Colectores/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Receptores de Superficie Celular/metabolismo , Receptores de Factores de Crecimiento , Factor de Crecimiento Transformador beta , Receptores de Activinas , Animales , Proteína Morfogenética Ósea 2 , Receptores de Proteínas Morfogenéticas Óseas , Proteínas Morfogenéticas Óseas/farmacología , Línea Celular , Proteínas de Unión al ADN/metabolismo , Factor de Crecimiento de Hepatocito/farmacología , Túbulos Renales Colectores/citología , Modelos Biológicos , Morfogénesis/efectos de los fármacos , Fosforilación , Proteínas Serina-Treonina Quinasas/genética , Receptores de Superficie Celular/genética , Transducción de Señal , Proteínas Smad , Transactivadores/metabolismo , Transfección
20.
Sci STKE ; 2000(23): re1, 2000 Mar 14.
Artículo en Inglés | MEDLINE | ID: mdl-11752591

RESUMEN

The transforming growth factor-beta (TGF-beta) superfamily of secreted polypeptide growth factors exerts extensive control over all aspects of development and homeostasis, and components of this pathway are often mutated in cancers and in several hereditary disorders. Apart from TGF-beta, the superfamily also includes the activins and the bone morphogenetic proteins. These factors signal through heteromeric complexes of type II and type I serine-threonine kinase receptors, which activate the downstream Smad signal transduction pathway. Three classes of Smads have been defined: the receptor-regulated Smads (R-Smads), the common-mediator Smads (co-Smads), and the antagonistic or inhibitory Smads (I-Smads). Receptor complexes activate the Smad pathway by interacting and phosphorylating specific R-Smads. Phosphorylation of the R-Smads causes dissociation from the receptor and induces assembly into complexes with Smad4, a co-Smad. This heteromeric complex then translocates into the nucleus, where the Smads function as transcriptional comodulators by recruiting coactivators or corepressors to Smad DNA binding partners. Thus, Smads transmit signals directly from the receptor kinase into the nucleus. Crosstalk between Smads and other signaling pathways occurs both in the cytosol and in the nucleus. In the cytosol, Smad translocation might be inhibited by mitogen-activated protein kinase-dependent phosphorylation, whereas in the nucleus Smads interact with a number of transcription factors that themselves are primary targets of other signaling pathways. Furthermore, Smad-dependent regulation of these targets often requires input from the primary signaling pathway. In these examples, Smad signaling may represent a secondary signal that modifies the output of the primary pathway. Consequently, the transcriptional response to TGF-beta family ligands may be dependent on what other signals are being received by the cell. Crosstalk may thus provide one explanation for the long-standing observation that the biological response to TGF-beta is often dependent on the extracellular environment of the cell.


Asunto(s)
Proteínas de Unión al ADN/fisiología , Receptores de Factores de Crecimiento Transformadores beta/fisiología , Transducción de Señal , Transactivadores/fisiología , Factor de Crecimiento Transformador beta/fisiología , Animales , Humanos , Receptor Cross-Talk/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA