Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 4 de 4
Filter
Add more filters










Database
Language
Publication year range
1.
Neurobiol Dis ; 36(1): 181-90, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19632331

ABSTRACT

Myotonic dystrophy type 1 (DM1) and type II (DM2) are dominantly inherited multisystemic disorders. DM1 is triggered by the pathological expansion of a (CTG)(n) triplet repeat in the DMPK gene, whereas a (CCTG)(n) tetranucleotide repeat expansion in the ZNF9 gene causes DM2. Both forms of the disease share several features, even though the causative mutations and the loci involved differ. Important distinctions exist, such as the lack of a congenital form of DM2. The reason for these disparities is unknown. In this study, we characterized skeletal muscle satellite cells from adult DM2 patients to provide an in vitro model for the disease. We used muscle cells from DM1 biopsies as a comparison tool. Our main finding is that DM2 satellite cells differentiate normally in vitro. Myotube formation was similar to unaffected controls. In contrast, fetal DM1 cells were deficient in that ability. Consistent with this observation, the myogenic program in DM2 was intact but is compromised in fetal DM1 cells. Although expression of the ZNF9 gene was enhanced in DM2 during differentiation, the levels of the ZNF9 protein were substantially reduced. This suggests that the presence of a large CCTG tract impairs the translation of the ZNF9 mRNA. Additionally, DM2 muscle biopsies displayed the altered splicing of the insulin receptor mRNA, correlating with insulin resistance in the patients. Finally, CUGBP1 steady-state protein levels were unchanged in DM2 cultured muscle cells and in DM2 muscle biopsies relative to controls, whereas they are increased in DM1 muscle cells. Our findings suggest that the myogenic program throughout muscle development and tissue regeneration is intact in DM2.


Subject(s)
Cell Differentiation/genetics , Muscle, Skeletal/pathology , Myotonic Disorders/classification , Myotonic Disorders/pathology , Satellite Cells, Skeletal Muscle/physiology , Adult , Alternative Splicing/genetics , Analysis of Variance , CELF1 Protein , Cells, Cultured , Female , Fetus , Gene Expression Regulation/genetics , Humans , Male , Middle Aged , Myotonin-Protein Kinase , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Receptor, Insulin/genetics , Receptor, Insulin/metabolism , Trinucleotide Repeat Expansion
2.
BMC Mol Biol ; 9: 44, 2008 Apr 29.
Article in English | MEDLINE | ID: mdl-18445271

ABSTRACT

BACKGROUND: In mammals, sex determination is genetically controlled. The SRY gene, located on Y chromosome, functions as the dominant genetic switch for testis development. The SRY gene is specifically expressed in a subpopulation of somatic cells (pre-Sertoli cells) of the developing urogenital ridge for a brief period during gonadal differentiation. Despite this tight spatiotemporal expression pattern, the molecular mechanisms that regulate SRY transcription remain poorly understood. Sry expression has been shown to be markedly reduced in transgenic mice harboring a mutant GATA4 protein (a member of the GATA family of transcription factors) disrupted in its ability to interact with its transcriptional partner FOG2, suggesting that GATA4 is involved in SRY gene transcription. RESULTS: Although our results show that GATA4 directly targets the pig SRY promoter, we did not observe similar action on the mouse and human SRY promoters. In the mouse, Wilms' tumor 1 (WT1) is an important regulator of both Sry and Müllerian inhibiting substance (Amh/Mis) expression and in humans, WT1 mutations are associated with abnormalities of sex differentiation. GATA4 transcriptionally cooperated with WT1 on the mouse, pig, and human SRY promoters. Maximal GATA4/WT1 synergism was dependent on WT1 but not GATA4 binding to their consensus regulatory elements in the SRY promoter and required both the zinc finger and C-terminal regions of the GATA4 protein. Although both isoforms of WT1 synergized with GATA4, synergism was stronger with the +KTS rather than the -KTS isoform. WT1/GATA4 synergism was also observed on the AMH promoter. In contrast to SRY, WT1/GATA4 action on the mouse Amh promoter was specific for the -KTS isoform and required both WT1 and GATA4 binding. CONCLUSION: Our data therefore provide new insights into the molecular mechanisms that contribute to the tissue-specific expression of the SRY and AMH genes in both normal development and certain syndromes of abnormal sex differentiation.


Subject(s)
GATA4 Transcription Factor/metabolism , Gene Expression Regulation , Sex Determination Processes , Sex Differentiation/genetics , Transcription, Genetic , WT1 Proteins/metabolism , Animals , Binding Sites , Cell Line , GATA4 Transcription Factor/chemistry , Humans , Mice , Mullerian Ducts/metabolism , Mutant Proteins/metabolism , Promoter Regions, Genetic/genetics , Protein Binding , Protein Structure, Tertiary , Receptors, Peptide/metabolism , Receptors, Transforming Growth Factor beta/metabolism , Sex-Determining Region Y Protein/genetics , Species Specificity , Swine/genetics , Zinc Fingers
3.
Biol Reprod ; 68(4): 1098-106, 2003 Apr.
Article in English | MEDLINE | ID: mdl-12606467

ABSTRACT

To study the process of mammalian sex determination and in particular to further understand the mechanisms of transcriptional regulation of the SRY gene, we have isolated a 4.5-kilobase (kb) pig SRY 5' flanking sequence. To facilitate the in vitro analysis of these sequences, we have generated a porcine genital ridge (PGR) cell line (9E11) that expresses SRY as well as SOX9, steroidogenic factor-1 (SF-1), and DAX1. Via primer extension analysis on RNA from this cell line, a transcription start site for porcine SRY was identified at -661 base pairs (bps) 5' from the translation initiation site. Deletion studies of the SRY 5' flanking sequences in PGR 9E11 cells demonstrated that -1.4 kb of 5' flanking sequences retained full transcriptional activity compared with the -4.5 kb fragment, but that transcriptional activity fell when further deletions were made. Sequences downstream of the transcriptional start site are important for promoter activity, because deleting transcribed but not translated sequences eliminated promoter activity. Sequence analysis of the -1.4 kb fragment identified two potential binding sites for SF-1, at -1369 and at -290 from the ATG. To address the role of SF-1 transactivation in SRY promoter activity, mutagenesis studies of the potential SF-1 binding sites were performed and revealed that these sites were indeed important for SRY promoter activity. Cotransfection studies in a heterologous cell system (mouse CV-1 cells) demonstrated that pig SF-1 was able to transactivate the pig SRY promoter. Gel shift assays confirmed that the upstream site was recognized by mouse SF-1 protein. We conclude that two sites for SF-1 transactivation exist within the pig SRY promoter, at -1369 bp and at -290 bp, and that the site at -1369 bp is quantitatively the most important.


Subject(s)
DNA-Binding Proteins/genetics , DNA-Binding Proteins/physiology , Nuclear Proteins , Promoter Regions, Genetic , Swine/genetics , Transcription Factors/physiology , Animals , Base Sequence/genetics , Binding Sites/genetics , Cell Line , DNA-Binding Proteins/metabolism , Embryo, Mammalian/cytology , Fushi Tarazu Transcription Factors , Gene Deletion , Genitalia/embryology , Homeodomain Proteins , Molecular Sequence Data , Mutation , Promoter Regions, Genetic/genetics , Receptors, Cytoplasmic and Nuclear , Sex-Determining Region Y Protein , Steroidogenic Factor 1 , Swine/embryology , Transcription Factors/metabolism , Transcription, Genetic/physiology
4.
Endocrinology ; 143(10): 3935-45, 2002 Oct.
Article in English | MEDLINE | ID: mdl-12239105

ABSTRACT

Steroidogenic acute regulatory protein (StAR) is an essential cholesterol transporter in steroidogenic tissues. Hormone-induced StAR expression is regulated through the cAMP-dependent pathway involving activation of protein kinase A (PKA). The StAR promoter contains several conserved DNA regulatory elements. These include binding sites for steroidogenic factor 1, CCAAT/enhancer-binding protein (C/EBP), and GATA transcription factors. Although these elements are important for StAR promoter activity, how the various transcription factors that bind these elements cooperate to confer cAMP responsiveness remains poorly understood. As induction of StAR transcription by cAMP in steroidogenic MA-10 cells does not require de novo protein synthesis, this suggests that all essential transcription factors are present and that posttranslational modifications of the factors are involved. We now report that GATA-4 is phosphorylated in MA-10 cells in response to cAMP and in heterologous CV-1 cells, GATA-4 transcriptional activity is stimulated by PKA. Moreover, we show that GATA-4 and C/EBPbeta directly interact in vitro and in vivo and synergistically activate the StAR promoter in CV-1 cells exclusively in the presence of PKA. As PKA-dependent synergy was also observed with other GATA and C/EBP family members, this transcriptional cooperation may contribute to hormone-stimulated StAR expression in all steroidogenic tissues.


Subject(s)
CCAAT-Enhancer-Binding Proteins/physiology , Cyclic AMP-Dependent Protein Kinases/physiology , DNA-Binding Proteins/physiology , Phosphoproteins/genetics , Promoter Regions, Genetic/physiology , Transcription Factors/physiology , Animals , Cell Line , Chlorocebus aethiops , Cyclic AMP-Dependent Protein Kinases/metabolism , DNA/metabolism , GATA4 Transcription Factor , Mice , Phosphorylation , Protein Structure, Tertiary/physiology , Steroidogenic Factor 1
SELECTION OF CITATIONS
SEARCH DETAIL
...