Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
1.
Clin Cancer Res ; 23(5): 1227-1235, 2017 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-27582484

RESUMEN

Purpose: Neuropathy is the dose-limiting toxicity of paclitaxel and a major cause for decreased quality of life. Genetic factors have been shown to contribute to paclitaxel neuropathy susceptibility; however, the major causes for interindividual differences remain unexplained. In this study, we identified genetic markers associated with paclitaxel-induced neuropathy through massive sequencing of candidate genes.Experimental Design: We sequenced the coding region of 4 EPHA genes, 5 genes involved in paclitaxel pharmacokinetics, and 30 Charcot-Marie-Tooth genes, in 228 cancer patients with no/low neuropathy or high-grade neuropathy during paclitaxel treatment. An independent validation series included 202 paclitaxel-treated patients. Variation-/gene-based analyses were used to compare variant frequencies among neuropathy groups, and Cox regression models were used to analyze neuropathy along treatment.Results: Gene-based analysis identified EPHA6 as the gene most significantly associated with paclitaxel-induced neuropathy. Low-frequency nonsynonymous variants in EPHA6 were present exclusively in patients with high neuropathy, and all affected the ligand-binding domain of the protein. Accumulated dose analysis in the discovery series showed a significantly higher neuropathy risk for EPHA5/6/8 low-frequency nonsynonymous variant carriers [HR, 14.60; 95% confidence interval (CI), 2.33-91.62; P = 0.0042], and an independent cohort confirmed an increased neuropathy risk (HR, 2.07; 95% CI, 1.14-3.77; P = 0.017). Combining the series gave an estimated 2.5-fold higher risk of neuropathy (95% CI, 1.46-4.31; P = 9.1 × 10-4).Conclusions: This first study sequencing EPHA genes revealed that low-frequency variants in EPHA6, EPHA5, and EPHA8 contribute to the susceptibility to paclitaxel-induced neuropathy. Furthermore, EPHA's neuronal injury repair function suggests that these genes might constitute important neuropathy markers for many neurotoxic drugs. Clin Cancer Res; 23(5); 1227-35. ©2016 AACR.


Asunto(s)
Paclitaxel/efectos adversos , Enfermedades del Sistema Nervioso Periférico/genética , Receptor EphA5/genética , Receptor EphA6/genética , Receptor EphA8/genética , Adulto , Anciano , Biomarcadores Farmacológicos/análisis , Neoplasias de la Mama/complicaciones , Neoplasias de la Mama/tratamiento farmacológico , Femenino , Estudios de Asociación Genética , Humanos , Persona de Mediana Edad , Neoplasias Ováricas/complicaciones , Neoplasias Ováricas/tratamiento farmacológico , Paclitaxel/administración & dosificación , Enfermedades del Sistema Nervioso Periférico/inducido químicamente , Enfermedades del Sistema Nervioso Periférico/patología , Polimorfismo de Nucleótido Simple , Modelos de Riesgos Proporcionales , Calidad de Vida
2.
FEBS Lett ; 589(6): 756-65, 2015 Mar 12.
Artículo en Inglés | MEDLINE | ID: mdl-25683004

RESUMEN

MicroRNAs (miRNAs) play a critical role in the development of cancers. However, the role of miRNAs in glioma is still poorly understood. In this study, we demonstrate that microRNA-10a (miR-10a) promotes cell migration and invasion by negatively regulating the expression of Eph tyrosine kinase receptor A8 (EphA8). Ectopic expression of EphA8 counteracts the promotion of migration and invasion induced by miR-10a. We further demonstrate that miR-10a and EphA8 regulate epithelial-mesenchymal transition (EMT) to affect cell migration and invasion. Collectively, we unveil a branch of the miR-10a/EphA8 pathway that regulates the progression of glioma.


Asunto(s)
Movimiento Celular , Transición Epitelial-Mesenquimal , Glioma/patología , MicroARNs/fisiología , Receptor EphA8/genética , Regiones no Traducidas 3' , Secuencia de Bases , Sitios de Unión , Línea Celular Tumoral , Expresión Génica , Regulación Neoplásica de la Expresión Génica , Humanos , Datos de Secuencia Molecular , Invasividad Neoplásica , Interferencia de ARN , Receptor EphA8/metabolismo , Regulación hacia Arriba
3.
Exp Mol Pathol ; 96(3): 346-53, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24613686

RESUMEN

Chronic occupational benzene exposure is associated with an increased risk of hematological malignancies such as aplastic anemia and leukemia. The new biomarker and action mechanisms of chronic benzene poisoning are still required to be explored. Aberrant DNA methylation, which may lead to genomic instability and the altered gene expression, is frequently observed in hematological cancers. To gain an insight into the new biomarkers and molecular mechanisms of chronic benzene poisoning, DNA methylation profiles and mRNA expression pattern from the peripheral blood mononuclear cells of four chronic benzene poisoning patients and four health controls that matched age and gender without benzene exposure were performed using the high resolution Infinium 450K methylation array and Gene Chip Human Gene 2.0ST Arrays, respectively. By integrating DNA methylation and mRNA expression data, we identified 3 hypermethylated genes showing concurrent down-regulation (PRKG1, PARD3, EPHA8) and 2 hypomethylated genes showing increased expression (STAT3, IFNGR1). Signal net analysis of differential methylation genes associated with chronic benzene poisoning showed that two key hypomethylated STAT3 and hypermethylated GNAI1 were identified. Further GO analysis and pathway analysis indicated that hypomethylated STAT3 played central roles through regulation of transcription, DNA-dependent, positive regulation of transcription from RNA polymerase II promoter, JAK-STAT cascade and adipocytokine signaling pathway, Acute myeloid leukemia, and JAK-STAT signaling pathway. In conclusion, the aberrant hypomethylated STAT3 might be a potential biomarker of chronic benzene poisoning.


Asunto(s)
Benceno/envenenamiento , Metilación de ADN , Leucocitos Mononucleares/metabolismo , Factor de Transcripción STAT3/metabolismo , Proteínas Adaptadoras Transductoras de Señales , Adipoquinas/genética , Adulto , Biomarcadores/metabolismo , Estudios de Casos y Controles , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Proteína Quinasa Dependiente de GMP Cíclico Tipo I/genética , Proteína Quinasa Dependiente de GMP Cíclico Tipo I/metabolismo , Regulación hacia Abajo , Subunidades alfa de la Proteína de Unión al GTP Gi-Go/genética , Subunidades alfa de la Proteína de Unión al GTP Gi-Go/metabolismo , Expresión Génica , Humanos , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Persona de Mediana Edad , Exposición Profesional/efectos adversos , Análisis de Secuencia por Matrices de Oligonucleótidos , Regiones Promotoras Genéticas , ARN Mensajero/genética , Receptor EphA8/genética , Receptor EphA8/metabolismo , Receptores de Interferón/genética , Receptores de Interferón/metabolismo , Factor de Transcripción STAT3/genética , Transducción de Señal , Receptor de Interferón gamma
4.
Clin Exp Immunol ; 173(1): 76-83, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23607884

RESUMEN

Rodent models for arthritis implicate a role for complement in disease development and progression. In humans, complement deposition has been observed in inflamed synovia of rheumatoid arthritis (RA) patients. In this study we analysed whether genetic variants of complement component C1q predispose to RA. We genotyped single nucleotide polymorphisms (SNPs) in and around the C1q genes, C1qA, C1qB and C1qC, in a Dutch set of 845 RA cases and 1046 controls. Replication was sought in a sample set from North America (868 cases/1193 controls), and a meta-analysis was performed in a combined samples set of 8000 cases and 23 262 controls of European descent. We determined C1q serum levels in relation to C1q genotypes. In the discovery phase, five of the 13 SNPs tested in the C1q genes showed a significant association with RA. Additional analysis of the genomic area around the C1q genes revealed that the strongest associating SNPs were confined to the C1q locus. Within the C1q locus we observed no additional signal independent of the strongest associating SNP, rs292001 [odds ratio (OR) = 0·72 (0·58-0·88), P = 0·0006]. The variants of this SNP were associated with different C1q serum levels in healthy controls (P = 0·006). Interestingly, this SNP was also associated significantly in genome-wide association studies (GWAS) from the North American Rheumatoid Arthritis Consortium study, confirming the association with RA [OR = 0·83 (0·69-1·00), P = 0·043]. Combined analysis, including integrated data from six GWAS studies, provides support for the genetic association. Genetic variants in C1q are correlated with C1q levels and may be a risk for the development of RA.


Asunto(s)
Artritis Reumatoide/genética , Complemento C1q/genética , Polimorfismo de Nucleótido Simple , Artritis Reumatoide/epidemiología , Canadá/epidemiología , Estudios de Cohortes , Predisposición Genética a la Enfermedad , Estudio de Asociación del Genoma Completo , Genotipo , Grecia/epidemiología , Humanos , Países Bajos/epidemiología , ARN Mensajero/genética , Receptor EphA8/genética , Receptor EphB2/genética , Estados Unidos/epidemiología
5.
PLoS One ; 7(1): e30575, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22291991

RESUMEN

The Rab family of small guanosine triphosphatases (GTPases) plays a vital role in membrane trafficking. Its active GTP-bound state is driven by guanine nucleotide-exchange factors (GEFs). Ras and Rab interactor (or Ras interaction/interference)-like (RINL), which contains a conserved VPS9 domain critical for GEF action, was recently identified as a new Rab5 subfamily GEF in vitro. However, its detailed function and interacting molecules have not yet been fully elucidated. Here we found that RINL has GEF activity for the Rab5 subfamily proteins by measuring their GTP-bound forms in cultured cells. We also found that RINL interacts with odin, a member of the ankyrin-repeat and sterile-alpha motif (SAM) domain-containing (Anks) protein family. In addition, the Eph tyrosine kinase receptor EphA8 formed a ternary complex with both RINL and odin. Interestingly, RINL expression in cultured cells reduced EphA8 levels in a manner dependent on both its GEF activity and interaction with odin. In addition, knockdown of RINL increased EphA8 level in HeLa cells. Our findings suggest that RINL, as a GEF for Rab5 subfamily, is implicated in the EphA8-degradation pathway via its interaction with odin.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/fisiología , Factores de Intercambio de Guanina Nucleótido/fisiología , Proteolisis , Receptor EphA8/metabolismo , Proteínas de Unión al GTP rab/fisiología , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Animales , Células Cultivadas , Factores de Intercambio de Guanina Nucleótido/genética , Factores de Intercambio de Guanina Nucleótido/metabolismo , Células HEK293 , Células HeLa , Humanos , Modelos Biológicos , Familia de Multigenes , Receptor EphA8/genética , Transducción de Señal/genética , Spodoptera , Transfección , Proteínas de Unión al GTP rab/genética , Proteínas de Unión al GTP rab/metabolismo , Proteínas de Unión al GTP rab5/genética , Proteínas de Unión al GTP rab5/metabolismo
6.
Mol Cells ; 29(6): 603-9, 2010 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-20496116

RESUMEN

Recent studies indicate that endocytosis of Eph-ephrin complexes may be one of the mechanisms by which a high affinity cell-cell adhesion is converted to a repulsive interaction. In this study, we show that EphA8 undergoes clathrin-mediated endocytosis upon treatment with ephrin-A5, and that EphA8 is associated tightly with Tiam-1, a Rac-specific guanine nucleotide exchange factor. Analysis of EphA8 deletion mutants revealed that a juxtamembrane region in EphA8 is critically involved in endocytosis of EphA8-ephrinA5 complexes. An EphA8 mutant lacking this juxtamembrane portion was defective for endocytosis with ephrinA5, and also displayed a weak association with Tiam-1. Expression of an endocytosis-defective version of EphA8 resulted in a low level of Rac activity in response to ephrin-A5 stimulation. More importantly, down-regulation of Tiam-1 resulted in inefficient endocytosis of EphA8-ephrinA5 complexes. These results suggest that Tiam-1 plays a role in clathrin-dependent endocytosis of EphA8-ephrinA5 complexes.


Asunto(s)
Adhesión Celular , Factores de Intercambio de Guanina Nucleótido/metabolismo , Receptor EphA8/metabolismo , Proteínas de Unión al GTP rac/metabolismo , Citoesqueleto de Actina/enzimología , Línea Celular Transformada , Clatrina/metabolismo , Endocitosis/efectos de los fármacos , Endocitosis/genética , Activación Enzimática/efectos de los fármacos , Activación Enzimática/genética , Efrina-A5/farmacología , Factores de Intercambio de Guanina Nucleótido/genética , Humanos , Unión Proteica/efectos de los fármacos , Unión Proteica/genética , Estructura Terciaria de Proteína/genética , Receptor EphA8/genética , Eliminación de Secuencia/genética , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Proteína 1 de Invasión e Inducción de Metástasis del Linfoma-T , Proteínas de Unión al GTP rac/genética
7.
Mol Cell Biol ; 30(7): 1582-92, 2010 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-20100865

RESUMEN

We recently reported that the phosphotyrosine-binding (PTB) domain of Anks family proteins binds to EphA8, thereby positively regulating EphA8-mediated signaling pathways. In the current study, we identified a potential role for the SAM domains of Anks family proteins in EphA signaling. We found that SAM domains of Anks family proteins directly bind to ubiquitin, suggesting that Anks proteins regulate the degradation of ubiquitinated EphA receptors. Consistent with the role of Cbl ubiquitin ligases in the degradation of Eph receptors, our results revealed that the ubiquitin ligase c-Cbl induced the ubiquitination and degradation of EphA8 upon ligand binding. Ubiquitinated EphA8 also bound to the SAM domains of Odin, a member of the Anks family proteins. More importantly, the overexpression of wild-type Odin protected EphA8 and EphA2 from undergoing degradation following ligand stimulation and promoted EphA-mediated inhibition of cell migration. In contrast, a SAM domain deletion mutant of Odin strongly impaired the function of endogenous Odin, suggesting that the mutant functions in a dominant-negative manner. An analysis of Odin-deficient primary embryonic fibroblasts indicated that Odin levels play a critical role in regulating the stability of EphA2 in response to ligand stimulation. Taken together, our studies suggest that the SAM domains of Anks family proteins play a pivotal role in enhancing the stability of EphA receptors by modulating the ubiquitination process.


Asunto(s)
Proteínas Portadoras/metabolismo , Receptor EphA2/metabolismo , Receptor EphA8/metabolismo , Proteínas Adaptadoras Transductoras de Señales , Animales , Proteínas Portadoras/genética , Células Cultivadas , Fibroblastos/citología , Fibroblastos/metabolismo , Humanos , Ratones , Ratones Noqueados , Unión Proteica , Estructura Terciaria de Proteína , Proteínas Proto-Oncogénicas c-cbl/metabolismo , Receptor EphA2/genética , Receptor EphA8/química , Receptor EphA8/genética , Transducción de Señal/fisiología , Técnicas del Sistema de Dos Híbridos , Ubiquitina/metabolismo
8.
J Biochem Mol Biol ; 40(5): 656-61, 2007 Sep 30.
Artículo en Inglés | MEDLINE | ID: mdl-17927897

RESUMEN

In this report, we describe an optimized method for generation of ephA8 BAC transgenic mice expressing the lacZ reporter gene under ephA8 regulatory sequences. First, we constructed a targeting vector that carries a 1.2 kb ephA8 DNA upstream of its first exon, a lacZ expression cassette, a kanamycin cassette, and a 0.7 kb ephA8 DNA downstream of its first exon. Second, the targeting vector was electroporated into cells containing the ephA8 BAC and pKOBEGA, in which recombinases induce a homologous recombination between the ephA8 BAC DNA and the targeting vector. Third, the FLP plasmid expressing the Flipase was electroporated into these bacteria to eliminate a kanamycin cassette from the recombinant BAC DNA. The appropriate structures of the modified ephA8 BAC DNA were confirmed by Southern analysis. Finally, BAC transgenic mouse embryos were generated by pronuclear injection of the recombinant BAC DNA. Whole mount X-gal staining revealed that the lacZ reporter expression is restricted to the anterior region of the developing midbrain in each transgenic embryo. These results indicate that the ephA8 BAC DNA contains most, if not all, regulatory sequences to direct temporal and spatial expression of the lacZ gene in vivo.


Asunto(s)
Cromosomas Artificiales Bacterianos/genética , Operón Lac/genética , Receptor EphA8/genética , Recombinación Genética/genética , Animales , Southern Blotting , Embrión de Mamíferos/metabolismo , Femenino , Ingeniería Genética/métodos , Vectores Genéticos/genética , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Modelos Genéticos , Embarazo
9.
Mol Cell Biol ; 27(5): 1614-30, 2007 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-17178831

RESUMEN

The mouse ephA8 gene is expressed in a rostral-to-caudal gradient in the developing superior colliculus, and these EphA gradients may contribute to the proper development of the retinocollicular projection. Thus, it is of considerable interest to elucidate how the ephA8 gene expression is controlled by upstream regulators during the development of the mesencephalon. In this study, we employed in vivo expression analysis in transgenic mouse embryos to dissect the cis-acting DNA regulatory region, leading to the identification of a CGGTCA sequence critical for the ephA8 enhancer activity. Using this element as the target in a yeast one-hybrid system, we identified a Meis homeobox transcription factor. Significantly, DNA binding sites for Pbx, another TALE homeobox transcription factor, were also identified in the ephA8 enhancer region. Meis2 and Pbx1/2 are specifically expressed in the entire region of the dorsal mesencephalon, where specific colocalization of EphA8 and Meis is restricted to a subset of cells. Meis2 and Pbx2 synergistically bind the ephA8 regulatory sequence in vitro, and this interaction is critical for the transcriptional activation of a reporter construct bearing the ephA8 regulatory region in the presence of histone deacetylase inhibitor. More importantly, when expressed in the embryonic midbrain, the dominant-negative form of Meis down-regulates endogenous ephA8. Interestingly, we found that both Meis2 and Pbx2 are constitutively bound in the ephA8 regulatory region in the dorsal mesencephalon. These studies strongly suggest that Meis and Pbx homeobox transcription factors tightly associate with the ephA8 regulatory sequence and require an additional unidentified regulator to ensure the specific activation of ephA8.


Asunto(s)
Regulación del Desarrollo de la Expresión Génica/fisiología , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Mesencéfalo/embriología , Mesencéfalo/fisiología , Receptor EphA8/genética , Animales , Elementos de Facilitación Genéticos , Ratones , Ratones Transgénicos , Factores de Transcripción/genética , Transgenes
10.
Oncogene ; 24(26): 4243-56, 2005 Jun 16.
Artículo en Inglés | MEDLINE | ID: mdl-15782114

RESUMEN

Recent studies in our laboratory demonstrate that ligand-mediated activation of the EphA8 receptor critically regulates cell adhesion and migration. In this report, we show that the EphA8 receptor induces neurite outgrowth in NG108-15 cells in the absence of ligand stimulation. Using various deletion mutants lacking specific intracytoplasmic regions, we confirm that the tyrosine kinase domain of EphA8 is important for inducing neurite outgrowth. However, the tyrosine kinase activity of EphA8 is not crucial for neurite outgrowth induction. Treatment with various inhibitors further reveals that the mitogen-activated protein kinase (MAPK) signaling pathway is critical for neurite outgrowth induced by EphA8. Consistent with these results, EphA8 expression induced a sustained increase in the activity of MAPK, whereas ligand-mediated EphA8 activation had no further modulatory effects on MAP kinase activity. Additionally, activated MAPK relocalized from the cytoplasm to the nucleus in response to EphA8 transfection. These results collectively suggest that the EphA8 receptor is capable of inducing a sustained increase in MAPK activity, thereby promoting neurite outgrowth in neuronal cells.


Asunto(s)
Proteínas Quinasas Activadas por Mitógenos/biosíntesis , Proteínas Quinasas Activadas por Mitógenos/farmacología , Neuritas , Receptor EphA8/genética , Receptor EphA8/fisiología , Animales , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/patología , Neoplasias Encefálicas/veterinaria , Activación Enzimática , Glioma/genética , Glioma/patología , Glioma/veterinaria , Ratones , Proteínas Quinasas Activadas por Mitógenos/genética , Neuroblastoma/genética , Neuroblastoma/patología , Neuroblastoma/veterinaria , Neuronas , Proteínas Tirosina Quinasas/genética , Ratas , Transducción de Señal , Células Tumorales Cultivadas
11.
J Immunol ; 172(6): 3758-65, 2004 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-15004180

RESUMEN

LL-37 is a cationic peptide that is found in the granules of neutrophils and is secreted by epithelial cells from a variety of tissues. Levels of LL-37 in vivo increase upon infection, and its production and secretion are increased upon stimulation with proinflammatory mediators. It has been postulated that LL-37 modulates the immune response by interacting with the effector cells of innate immunity; however, the mechanism of this interaction is unknown. LL-37 induced phosphorylation and activation of the mitogen-activated protein kinases, extracellular signal-regulated kinase 1/2 (ERK1/2) and p38, in human peripheral blood-derived monocytes and a human bronchial epithelial cell line, but not in B or T lymphocytes. Phosphorylation was not dependent on the G protein-coupled formyl peptide-like receptor 1, which was previously proposed to be the receptor for LL-37-induced chemotaxis on human monocytes and T cells. Activation of ERK1/2 and p38 was markedly increased by the presence of GM-CSF, but not M-CSF. Exposure to LL-37 also led to the activation of Elk-1, a transcription factor that is downstream of and activated by phosphorylated ERK1/2, the up-regulation of various Elk-1-controlled genes, and the transcription and secretion of IL-8. Inhibition of either p38 or ERK1/2 kinases led to a reduction in LL-37-induced IL-8 secretion and inhibition of the transcription of various chemokine genes. The ability of LL-37 to signal through these pathways has broad implications in immunity, monocyte activation, proliferation, and differentiation.


Asunto(s)
Péptidos Catiónicos Antimicrobianos/fisiología , Sistema de Señalización de MAP Quinasas/inmunología , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Monocitos/enzimología , Péptidos Catiónicos Antimicrobianos/sangre , Catelicidinas , Células Cultivadas , Quimiocinas/genética , Citotoxicidad Inmunológica , Relación Dosis-Respuesta Inmunológica , Sinergismo Farmacológico , Activación Enzimática/inmunología , Células Epiteliales/enzimología , Proteínas de Unión al GTP/fisiología , Regulación de la Expresión Génica/inmunología , Factor Estimulante de Colonias de Granulocitos y Macrófagos/fisiología , Humanos , Interleucina-8/metabolismo , Activación de Macrófagos/inmunología , Proteína Quinasa 1 Activada por Mitógenos/sangre , Proteína Quinasa 3 Activada por Mitógenos , Proteínas Quinasas Activadas por Mitógenos/sangre , Proteínas Quinasas Activadas por Mitógenos/fisiología , Monocitos/inmunología , Receptor EphA8/genética , Transcripción Genética/inmunología , Proteínas Quinasas p38 Activadas por Mitógenos
12.
Dev Dyn ; 226(4): 596-603, 2003 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-12666197

RESUMEN

Eph receptors and ephrins are dynamically expressed in a wide range of regions of the vertebrate during embryogenesis. The dorsal mesencephalon appears to be segmented into two broad regions demarcated by the mutually exclusive expression of EphA receptors and ephrinA ligands. It is of considerable interest to elucidate how these expression domains are established in the development of the mesencephalon. In this study, we used a transgenic approach to define the cis-acting DNA regulatory elements involved in the anterior mesencephalon-specific expression of the mouse ephA8 gene. Our analyses of the temporal and spatial expression patterns of various ephA8/lacZ gene fusions in transgenic mice revealed that the 10-kb genomic DNA 5' immediately upstream of the ephA8 coding sequence is capable of directing lacZ expression in an ephA8-specific manner. Further deletion analyses of the ephA8 genomic region led to the identification of a 1-kb enhancer region, which directs expression in the embryo to the anterior region of the developing midbrain. This ephA8-specific regulatory DNA sequences can now be used in biochemical analyses to identify proteins modulating the anterior differentiation of the optic tectum, and in functional analyses to direct the expression of other developmentally important genes to this region.


Asunto(s)
Elementos de Facilitación Genéticos/genética , Regulación del Desarrollo de la Expresión Génica/fisiología , Mesencéfalo/embriología , Mesencéfalo/fisiología , Receptor EphA8/genética , Región de Flanqueo 5'/genética , Animales , Ligandos , Ratones , Ratones Transgénicos , Receptores de la Familia Eph/genética , Receptores de la Familia Eph/metabolismo , Colículos Superiores/embriología , Colículos Superiores/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...