Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
PLoS One ; 7(9): e45369, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-23028966

RESUMEN

The homeobox gene SHOX encodes for a transcription factor that plays an important role during limb development. Mutations or deletions of SHOX in humans cause short stature in Turner, Langer and Leri-Weill syndrome as well as idiopathic short stature. During embryonic development, SHOX is expressed in a complex spatio-temporal pattern that requires the presence of specific regulatory mechanisms. Up to now, it was known that SHOX is regulated by two upstream promoters and several enhancers on either side of the gene, but no regulators have been identified that can activate or repress the transcription of SHOX by binding to these regulatory elements. We have now identified the homeodomain protein HOXA9 as a positive regulator of SHOX expression in U2OS cells. Using luciferase assays, chromatin immunoprecipitation and electrophoretic mobility shift assays, we could narrow down the HOXA9 binding site to two AT-rich sequences of 31 bp within the SHOX promoter 2. Virus-induced Hoxa9 overexpression in a chicken micromass model validated the regulation of Shox by Hoxa9 (negative regulation). As Hoxa9 and Shox are both expressed in overlapping regions of the developing limb buds, a regulatory relationship of Hoxa9 and Shox during the process of limb development can be assumed.


Asunto(s)
Proteínas de Homeodominio/metabolismo , Animales , Sitios de Unión , Línea Celular Tumoral , Embrión de Pollo , Pollos , Inmunoprecipitación de Cromatina , Islas de CpG/genética , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Ensayo de Cambio de Movilidad Electroforética , Proteínas de Homeodominio/genética , Humanos , Hibridación in Situ , Proteína 1 del Sitio de Integración Viral Ecotrópica Mieloide , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Factor de Transcripción 1 de la Leucemia de Células Pre-B , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Proteína de la Caja Homeótica de Baja Estatura
2.
BMC Cancer ; 11: 102, 2011 Mar 22.
Artículo en Inglés | MEDLINE | ID: mdl-21426551

RESUMEN

BACKGROUND: DNA methylation in the SHOX2 locus was previously used to reliably detect lung cancer in a group of critical controls, including 'cytologically negative' samples with no visible tumor cell content, at a high specificity based on the analysis of bronchial lavage samples. This study aimed to investigate, if the methylation correlates with SHOX2 gene expression and/or copy number alterations. An amplification of the SHOX2 gene locus together with the observed tumor-specific hypermethylation might explain the good performance of this marker in bronchial lavage samples. METHODS: SHOX2 expression, gene copy number and DNA methylation were determined in lung tumor tissues and matched morphologically normal adjacent tissues (NAT) from 55 lung cancer patients. Quantitative HeavyMethyl (HM) real-time PCR was used to detect SHOX2 DNA methylation levels. SHOX2 expression was assayed with quantitative real-time PCR, and copy numbers alterations were measured with conventional real-time PCR and array CGH. RESULTS: A hypermethylation of the SHOX2 locus in tumor tissue as compared to the matched NAT from the same patient was detected in 96% of tumors from a group of 55 lung cancer patients. This correlated highly significantly with the frequent occurrence of copy number amplification (p < 0.0001), while the expression of the SHOX2 gene showed no difference. CONCLUSIONS: Frequent gene amplification correlated with hypermethylation of the SHOX2 gene locus. This concerted effect qualifies SHOX2 DNA methylation as a biomarker for lung cancer diagnosis, especially when sensitive detection is needed, i.e. in bronchial lavage or blood samples.


Asunto(s)
Carcinoma/genética , Metilación de ADN , Amplificación de Genes/fisiología , Proteínas de Homeodominio/genética , Neoplasias Pulmonares/genética , Biomarcadores de Tumor/genética , Biomarcadores de Tumor/metabolismo , Biomarcadores de Tumor/fisiología , Carcinoma/diagnóstico , Carcinoma/metabolismo , Carcinoma/patología , Hibridación Genómica Comparativa , Metilación de ADN/fisiología , Análisis Mutacional de ADN/métodos , Dosificación de Gen/fisiología , Proteínas de Homeodominio/metabolismo , Proteínas de Homeodominio/fisiología , Humanos , Neoplasias Pulmonares/diagnóstico , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patología , Análisis por Apareamiento , Polimorfismo Genético , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
3.
Hum Mol Genet ; 20(8): 1524-35, 2011 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-21273290

RESUMEN

The short stature homeobox gene SHOX encodes a transcription factor which is important for normal limb development. In humans, SHOX deficiency has been associated with various short stature syndromes including Leri-Weill dyschondrosteosis (LWD), Langer mesomelic dysplasia and Turner syndrome as well as non-syndromic idiopathic short stature. A common feature of these syndromes is disproportionate short stature with a particular shortening of the forearms and lower legs. In our studies employing microarray analyses and cell culture experiments, we revealed a strong positive effect of SHOX on the expression of the fibroblast growth factor receptor gene FGFR3, another well-known factor for limb development. Luciferase reporter gene assays show that SHOX activates the extended FGFR3 promoter, and results from chromatin immunoprecipitation (ChIP)-sequencing, ChIP and electrophoretic mobility shift assay experiments suggest a direct binding of SHOX to multiple upstream sequences of FGFR3. To further investigate these regulations in a cellular system for limb development, the effect of viral overexpression of Shox in limb bud derived chicken micromass cultures was tested. We found that Fgfr3 was negatively regulated by Shox, as demonstrated by quantitative real-time polymerase chain reaction and in situ hybridization. This repressive effect might explain the almost mutually exclusive expression patterns of Fgfr3 and Shox in embryonic chicken limbs. A negative regulation that occurs mainly in the mesomelic segments, a region where SHOX is known to be strongly expressed, offers a possible explanation for the phenotypes seen in patients with FGFR3 (e.g. achondroplasia) and SHOX defects (e.g. LWD). In summary, these data present a link between two frequent short stature phenotypes.


Asunto(s)
Miembro Posterior/embriología , Proteínas de Homeodominio/metabolismo , Receptor Tipo 3 de Factor de Crecimiento de Fibroblastos/metabolismo , Alas de Animales/embriología , Animales , Secuencia de Bases , Línea Celular Tumoral , Embrión de Pollo , Condrocitos/metabolismo , Regulación hacia Abajo , Ectodermo/metabolismo , Genes Reporteros , Miembro Posterior/citología , Miembro Posterior/metabolismo , Proteínas de Homeodominio/genética , Humanos , Luciferasas de Luciérnaga/biosíntesis , Luciferasas de Luciérnaga/genética , Mesodermo/metabolismo , Datos de Secuencia Molecular , Mutación Missense , Fenotipo , Regiones Promotoras Genéticas , Interferencia de ARN , Receptor Tipo 3 de Factor de Crecimiento de Fibroblastos/genética , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Proteína de la Caja Homeótica de Baja Estatura , Transcripción Genética , Alas de Animales/citología , Alas de Animales/metabolismo
4.
Hum Mol Genet ; 19(23): 4625-33, 2010 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-20858598

RESUMEN

Heart formation requires a highly balanced network of transcriptional activation of genes. The homeodomain transcription factor, Shox2, is essential for the formation of the sinoatrial valves and for the development of the pacemaking system. The elucidation of molecular mechanisms underlying the development of pacemaker tissue has gained clinical interest as defects in its patterning can be related to atrial arrhythmias. We have analyzed putative targets of Shox2 and identified the Bmp4 gene as a direct target. Shox2 interacts directly with the Bmp4 promoter in chromatin immunoprecipitation assays and activates transcription in luciferase-reporter assays. In addition, ectopic expression of Shox2 in Xenopus embryos stimulates transcription of the Bmp4 gene, and silencing of Shox2 in cardiomyocytes leads to a reduction in the expression of Bmp4. In Tbx5(del/+) mice, a model for Holt-Oram syndrome, and Shox2(-/-) mice, we show that the T-box transcription factor Tbx5 is a regulator of Shox2 expression in the inflow tract and that Bmp4 is regulated by Shox2 in this compartment of the embryonic heart. In addition, we could show that Tbx5 acts cooperatively with Nkx2.5 to regulate the expression of Shox2 and Bmp4. This work establishes a link between Tbx5, Shox2 and Bmp4 in the pacemaker region of the developing heart and thus contributes to the unraveling of the intricate interplay between the heart-specific transcriptional machinery and developmental signaling pathways.


Asunto(s)
Proteína Morfogenética Ósea 4/genética , Proteína Morfogenética Ósea 4/metabolismo , Corazón/embriología , Proteínas de Homeodominio/genética , Proteínas de Dominio T Box/genética , Animales , Células COS , Chlorocebus aethiops , Inmunoprecipitación de Cromatina , Regulación del Desarrollo de la Expresión Génica , Genes Reporteros , Células HEK293 , Frecuencia Cardíaca , Proteína Homeótica Nkx-2.5 , Humanos , Hibridación in Situ , Ratones , Modelos Animales , Miocardio/metabolismo , Miocitos Cardíacos/metabolismo , Reacción en Cadena de la Polimerasa , Transducción de Señal , Factores de Transcripción/genética , Activación Transcripcional , Xenopus
5.
Arch Physiol Biochem ; 113(3): 116-23, 2007 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-17922307

RESUMEN

The Short Stature Homeobox-containing Gene SHOX was identified as the genetic cause of the short stature phenotype in patients with Turner Syndrome and in certain patients with idiopathic short stature. Shortly after, SHOX mutations were also associated with the growth failure and skeletal deformities seen in patients with Léri - Weill dyschondrosteosis and Langer mesomelic dysplasia. Today it is estimated that SHOX mutations occur with an incidence of roughly 1:1,000 in newborns, making mutations of this gene one of the most common genetic defects leading to growth failure in humans. This review summarises the involvement of SHOX in several short stature syndromes and describes recent advances in our understanding of SHOX functions and regulation. We also discuss the current evidence in the literature that points to a role of this protein in growth and bone development. These studies have improved our knowledge of the SHOX gene and protein functions, and have given insight into the etiopathogenesis of short stature. However, the exact role of SHOX in bone development still remains elusive and poses the next major challenge for researchers in this field.


Asunto(s)
Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Humanos , Mutación , Proteína de la Caja Homeótica de Baja Estatura
6.
J Mol Biol ; 355(3): 590-603, 2006 Jan 20.
Artículo en Inglés | MEDLINE | ID: mdl-16325853

RESUMEN

Mutations within the homeobox SHOX gene have been associated with short stature and the skeletal deformities found in Léri-Weill, Turner and Langer syndromes implying an involvement of SHOX in growth and bone formation. Despite its clinical significance, the precise role of SHOX and the mechanisms that modulate its functions remain unknown. We reported previously that SHOX is a nuclear protein that specifically binds DNA and acts as a transcriptional activator. We have shown that ectopic expression of SHOX leads to cell-cycle arrest and apoptosis in osteosarcoma and primary cells. To further characterize SHOX, we investigated whether the protein could be a target for phosphorylation. Here, we report that SHOX is phosphorylated exclusively on serine residues in vivo. Two-dimensional phospho-peptide mapping showed that SHOX is phosphorylated to various extents on multiple sites. Site-directed mutagenesis demonstrated that serine 106 is the major SHOX phosphorylation site. We show also that casein kinase II phosphorylates SHOX on serine 106 efficiently in vitro and specific casein kinase II inhibitors reduce SHOX phosphorylation strongly in vivo. Finally, we provide evidence that phosphorylation may play an important role in modulating SHOX biological activities, since a S106A SHOX mutant, defective in phosphorylation, does not activate transcription and fails to induce cell-cycle arrest and apoptosis.


Asunto(s)
Proteínas de Homeodominio/metabolismo , Serina/metabolismo , Secuencia de Aminoácidos , Animales , Apoptosis , Quinasa de la Caseína II/antagonistas & inhibidores , Quinasa de la Caseína II/metabolismo , Ciclo Celular , Línea Celular Tumoral , Proteínas de Homeodominio/genética , Humanos , Datos de Secuencia Molecular , Mutagénesis Sitio-Dirigida , Mutación , Fosforilación , Proteína de la Caja Homeótica de Baja Estatura , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción
7.
Am J Hum Genet ; 77(1): 89-96, 2005 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-15931595

RESUMEN

Human growth is influenced not only by environmental and internal factors but also by a large number of different genes. One of these genes, SHOX, is believed to play a major role in growth, since defects in this homeobox-containing gene on the sex chromosomes lead to syndromal short stature (Leri-Weill dyschondrosteosis, Langer mesomelic dysplasia, and Turner syndrome) as well as to idiopathic short stature. We have analyzed 118 unrelated patients with Leri-Weill dyschondrosteosis and >1,500 patients with idiopathic short stature for deletions encompassing SHOX. Deletions were detected in 34% of the patients with Leri-Weill dyschondrosteosis and in 2% of the patients with idiopathic short stature. For 27 patients with Leri-Weill dyschondrosteosis and for 6 with idiopathic short stature, detailed deletion mapping was performed. Analysis was performed by polymerase chain reaction with the use of pseudoautosomal polymorphic markers and by fluorescence in situ hybridization with the use of cosmid clones. Here, we show that, although the identified deletions vary in size, the vast majority (73%) of patients tested share a distinct proximal deletion breakpoint. We propose that the sequence present within this proximal deletion breakpoint "hotspot" region predisposes to recurrent breaks.


Asunto(s)
Eliminación de Gen , Trastornos del Crecimiento/genética , Proteínas de Homeodominio/genética , Factores de Transcripción/genética , Deleción Cromosómica , Humanos , Hibridación in Situ , Repeticiones de Microsatélite , Osteocondrodisplasias/genética , Reacción en Cadena de la Polimerasa , Polimorfismo de Nucleótido Simple , Recombinación Genética , Proteína de la Caja Homeótica de Baja Estatura
8.
Hum Mutat ; 26(1): 44-52, 2005 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-15931687

RESUMEN

Haploinsufficiency of the short stature homeobox gene SHOX has been found in patients with idiopathic short stature (ISS) and Leri-Weill dyschondrosteosis (LWD). In addition to complete gene deletions and nonsense mutations, several missense mutations have been identified in both patient groups, leading to amino acid substitutions in the SHOX protein. The majority of missense mutations were found to accumulate in the region encoding the highly conserved homeodomain of the paired-like type. In this report, we investigated nine different amino acid exchanges in the homeodomain of SHOX patients with ISS and LWD. We were able show that these mutations cause an alteration of the biological function of SHOX by loss of DNA binding, reduced dimerization ability, and/or impaired nuclear translocation. Additionally, one of the mutations (c.458G>T, p.R153L) is defective in transcriptional activation even though it is still able to bind to DNA, dimerize, and translocate to the nucleus. Thus, we demonstrate that single missense mutations in the homeodomain fundamentally impair SHOX key functions, thereby leading to the phenotype observed in patients with LWD and ISS.


Asunto(s)
Estatura/genética , Núcleo Celular/metabolismo , ADN/metabolismo , Trastornos del Crecimiento/genética , Proteínas de Homeodominio/química , Proteínas de Homeodominio/metabolismo , Mutación/genética , Factores de Transcripción/química , Factores de Transcripción/metabolismo , Transporte Activo de Núcleo Celular , Secuencia de Aminoácidos , Ciclo Celular , Dimerización , Genes Homeobox/genética , Proteínas de Homeodominio/genética , Humanos , Datos de Secuencia Molecular , Mutación Missense/genética , Proteína de la Caja Homeótica de Baja Estatura , Factores de Transcripción/genética , Activación Transcripcional
9.
J Cell Sci ; 117(Pt 14): 3041-8, 2004 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-15173321

RESUMEN

We report the characterization of the nuclear localization signal (NLS) of the short stature homeobox gene SHOX. Mutations within the SHOX gene cause Léri-Weill dyschondrosteosis (LWD) and Langer mesomelic dysplasia (LD) as well as idiopathic short stature (ISS). Furthermore, haploinsufficiency of SHOX has also been implicated in Turner syndrome. SHOX has been shown to be a cell-type-specific transcriptional activator that localizes to the nucleus. The SHOX protein contains a central homeodomain that together with its transactivation domain regulates the transcription of its target sequences within the nucleus. The sequences for its nuclear localization have not been identified yet. Experimental characterization of SHOX-NLS by deletion mapping identified a non-classic type basic signal, AKCRK, in the recognition helix of the homeodomain. Fusion of this stretch of five amino acids to a cytoplasmic reporter protein resulted in its nuclear translocation. Functional analysis of a missense mutation R173C (C517T) affecting the identified SHOX-NLS in two families with LWS and LD showed that the mutated SHOX protein is unable to enter the nucleus. Conversely, we can demonstrate that insertion of the identified signal adjacent to the mutant site can restore its nuclear translocation. These results establish impairment of nuclear localization as a mechanistic basis for SHOX-related diseases.


Asunto(s)
Núcleo Celular/metabolismo , Proteínas de Homeodominio/metabolismo , Señales de Localización Nuclear , Secuencia de Aminoácidos , Línea Celular , Línea Celular Tumoral , Núcleo Celular/genética , Análisis Mutacional de ADN , Ligamiento Genético , Proteínas de Homeodominio/genética , Humanos , Inmunohistoquímica , Datos de Secuencia Molecular , Osteocondrodisplasias/genética , Homología de Secuencia de Aminoácido , Proteína de la Caja Homeótica de Baja Estatura , Síndrome , Síndrome de Turner/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...