Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
Hum Mol Genet ; 19(23): 4625-33, 2010 Dec 01.
Article in English | MEDLINE | ID: mdl-20858598

ABSTRACT

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.


Subject(s)
Bone Morphogenetic Protein 4/genetics , Bone Morphogenetic Protein 4/metabolism , Heart/embryology , Homeodomain Proteins/genetics , T-Box Domain Proteins/genetics , Animals , COS Cells , Chlorocebus aethiops , Chromatin Immunoprecipitation , Gene Expression Regulation, Developmental , Genes, Reporter , HEK293 Cells , Heart Rate , Homeobox Protein Nkx-2.5 , Humans , In Situ Hybridization , Mice , Models, Animal , Myocardium/metabolism , Myocytes, Cardiac/metabolism , Polymerase Chain Reaction , Signal Transduction , Transcription Factors/genetics , Transcriptional Activation , Xenopus
2.
Circulation ; 115(14): 1830-8, 2007 Apr 10.
Article in English | MEDLINE | ID: mdl-17372176

ABSTRACT

BACKGROUND: Identifying molecular pathways regulating the development of pacemaking and coordinated heartbeat is crucial for a comprehensive mechanistic understanding of arrhythmia-related diseases. Elucidation of these pathways has been complicated mainly by an insufficient definition of the developmental structures involved in these processes and the unavailability of animal models specifically targeting the relevant tissues. Here, we report on a highly restricted expression pattern of the homeodomain transcription factor Shox2 in the sinus venosus myocardium, including the sinoatrial nodal region and the venous valves. METHODS AND RESULTS: To investigate its function in vivo, we have generated mouse lines carrying a targeted mutation of the Shox2 gene. Although heterozygous animals did not exhibit obvious defects, homozygosity of the targeted allele led to embryonic lethality at 11.5 to 13.5 dpc. Shox2-/- embryos exhibited severe hypoplasia of the sinus venosus myocardium in the posterior heart field, including the sinoatrial nodal region and venous valves. We furthermore demonstrate aberrant expression of connexin 40 and connexin 43 and the transcription factor Nkx2.5 in vivo specifically within the sinoatrial nodal region and show that Shox2 deficiency interferes with pacemaking function in zebrafish embryos. CONCLUSIONS: From these results, we postulate a critical function of Shox2 in the recruitment of sinus venosus myocardium comprising the sinoatrial nodal region.


Subject(s)
Gene Expression Regulation, Developmental , Heart/embryology , Homeodomain Proteins/physiology , Transcription Factors/physiology , Zebrafish Proteins/physiology , Animals , Bradycardia/embryology , Bradycardia/genetics , Connexin 43/analysis , Connexins/analysis , Embryonic Development/genetics , Fetal Heart/pathology , Gene Targeting , Genes, Lethal , Heart Conduction System/embryology , Heart Conduction System/physiopathology , Heart Defects, Congenital/embryology , Heart Defects, Congenital/genetics , Heart Valves/embryology , Homeobox Protein Nkx-2.5 , Homeodomain Proteins/analysis , Homeodomain Proteins/genetics , Mice/embryology , Mice, Inbred C57BL , Mice, Knockout , Myocardium/pathology , Myocytes, Cardiac/cytology , Phenotype , Sinoatrial Node/embryology , Transcription Factors/analysis , Transcription Factors/genetics , Zebrafish/embryology , Zebrafish Proteins/deficiency , Zebrafish Proteins/genetics , Gap Junction alpha-5 Protein
3.
Dev Biol ; 298(2): 585-96, 2006 Oct 15.
Article in English | MEDLINE | ID: mdl-16904661

ABSTRACT

SHOX is a homeobox-containing gene, highly conserved among species as diverse as fish, chicken and humans. SHOX gene mutations have been shown to cause idiopathic short stature and skeletal malformations frequently observed in human patients with Turner, Leri-Weill and Langer syndromes. We cloned the chicken orthologue of SHOX, studied its expression pattern and compared this with expression of the highly related Shox2. Shox is expressed in central regions of early chick limb buds and proximal two thirds of later limbs, whereas Shox2 is expressed more posteriorly in the proximal third of the limb bud. Shox expression is inhibited distally by signals from the apical ectodermal ridge, both Fgfs and Bmps, and proximally by retinoic acid signaling. We tested Shox functions by overexpression in embryos and micromass cultures. Shox-infected chick limbs had normal proximo-distal patterning but the length of skeletal elements was consistently increased. Primary chick limb bud cell cultures infected with Shox showed an initial increase in cartilage nodules but these did not enlarge. These results fit well with the proposed role of Shox in cartilage and bone differentiation and suggest chick embryos as a useful model to study further the role of Shox in limb development.


Subject(s)
Gene Expression Regulation, Developmental , Genes, Homeobox , Homeodomain Proteins/metabolism , Limb Buds/metabolism , Signal Transduction , Amino Acid Sequence , Animals , Cells, Cultured , Chick Embryo , Homeodomain Proteins/genetics , Limb Buds/anatomy & histology , Limb Buds/cytology , Limb Buds/embryology , Models, Anatomic , Molecular Sequence Data , Organ Culture Techniques , Sequence Homology, Amino Acid
4.
Curr Opin Genet Dev ; 16(3): 233-9, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16650979

ABSTRACT

The pseudoautosomal regions represent blocks of sequence identity between the mammalian sex chromosomes. In humans, they reside at the ends of the X and Y chromosomes and encompass roughly 2.7 Mb (PAR1) and 0.33 Mb (PAR2). As a major asset of recently available sequence data, our view of their structural characteristics could be refined considerably. While PAR2 resembles the overall sequence composition of the X chromosome and exhibits only slightly elevated recombination rates, PAR1 is characterized by a significantly higher GC content and a completely different repeat structure. In addition, it exhibits one of the highest recombination frequencies throughout the entire human genome and, probably as a consequence of its structural features, displays a significantly faster rate of evolution. It therefore represents an exceptional model to explore the correlation between meiotic recombination and evolutionary forces such as gene mutation and conversion. At least twenty-nine genes lie within the human pseudoautosomal regions, and these genes exhibit 'autosomal' rather than sex-specific inheritance. All genes within PAR1 escape X inactivation and are therefore candidates for the etiology of haploinsufficiency disorders including Turner syndrome (45,X). However, the only known disease gene within the pseudoautosomal regions is the SHORT STATURE HOMEBOX (SHOX) gene, functional loss of which is causally related to various short stature conditions and disturbed bone development. Recent analyses have furthermore revealed that the phosphorylation-sensitive function of SHOX is directly involved in chondrocyte differentiation and maturation.


Subject(s)
Chromosomes/genetics , Disease , Homeodomain Proteins/genetics , Animals , Disease Susceptibility , Humans , Mutation/genetics , Short Stature Homeobox Protein
5.
J Biol Chem ; 281(2): 695-704, 2006 Jan 13.
Article in English | MEDLINE | ID: mdl-16275643

ABSTRACT

The myeov gene has been isolated by the tumorigenicity assay and is localized at chromosome 11q13, a frequent site for chromosomal rearrangements in various carcinomas and B-cell neoplasms. In addition, myeov is coamplified with cyclin D1 and overexpressed in carcinomas of various organs. The mechanisms of myeov regulation remain enigmatic. The 5'-untranslated region (5'-UTR) of the myeov gene is long, encompasses several upstream AUGs, and is predicted to fold in a strong secondary structure, suggesting that its translation might be regulated by an internal ribosomal entry site. Here we show that initial experiments using monocistronic and dicistronic reporter constructs supported this assumption. However, the application of in vitro transcription/translation assays, Northern blot analysis, and promoterless dicistronic constructs revealed promoter activity of the myeov 5'-UTR. DNA transfection of dicistronic DNA constructs, normal and mutated forms of myeov cDNA fragments cloned in a eukaryotic expression vector, and direct RNA transfection analysis revealed that upstream AUG triplets in the 5'-UTR of the myeov transcript abrogate translation. Alternative splicing mechanisms in specific cell types and/or developmental stage may evade this translation control. Control experiments suggest that the 5'-UTR from encephalomyocarditis virus, when inserted at the midpoint of a dicistronic vector, is also able to function as a cryptic promoter.


Subject(s)
Gene Expression Regulation, Neoplastic , Oncogene Proteins/biosynthesis , Oncogene Proteins/genetics , Open Reading Frames , 5' Untranslated Regions , Base Sequence , Blotting, Northern , Cell Line , Chromosomes, Human, Pair 11 , Cyclin D1/metabolism , DNA/metabolism , DNA, Complementary/metabolism , Encephalomyocarditis virus/genetics , Genes, Reporter , Genetic Vectors , Humans , Immunoblotting , Luciferases/metabolism , Models, Genetic , Molecular Sequence Data , Mutation , Promoter Regions, Genetic , Protein Biosynthesis , Protein Structure, Secondary , Proto-Oncogene Proteins , RNA/metabolism , Transcription, Genetic , Transfection
6.
J Mol Biol ; 355(3): 590-603, 2006 Jan 20.
Article in English | MEDLINE | ID: mdl-16325853

ABSTRACT

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.


Subject(s)
Homeodomain Proteins/metabolism , Serine/metabolism , Amino Acid Sequence , Animals , Apoptosis , Casein Kinase II/antagonists & inhibitors , Casein Kinase II/metabolism , Cell Cycle , Cell Line, Tumor , Homeodomain Proteins/genetics , Humans , Molecular Sequence Data , Mutagenesis, Site-Directed , Mutation , Phosphorylation , Short Stature Homeobox Protein , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization
7.
Hum Mutat ; 26(1): 44-52, 2005 Jul.
Article in English | MEDLINE | ID: mdl-15931687

ABSTRACT

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.


Subject(s)
Body Height/genetics , Cell Nucleus/metabolism , DNA/metabolism , Growth Disorders/genetics , Homeodomain Proteins/chemistry , Homeodomain Proteins/metabolism , Mutation/genetics , Transcription Factors/chemistry , Transcription Factors/metabolism , Active Transport, Cell Nucleus , Amino Acid Sequence , Cell Cycle , Dimerization , Genes, Homeobox/genetics , Homeodomain Proteins/genetics , Humans , Molecular Sequence Data , Mutation, Missense/genetics , Short Stature Homeobox Protein , Transcription Factors/genetics , Transcriptional Activation
8.
J Cell Sci ; 117(Pt 14): 3041-8, 2004 Jun 15.
Article in English | MEDLINE | ID: mdl-15173321

ABSTRACT

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.


Subject(s)
Cell Nucleus/metabolism , Homeodomain Proteins/metabolism , Nuclear Localization Signals , Amino Acid Sequence , Cell Line , Cell Line, Tumor , Cell Nucleus/genetics , DNA Mutational Analysis , Genetic Linkage , Homeodomain Proteins/genetics , Humans , Immunohistochemistry , Molecular Sequence Data , Osteochondrodysplasias/genetics , Sequence Homology, Amino Acid , Short Stature Homeobox Protein , Syndrome , Turner Syndrome/genetics
9.
J Biol Chem ; 279(35): 37103-14, 2004 Aug 27.
Article in English | MEDLINE | ID: mdl-15145945

ABSTRACT

Mutations in the homeobox gene SHOX cause growth retardation and the skeletal abnormalities associated with Léri-Weill, Langer, and Turner syndromes. Little is known about the mechanism underlying these SHOX-related inherited disorders of bone formation. Here we demonstrate that SHOX expression in osteogenic stable cell lines, primary oral fibroblasts, and primary chondrocytes leads to cell cycle arrest and apoptosis. These events are associated with alterations in the expression of several cellular genes, including pRB, p53, and the cyclin kinase inhibitors p21(Cip1) and p27(Kip1). A SHOX mutant, such as seen in Léri-Weill syndrome patients, does not display these activities of the wild type protein. We have also shown that endogenous SHOX is mainly expressed in hypertrophic/apoptotic chondrocytes of the growth plate, strongly suggesting that the protein plays a direct role in regulating the differentiation of these cells. This study provides the first insight into the biological function of SHOX as regulator of cellular proliferation and viability and relates these cellular events to the phenotypic consequences of SHOX deficiency.


Subject(s)
Apoptosis , Chondrocytes/metabolism , Growth Plate/metabolism , Homeodomain Proteins/genetics , Homeodomain Proteins/physiology , Mutation , Transcription Factors/genetics , Transcription Factors/physiology , Antimetabolites, Antineoplastic/pharmacology , Blotting, Western , Body Height , Bromodeoxyuridine/pharmacology , Cell Cycle , Cell Cycle Proteins/metabolism , Cell Division , Cell Line, Tumor , Cell Separation , Cells, Cultured , Cyclin-Dependent Kinase Inhibitor p21 , Cyclin-Dependent Kinase Inhibitor p27 , Cyclins/metabolism , Fibroblasts/metabolism , Flow Cytometry , Gene Deletion , Homeodomain Proteins/chemistry , Humans , Immunohistochemistry , In Situ Nick-End Labeling , Mouth/metabolism , Protein Structure, Tertiary , Retinoblastoma Protein/metabolism , Retroviridae/genetics , Short Stature Homeobox Protein , Time Factors , Tumor Suppressor Protein p53/metabolism , Tumor Suppressor Proteins/metabolism
10.
J Biol Chem ; 278(48): 47820-6, 2003 Nov 28.
Article in English | MEDLINE | ID: mdl-12960152

ABSTRACT

Regulation of gene expression is particularly important for gene dosage-dependent diseases and the phenomenon of clinical heterogeneity frequently associated with these phenotypes. We here report on the combined transcriptional and translational regulatory mechanisms controlling the expression of the Léri-Weill and Turner syndrome gene SHOX. We define an alternative promotor within exon 2 of the SHOX gene by transient transfections of mono- and bicistronic reporter constructs and demonstrate substantial differences in the translation efficiency of the mRNAs transcribed from these alternative promotors by in vitro translation assays and direct mRNA transfections into different cell lines. Although transcripts generated from the intragenic promotor (P2) are translated with high efficiencies, mRNA originating from the upstream promotor (P1) exhibit significant translation inhibitory effects due to seven AUG codons upstream of the main open reading frame (uAUGs). Site-directed mutagenesis of these uAUGs confers full translation efficiency to reporter mRNAs in different cell lines and after injection of Xenopus embryos. In conclusion, our data support a model where functional SHOX protein levels are regulated by a combination of transcriptional and translational control mechanisms.


Subject(s)
Homeodomain Proteins/genetics , Protein Biosynthesis , Transcription Factors/genetics , Transcription, Genetic , Turner Syndrome/metabolism , 5' Untranslated Regions , Animals , COS Cells , Cell Line , Codon , Down-Regulation , Exons , Gene Expression Regulation , Genes, Reporter , Humans , Luciferases/metabolism , Models, Genetic , Mutagenesis, Site-Directed , Open Reading Frames , Phenotype , Plasmids/metabolism , Promoter Regions, Genetic , RNA/metabolism , RNA, Messenger/metabolism , Short Stature Homeobox Protein , Time Factors , Transfection , Xenopus
SELECTION OF CITATIONS
SEARCH DETAIL
...