Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 218
Filtrar
1.
Sci Total Environ ; 777: 144579, 2021 Jul 10.
Artículo en Inglés | MEDLINE | ID: mdl-33677295

RESUMEN

The Guadalquivir Valley is one of three major O3 hotspots in Spain. An airborne and surface measurement campaign was carried out from July 9th to 11th, 2019 to quantify the local/regional O3 contributions using experimental approaches. Air quality and meteorology data from surface measurements, a microlight aircraft, a helium balloon, and remote sensing data (TROPOMI-NO2-ESA) were used to obtain the 3D distribution of O3 and various tracer pollutants. O3 accumulation over 2.5 days started with inputs from oceanic air masses transported inland by sea breezes, which drew O3 and its precursors from a local/regional origin to the northeastern end of the basin. The orographic-meteorological setting of the valley caused vertical recirculation of the air masses inside the valley that caused the accumulation by increasing regional background O3 concentration by 25-30 ppb. Furthermore, possible Mediterranean O3 contributions and additional vertical recirculation through the entrainment zone of the convective boundary layer also contributed. Using particulate matter finer than 2.5 µm (PM2.5), ultrafine particles (UFP), and black carbon (BC) as tracers of local sources, we calculated that local contributions increased regional O3 levels by 20 ppb inside specific pollution plumes transported by the breeze into the valley, and by 10 ppb during midday when flying over an area with abundant agricultural burning during the morning. Air masses that crossed the southern boundaries of the Betic system at mid-altitude (400-1850 m a.s.l.) on July 10th and 11th may have provided additional O3. Meanwhile, a decreasing trend at high altitudes (3000-5000 m a.s.l.) was observed, signifying that the impact of stratospheric O3 intrusion decreased during the campaign.

2.
Artículo en Inglés | MEDLINE | ID: mdl-16869748

RESUMEN

Metastasis is a multistep and multifunctional biological cascade that is the final and most life-threatening stage of cancer progression. Understanding the biological underpinnings of this complex process is of extreme clinical relevance and requires unbiased and comprehensive biological scrutiny. In recent years, we have utilized a xenograft model of breast cancer metastasis to discover genes that mediate organ-specific patterns of metastatic colonization. Examination of transcriptomic data from cohorts of primary breast cancers revealed a subset of site-specific metastasis genes that are selected for early in tumor progression. High expression of these genes predicts the propensity for lung metastasis independently of several classic markers of poor prognosis. These genes fulfill dual functions-enhanced primary tumorigenicity and augmented organ-specific metastatic activity. Other metastasis genes fulfill functions specialized for the microenvironment of the metastatic site and are consequently not selected for in primary tumors. These findings improve our understanding of metastatic progression, facilitate the interpretation of primary tumor gene expression data, and open several important possibilities for future clinical application.


Asunto(s)
Metástasis de la Neoplasia/genética , Oncogenes , Animales , Neoplasias Óseas/genética , Neoplasias Óseas/patología , Neoplasias Óseas/secundario , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Línea Celular Tumoral , Femenino , Perfilación de la Expresión Génica , Humanos , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patología , Neoplasias Pulmonares/secundario , Ratones , Metástasis de la Neoplasia/patología , Trasplante de Neoplasias , Especificidad de Órganos , Pronóstico , Trasplante Heterólogo
3.
Mol Cell ; 8(3): 671-82, 2001 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-11583628

RESUMEN

The type I TGF beta receptor (T beta R-I) is activated by phosphorylation of the GS region, a conserved juxtamembrane segment located just N-terminal to the kinase domain. We have studied the molecular mechanism of receptor activation using a homogeneously tetraphosphorylated form of T beta R-I, prepared using protein semisynthesis. Phosphorylation of the GS region dramatically enhances the specificity of T beta R-I for the critical C-terminal serines of Smad2. In addition, tetraphosphorylated T beta R-I is bound specifically by Smad2 in a phosphorylation-dependent manner and is no longer recognized by the inhibitory protein FKBP12. Thus, phosphorylation activates T beta R-I by switching the GS region from a binding site for an inhibitor into a binding surface for substrate. Our observations suggest that phosphoserine/phosphothreonine-dependent localization is a key feature of the T beta R-I/Smad activation process.


Asunto(s)
Receptores de Activinas Tipo I , Proteínas de Ciclo Celular , Proteínas de Unión al ADN/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Receptores de Factores de Crecimiento Transformadores beta/metabolismo , Proteínas de Saccharomyces cerevisiae , Transactivadores/metabolismo , Secuencia de Aminoácidos , Quinasa de Punto de Control 2 , Cristalografía por Rayos X , Proteínas de Unión al ADN/química , Proteínas de Unión al ADN/genética , Immunoblotting , Modelos Biológicos , Modelos Moleculares , Datos de Secuencia Molecular , Estructura Molecular , Fosforilación , Unión Proteica , Proteínas Serina-Treonina Quinasas/síntesis química , Proteínas Serina-Treonina Quinasas/genética , Estructura Secundaria de Proteína , Estructura Terciaria de Proteína , Receptor Tipo I de Factor de Crecimiento Transformador beta , Receptores de Factores de Crecimiento Transformadores beta/genética , Proteínas Recombinantes de Fusión/metabolismo , Proteína Smad2 , Proteína 1A de Unión a Tacrolimus/metabolismo , Transactivadores/química , Transactivadores/genética , Factor de Crecimiento Transformador beta/antagonistas & inhibidores , Factor de Crecimiento Transformador beta/metabolismo
4.
Cell Growth Differ ; 12(9): 457-63, 2001 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-11571228

RESUMEN

The homeodomain protein TG-interacting factor (TGIF) represses transcription by histone deacetylase-dependent and -independent means. Heterozygous mutations in human TGIF result in holoprosencephaly, a severe genetic disorder affecting craniofacial development, suggesting that TGIF is critical for normal development. After transforming growth factorbeta (TGFbeta) stimulation, Smad proteins enter the nucleus and form transcriptional activation complexes or interact with TGIF, which functions as a corepressor. The relative levels of Smad corepressors and coactivators present within the cell may determine the outcome of a TGFbeta response. We show that TGIF interacts directly with the paired amphipathic alpha-helix 2 domain of the mSin3 corepressor, and TGIF recruits mSin3 to a TGFbeta-activated Smad complex. The mSin3 interaction domain of TGIF has been shown to be essential for repression of a TGFbeta transcriptional response. Thus, TGIF represents a targeting component of the mSin3 corepressor complex.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Proteínas de Homeodominio/metabolismo , Proteínas Represoras/metabolismo , Proteínas de Saccharomyces cerevisiae , Transactivadores/metabolismo , Factores de Transcripción/metabolismo , Animales , Células COS , Chlorocebus aethiops , Proteínas de Unión al ADN/genética , Histona Desacetilasas , Proteínas de Homeodominio/genética , Humanos , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Proteínas Represoras/genética , Proteína Smad2 , Transactivadores/genética , Transcripción Genética , Factor de Crecimiento Transformador beta/farmacología , Células Tumorales Cultivadas
5.
Nat Cell Biol ; 3(4): 392-9, 2001 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-11283613

RESUMEN

Deregulated expression of c-myc can induce cell proliferation in established cell lines and in primary mouse embryonic fibroblasts (MEFs), through a combination of both transcriptional activation and repression by Myc. Here we show that a Myc-associated transcription factor, Miz-1, arrests cells in G1 phase and inhibits cyclin D-associated kinase activity. Miz-1 upregulates expression of the cyclin-dependent kinases (CDK) inhibitor p15INK4b by binding to the initiator element of the p15INK4b promoter. Myc and Max form a complex with Miz-1 at the p15 initiator and inhibit transcriptional activation by Miz-1. Expression of Myc in primary cells inhibits the accumulation of p15INK4b that is associated with cellular senescence; conversely, deletion of c-myc in an established cell line activates p15INK4b expression. Alleles of c-myc that are unable to bind to Miz-1 fail to inhibit accumulation of p15INK4b messenger RNA in primary cells and are, as a consequence, deficient in immortalization.


Asunto(s)
Proteínas Portadoras/biosíntesis , Proteínas de Ciclo Celular , Inhibidor p16 de la Quinasa Dependiente de Ciclina , Proteínas de Unión al ADN/metabolismo , Proteínas Proto-Oncogénicas c-myc/metabolismo , Factores de Transcripción/metabolismo , Proteínas Supresoras de Tumor , Dedos de Zinc , Células 3T3 , Animales , Inhibidor p15 de las Quinasas Dependientes de la Ciclina , Proteínas de Unión al ADN/genética , Regulación de la Expresión Génica , Células HeLa , Humanos , Factores de Transcripción de Tipo Kruppel , Ratones , Proteínas Nucleares/metabolismo , Regiones Promotoras Genéticas , Ratas , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Transactivadores/metabolismo , Factores de Transcripción/genética
6.
Nat Cell Biol ; 3(4): 400-8, 2001 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-11283614

RESUMEN

Transforming growth factor-beta (TGFbeta) is a cytokine that arrests epithelial cell division by switching off the proto-oncogene c-myc and rapidly switching on cyclin-dependent kinase (CDK) inhibitors such as p15INK4b. Gene responses to TGFbeta involve Smad transcription factors that are directly activated by the TGFbeta receptor. Why downregulation of c-myc expression by TGFbeta is required for rapid activation of p15INK4b has remained unknown. Here we provide evidence that TGFbeta signalling prevents recruitment of Myc to the p15INK4b transcriptional initiator by Myc-interacting zinc-finger protein 1 (Miz-1). This relieves repression and enables transcriptional activation by a TGFbeta-induced Smad protein complex that recognizes an upstream p15INK4b promoter region and contacts Miz-1. Thus, two separate TGFbeta-dependent inputs - Smad-mediated transactivation and relief of repression by Myc - keep tight control over p15INK4b activation.


Asunto(s)
Proteínas Portadoras/genética , Proteínas de Ciclo Celular , Inhibidor p16 de la Quinasa Dependiente de Ciclina , Quinasas Ciclina-Dependientes/antagonistas & inhibidores , Proteínas de Unión al ADN/metabolismo , Proteínas Proto-Oncogénicas c-myc/metabolismo , Transactivadores/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Proteínas Supresoras de Tumor , Dedos de Zinc , Animales , Secuencia de Bases , Células COS , Chlorocebus aethiops , Inhibidor p15 de las Quinasas Dependientes de la Ciclina , Proteínas de Unión al ADN/genética , Silenciador del Gen , Humanos , Factores de Transcripción de Tipo Kruppel , Ratones , Datos de Secuencia Molecular , Regiones Promotoras Genéticas , Proto-Oncogenes Mas , Proteínas Proto-Oncogénicas c-myc/genética , Elementos de Respuesta , Proteína Smad2 , Proteína smad3 , Proteína Smad4 , Proteína smad7 , Transactivadores/genética , Factores de Transcripción , Activación Transcripcional
7.
Cancer Res ; 61(1): 256-60, 2001 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-11196171

RESUMEN

Transforming growth factor-beta (TGF-beta) inhibits growth and induces apoptosis of colon epithelial cells. Binding of TGF-beta to its receptor induces phosphorylation of the Smad proteins Smad2 and Smad3, which then form heteromeric complexes with Smad4, translocate to the nucleus, and activate gene transcription. Smad4 function has been considered an obligate requirement for TGF-beta signaling, and Smad4 mutations present in some cancers have been considered sufficient to inactivate TGF-beta signaling. In this work, we describe studies with a nontransformed human colon epithelial cell line that is mutant for Smad4 but remains growth-inhibited by TGF-beta. The colon cell line VACO-235 has lost one of its Smad4 alleles via a chromosome 18q deletion. The remaining allele bears two missense point mutations located in regions important for Smad4 trimer formation, which is thought necessary for Smad4 function. As expected, pSBE4-BV/Luc, a Smad4-activated transcriptional reporter, was inactive in VACO-235. Nonetheless, VACO-235 demonstrated 80% growth inhibition in response to TGF-beta, as well as retention of some TGF-beta-mediated activation of the p3TP-Lux transcriptional reporter. Transient transfection of the VACO-235 Smad4 mutant allele into a Smad4-null cell line confirmed that this allele is functionally inactive as assayed by both the pSBE4-BV and p3TP-Lux reporters. The simplest explanation of these results is that there is a non-Smad4-dependent pathway for TGF-beta-mediated signaling and growth inhibition in VACO-235 cells.


Asunto(s)
Adenoma/genética , Adenoma/patología , Neoplasias del Colon/genética , Neoplasias del Colon/patología , Proteínas de Unión al ADN/genética , Inhibidores de Crecimiento/farmacología , Mutación , Transactivadores/genética , Factor de Crecimiento Transformador beta/farmacología , División Celular/efectos de los fármacos , División Celular/genética , Genes Reporteros , Humanos , Luciferasas/biosíntesis , Luciferasas/genética , Proteína Smad4 , Activación Transcripcional/genética , Células Tumorales Cultivadas
8.
Curr Top Microbiol Immunol ; 254: 145-64, 2001.
Artículo en Inglés | MEDLINE | ID: mdl-11190572

RESUMEN

The known Smad transcriptional repressors appear to play multiple roles in modulating TGF beta-activated transcriptional responses. As detailed in Fig. 4, in the [figure: see text] absence of TGF beta signals, Ski/Sno prevent the activation of transcription by Smad proteins that find their way to the nucleus. Following TGF beta stimulation, the interaction with Ski/Sno is lost and these proteins are degraded. The free, activated Smad complex then enters the nucleus, where it can form two different kinds of transcriptional complexes: one involving interactions with general transcriptional coactivators, resulting in transcriptional activation, and the alternate complex, in which coactivators are displaced by a complex of corepressors recruited via a protein such as TGIF. The relative levels of these two complexes formed appear to be determined by the levels of available Smad coactivators and corepressors present within the cell. Once Smad transcriptional complexes have been formed, they can be further modulated by corepressors in at least two ways. TGF beta itself appears to upregulate SnoN expression, perhaps resulting in negative feedback on the activating Smad complexes. The balance between coactivators and corepressors within the cell can also be altered by other signaling inputs, and it appears that the stabilization of TGIF in response to activation of the MAP kinase pathway is able to shift the balance towards transcriptional repression. The scheme of action of Smad corepressors, represented in Fig. 4, is based on the initial analyses of these factors, and the challenge for the future is to more fully understand the precise physiological roles of Smad corepressors. Determining the roles they play in modulating responses to TGF beta family ligands during development, together with an analysis of the contributions of mutations that affect Smad corepressor function to genetic diseases such as HPE and to cancer will also be of great interest. Additionally, a better understanding of the events within the nucleus following BMP signaling may reveal the presence not only of more BMP-specific Smad recruiters, but also of BMP Smad-specific corepressors.


Asunto(s)
Proteínas Represoras/metabolismo , Transducción de Señal , Transactivadores/metabolismo , Transcripción Genética , Factor de Crecimiento Transformador beta/metabolismo , Animales , Núcleo Celular/metabolismo , Regulación de la Expresión Génica , Humanos , Ratones , Proteínas Represoras/genética , Transactivadores/genética , Factor de Crecimiento Transformador beta/genética
9.
EMBO J ; 20(1-2): 128-36, 2001 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-11226163

RESUMEN

Smad transcription factors mediate the actions of transforming growth factor-beta (TGF-beta) cytokines during development and tissue homeostasis. TGF-beta receptor-activated Smad2 regulates gene expression by associating with transcriptional co-activators or co-repressors. The Smad co-repressor TGIF competes with the co-activator p300 for Smad2 association, such that TGIF abundance helps determine the outcome of a TGF-beta response. Small alterations in the physiological levels of TGIF can have profound effects on human development, as shown by the devastating brain and craniofacial developmental defects in heterozygotes carrying a hypomorphic TGIF mutant allele. Here we show that TGIF levels modulate sensitivity to TGF-beta-mediated growth inhibition, that TGIF is a short-lived protein and that epidermal growth factor (EGF) signaling via the Ras-Mek pathway causes the phosphorylation of TGIF at two Erk MAP kinase sites, leading to TGIF stabilization and favoring the formation of Smad2-TGIF co-repressor complexes in response to TGF-beta. These results identify the first mechanism for regulating TGIF levels and suggest a potential link for Smad and Ras pathway convergence at the transcriptional level.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Proteínas de Homeodominio/metabolismo , Transactivadores/metabolismo , Factor de Crecimiento Transformador beta/farmacología , Proteínas ras/metabolismo , Animales , Células COS , División Celular/efectos de los fármacos , Línea Celular , Chlorocebus aethiops , Humanos , Cinética , Fosfatos/metabolismo , Fosforilación , Proteínas Recombinantes/metabolismo , Proteínas Represoras/metabolismo , Proteína Smad2 , Transfección
10.
Proc Natl Acad Sci U S A ; 98(3): 992-9, 2001 Jan 30.
Artículo en Inglés | MEDLINE | ID: mdl-11158583

RESUMEN

Loss of growth inhibitory responses to the cytokine transforming growth factor beta (TGF-beta) in cancer cells may result from mutational inactivation of TGF-beta receptors or their signal transducers, the Smad transcription factors. In breast cancer, however, loss of TGF-beta growth inhibition often occurs without a loss of these signaling components. A genome-wide analysis of rapid TGF-beta gene responses in MCF-10A human mammary epithelial cells and MDA-MB-231 breast cancer cells shows that c-myc repression, a response that is key to the TGF-beta program of cell cycle arrest, is selectively lost in the cancer cell line. Transformation of MCF-10A cells with c-Ha-ras and c-erbB2 oncogenes also led to a selective loss of c-myc repression and cell cycle arrest response. TGF-beta stimulation of epithelial cells rapidly induces the formation of a Smad complex that specifically recognizes a TGF-beta inhibitory element in the c-myc promoter. Formation of this complex is deficient in the oncogenically transformed breast cells. These results suggest that a Smad complex that specifically mediates c-myc repression is a target of oncogenic signals in breast cancer.


Asunto(s)
Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Regulación Neoplásica de la Expresión Génica , Regulación de la Expresión Génica , Genes myc , Factor de Crecimiento Transformador beta/genética , Factor de Crecimiento Transformador beta/farmacología , Secuencia de Bases , Mama , Ciclo Celular/efectos de los fármacos , Células Epiteliales/citología , Células Epiteliales/efectos de los fármacos , Células Epiteliales/fisiología , Femenino , Humanos , Datos de Secuencia Molecular , Proteínas Proto-Oncogénicas c-myc/metabolismo , Transcripción Genética , Células Tumorales Cultivadas
11.
Mol Cell ; 8(6): 1277-89, 2001 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-11779503

RESUMEN

Ligand-induced phosphorylation of the receptor-regulated Smads (R-Smads) is essential in the receptor Ser/Thr kinase-mediated TGF-beta signaling. The crystal structure of a phosphorylated Smad2, at 1.8 A resolution, reveals the formation of a homotrimer mediated by the C-terminal phosphoserine (pSer) residues. The pSer binding surface on the MH2 domain, frequently targeted for inactivation in cancers, is highly conserved among the Co- and R-Smads. This finding, together with mutagenesis data, pinpoints a functional interface between Smad2 and Smad4. In addition, the pSer binding surface on the MH2 domain coincides with the surface on R-Smads that is required for docking interactions with the serine-phosphorylated receptor kinases. These observations define a bifunctional role for the MH2 domain as a pSer-X-pSer binding module in receptor Ser/Thr kinase signaling pathways.


Asunto(s)
Proteínas de Unión al ADN/química , Proteínas de Unión al ADN/metabolismo , Fosfoserina/metabolismo , Transducción de Señal/efectos de los fármacos , Transactivadores/química , Transactivadores/metabolismo , Factor de Crecimiento Transformador beta/farmacología , Secuencias de Aminoácidos , Secuencia de Aminoácidos , Animales , Sitios de Unión , Cristalografía por Rayos X , Proteínas de Unión al ADN/genética , Humanos , Modelos Biológicos , Modelos Moleculares , Datos de Secuencia Molecular , Mutación/genética , Neoplasias/genética , Fosforilación , Proteínas Serina-Treonina Quinasas/metabolismo , Estructura Cuaternaria de Proteína , Estructura Terciaria de Proteína , Alineación de Secuencia , Proteína Smad2 , Proteína Smad4 , Relación Estructura-Actividad , Transactivadores/genética
13.
J Biol Chem ; 275(51): 40282-7, 2000 Dec 22.
Artículo en Inglés | MEDLINE | ID: mdl-11032826

RESUMEN

The t(8;21) translocation, found in adult acute myelogenous leukemia, results in the formation of an AML1/ETO chimeric transcription factor. AML1/ETO expression leads to alterations in hematopoietic progenitor cell differentiation, although its role in leukemic transformation is not clear. The N-terminal portion of AML1, which is retained in AML1/ETO, contains a region of homology to the FAST proteins, which cooperate with Smads to regulate transforming growth factor beta1 (TGF-beta1) target genes. We have demonstrated the physical association of Smad proteins with AML1 and AML1/ETO by immunoprecipitation and have mapped the region of interaction to the runt homology domain in these AML1 proteins. Using confocal microscopy, we demonstrated that AML1, and ETO and/or AML1/ETO, colocalize with Smads in the nucleus of t(8;21)-positive Kasumi-1 cells, in the presence but not the absence of TGF-beta1. Using transient transfection assays and a reporter gene construct that contains both Smad and AML1 consensus binding sequences, we demonstrated that overexpression of AML1B cooperates with TGF-beta1 in stimulating reporter gene activity, whereas AML1/ETO represses basal promoter activity and blocks the response to TGF-beta1. Considering the critical role of TGF-beta1 in the growth and differentiation of hematopoietic cells, interference with TGF-beta1 signaling by AML1/ETO may contribute to leukemogenesis.


Asunto(s)
Proteínas de Fusión Oncogénica/metabolismo , Transducción de Señal , Factores de Transcripción/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Células 3T3 , Animales , Secuencia de Bases , Células COS , Subunidad alfa 2 del Factor de Unión al Sitio Principal , Cartilla de ADN , Ratones , Proteína 1 Compañera de Translocación de RUNX1
14.
J Biol Chem ; 275(52): 40710-7, 2000 Dec 29.
Artículo en Inglés | MEDLINE | ID: mdl-11018029

RESUMEN

Protein interactions are critical for the function of SMADs as mediators of transforming growth factor-beta (TGF-beta) signals. TGF-beta receptor phosphorylation of SMAD2 or SMAD3 causes their association with SMAD4 and accumulation in the nucleus where the SMAD complex binds cofactors that determine the choice of target genes. We provide evidence that in the basal state, SMADs 2, 3, and 4 form separate, strikingly different complexes. SMAD2 is found mostly as monomer, whereas the closely related SMAD3 exists in multiple oligomeric states. This difference is due to a unique structural element in the MH1 domain of SMAD2 that inhibits protein-protein interactions in the basal state. In contrast to SMAD2 and SMAD3, SMAD4 in the basal state is found mostly as a homo-oligomer, most likely a trimer. Upon cell stimulation with TGF-beta, SMAD proteins become engaged in a multitude of complexes ranging in size from SMAD2-SMAD4 heterodimers to assemblies of >650 kDa. The latter display the highest DNA binding affinity for the TGF-beta-response elements of JUNB and collagen 7. These observations, all validated with endogenous SMAD proteins, modify previous models regarding the assembly and activity of SMAD complexes in the TGF-beta pathway.


Asunto(s)
Proteínas de Unión al ADN/química , Transactivadores/química , Factor de Crecimiento Transformador beta/farmacología , Animales , Células COS , ADN/metabolismo , Proteínas de Unión al ADN/metabolismo , Dimerización , Elementos de Respuesta , Proteína Smad2 , Proteína smad3 , Transactivadores/metabolismo
16.
Mol Cell Biol ; 20(17): 6201-11, 2000 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-10938097

RESUMEN

The winged-helix (WH) BF-1 gene, which encodes brain factor 1 (BF-1) (also known as foxg1), is essential for the proliferation of the progenitor cells of the cerebral cortex. Here we show that BF-1-deficient telencephalic progenitor cells are more apt to leave the cell cycle in response to transforming growth factor beta (TGF-beta) and activin. We found that ectopic expression of BF-1 in vitro inhibits TGF-beta mediated growth inhibition and transcriptional activation. Surprisingly, we found that the ability of BF-1 to function as a TGF-beta antagonist does not require its DNA binding activity. Therefore, we investigated whether BF-1 can inhibit Smad-dependent transcriptional responses by interacting with Smads or Smad binding partners. We found that BF-1 does not interact with Smads. Because the identities of the Smad partners mediating growth inhibition by TGF-beta are not clearly established, we examined a model reporter system which is known to be activated by activin and TGF-beta through Smads and the WH factor FAST-2. We demonstrate that BF-1 associates with FAST-2. This interaction is dependent on the same region of protein which mediates its ability to interfere with the antiproliferative activity of TGF-beta and with TGF-beta-dependent transcriptional activation. Furthermore, the interaction of FAST-2 with BF-1 is mediated by the same domain which is required for FAST-2 to interact with Smad2. We propose a model in which BF-1 interferes with transcriptional responses to TGF-beta by interacting with FAST-2 or with other DNA binding proteins which function as Smad2 partners and which have a common mode of interaction with Smad2.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Transducción de Señal , Transactivadores/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Activinas , Animales , Células COS , División Celular/efectos de los fármacos , Línea Celular , Proteínas de Unión al ADN/química , Proteínas de Unión al ADN/genética , Relación Dosis-Respuesta a Droga , Factores de Transcripción Forkhead , Immunoblotting , Inhibinas/metabolismo , Luciferasas/metabolismo , Ratones , Ratones Endogámicos C57BL , Visón , Modelos Biológicos , Mutagénesis Sitio-Dirigida , Proteínas del Tejido Nervioso/química , Proteínas del Tejido Nervioso/genética , Pruebas de Precipitina , Unión Proteica , Estructura Terciaria de Proteína , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Proteína Smad2 , Factores de Transcripción/metabolismo , Activación Transcripcional/efectos de los fármacos , Factor de Crecimiento Transformador beta/química , Factor de Crecimiento Transformador beta/farmacología , beta-Galactosidasa/metabolismo
17.
J Biol Chem ; 275(41): 32066-70, 2000 Oct 13.
Artículo en Inglés | MEDLINE | ID: mdl-10906337

RESUMEN

Recently, the oncoprotein MDM-2 was implicated in the transforming growth factor-beta (TGF-beta) growth inhibitory pathway by the finding that prolonged, constitutive expression of MDM-2 in mink lung epithelial cells could overcome the antiproliferative effect of TGF-beta (Sun, P., Dong, P., Dai, K., Hannon, G. J., and Beach, D. (1998) Science 282, 2270-2272). However, using Mv1Lu cells conditionally expressing MDM-2, we found that MDM-2 does not overcome TGF-beta-mediated growth arrest. No detectable changes were observed in various TGF-beta responses, including cell cycle arrest, activation of transcriptional reporters, and TGF-beta-dependent Smad2/3 nuclear accumulation. This finding was in direct contrast to the effect of forcing c-Myc expression, a bona fide member of the TGF-beta growth inhibitory pathway, which renders cells refractory to TGF-beta-induced cell cycle arrest. Our results suggest that an MDM-2-dependent increase in cell cycle progression may allow the acquisition of additional mutations over time and that these alterations then allow cells to evade a TGF-beta-mediated growth arrest. Our conclusion is that, whereas c-Myc down-regulation by TGF-beta is a required event in the cell cycle arrest response of epithelial cells, MDM-2 is not a direct participant in the normal TGF-beta antiproliferative response.


Asunto(s)
Ciclo Celular/efectos de los fármacos , Proteínas Nucleares , Proteínas Proto-Oncogénicas c-myc/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Factor de Crecimiento Transformador beta/farmacología , Animales , División Celular/efectos de los fármacos , Línea Celular , Núcleo Celular/efectos de los fármacos , Núcleo Celular/metabolismo , Proteínas de Unión al ADN/metabolismo , Regulación hacia Abajo/efectos de los fármacos , Técnica del Anticuerpo Fluorescente , Genes Reporteros , Pulmón , Visón , Mutagénesis , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas c-mdm2 , Proteínas Proto-Oncogénicas c-myc/genética , Transducción de Señal/efectos de los fármacos , Proteína Smad2 , Proteína smad3 , Transactivadores/metabolismo , Activación Transcripcional/efectos de los fármacos , Transfección
18.
Nat Genet ; 25(2): 205-8, 2000 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-10835638

RESUMEN

Holoprosencephaly (HPE) is the most common structural defect of the developing forebrain in humans (1 in 250 conceptuses, 1 in 16,000 live-born infants). HPE is aetiologically heterogeneous, with both environmental and genetic causes. So far, three human HPE genes are known: SHH at chromosome region 7q36 (ref. 6); ZIC2 at 13q32 (ref. 7); and SIX3 at 2p21 (ref. 8). In animal models, genes in the Nodal signalling pathway, such as those mutated in the zebrafish mutants cyclops (refs 9,10), squint (ref. 11) and one-eyed pinhead (oep; ref. 12), cause HPE. Mice heterozygous for null alleles of both Nodal and Smad2 have cyclopia. Here we describe the involvement of the TG-interacting factor (TGIF), a homeodomain protein, in human HPE. We mapped TGIF to the HPE minimal critical region in 18p11.3. Heterozygous mutations in individuals with HPE affect the transcriptional repression domain of TGIF, the DNA-binding domain or the domain that interacts with SMAD2. (The latter is an effector in the signalling pathway of the neural axis developmental factor NODAL, a member of the transforming growth factor-beta (TGF-beta) family.) Several of these mutations cause a loss of TGIF function. Thus, TGIF links the NODAL signalling pathway to the bifurcation of the human forebrain and the establishment of ventral midline structures.


Asunto(s)
Tipificación del Cuerpo/genética , Holoprosencefalia/genética , Proteínas de Homeodominio/genética , Transducción de Señal , Factor de Crecimiento Transformador beta/fisiología , Animales , Secuencia de Bases , Células COS , Cromosomas Humanos Par 18/genética , ADN/genética , ADN/metabolismo , Análisis Mutacional de ADN , Proteínas de Unión al ADN/metabolismo , Exones/genética , Regulación de la Expresión Génica/genética , Proteínas de Homeodominio/química , Proteínas de Homeodominio/metabolismo , Humanos , Ratones , Mutación , Proteína Nodal , Mapeo Físico de Cromosoma , Prosencéfalo/anomalías , Prosencéfalo/embriología , Prosencéfalo/metabolismo , Unión Proteica , ARN Mensajero/análisis , ARN Mensajero/genética , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Proteínas Represoras/química , Proteínas Represoras/genética , Proteínas Represoras/metabolismo , Proteína Smad2 , Transactivadores/metabolismo
19.
J Biol Chem ; 275(36): 27973-8, 2000 Sep 08.
Artículo en Inglés | MEDLINE | ID: mdl-10854429

RESUMEN

Anti-Müllerian hormone induces the regression of fetal Müllerian ducts and inhibits the transcription of gonadal steroidogenic enzymes. It belongs to the transforming growth factor-beta family whose members signal through a pair of serine/threonine kinase receptors and Smad effectors. Only the anti-Müllerian hormone type II receptor has been identified. Our goal was to determine whether anti-Müllerian hormone could share a type I receptor with another family member. Co-immunoprecipitation of known type I receptors with anti-Müllerian hormone type II receptor clearly showed that the bone morphogenetic protein type IB receptor was the only cloned type I receptor interacting in a ligand-dependent manner with this type II receptor. Anti-Müllerian hormone also activates the bone morphogenetic protein-specific Smad1 pathway and the XVent2 reporter gene, an anti-Müllerian hormone type II receptor-dependent effect abrogated by a dominant negative version of bone morphogenetic protein type IB receptor. Reverse amplification experiments showed that bone morphogenetic protein type IB receptor is co-expressed with anti-Müllerian hormone type II receptor in most anti-Müllerian hormone target tissues. Our data support a model in which a ligand, anti-Müllerian hormone, gains access to a shared type I receptor and Smad1 system through a highly restricted type II receptor.


Asunto(s)
Proteínas de Unión al ADN , Glicoproteínas , Inhibidores de Crecimiento/farmacología , Proteínas Serina-Treonina Quinasas/fisiología , Proteínas/metabolismo , Receptores de Factores de Crecimiento/fisiología , Receptores de Péptidos/fisiología , Hormonas Testiculares/farmacología , Transactivadores , Animales , Hormona Antimülleriana , Receptores de Proteínas Morfogenéticas Óseas de Tipo 1 , Receptores de Proteínas Morfogenéticas Óseas de Tipo II , Células CHO , Línea Celular , Cricetinae , Genes Reporteros , Humanos , Ratones , Proteínas Serina-Treonina Quinasas/efectos de los fármacos , Proteínas Serina-Treonina Quinasas/genética , Receptores de Superficie Celular/fisiología , Receptores de Factores de Crecimiento/efectos de los fármacos , Receptores de Factores de Crecimiento/genética , Receptores de Péptidos/efectos de los fármacos , Receptores de Péptidos/genética , Receptores de Factores de Crecimiento Transformadores beta , Proteínas Recombinantes/metabolismo , Proteínas Recombinantes/farmacología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal/efectos de los fármacos , Proteínas Smad , Proteína Smad1 , Transfección , Células Tumorales Cultivadas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA