Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 54
Filtrar
1.
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
2.
Br J Cancer ; 89(8): 1538-44, 2003 Oct 20.
Artículo en Inglés | MEDLINE | ID: mdl-14562029

RESUMEN

The breast cancer-associated T2A10 clone was originally isolated from a cDNA library enriched for tumour messenger ribonucleic acids. Our survey of 125 microarrayed primary tumour tissues using affinity purified polyclonal antibodies has revealed that corresponding protein is overexpressed in invasive breast cancer and is weakly expressed in kidney and prostate tumours. Now known as RNF11, the gene encodes a RING-H2 domain and a PY motif, both of which mediate protein-protein interactions. In particular, the PPPPY sequence of RNF11 PY motif is identical to that of Smad7, which has been shown to bind to WW domains of Smurf2, an E3 ubiquitin ligase that mediates the ubiquitination and degradation of the TGFbeta receptor complex. Using various mutants of RNF11 in GST pulldown and immunoprecipitation assays, we found that RNF11 interacts with Smurf2 through the PY motif, leading to ubiquitination of both proteins. Smurf2 plays an active role in the repression of TGFbeta signalling, and our data indicate that overexpression of RNF11, through its interaction with Smurf2, can restore TGFbeta responsiveness in transfected cells.


Asunto(s)
Neoplasias de la Mama/genética , Proteínas Portadoras/biosíntesis , Regulación Neoplásica de la Expresión Génica , Neoplasias de la Mama/patología , Proteínas de Unión al ADN , Humanos , Inmunohistoquímica , Invasividad Neoplásica , Análisis de Secuencia por Matrices de Oligonucleótidos , Receptores de Factores de Crecimiento Transformadores beta/genética , Receptores de Factores de Crecimiento Transformadores beta/fisiología , Transducción de Señal , Transfección , Células Tumorales Cultivadas , Ubiquitina-Proteína Ligasas/farmacología , Dedos de Zinc
3.
Genome Biol ; 2(8): REVIEWS3010, 2001.
Artículo en Inglés | MEDLINE | ID: mdl-11532220

RESUMEN

SUMMARY: The large transforming growth factor-beta (TGFbeta) superfamily of secreted proteins regulate the growth, development and differentiation of cells in diverse organisms, including nematode worms, flies, mice and humans. Signals are initiated upon binding of TGFbeta superfamily members to cell-surface serine/threonine kinase receptors and are then propagated by the intracellular mediators known as Smads. Activation of Smads results in their translocation from the cytoplasm into the nucleus, where they activate or repress transcription together with transcription factors so as to regulate target gene expression. Most Smads consist of two conserved domains. Mad homology (MH) domains I and 2, which are separated by a non-conserved linker region. These domains lack enzymatic activity and, instead, Smads mediate their effects through protein-protein and protein-DNA interactions. Targeted disruption of Smad genes in mice has revealed their importance in embryonic development, and a tumor-suppressor role for Smads in human cancers has been described. Smads therefore play an essential role in mediating TGFbeta-superfamily signals in development and disease.


Asunto(s)
Proteínas de Unión al ADN/química , Proteínas de Unión al ADN/metabolismo , Transactivadores/química , Transactivadores/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Animales , Mapeo Cromosómico , Proteínas de Unión al ADN/deficiencia , Proteínas de Unión al ADN/genética , Evolución Molecular , Regulación del Desarrollo de la Expresión Génica , Humanos , Modelos Moleculares , Mutación , Fenotipo , Estructura Terciaria de Proteína , Transducción de Señal , Proteínas Smad , Transactivadores/deficiencia , Transactivadores/genética
4.
J Biol Chem ; 276(42): 38527-35, 2001 Oct 19.
Artículo en Inglés | MEDLINE | ID: mdl-11486006

RESUMEN

Signaling by transforming growth factor (TGF)-beta family members is mediated by Smad proteins that regulate gene transcription through functional cooperativity and association with other DNA-binding proteins. The hypoxia-inducible factor (HIF)-1 is a transcriptional complex that plays a key role in oxygen-regulated gene expression. We demonstrate that hypoxia and TGF-beta cooperate in the induction of the promoter activity of vascular endothelial growth factor (VEGF), which is a major stimulus in the promotion of angiogenesis. This cooperation has been mapped on the human VEGF promoter within a region at -1006 to -954 that contains functional DNA-binding sequences for HIF-1 and Smads. Optimal HIF-1alpha-dependent induction of the VEGF promoter was obtained in the presence of Smad3, suggesting an interaction between these proteins. Consistent with this, co-immunoprecipitation experiments revealed that HIF-1alpha physically associates with Smad3. These results demonstrate that both TGF-beta and hypoxia signaling pathways can synergize in the regulation of VEGF gene expression at the transcriptional level.


Asunto(s)
Factores de Crecimiento Endotelial/biosíntesis , Factores de Crecimiento Endotelial/genética , Regulación de la Expresión Génica , Hipoxia , Linfocinas/biosíntesis , Linfocinas/genética , Factores de Transcripción , Factor de Crecimiento Transformador beta/metabolismo , Animales , Secuencia de Bases , Northern Blotting , Células COS , Línea Celular , Proteínas de Unión al ADN/metabolismo , Ensayo de Inmunoadsorción Enzimática , Células HeLa , Humanos , Factor 1 Inducible por Hipoxia , Subunidad alfa del Factor 1 Inducible por Hipoxia , Modelos Genéticos , Datos de Secuencia Molecular , Neovascularización Fisiológica , Proteínas Nucleares/metabolismo , Oxígeno/metabolismo , Plásmidos/metabolismo , Pruebas de Precipitina , Regiones Promotoras Genéticas , Unión Proteica , Estructura Terciaria de Proteína , Ratas , Transducción de Señal , Proteína smad3 , Factores de Tiempo , Transactivadores/metabolismo , Transcripción Genética , Transfección , Células Tumorales Cultivadas , Factor A de Crecimiento Endotelial Vascular , Factores de Crecimiento Endotelial Vascular
5.
J Biol Chem ; 276(36): 33986-94, 2001 Sep 07.
Artículo en Inglés | MEDLINE | ID: mdl-11448947

RESUMEN

Furin, a predominant convertase of the cellular constitutive secretory pathway, is known to be involved in the maturation of a number of growth/differentiation factors, but the mechanisms governing its expression remain elusive. We have previously demonstrated that transforming growth factor (TGF) beta 1, through the activation of Smad transducers, regulates its own converting enzyme, furin, creating a unique activation/regulation loop of potential importance in a variety of cell fate and functions. Here we studied the involvement of the p42/p44 MAPK pathway in such regulation. Using HepG2 cells transfected with fur P1 LUC (luciferase) promoter construct, we observed that forced expression of a dominant negative mutant form of the small G protein p21(ras) (RasN17) inhibited TGF beta 1-induced fur gene transcription, suggesting the involvement of the p42/p44 MAPK cascade. In addition, TGF beta induced sustained activation/phosphorylation of endogenous p42/p44 MAPK. Further-more, the role of MAPK cascade in fur gene transcription was highlighted by the use of the MEK1/2 inhibitors, PD98059 or U0126, or co-expression of a p44 antisense construct that repressed the induction of fur promoter transactivation. Conversely, overexpression of a constitutively active form of MEK1 increased unstimulated, TGF beta 1-stimulated, and Smad2-stimulated promoter P1 transactivation, and the universal Smad inhibitor, Smad7, inhibited this effect. Activation of Smad2 by MEK1 or TGF beta 1 resulted in an enhanced nuclear localization of Smad2, which was inhibited upon blocking MEK1 activity. Our findings clearly show that the activation of the p42/p44 MAPK pathway is involved in fur gene expression and led us to propose a co-operative model whereby TGF beta 1-induced receptor activation stimulates not only a Smad pathway but also a parallel p42/p44 MAPK pathway that targets Smad2 for an increased nuclear translocation and enhanced fur gene transactivation. Such an uncovered mechanism may be a key determinant for the regulation of furin in embryogenesis and growth-related physiopathological conditions.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Subtilisinas/metabolismo , Transactivadores/metabolismo , Activación Transcripcional , Factor de Crecimiento Transformador beta/metabolismo , Transporte Activo de Núcleo Celular , Northern Blotting , Butadienos/farmacología , División Celular , Línea Celular , Inhibidores Enzimáticos/farmacología , Flavonoides/farmacología , Técnica del Anticuerpo Fluorescente Indirecta , Furina , Humanos , Immunoblotting , Luciferasas/metabolismo , MAP Quinasa Quinasa 1 , Quinasas de Proteína Quinasa Activadas por Mitógenos/metabolismo , Modelos Biológicos , Nitrilos/farmacología , Fosforilación , Regiones Promotoras Genéticas , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Proto-Oncogénicas p21(ras)/metabolismo , Proteínas Recombinantes/metabolismo , Proteínas Smad , Proteína smad7 , Factor de Crecimiento Transformador beta1
6.
Mol Cell Endocrinol ; 180(1-2): 3-11, 2001 Jun 30.
Artículo en Inglés | MEDLINE | ID: mdl-11451566

RESUMEN

The Smad family of proteins are critical components of the TGFbeta superfamily signalling pathway. Ligand addition induces phosphorylation of specific receptor-regulated Smads, which then form heteromeric complexes with the common mediator Smad, Smad4. This complex then translocates from the cytoplasm into the nucleus. Once there, the R-Smad/Smad4 complex interacts with a variety of DNA binding proteins and is thereby targetted to a diverse array of gene promoters. The Smad-containing DNA binding complex can then positively or negatively regulate gene expression through the recruitment of co-activators and co-repressors. Xenopus FAST (now known as FoxH1) was the first Smad DNA binding partner identified and the FoxH1 family now includes related proteins from mouse, human and Zebrafish. In all organisms examined, FoxH1 is expressed primarily during the earliest stages of development and thus FoxH1 is thought to play a critical role in mediating TGFbeta superfamily signals during these early developmental stages. Other Smad partners range from those that are ubiquitously expressed to others that are present only in specific cell types or developmental stages. Thus, it is the interaction of Smads with a wide range of specific transcriptional partners that is important for the generation of diverse biological responses to TGFbeta superfamily members.


Asunto(s)
Activinas/farmacología , Proteínas de Unión al ADN/farmacología , Transactivadores/farmacología , Factores de Transcripción/farmacología , Transcripción Genética/efectos de los fármacos , Factor de Crecimiento Transformador beta/farmacología , Proteínas de Xenopus , Proteínas de Pez Cebra , Activinas/fisiología , Animales , Proteínas de Unión al ADN/metabolismo , Proteínas de Unión al ADN/fisiología , Factores de Transcripción Forkhead , Humanos , Factores de Crecimiento Nervioso , Fosfoproteínas , Unión Proteica , Transducción de Señal/efectos de los fármacos , Proteínas Smad , Proteína Smad1 , Proteína Smad2 , Proteína smad3 , Proteína Smad4 , Proteína Smad5 , Proteína smad6 , Proteína smad7 , Proteína Smad8 , Transactivadores/metabolismo , Transactivadores/fisiología , Factores de Transcripción/metabolismo , Factores de Transcripción/fisiología , Transcripción Genética/fisiología
7.
J Cell Physiol ; 188(2): 264-73, 2001 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-11424093

RESUMEN

Furin is recognized as being one of the main convertases of the cellular constitutive secretion pathway but the mechanisms regulating its expression are still unknown. We have previously demonstrated that TGFbeta1 up-regulates its own converting enzyme, furin, creating a novel activation/regulation cycle of potential importance in a variety of physiological and pathophysiological conditions. The fur (fes upstream region) gene is regulated via three alternative promoters; P1, P1A, and P1B. To gain insight into the molecular mechanism(s) underlying this up-regulation, we performed transient cell transfections with P1, P1A, and P1B promoter luciferase constructs. Transfection experiments in HepG2 cells revealed that fur P1 promoter is the strongest and the most sensitive to TGFbeta1 stimulation (5 ng/ml) (3.2-fold vs. 2.4-fold for P1A and 2.1-fold for P1B). Cotransfection with either a dominant negative mutant form of Smad2 [Smad2(3SA)] or a known Smad inhibitor [Smad7] inhibit constitutive and TGFbeta1-induced luciferase activity indicating the participation of endogenous Smads. Increased levels of TGFbeta1-induced transcriptional activation of the P1 promoter by overexpression of Smad2 and/or Smad4 is greatly reduced in the presence of Smad2(3SA) and completely inhibited by Smad7, suggesting the participation of endogenous Smad2/Smad4 complexes. Furthermore, the fork-head activin signal transducer (FAST-1), known to interact with Smad2/Smad4 complexes, is a potent stimulator of TGFbeta1-induced transactivation of the fur P1 promoter. Five prime-deletion analysis of this promoter identified the proximal region (between positions -8734 and -7925), as the nucleotide stretch that carries most of the transcriptional activation of fur P1 promoter by Smad2. Overall, the present data demonstrate that Smad2 and Smad4 possibly in complex with FAST-1 or other DNA binding partners participate in the constitutive and inducible transactivation of the fur P1 promoter. This represents the first detailed study of the transcriptional regulation of the fur gene.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Subtilisinas/genética , Transactivadores/metabolismo , Factor de Crecimiento Transformador beta/farmacología , Northern Blotting , Proteínas de Unión al ADN/genética , Furina , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Regulación Enzimológica de la Expresión Génica/fisiología , Hepatoblastoma , Humanos , Neoplasias Hepáticas , Luciferasas/genética , Regiones Promotoras Genéticas/fisiología , ARN Mensajero/análisis , Proteína Smad2 , Proteína Smad4 , Proteína smad7 , Transactivadores/genética , Transcripción Genética/efectos de los fármacos , Transcripción Genética/fisiología , Transfección , Factor de Crecimiento Transformador beta1 , Células Tumorales Cultivadas
8.
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
9.
J Bone Joint Surg Am ; 83-A Suppl 1(Pt 1): S31-9, 2001.
Artículo en Inglés | MEDLINE | ID: mdl-11263663

RESUMEN

BACKGROUND: Several studies have shown that cooperation between transforming growth factor beta (TGF-beta) and Wnt/wingless signaling pathways plays a role in controlling certain developmental events. These factors elicit their biological effects through distinct pathways in which TGF-beta and Wnt signaling induce activation of the transcriptional regulators Smads and lymphoid enhancer binding factor/T-cell-specific factor (LEF/TCF), respectively. To understand the mechanism for cooperativity between these pathways, we have investigated the molecular mechanism for this synergistic effect. METHODS: Transcriptional assays were conducted by transient transfection of HepG2 cells with use of luciferase reporter constructs. Protein/protein interaction studies were conducted in vitro with the use of glutathione-S-transferase pull-down assays and in intact cells by immunoprecipitation and immunoblotting. RESULTS: We show that Smads physically interact with LEF1/TCF transcription factors and that specific DNA binding sites in the Xenopus twin promoter are required for synergistic activation by TGF-beta and Wnt pathways. In addition, we demonstrate that TGF-beta-dependent activation of LEF1/TCF target genes can occur independently of beta-catenin, an essential component of the Wnt signaling pathway. CONCLUSIONS: TGF-beta and Wnt signaling pathways can independently or cooperatively regulate LEF1/TCF target genes. This suggests that the cooperation between these pathways may be important for the specification of cell fates during development.


Asunto(s)
Proteínas de Unión al ADN/fisiología , Regulación de la Expresión Génica , Proteínas Tirosina Quinasas/fisiología , Proteínas Proto-Oncogénicas/fisiología , Transducción de Señal/fisiología , Transactivadores/fisiología , Transcripción Genética , Factor de Crecimiento Transformador beta/fisiología , Proteínas de Pez Cebra , Animales , Células Cultivadas , Proteínas del Citoesqueleto/fisiología , Humanos , Regiones Promotoras Genéticas , Proteínas Smad , Factores de Transcripción/fisiología , Transfección , Proteínas Wnt , beta Catenina
10.
Proc Natl Acad Sci U S A ; 97(15): 8358-63, 2000 Jul 18.
Artículo en Inglés | MEDLINE | ID: mdl-10890911

RESUMEN

The transforming growth factor-beta (TGFbeta) and Wnt/wingless pathways play pivotal roles in tissue specification during development. Activation of Smads, the effectors of TGFbeta superfamily signals, results in Smad translocation from the cytoplasm into the nucleus where they act as transcriptional comodulators to regulate target gene expression. Wnt/wingless signals are mediated by the DNA-binding HMG box transcription factors lymphoid enhancer binding factor 1/T cell-specific factor (LEF1/TCF) and their coactivator beta-catenin. Herein, we show that Smad3 physically interacts with the HMG box domain of LEF1 and that TGFbeta and Wnt pathways synergize to activate transcription of the Xenopus homeobox gene twin (Xtwn). Disruption of specific Smad and LEF1/TCF DNA-binding sites in the promoter abrogates synergistic activation of the promoter. Consistent with this observation, introduction of Smad sites into a TGFbeta-insensitive LEF1/TCF target gene confers cooperative TGFbeta and Wnt responsiveness to the promoter. Furthermore, we demonstrate that TGFbeta-dependent activation of LEF1/TCF target genes can occur in the absence of beta-catenin binding to LEF1/TCF and requires both Smad and LEF1/TCF DNA-binding sites in the Xtwn promoter. Thus, our results show that TGFbeta and Wnt signaling pathways can independently or cooperatively regulate LEF1/TCF target genes and suggest a model for how these pathways can synergistically activate target genes.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Transducción de Señal/fisiología , Transactivadores/metabolismo , Factores de Transcripción/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Proteínas de Xenopus , Proteínas de Pez Cebra , Animales , Sitios de Unión , Células COS , Línea Celular Transformada , Proteínas del Citoesqueleto/metabolismo , Proteínas de Unión al ADN/genética , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Humanos , Factor de Unión 1 al Potenciador Linfoide , Ratones , Factores de Crecimiento Nervioso , Regiones Promotoras Genéticas , Elementos de Respuesta , Proteínas Smad , Proteína Smad2 , Proteína smad3 , Proteína Smad4 , Factores de Transcripción/genética , Células Tumorales Cultivadas , Proteínas Wnt , Xenopus laevis , beta Catenina
12.
Proc Natl Acad Sci U S A ; 97(9): 4820-5, 2000 Apr 25.
Artículo en Inglés | MEDLINE | ID: mdl-10781087

RESUMEN

Biological signals for transforming growth factor beta (TGF-beta) are transduced through transmembrane serine/threonine kinase receptors that signal to a family of intracellular mediators known as Smads. Smad2 and Smad4 are important for transcriptional and antiproliferative responses to TGF-beta, and their inactivation in human cancers indicates that they are tumor suppressors. A missense mutation at a conserved arginine residue in the amino-terminal MH1 domain of both Smad2 and Smad4 has been identified in tumors from patients with colorectal and pancreatic cancers, respectively. However, the mechanism whereby this mutation interferes with Smad activity is uncertain. Here we show that these mutations do not disrupt activation of Smads, including receptor-mediated phosphorylation of Smad2, Smad2/Smad4 heteromeric complex formation, and Smad nuclear translocation. In contrast, we demonstrate that the mutant Smads are degraded rapidly in comparison with their wild-type counterparts. We show that this decrease in Smad protein stability occurs through induction of Smad ubiquitination by pathways involving the UbcH5 family of ubiquitin ligases. These studies thus reveal a mechanism for tumorigenesis whereby genetic defects in Smads induce their degradation through the ubiquitin-mediated pathway.


Asunto(s)
Cisteína Endopeptidasas/metabolismo , Proteínas de Unión al ADN/genética , Genes Supresores de Tumor , Complejos Multienzimáticos/metabolismo , Transactivadores/genética , Factor de Crecimiento Transformador beta/fisiología , Ubiquitinas/metabolismo , Adenosina Trifosfatasas/metabolismo , Secuencia de Aminoácidos , Arginina , Núcleo Celular/metabolismo , Neoplasias Colorrectales/genética , Secuencia Conservada , Proteínas de Unión al ADN/química , Proteínas de Unión al ADN/metabolismo , Humanos , Mutación Missense , Neoplasias Pancreáticas/genética , Regiones Promotoras Genéticas , Complejo de la Endopetidasa Proteasomal , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Transducción de Señal , Proteína Smad2 , Proteína Smad4 , Transactivadores/química , Transactivadores/metabolismo , Activación Transcripcional , Factor de Crecimiento Transformador beta/genética
13.
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
14.
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
15.
J Immunol ; 162(10): 5917-23, 1999 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-10229828

RESUMEN

The mouse NK1.1 Ag originally defined as NK cell receptor (NKR)-P1C (CD161) mediates NK cell activation. Here, we show that another member of the mouse CD161 family, NKR-P1B, represents a novel NK1.1 Ag. In contrast to NKR-P1C, which functions as an activating receptor, NKR-P1B inhibits NK cell activation. Association of NKR-P1B with Src homology 2-containing protein tyrosine phosphatase-1 provides a molecular mechanism for this inhibition. The existence of these two NK1.1 Ags with opposite functions suggests a potential role for NKR-P1 molecules, such as those of the Ly-49 gene family, in regulating NK cell function.


Asunto(s)
Antígenos de Superficie/metabolismo , Antígenos/metabolismo , Citotoxicidad Inmunológica , Células Asesinas Naturales/inmunología , Lectinas Tipo C , Proteínas/metabolismo , Receptores Inmunológicos/metabolismo , Secuencia de Aminoácidos , Animales , Antígenos/genética , Antígenos Ly , Antígenos de Superficie/genética , Células Sanguíneas/inmunología , Sangre Fetal/inmunología , Péptidos y Proteínas de Señalización Intracelular , Ratones , Ratones Endogámicos C57BL , Modelos Inmunológicos , Datos de Secuencia Molecular , Familia de Multigenes , Subfamilia B de Receptores Similares a Lectina de Células NK , Fosforilación , Unión Proteica , Proteína Fosfatasa 1 , Proteína Tirosina Fosfatasa no Receptora Tipo 11 , Proteína Tirosina Fosfatasa no Receptora Tipo 6 , Proteínas Tirosina Fosfatasas/metabolismo , Proteínas/genética , Receptores Inmunológicos/genética , Proteínas Tirosina Fosfatasas con Dominio SH2 , Homología de Secuencia de Aminoácido , Especificidad de la Especie , Dominios Homologos src
16.
J Biol Chem ; 274(15): 9984-92, 1999 Apr 09.
Artículo en Inglés | MEDLINE | ID: mdl-10187774

RESUMEN

Germ line mutations in one of two distinct genes, endoglin or ALK-1, cause hereditary hemorrhagic telangiectasia (HHT), an autosomal dominant disorder of localized angiodysplasia. Both genes encode endothelial cell receptors for the transforming growth factor beta (TGF-beta) ligand superfamily. Endoglin has homology to the type III receptor, betaglycan, although its exact role in TGF-beta signaling is unclear. Activin receptor-like kinase 1 (ALK-1) has homology to the type I receptor family, but its ligand and corresponding type II receptor are unknown. In order to identify the ligand and type II receptor for ALK-1 and to investigate the role of endoglin in ALK-1 signaling, we devised a chimeric receptor signaling assay by exchanging the kinase domain of ALK-1 with either the TGF-beta type I receptor or the activin type IB receptor, both of which can activate an inducible PAI-1 promoter. We show that TGF-beta1 and TGF-beta3, as well as a third unknown ligand present in serum, can activate chimeric ALK-1. HHT-associated missense mutations in the ALK-1 extracellular domain abrogate signaling. The ALK-1/ligand interaction is mediated by the type II TGF-beta receptor for TGF-beta and most likely through the activin type II or type IIB receptors for the serum ligand. Endoglin is a bifunctional receptor partner since it can bind to ALK-1 as well as to type I TGF-beta receptor. These data suggest that HHT pathogenesis involves disruption of a complex network of positive and negative angiogenic factors, involving TGF-beta, a new unknown ligand, and their corresponding receptors.


Asunto(s)
Proteínas Serina-Treonina Quinasas/metabolismo , Telangiectasia Hemorrágica Hereditaria/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Molécula 1 de Adhesión Celular Vascular/metabolismo , Receptores de Activinas , Activinas , Animales , Antígenos CD , Proteínas Morfogenéticas Óseas/metabolismo , Células COS , Línea Celular , Endoglina , Inhibinas/metabolismo , Ligandos , Proteoglicanos/química , Proteoglicanos/metabolismo , Conejos , Receptores de Superficie Celular , Receptores de Factores de Crecimiento Transformadores beta/química , Receptores de Factores de Crecimiento Transformadores beta/metabolismo , Proteínas Recombinantes de Fusión/metabolismo , Homología de Secuencia de Aminoácido , Transducción de Señal , Molécula 1 de Adhesión Celular Vascular/química
17.
Dev Biol ; 207(2): 364-79, 1999 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-10068469

RESUMEN

Smads are central mediators of signal transduction for the TGFbeta superfamily. However, the precise functions of Smad-mediated signaling pathways in early development are unclear. Here we demonstrate a requirement for Smad2 signaling in dorsoanterior axis formation during Xenopus development. Using two point mutations of Smad2 previously identified in colorectal carcinomas, we show that Smad2 ushers Smad4 to the nucleus to form a transcriptional activation complex with the nuclear DNA-binding protein FAST-1 and that the mutant proteins interact normally with FAST-1 but fail to recruit Smad4 into the nucleus. This mechanism of inhibition specifically restricts the dominant-negative activity of these mutants to the activin/Vg1 signaling pathway without inhibiting BMPs. Furthermore, expression of these mutants in Xenopus animal caps inhibits but does not abolish activin and Vg1 induction of mesoderm and in the embryo results in a truncated dorsoanterior axis. These studies define a mechanism through which mutations in Smad2 may block TGFbeta-dependent signaling and suggest a critical role for inductive signaling mediated by the Smad2 pathway in Xenopus organizer function.


Asunto(s)
Proteínas de Unión al ADN/genética , Inhibinas/genética , Transducción de Señal/genética , Transactivadores/genética , Proteínas de Xenopus , Xenopus/embriología , Receptores de Activinas Tipo I , Activinas , Animales , Células COS , Desarrollo Embrionario , Factores de Transcripción Forkhead , Regulación del Desarrollo de la Expresión Génica/genética , Morfogénesis , Mutación , Factores de Crecimiento Nervioso , Fosforilación , ARN Mensajero/metabolismo , Receptores de Factores de Crecimiento/genética , Proteínas Smad , Proteína Smad2 , Proteína Smad4 , Factores de Transcripción/genética , Activación Transcripcional/genética , Transfección , Factor de Crecimiento Transformador beta/genética
18.
Cell ; 95(6): 779-91, 1998 Dec 11.
Artículo en Inglés | MEDLINE | ID: mdl-9865696

RESUMEN

Smads transmit signals from transmembrane ser/thr kinase receptors to the nucleus. We now identify SARA (for Smad anchor for receptor activation), a FYVE domain protein that interacts directly with Smad2 and Smad3. SARA functions to recruit Smad2 to the TGFbeta receptor by controlling the subcellular localization of Smad2 and by interacting with the TGFbeta receptor complex. Phosphorylation of Smad2 induces dissociation from SARA with concomitant formation of Smad2/Smad4 complexes and nuclear translocation. Furthermore, mutations in SARA that cause mislocalization of Smad2 inhibit TGFbeta-dependent transcriptional responses, indicating that the regulation of Smad localization is important for TGFbeta signaling. These results thus define SARA as a component of the TGFbeta pathway that brings the Smad substrate to the receptor.


Asunto(s)
Proteínas Portadoras/metabolismo , Proteínas de Unión al ADN/metabolismo , Péptidos y Proteínas de Señalización Intracelular , Receptores de Factores de Crecimiento Transformadores beta/metabolismo , Serina Endopeptidasas , Transactivadores/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Proteínas de Xenopus , Dedos de Zinc , Células 3T3 , Adulto , Secuencia de Aminoácidos , Animales , Sitios de Unión , Células COS , Proteínas Portadoras/genética , Humanos , Ratones , Datos de Secuencia Molecular , Factores de Crecimiento Nervioso , Fosforilación , Proteínas Serina-Treonina Quinasas , Conejos , Receptor Tipo II de Factor de Crecimiento Transformador beta , Proteínas Recombinantes de Fusión/metabolismo , Transducción de Señal , Proteínas Smad , Proteína Smad2 , Proteína smad3 , Proteína Smad4 , Fracciones Subcelulares , Células Tumorales Cultivadas , Xenopus
19.
J Biol Chem ; 273(49): 33011-9, 1998 Dec 04.
Artículo en Inglés | MEDLINE | ID: mdl-9830054

RESUMEN

Endoglin (CD105) is the target gene for the hereditary hemorrhagic telangiectasia type I (HHT1), a dominantly inherited vascular disorder. It shares with betaglycan a limited amino acid sequence homology and being components of the membrane transforming growth factor-beta (TGF-beta) receptor complex. Using rat myoblasts as a model system, we found that overexpression of endoglin led to a decreased TGF-beta response to cellular growth inhibition and plasminogen activator inhibitor-1 synthesis, whereas overexpression of betaglycan resulted in an enhanced response to inhibition of cellular proliferation and plasminogen activator inhibitor-1 induced expression in the presence of TGF-beta. The regulation by endoglin of TGF-beta responses seems to reside on the extracellular domain, as evidenced by the functional analysis of two chimeric proteins containing different combinations of endoglin and betaglycan domains. Binding followed by cross-linking with 125I-TGF-beta1 demonstrated that betaglycan expressing cells displayed a clear increase (about 3. 5-fold), whereas endoglin expressing cells only displayed an slight increment (about 1.6-fold) in ligand binding with respect to mock transfectants. SDS-polyacrylamide gel electrophoresis analysis of radiolabeled receptors demonstrated that expression of endoglin or betaglycan is associated with an increased TGF-beta binding to the signaling receptor complex; however, while endoglin increased binding to types I and II receptors, betaglycan increased the binding to the type II receptor. Conversely, we found that TGF-beta binding to endoglin required the presence of receptor type II as evidenced by transient transfections experiments in COS cells. These findings suggest a role for endoglin in TGF-beta responses distinct from that of betaglycan.


Asunto(s)
Proteoglicanos/fisiología , Receptores de Factores de Crecimiento Transformadores beta/fisiología , Factor de Crecimiento Transformador beta/fisiología , Molécula 1 de Adhesión Celular Vascular/fisiología , Animales , Antígenos CD , Secuencia de Bases , Línea Celular , Cartilla de ADN , Endoglina , Músculos/citología , Músculos/metabolismo , Ratas , Receptores de Superficie Celular , Transducción de Señal , Transfección , Factor de Crecimiento Transformador beta/genética
20.
Mol Cell ; 2(1): 109-20, 1998 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-9702197

RESUMEN

We identify a mammalian forkhead domain protein, FAST2, that is required for induction of the goosecoid (gsc) promoter by TGF beta or activin signaling. FAST2 binds to a sequence in the gsc promoter, but efficient transcriptional activation and assembly of a DNA-binding complex of FAST2, Smad2, and Smad4 requires an adjacent Smad4 site. Smad3 is closely related to Smad2 but suppresses activation of the gsc promoter. Inhibitory activity is conferred by the MH1 domain, which unlike that of Smad2, binds to the Smad4 site. Through competition for this shared site, Smad3 may prevent transcription by altering the conformation of the DNA-binding complex. Thus, we describe a mechanism whereby Smad2 and Smad3 positively and negatively regulate a TGF beta/activin target gene.


Asunto(s)
Proteínas de Unión al ADN/fisiología , Regulación de la Expresión Génica/fisiología , Proteínas de Homeodominio/genética , Proteínas Represoras , Transactivadores , Transcripción Genética/fisiología , Factor de Crecimiento Transformador beta/fisiología , Proteínas de Xenopus , Activinas , Secuencia de Aminoácidos , Animales , ADN Recombinante/genética , Proteínas de Unión al ADN/química , Proteínas de Unión al ADN/aislamiento & purificación , Factores de Transcripción Forkhead , Proteína Goosecoide , Humanos , Inhibinas/fisiología , Sustancias Macromoleculares , Ratones , Datos de Secuencia Molecular , Regiones Promotoras Genéticas , Alineación de Secuencia , Homología de Secuencia de Aminoácido , Transducción de Señal/fisiología , Proteína Smad2 , Proteína smad3 , Factores de Transcripción/química , Células Tumorales Cultivadas , Xenopus laevis/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...