Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
1.
Cell Physiol Biochem ; 51(2): 961-978, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30466106

RESUMEN

BACKGROUND/AIMS: Interferon consensus sequence-binding protein 8 (IRF8) belongs to a family of interferon (IFN) regulatory factors that modulates various important physiological processes including carcinogenesis. As reported by others and our group, IRF8 expression is silenced by DNA methylation in both human solid tumors and hematological malignancies. However, the role of IRF8 in lung carcinoma remains elusive. In this study, we determined IRF8 epigenetic regulation, biological functions, and the signaling pathway involved in non-small cell lung cancer (NSCLC). METHODS: IRF8 expression were determined by Q- PCR. MSP and A+T determined promotor methylation. MTS, clonogenic, Transwell assay, Flow cytometry, three-dimensional culture and AO/EB stain verified cell function. In vivo tumorigenesis examed the in vivo effects. By Chip-QPCR, RT-PCR, Western blot and Immunofluorescence staining, the mechanisms were studied. RESULTS: IRF8 was significantly downregulated in lung tumor tissues compared with adjacent non-cancerous tissues. Furthermore, methylation-specific PCR analyses revealed that IRF8 methylation in NSCLC was a common event, and demethylation reagent treatment proved that downregulation of IRF8 was due to its promoter CpG hypermethylation. Clinical data showed that the IRF8 methylation was associated with tumor stage, lymph node metastasis status, patient outcome, and tumor histology. Exogenous expression of IRF8 in the silenced or downregulated lung cancer cell lines A549 and H1299 at least partially restored the sensitivity of lung cancer cells to apoptosis, and arrested cells at the G0/G1 phase. Cell viability, clonogenicity, and cell migration and invasive abilities were strongly inhibited by restored expression of IRF8. A three-dimensional culture system demonstrated that IRF8 changed the cells to a more spherical phenotype. Moreover, ectopic expression of IRF8 enhanced NSCLC chemosensitivity to cisplatin. Furthermore, as verified by Chip-qPCR, immunofluorescence staining, and western blotting, IRF8 bound to the T-cell factor/lymphoid enhancer factor (TCF /LEF) promoter, thus repressing ß-catenin nuclear translocation and its activation. IRF8 significantly disrupted the effects of Wnt agonist, bml284, further suggesting its involvement in the Wnt/ß-catenin pathway. CONCLUSION: IRF8 acted as a tumor suppressor gene through the transcriptional repression of ß-catenin-TCF/LEF in NSCLC. IRF8 methylation may serve as a potential biomarker in NSCLC prognosis.


Asunto(s)
Factores Reguladores del Interferón/metabolismo , Vía de Señalización Wnt , Anciano , Animales , Apoptosis/efectos de los fármacos , Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Carcinoma de Pulmón de Células no Pequeñas/patología , Movimiento Celular/efectos de los fármacos , Cisplatino/farmacología , Metilación de ADN , Femenino , Puntos de Control de la Fase G1 del Ciclo Celular/efectos de los fármacos , Humanos , Factores Reguladores del Interferón/genética , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/patología , Factor de Unión 1 al Potenciador Linfoide/química , Factor de Unión 1 al Potenciador Linfoide/genética , Factor de Unión 1 al Potenciador Linfoide/metabolismo , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Persona de Mediana Edad , Estadificación de Neoplasias , Regiones Promotoras Genéticas , Proteínas Wnt/agonistas , Proteínas Wnt/metabolismo , Vía de Señalización Wnt/efectos de los fármacos , beta Catenina/metabolismo
2.
Cell Physiol Biochem ; 48(1): 87-98, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30001529

RESUMEN

BACKGROUND/AIMS: Chemoresistance is largely responsible for relapses of bladder cancer during clinical therapy. However, the molecular mechanisms involved in the chemoresistance of bladder cancer are unclear. Growing evidence supports the theory that microRNAs (miRNAs) play an important role in chemotherapeutic drug resistance because they are downregulated in many malignancies that have been implicated in the regulation of diverse processes in cancer cells. More specifically, the extent and precise mechanism of the involvement of miR-34as in chemoresistance to epirubicin (EPI) in the treatment of bladder cancer remains unclear. METHODS: In this study, real-time quantitative polymerase chain reaction (PCR) was used to analyze the expression of miR-34a in bladder cancer cell line BIU87 and its EPI chemoresistant cell line BIU87/ADR. The miR-34a profiles in bladder cancer tissues were obtained from The Cancer Genome Atlas database. The effect of miR-34a on chemosensitivity was evaluated by cell viability assays, colony formation assays, and in vivo experimentation. Apoptosis and the cell cycle were examined by flow cytometry. A luciferase reporter assay was used to assess the target genes of miR-34a. Western blot and qPCR were used to analyze the expression of target proteins and downstream molecules. RESULTS: The downregulation of miR-34a in bladder cancer serves as an independent predictor of reduced patient survival. The CCK-8 assay showed that miR-34a overexpression resulted in increased sensitivity to EPI, while miR-34a downregulation resulted in chemoresistance to EPI in vitro. Moreover, it was found that miR-34a increased the sensitivity of BIU87/ADR cells to chemotherapy in vivo. The luciferase reporter assay ascertained that TCF1 and LEF1 are direct target genes of miR-34a. It was found that miR-34a increased chemosensitivity in BIU87/ADR cells by inhibiting the TCF1/LEF1 axis. CONCLUSIONS: The results of this study indicate that miR-34a contributes to the chemosensitivity of BIU87/ADR by inhibiting the TCF1/LEF1 axis. Consequently, miR-34a is a determinant of BIU87 chemosensitivity and may therefore serve as a potential therapeutic target in bladder cancer treatment.


Asunto(s)
Factor Nuclear 1-alfa del Hepatocito/metabolismo , Factor de Unión 1 al Potenciador Linfoide/metabolismo , MicroARNs/metabolismo , Neoplasias de la Vejiga Urinaria/patología , Regiones no Traducidas 3' , Animales , Antagomirs/metabolismo , Apoptosis , Línea Celular Tumoral , Bases de Datos Genéticas , Regulación hacia Abajo , Resistencia a Antineoplásicos/efectos de los fármacos , Epirrubicina/farmacología , Puntos de Control de la Fase G1 del Ciclo Celular , Factor Nuclear 1-alfa del Hepatocito/química , Factor Nuclear 1-alfa del Hepatocito/genética , Humanos , Estimación de Kaplan-Meier , Factor de Unión 1 al Potenciador Linfoide/química , Factor de Unión 1 al Potenciador Linfoide/genética , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , MicroARNs/antagonistas & inhibidores , MicroARNs/genética , Metástasis de la Neoplasia , Neoplasias de la Vejiga Urinaria/genética , Neoplasias de la Vejiga Urinaria/mortalidad
3.
Phys Chem Chem Phys ; 14(2): 538-45, 2012 Jan 14.
Artículo en Inglés | MEDLINE | ID: mdl-22089506

RESUMEN

Lymphoid enhancer-binding factor-1 (LEF-1) is a sequence-specific and cell type-specific transcription factor in regulation of the human T cell receptor α enhancer. It has been shown the minor groove of DNA can bind the intrinsic disordered LEF. To get an insight into the mechanism of how the intrinsic disordered LEF specifically recognizes DNA, we have performed explicit-solvent multiple molecular dynamics (MD) simulations to study the specific recognition between DNA and LEF. Room-temperature MD simulations suggest that the disordered C-tails of LEF have nonspecific and specific interactions with the minor groove of DNA. Kinetic analysis of high-temperature MD simulations shows that bound and apo-states unfold via a two-state process. The specific binding of the disordered C-tails of LEF can accelerate the formation of a complex. Gly38Ala and Met11Gly mutant simulations show that electrostatic interactions between DNA and LEF significantly decrease. Kolmogorov-Smirnov (KS) P test analysis illustrates that the specific recognition between DNA and LEF might follow an induced-fit mechanism. Furthermore, these methods can be used for the research of specific recognition between DNA and other intrinsic disordered proteins.


Asunto(s)
ADN/metabolismo , Factor de Unión 1 al Potenciador Linfoide/metabolismo , ADN/química , Humanos , Cinética , Factor de Unión 1 al Potenciador Linfoide/química , Simulación de Dinámica Molecular , Unión Proteica , Estructura Terciaria de Proteína , Desplegamiento Proteico , Temperatura
4.
Crit Rev Eukaryot Gene Expr ; 21(3): 207-36, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-22111711

RESUMEN

Wnt signaling controls cell specification and fate during development and adult tissue homeostasis by converging on a small family of DNA binding factors, the T-cell factor/lymphoid enhancer factor (TCF/LEF) family. In response to Wnt signals, TCF/LEF members undergo a transcriptional switch from repression to activation mediated in part by nuclear ß-catenin binding and recruitment of co-activator complexes. In mammals, the specificity and fine tuning of this transcriptional switch is also achieved by the cell-context-dependent expression of four members (TCF7, TCF7L1, TCF7L2, and LEF1) and numerous variants, which display differential DNA binding affinity and specificity, repression strength, activation potential, and regulators. TCF7/LEF1 variants are generated by alternative promoters, alternative exon cassettes, and alternative donor/acceptor splicing sites, allowing combinatorial insertion/exclusion of modular functional and regulatory domains. In this review we present mounting evidence for the interdependency of TCF7/LEF1 variant expression and functions with cell lineage and cell state. We also illustrate how the p53 and nuclear receptor family of transcription factors, known to control cell fate and to inhibit Wnt signaling, may participate in the fine tuning of TCF7/LEF1 repression/activation potentials.


Asunto(s)
Factor de Unión 1 al Potenciador Linfoide/genética , Factor de Unión 1 al Potenciador Linfoide/metabolismo , Factor 1 de Transcripción de Linfocitos T/genética , Factor 1 de Transcripción de Linfocitos T/metabolismo , Empalme Alternativo , Animales , Expresión Génica , Humanos , Factor de Unión 1 al Potenciador Linfoide/química , Mamíferos/embriología , Mamíferos/metabolismo , Neoplasias/metabolismo , Factor 1 de Transcripción de Linfocitos T/química , Factores de Transcripción TCF/química , Factores de Transcripción TCF/genética , Factores de Transcripción TCF/metabolismo , Vía de Señalización Wnt
5.
Blood ; 118(24): 6362-7, 2011 Dec 08.
Artículo en Inglés | MEDLINE | ID: mdl-22010100

RESUMEN

Aberrant activation of the Wnt pathway plays a pathogenetic role in various tumors and has been associated with adverse outcome in acute lymphoblastic leukemia (ALL). LEF1, a key mediator of Wnt signaling, has been linked to leukemic transformation, and recurrent mutations of LEF1 have been identified in pediatric T-ALL. Here we evaluated the prognostic significance of LEF1 expression in B-precursor ALL patients. LEF1 expression was determined by quantitative real-time RT-PCR in 282 adult B-precursor ALL patients treated on 06/99 and 07/03 GMALL trials. Patients were grouped into quartiles (Q1-Q4) according to LEF1 expression levels (LEF1 high, Q4; n = 71; LEF1 low, Q1-Q3; n = 211). Patients with high LEF1 expression had a significantly shorter relapse-free survival (RFS) compared with low LEF1 expressers (5-year RFS: LEF1 high, 27%; LEF1 low, 47%; P = .05). Importantly, high LEF1 expression was also associated with inferior RFS in standard-risk patients and was independently predictive for RFS (P = .02) in multivariate analyses for this subgroup. Thus, high LEF1 expression identifies B-precursor ALL patients with inferior RFS, supporting a pathogenetic role of Wnt signaling in ALL. Standard-risk patients with high LEF1 expression might benefit from early treatment modifications and new molecular therapies, including agents targeting the Wnt pathway.


Asunto(s)
Médula Ósea/metabolismo , Factor de Unión 1 al Potenciador Linfoide/metabolismo , Proteínas de Neoplasias/metabolismo , Leucemia-Linfoma Linfoblástico de Células Precursoras B/metabolismo , Regulación hacia Arriba , Adolescente , Adulto , Anciano , Biomarcadores de Tumor/metabolismo , Estudios de Cohortes , Exones , Femenino , Humanos , Factor de Unión 1 al Potenciador Linfoide/química , Factor de Unión 1 al Potenciador Linfoide/genética , Masculino , Persona de Mediana Edad , Proteínas Mutantes/química , Proteínas Mutantes/metabolismo , Proteínas de Neoplasias/química , Proteínas de Neoplasias/genética , Leucemia-Linfoma Linfoblástico de Células Precursoras B/diagnóstico , Leucemia-Linfoma Linfoblástico de Células Precursoras B/fisiopatología , Leucemia-Linfoma Linfoblástico de Células Precursoras B/terapia , Pronóstico , ARN Mensajero/metabolismo , Recurrencia , Inducción de Remisión , Análisis de Supervivencia , Adulto Joven
6.
J Mol Biol ; 405(2): 519-30, 2011 Jan 14.
Artículo en Inglés | MEDLINE | ID: mdl-21075118

RESUMEN

In the Wnt/ß-catenin signaling pathway, ß-catenin activates target genes through its interactions with the T-cell factor/lymphoid enhancer-binding factor (TCF/Lef) family of transcription factors. The crystal structures of complexes between the ß-catenin armadillo domain and the Lef-1 N-terminal domain show that the overall conformation and many of the interactions are similar to other published structures of TCFs bound to ß-catenin. However, a second salt bridge in other TCF-ß-catenin structures is absent in the structure of ß-catenin-Lef-1 complex, indicating that this feature is not obligatory for ß-catenin binding. Casein kinase II (CK2) has been shown to act as a positive regulator of Wnt signaling, and Lef-1 is a substrate of CK2. In vitro phosphorylation of purified Lef-1 was used to examine the effect of CK2 on the interaction of Lef-1 with ß-catenin. Mass spectrometry data show that CK2 phosphorylation of Lef-1 N-terminal domain results in a single phosphorylation site at Ser40. Isothermal titration calorimetry revealed that ß-catenin binds to nonphosphorylated or CK2-phosphorylated Lef-1 with the same affinity, which is consistent with the absence of phospho-Ser40 interactions in the crystal structure of phosphorylated Lef-1 N-terminal domain bound to ß-catenin. These data indicate that the effect of CK2 on the Wnt/ß-catenin pathway does not appear to be at the level of the Lef-1-ß-catenin interaction.


Asunto(s)
Quinasa de la Caseína II/química , Quinasa de la Caseína II/metabolismo , Factor de Unión 1 al Potenciador Linfoide/química , Factor de Unión 1 al Potenciador Linfoide/metabolismo , beta Catenina/química , beta Catenina/metabolismo , Secuencia de Aminoácidos , Animales , Células Cultivadas , Humanos , Ratones , Datos de Secuencia Molecular , Fosforilación , Unión Proteica , Conformación Proteica , Mapeo de Interacción de Proteínas , Homología de Secuencia de Aminoácido , Transducción de Señal , Factores de Transcripción TCF/metabolismo , Transcripción Genética
7.
J Neurosci ; 30(14): 4957-69, 2010 Apr 07.
Artículo en Inglés | MEDLINE | ID: mdl-20371816

RESUMEN

beta-Catenin, together with LEF1/TCF transcription factors, activates genes involved in the proliferation and differentiation of neuronal precursor cells. In mature neurons, beta-catenin participates in dendritogenesis and synaptic function as a component of the cadherin cell adhesion complex. However, the transcriptional activity of beta-catenin in these cells remains elusive. In the present study, we found that in the adult mouse brain, beta-catenin and LEF1 accumulate in the nuclei of neurons specifically in the thalamus. The particular electrophysiological properties of thalamic neurons depend on T-type calcium channels. Cav3.1 is the predominant T-type channel subunit in the thalamus, and we hypothesized that the Cacna1g gene encoding Cav3.1 is a target of the LEF1/beta-catenin complex. We demonstrated that the expression of Cacna1g is high in the thalamus and is further increased in thalamic neurons treated in vitro with LiCl or WNT3A, activators of beta-catenin. Luciferase reporter assays confirmed that the Cacna1G promoter is activated by LEF1 and beta-catenin, and footprinting analysis revealed four LEF1 binding sites in the proximal region of this promoter. Chromatin immunoprecipitation demonstrated that the Cacna1g proximal promoter is occupied by beta-catenin in vivo in the thalamus, but not in the hippocampus. Moreover, WNT3A stimulation enhanced T-type current in cultured thalamic neurons. Together, our data indicate that the LEF1/beta-catenin complex regulates transcription of Cacna1g and uncover a novel function for beta-catenin in mature neurons. We propose that beta-catenin contributes to neuronal excitability not only by a local action at the synapse but also by activating gene expression in thalamic neurons.


Asunto(s)
Canales de Calcio Tipo T/genética , Factor de Unión 1 al Potenciador Linfoide/fisiología , Neuronas/fisiología , Tálamo/fisiología , Activación Transcripcional/fisiología , beta Catenina/fisiología , Factores de Edad , Animales , Canales de Calcio Tipo T/biosíntesis , Canales de Calcio Tipo T/química , Células Cultivadas , Femenino , Células HeLa , Humanos , Factor de Unión 1 al Potenciador Linfoide/química , Masculino , Ratones , Ratas , Sinapsis/química , Sinapsis/genética , Sinapsis/metabolismo , beta Catenina/química
8.
Cytogenet Genome Res ; 124(2): 139-46, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-19420926

RESUMEN

The lymphoid enhancer-binding factor-1 (LEF1) belongs to a family of regulatory proteins that share homology with the high mobility group protein-1 (HMG1). The LEF1 gene is a mediator in the canonical Wnt-signalling pathway required for morphogenesis of early mammary gland during embryogenesis. Here we describe the molecular characterisation of the porcine LEF1 gene and its association with number of teats and inverted teats in experimental and commercial populations. The 2357-bp cDNA sequence contains an 1197-bp open reading frame encoding a protein of 398 amino acids. The porcine LEF1 protein shares high identity with LEF1 in other mammalian species. The LEF1 gene contains 12 exons and maps to pig chromosome 8 (SSC8). We identified two single nucleotide polymorphisms (SNPs), a T1351C transition and an A1666C transversion, in the 3' end of LEF1. Associations of the SNP A1666C with presence of inverted teats (P

Asunto(s)
Factor de Unión 1 al Potenciador Linfoide/genética , Glándulas Mamarias Animales/metabolismo , Sus scrofa/genética , Secuencia de Aminoácidos , Animales , Mapeo Cromosómico , Cromosomas de los Mamíferos/genética , ADN Complementario/genética , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Frecuencia de los Genes , Haplotipos , Humanos , Factor de Unión 1 al Potenciador Linfoide/química , Datos de Secuencia Molecular , Fenotipo , Carácter Cuantitativo Heredable
9.
J Am Chem Soc ; 131(9): 3342-8, 2009 Mar 11.
Artículo en Inglés | MEDLINE | ID: mdl-19216569

RESUMEN

The screening of a >9000 compound library of synthetic DNA binding molecules for selective binding to the consensus sequence of the transcription factor LEF-1 followed by assessment of the candidate compounds in a series of assays that characterized functional activity (disruption of DNA-LEF-1 binding) at the intended target and site (inhibition of intracellular LEF-1-mediated gene transcription) resulting in a desired phenotypic cellular change (inhibit LEF-1-driven cell transformation) provided two lead compounds: lefmycin-1 and lefmycin-2. The sequence of screens defining the approach assures that activity in the final functional assay may be directly related to the inhibition of gene transcription and DNA binding properties of the identified molecules. Central to the implementation of this generalized approach to the discovery of DNA binding small molecule inhibitors of gene transcription was (1) the use of a technically nondemanding fluorescent intercalator displacement (FID) assay for initial assessment of the DNA binding affinity and selectivity of a library of compounds for any sequence of interest, and (2) the technology used to prepare a sufficiently large library of DNA binding compounds.


Asunto(s)
Transformación Celular Neoplásica/efectos de los fármacos , Neoplasias del Colon/metabolismo , ADN/efectos de los fármacos , Descubrimiento de Drogas , Factor de Unión 1 al Potenciador Linfoide/antagonistas & inhibidores , Transcripción Genética/efectos de los fármacos , Animales , Línea Celular , Transformación Celular Neoplásica/genética , Pollos , Neoplasias del Colon/genética , ADN/química , Relación Dosis-Respuesta a Droga , Ensayos de Selección de Medicamentos Antitumorales , Ensayo de Cambio de Movilidad Electroforética , Ensayos Analíticos de Alto Rendimiento , Humanos , Factor de Unión 1 al Potenciador Linfoide/química , Factor de Unión 1 al Potenciador Linfoide/metabolismo , Estructura Molecular , Bibliotecas de Moléculas Pequeñas , Transcripción Genética/genética
10.
J Inorg Biochem ; 102(2): 242-50, 2008 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-17961652

RESUMEN

The lymphoid enhancer-binding factor 1 (LEF-1) recognizes a double-stranded 9 base-pairs (bp) long motif in DNA which is significantly bent upon binding. This bend is centered at two destacked adenines whose geometry closely resembles that of two adjacent guanines crosslinked by the antitumor drug cisplatin. It has been proposed that cisplatin-GG crosslinks could hijack high mobility group (HMG) box containing transcription factors such as LEF-1. In order to examine such a possibility, we used electrophoretic mobility shift assays to determine the affinity of the HMG box of LEF-1 for a series of 25 oligonucleotides containing a central GG sequence, free or site-specifically modified by cisplatin. The binding affinity of the GG-platinated oligonucleotides was 3-6-fold higher than that determined for the corresponding unplatinated oligonucleotides, however, the binding to all cisplatin-modified oligonucleotides was at least 1 order of magnitude weaker than that to the 25 bp oligonucleotide containing the recognition 9 bp motif. The binding affinity was dependent on the nature of bases flanking the cisplatin-crosslinked G(*)G(*) dinucleotide, the AG(*)G(*)T sequence displaying the strongest affinity and CG(*)G(*)T showing the strongest binding enhancement upon platination. In contrast, modification of the AGGT sequence with the third-generation platinum antitumor drug oxaliplatin did not enhance the affinity significantly. These results suggest that the cisplatin-caused bending of DNA does produce a target for LEF-1 binding, however, the cisplatinated DNA does not appear to be a strong competitor for the LEF-1 recognition sequence.


Asunto(s)
Cisplatino/metabolismo , Aductos de ADN/metabolismo , ADN/metabolismo , Proteínas del Grupo de Alta Movilidad/metabolismo , Factor de Unión 1 al Potenciador Linfoide/metabolismo , Secuencia de Bases , ADN/química , Aductos de ADN/química , Proteínas de Unión al ADN/química , Proteínas de Unión al ADN/metabolismo , Guanina/metabolismo , Factor de Unión 1 al Potenciador Linfoide/química , Oligonucleótidos/metabolismo , Platino (Metal)/metabolismo
11.
Mol Cell Biol ; 27(21): 7560-73, 2007 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-17785445

RESUMEN

Lef-1 and PITX2 function in the Wnt signaling pathway by recruiting and interacting with beta-catenin to activate target genes. Chromatin immunoprecipitation (ChIP) assays identified the Lef-1 promoter as a PITX2 downstream target. Transgenic mice expressing LacZ driven by the 2.5-kb LEF-1 promoter demonstrated expression in the tooth epithelium correlated with endogenous Lef-1 FL epithelial expression. PITX2 isoforms regulate the LEF-1 promoter, and beta-catenin synergistically enhanced activation of the LEF-1 promoter in combination with PITX2 and Lef-1 isoforms. PITX2 enhances endogenous expression of the full-length beta-catenin-dependent Lef-1 isoform (Lef-1 FL) while decreasing expression of the N-terminally truncated beta-catenin-independent isoform. Our research revealed a novel interaction between PITX2, Lef-1, and beta-catenin in which the Lef-1 beta-catenin binding domain is dispensable for its interaction with PITX2. PITX2 interacts with two sites within the Lef-1 protein. Furthermore, beta-catenin interacts with the PITX2 homeodomain and Lef-1 interacts with the PITX2 C-terminal tail. Lef-1 and beta-catenin interact simultaneously and independently with PITX2 through two different sites to regulate PITX2 transcriptional activity. These data support a role for PITX2 in cell proliferation, migration, and cell division through differential Lef-1 isoform expression and interactions with Lef-1 and beta-catenin.


Asunto(s)
Proteínas de Homeodominio/metabolismo , Factor de Unión 1 al Potenciador Linfoide/metabolismo , Factores de Transcripción/metabolismo , beta Catenina/metabolismo , Animales , Células CHO , Cricetinae , Cricetulus , Femenino , Regulación del Desarrollo de la Expresión Génica , Proteínas de Homeodominio/genética , Humanos , Incisivo/embriología , Incisivo/metabolismo , Factor de Unión 1 al Potenciador Linfoide/química , Factor de Unión 1 al Potenciador Linfoide/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Mutación/genética , Regiones Promotoras Genéticas/genética , Unión Proteica , Isoformas de Proteínas/química , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Estructura Terciaria de Proteína , Factores de Transcripción/genética , Transcripción Genética , Proteína del Homeodomínio PITX2
12.
Nucleic Acids Res ; 34(20): 5951-65, 2006.
Artículo en Inglés | MEDLINE | ID: mdl-17068080

RESUMEN

Dlx2, Lymphoid Enhancer Factor (Lef-1) and Msx2 transcription factors are required for several developmental processes. To understand the control of gene expression by these factors, chromatin immunoprecipitation (ChIP) assays identified Msx2 as a downstream target of Dlx2 and Lef-1. Dlx2 activates the Msx2 promoter in several cell lines and binds DNA as a monomer and dimer. A Lef-1 beta-catenin-dependent isoform minimally activates the Msx2 promoter and a Lef-1 beta-catenin-independent isoform is inactive, however co-expression of Dlx2 and both Lef-1 isoforms synergistically activate the Msx2 promoter. Co-immunoprecipitation and protein pull-down experiments demonstrate Lef-1 physically interacts with Dlx2. Deletion analyses of the Lef-1 protein reveal specific regions required for synergism with Dlx2. The Lef-1 beta-catenin binding domain (betaDB) is not required for its interaction with Dlx2. Msx2 can auto-regulate its promoter and repress Dlx2 activation. Msx2 repression of Dlx2 activation is dose-specific and both bind a common DNA-binding element. These transcriptional mechanisms correlate with the temporal and spatial expression of these factors and may provide a mechanism for the control of several developmental processes. We demonstrate new transcriptional activities for Dlx2, Msx2 and Lef-1 through protein interactions and identification of downstream targets.


Asunto(s)
Proteínas de Unión al ADN/genética , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Factor de Unión 1 al Potenciador Linfoide/metabolismo , Factores de Transcripción/metabolismo , Activación Transcripcional , Animales , Sitios de Unión , Línea Celular , Inmunoprecipitación de Cromatina , Proteínas de Unión al ADN/metabolismo , Factor de Unión 1 al Potenciador Linfoide/química , Regiones Promotoras Genéticas , Estructura Terciaria de Proteína
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA