Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 44
Filter
Add more filters










Publication year range
1.
Cell Death Dis ; 4: e729, 2013 Jul 18.
Article in English | MEDLINE | ID: mdl-23868062

ABSTRACT

The transcription factor Pax8, a member of the Paired-box gene family, is a critical regulator required for proper development and differentiation of thyroid follicular cells. Despite being Pax8 well characterized with respect to its role in regulating genes responsible for thyroid differentiation, its involvement in cell survival and proliferation has been hypothesized but remains unclear. Here, we show that Pax8 overexpression significantly increases proliferation and colony-forming efficiency of Fischer rat thyroid line 5 epithelial cells, although it is not sufficient to overcome their hormone dependence. More interestingly, we show that Pax8-specific silencing induces apoptosis through a p53-dependent pathway that involves caspase-3 activation and cleavage of poly(ADP)ribose polymerase. Our data indicate that tumor protein 53 induced nuclear protein 1 (tp53inp1), a positive regulator of p53-dependent cell cycle arrest and apoptosis, is a transcriptional target of Pax8 and is upregulated by Pax8 knockdown. Remarkably, tp53inp1 silencing significantly abolishes Pax8-induced apoptosis thus suggesting that tp53inp1 may be the mediator of the observed effects. In conclusion, our data highlight that Pax8 is required for the survival of differentiated epithelial cells and its expression levels are able to modulate the proliferation rate of such cells.


Subject(s)
Cell Proliferation , Cell Survival , Epithelial Cells/physiology , Paired Box Transcription Factors/physiology , Animals , Apoptosis , Apoptosis Regulatory Proteins , Cell Cycle , Cell Line , Gene Knockdown Techniques , Heat-Shock Proteins/metabolism , Mice , Nuclear Proteins/metabolism , PAX8 Transcription Factor , RNA Interference , Rats
2.
J Endocrinol Invest ; 32(3): 238-41, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19542741

ABSTRACT

AIM: In 80-85% of cases, congenital hypothyroidism is associated with thyroid dysgenesis (TD), but only in a small percentage of cases mutations in thyroid transcription factors (NKX2.1, PAX8, FOXE1, and NKX2.5) have been associated with the disease. Several studies demonstrated that the activity of the transcription factors can be modulated by the interaction with other proteins, such as coactivators and co-repressors, and TAZ (transcriptional co-activator with PDZ-binding motif or WWTR1) is a co-activator interacting with both NKX2.1 and PAX8. In the present study we investigate the role of TAZ in the pathogenesis of TD. MATERIAL AND METHODS: By Single Stranded Conformational Polymorphism, we screened the entire TAZ coding sequence for mutations in 96 patients with TD and in 96 normal controls. RESULTS: No mutations were found in patients and controls, but we found several polymorphisms in both groups. No significant differences could be demonstrated in the prevalence of the mutations between patients and controls. CONCLUSIONS: Our data indicate that TAZ mutations are not a cause of TD in the series of patients studied.


Subject(s)
Nuclear Proteins/metabolism , Paired Box Transcription Factors/metabolism , Thyroid Dysgenesis/genetics , Transcription Factors/genetics , Transcription Factors/metabolism , Acyltransferases , Case-Control Studies , DNA Mutational Analysis , Gene Frequency , Genetic Testing , Humans , Mutation/physiology , PAX8 Transcription Factor , Polymorphism, Single-Stranded Conformational , Thyroid Nuclear Factor 1 , Trans-Activators/genetics , Trans-Activators/metabolism
3.
Mol Endocrinol ; 18(11): 2817-29, 2004 Nov.
Article in English | MEDLINE | ID: mdl-15319451

ABSTRACT

We previously demonstrated that transcription of the rat sodium/iodide symporter (NIS) gene is regulated by NUE, an upstream enhancer located between nucleotides -2264 and -2495 of the 5'-flanking region. To elucidate the mechanism of TSH/cAMP-mediated regulation of NIS gene expression, we have characterized the putative cAMP response element (CRE)/activator protein (AP)-1 site (termed NUC) that is closely located between the two Pax-8 (paired box domain transcription factor-8) binding sites within NUE. In two different approaches using either gel supershift analyses or dominant-negative inhibitors of b-Zip molecules, we have shown that NUC can be recognized by several members of the AP-1 and CREB family transcription factors that modulate the transcriptional activity of NUE. Using tethered dimers of b-Zip molecules, we have also demonstrated that specific homo- or heterodimers of AP-1 can synergistically stimulate NUE activity in concert with Pax-8. To demonstrate further that NUC is a bona fide CRE, we made an artificial promoter with the five-time tandem repeat of this sequence (5xNUC). In comparison to the canonical CRE (5xCRE), 5xNUC manifested greater transcriptional activity and broader response to cAMP signaling. Hence, we postulate that the significance of this evolutionally conserved CRE-like site may lie in its broader cell type specificity.


Subject(s)
Cyclic AMP Response Element-Binding Protein/metabolism , DNA-Binding Proteins/metabolism , Gene Expression Regulation , Nuclear Proteins/metabolism , Response Elements/genetics , Symporters/genetics , Trans-Activators/metabolism , Transcription Factors/metabolism , 5' Flanking Region/genetics , Animals , Binding Sites/genetics , Cell Line , Conserved Sequence , Cyclic AMP/metabolism , Cyclic AMP/pharmacology , Dimerization , Electrophoretic Mobility Shift Assay , Mutation/genetics , PAX8 Transcription Factor , Paired Box Transcription Factors , Promoter Regions, Genetic/genetics , Rats , Transcription Factor AP-1/metabolism
4.
Proc Natl Acad Sci U S A ; 99(24): 15462-7, 2002 Nov 26.
Article in English | MEDLINE | ID: mdl-12432093

ABSTRACT

The thyroid-stimulating hormone/thyrotropin (TSH) is the most relevant hormone in the control of thyroid gland physiology in adulthood. TSH effects on the thyroid gland are mediated by the interaction with a specific TSH receptor (TSHR). We studied the role of TSHTSHR signaling on gland morphogenesis and differentiation in the mouse embryo using mouse lines deprived either of TSH (pit(dw)pit(dw)) or of a functional TSHR (tshr(hyt)tshr(hyt) and TSHR-knockout lines). The results reported here show that in the absence of either TSH or a functional TSHR, the thyroid gland develops to a normal size, whereas the expression of thyroperoxidase and the sodium/iodide symporter are reduced greatly. Conversely, no relevant changes are detected in the amounts of thyroglobulin and the thyroid-enriched transcription factors TTF-1, TTF-2, and Pax8. These data suggest that the major role of the TSH/TSHR pathway is in controlling genes involved in iodide metabolism such as sodium/iodide symporter and thyroperoxidase. Furthermore, our data indicate that in embryonic life TSH does not play an equivalent role in controlling gland growth as in the adult thyroid.


Subject(s)
Gene Expression Regulation, Developmental/physiology , Receptors, Thyrotropin/physiology , Signal Transduction/physiology , Thyroid Gland/embryology , Thyrotropin/physiology , Animals , Antigens, Differentiation/biosynthesis , Antigens, Differentiation/genetics , Cell Differentiation , Crosses, Genetic , DNA-Binding Proteins/biosynthesis , DNA-Binding Proteins/genetics , Forkhead Transcription Factors , Gestational Age , Humans , Hypothyroidism/embryology , Hypothyroidism/genetics , Iodide Peroxidase/biosynthesis , Iodide Peroxidase/genetics , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Morphogenesis , Nuclear Proteins/biosynthesis , Nuclear Proteins/genetics , Organ Size , PAX8 Transcription Factor , Paired Box Transcription Factors , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Receptors, Thyrotropin/deficiency , Receptors, Thyrotropin/genetics , Recombinant Fusion Proteins/physiology , Repressor Proteins/biosynthesis , Repressor Proteins/genetics , Symporters/biosynthesis , Symporters/genetics , Thyroglobulin/biosynthesis , Thyroglobulin/genetics , Thyroid Gland/growth & development , Thyroid Gland/pathology , Thyroid Nuclear Factor 1 , Thyrotropin/deficiency , Thyrotropin/genetics , Trans-Activators/biosynthesis , Trans-Activators/genetics , Transcription Factors/biosynthesis , Transcription Factors/genetics
5.
J Endocrinol ; 172(1): 163-76, 2002 Jan.
Article in English | MEDLINE | ID: mdl-11786384

ABSTRACT

The transcription factor Pax8 plays an important role in the expression of the differentiated phenotype of thyroid follicular cells. It has recently been shown that Pax8 is necessary for thyroglobulin (Tg) gene expression in the fully differentiated rat thyroid cell line PC. We have used the PC model system to investigate the role of Pax8 as a mediator of TSH regulation of Tg gene expression. We have demonstrated that Pax8 expression, as well as Tg expression, is severely reduced in cells grown in the absence of hormones and serum. The re-addition of TSH or forskolin to the culture medium is able to restore to wild-type levels the expression of both Pax8 and Tg. We have determined that the action of TSH/forskolin on Pax8 is at the transcriptional level. However, the re-expression of Pax8 can be observed several hours before that of Tg, suggesting that either another factor is needed or that Pax8 itself must be post-translationally modified by a newly synthesized protein to become active. To distinguish between these two possibilities we have stably transfected into PC cells an exogenous Pax8 that is expressed independently of TSH. Our results indicate that in these cells the Tg promoter is still dependent on TSH despite the constitutive presence of Pax8. Furthermore, we also show that in this condition Tg gene transcription requires de novo protein synthesis. In conclusion, TSH regulates the expression of Pax8 at a transcriptional level and also regulates the activity of Pax8 by controlling the expression of one or more as yet unknown factors.


Subject(s)
DNA-Binding Proteins/physiology , Gene Expression Regulation , Nuclear Proteins , Thyroglobulin/genetics , Thyroid Gland/metabolism , Thyrotropin/metabolism , Trans-Activators/physiology , Animals , Blotting, Northern/methods , Blotting, Western/methods , Cell Line , DNA-Binding Proteins/analysis , DNA-Binding Proteins/genetics , Fluorescent Antibody Technique , Gene Expression , PAX8 Transcription Factor , Paired Box Transcription Factors , Rats , Reverse Transcriptase Polymerase Chain Reaction , Thyroglobulin/analysis , Thyroglobulin/metabolism , Trans-Activators/analysis , Trans-Activators/genetics , Transfection/methods
6.
Proc Natl Acad Sci U S A ; 97(24): 13144-9, 2000 Nov 21.
Article in English | MEDLINE | ID: mdl-11069301

ABSTRACT

Transformation of rat thyroid cells with polyoma virus middle T antigen results in loss of the thyroid-differentiated phenotype, measured as the expression of the thyroglobulin (Tg), thyroperoxidase (TPO), and sodium/iodide symporter (NIS) genes. Among the transcription factors involved in the regulation of these genes, TTF-1 and TTF-2 were still detected at nearly wild-type levels, while a specific loss of the paired domain transcription factor Pax8 was observed. In this study, we used the PCPy cell line as a model system to study the role of Pax8 in thyroid differentiation. We demonstrate that the reintroduction of Pax8 in PCPy cells is sufficient to activate expression of the endogenous genes encoding thyroglobulin, thyroperoxidase, and sodium/iodide symporter. Thus, this cell system provides direct evidence for the ability of Pax8 to activate transcription of thyroid-specific genes at their chromosomal locus and strongly suggests a fundamental role of this transcription factor in the maintenance of functional differentiation in thyroid cells. Moreover, we show that Pax8 and TTF-1 cooperate in the activation of the thyroglobulin promoter and that additional thyroid-specific mechanism(s) are involved in such a cooperation. To identify the Pax8 domain able to mediate the specific activation of the thyroglobulin promoter, we transfected in PCPy cells three different Pax8 isoforms. The results of such experiments indicate that for the transcriptional activation of thyroid-specific genes, Pax8 uses an as yet unidentified functional domain.


Subject(s)
Cell Differentiation/physiology , DNA-Binding Proteins/metabolism , Gene Expression Regulation , Thyroid Gland/cytology , Trans-Activators/metabolism , Animals , Cell Division , Cell Line , DNA-Binding Proteins/genetics , Forkhead Transcription Factors , Iodide Peroxidase/genetics , Mice , Nuclear Proteins/metabolism , PAX8 Transcription Factor , Paired Box Transcription Factors , Rats , Recombinant Proteins/metabolism , Repressor Proteins/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Thyroglobulin/genetics , Thyroid Gland/physiology , Thyroid Nuclear Factor 1 , Trans-Activators/genetics , Transcription Factors/metabolism , Transfection
7.
EMBO J ; 19(20): 5406-17, 2000 Oct 16.
Article in English | MEDLINE | ID: mdl-11032808

ABSTRACT

Treatment of human prostate carcinoma-derived LNCaP cells with androgen or oestradiol triggers simultaneous association of androgen receptor and oestradiol receptor beta with Src, activates the Src/Raf-1/Erk-2 pathway and stimulates cell proliferation. Surprisingly, either androgen or oestradiol action on each of these steps is inhibited by both anti-androgens and anti-oestrogens. Similar findings for oestradiol receptor alpha were observed in MCF-7 or T47D cells stimulated by either oestradiol or androgens. Microinjection of LNCaP, MCF-7 and T47D cells with SrcK(-) abolishes steroid-stimulated S-phase entry. Data from transfected Cos cells confirm and extend the findings from these cells. Hormone-stimulated Src interaction with the androgen receptor and oestradiol receptor alpha or beta is detected using glutathione S:-transferase fusion constructs. Src SH2 interacts with phosphotyrosine 537 of oestradiol receptor alpha and the Src SH3 domain with a proline-rich stretch of the androgen receptor. The role of this phosphotyrosine is stressed by its requirement for association of oestradiol receptor alpha with Src and consequent activation of Src in intact Cos cells.


Subject(s)
Prostatic Neoplasms/pathology , Receptors, Androgen/metabolism , Receptors, Estradiol/metabolism , Steroids/pharmacology , src-Family Kinases/metabolism , Androgen Antagonists/pharmacology , Androgens/pharmacology , Animals , COS Cells , Cell Division/drug effects , Estradiol/pharmacology , Estrogen Antagonists/pharmacology , Humans , MAP Kinase Signaling System/drug effects , Macromolecular Substances , Male , Metribolone/pharmacology , Models, Biological , Protein Binding/drug effects , Receptors, Androgen/genetics , Receptors, Estradiol/genetics , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Transfection , Tumor Cells, Cultured , src Homology Domains/drug effects , src-Family Kinases/genetics
8.
Cancer Res ; 60(14): 3916-20, 2000 Jul 15.
Article in English | MEDLINE | ID: mdl-10919669

ABSTRACT

The Akt/protein kinase B serine/threonine kinase is a downstream effector of phosphoinositide 3-kinase (PI3K). Akt is an important component of mitogenic and antiapoptotic signaling pathways and is implicated in neoplastic transformation. Thyroid cells in culture retain a differentiated phenotype consisting of epithelial cell morphology and the expression of several tissue-specific genes. The survival and proliferation of these cells depend on thyrotropin and a mixture of five additional hormones that includes insulin. The regulation of proliferation and the expression of the thyroid differentiation program are intimately connected processes. As a result, oncogenes that induce hormone-independent proliferation invariably impair the expression of the thyroid-specific differentiation markers. Given that thyrotropin and insulin stimulate Akt activation in thyroid cells, we set out to determine the effects of Akt on thyroid cell proliferation, survival, and differentiation. To this end, we expressed constitutively active myristylated Akt (myrAkt) in PC Cl 3 thyroid cells. The myrAkt-expressing cells continued to proliferate, even in the absence of hormones, and they were resistant to programmed cell death induced by starvation. These effects were paralleled by the induction of the G1 cyclins D3 and E and by the inhibition of induction of the proapoptotic Fas, Fas ligand, and BAD genes in starved cells. However, in marked contrast with several other oncogenes, myrAkt did not interfere with the expression of thyroid differentiation functions. These results unveil the existence of an Akt-triggered thyroid cell pathway that modulates proliferation and survival without affecting the expression of the thyroid cell differentiated phenotype.


Subject(s)
Protein Serine-Threonine Kinases , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins/physiology , Thyroid Gland/cytology , Animals , Apoptosis/genetics , Carrier Proteins/metabolism , Cell Division/genetics , Cell Line , Cell Survival/genetics , Cell Transformation, Neoplastic , Cyclin D3 , Cyclins/metabolism , DNA Fragmentation , DNA, Complementary/metabolism , Fas Ligand Protein , In Situ Nick-End Labeling , Membrane Glycoproteins/metabolism , Phenotype , Plasmids , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins c-akt , Rats , Rats, Inbred F344 , Signal Transduction , Transfection , bcl-Associated Death Protein , fas Receptor/metabolism
9.
Biochem Biophys Res Commun ; 275(1): 203-8, 2000 Aug 18.
Article in English | MEDLINE | ID: mdl-10944465

ABSTRACT

The thyroid transcription factor TTF-2 is a forkhead-containing protein involved in thyroid-specific gene expression and necessary for thyroid morphogenesis. In this paper, we demonstrate that TTF-2 is able to inhibit the activity of the thyroid-specific transcription factors TTF-1 and Pax-8 only on certain promoters. We identified the minimal protein domain responsible for repressor activity, which behaves as an independent functional domain, and we show that repression by TTF-2 is DNA-binding independent. We suggest that TTF-2 is able to interfere with a specific cofactor required for TTF-1 and Pax-8 activity.


Subject(s)
DNA-Binding Proteins/metabolism , Promoter Regions, Genetic/genetics , Repressor Proteins/metabolism , Thyroid Gland/metabolism , DNA/genetics , DNA/metabolism , DNA-Binding Proteins/antagonists & inhibitors , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/genetics , Enhancer Elements, Genetic/genetics , Forkhead Transcription Factors , Genes, Reporter/genetics , HeLa Cells , Humans , Iodide Peroxidase/genetics , Mutation/genetics , Nuclear Proteins/antagonists & inhibitors , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , PAX8 Transcription Factor , Paired Box Transcription Factors , Protein Binding , Protein Structure, Tertiary , Repressor Proteins/chemistry , Repressor Proteins/genetics , Substrate Specificity , Thyroglobulin/genetics , Thyroid Nuclear Factor 1 , Trans-Activators/antagonists & inhibitors , Trans-Activators/genetics , Trans-Activators/metabolism , Transcription Factors/antagonists & inhibitors , Transcription Factors/genetics , Transcription Factors/metabolism , Transfection
10.
J Biol Chem ; 274(21): 15213-21, 1999 May 21.
Article in English | MEDLINE | ID: mdl-10329730

ABSTRACT

The forkhead thyroid-specific transcription factor TTF-2 is the main mediator of thyrotropin and insulin regulation of thyroperoxidase (TPO) gene expression. This function depends on multimerization and specific orientation of its DNA-binding site, suggesting that TTF-2 is part of a complex interaction network within the TPO promoter. This was confirmed by transfection experiments and by protein-DNA interaction studies, which demonstrated that CTF/NF1 proteins bind 10 base pairs upstream of the TTF-2-binding site to enhance its action in hormone-induced expression of the TPO gene. GST pull-down assays showed that TTF-2 physically interacts with CTF/NF1 proteins. In addition, we demonstrate that increasing the distance between both transcription factors binding sites by base pair insertion results in loss of promoter activity and in a drastic decrease on the ability of the promoter to respond to the hormones. CTF/NF1 is a family of transcription factors that contributes to constitutive and cell-type specific gene expression. Originally identified as factors implicated in the replication of adenovirus, this group of proteins (CTF/NF1-A, -B, -C, and -X) is now known to be involved in the regulation of several genes. In contrast to other reports regarding the involvement of these proteins in inducible gene expression, we show here that members of this family of transcription factors are regulated by hormones. With the use of specific CTF/NF1 DNA probes and antibodies we demonstrate that CTF/NF1-C is a thyrotropin-, cAMP-, and insulin-inducible protein. Thus CTF/NF1 proteins do not only mediate hormone-induced gene expression cooperating with TTF-2, but are themselves hormonally regulated. All these findings are clearly of important value in understanding the mechanisms governing the transcription regulation of RNA polymerase II promoters, which often contain binding sites for multiple transcription factors.


Subject(s)
CCAAT-Enhancer-Binding Proteins , DNA-Binding Proteins/physiology , Iodide Peroxidase/genetics , Repressor Proteins/physiology , Thyroid Gland/enzymology , Transcription Factors/physiology , Base Sequence , Binding Sites , Cells, Cultured , Forkhead Transcription Factors , HeLa Cells , Humans , NFI Transcription Factors , Promoter Regions, Genetic , Transcription, Genetic
11.
J Cell Sci ; 112 ( Pt 6): 957-65, 1999 Mar.
Article in English | MEDLINE | ID: mdl-10036245

ABSTRACT

FRT thyroid epithelial cells synthesize fibronectin and organize a network of fibronectin fibrils at the basal surface of the cells. Fibronectin fibril formation is enhanced by the overexpression of the ubiquitous beta1A integrin and is inhibited by the expression of the dominant-negative beta1B subunit. We tested the hypotheses that RhoA activity might mediate the integrin-dependent fibronectin fibrillogenesis and might counteract beta1B integrin inhibitory effect. FRT-beta1A cells were transfected with a vector carrying a dominant negative form of RhoA (RhoAN19) or treated with the C3 transferase exoenzyme. Both treatments inhibited fibronectin assembly and caused loss of actin microfilaments and adhesion plaques. On the other hand, FRT-beta1B cells were transfected with the constitutively activated form of RhoA (RhoAV14) or treated with the E. coli cytotoxic necrotizing factor 1, which directly activates RhoA. Either treatment restored microfilament and adhesion plaque assembly and promoted fibronectin fibril organization. A great increase in fibronectin fibril assembly was also obtained by treatment of FRT-beta1B cells with TGF-beta. Our data indicate that RhoA is required to promote fibronectin matrix assembly in FRT cells and that the activation of the signal transduction pathway downstream of RhoA can overcome the inhibitory effect of beta1B integrin.


Subject(s)
Actin Cytoskeleton/physiology , Fibronectins/biosynthesis , Fibronectins/genetics , GTP-Binding Proteins/metabolism , Integrin beta1/physiology , Actin Cytoskeleton/ultrastructure , Actins/physiology , Animals , Cell Line , Epithelial Cells , Integrin beta1/chemistry , Integrin beta1/genetics , Macromolecular Substances , Recombinant Proteins/metabolism , Thyroid Gland , Transfection , rhoA GTP-Binding Protein
12.
Mol Cell Biol ; 19(3): 2051-60, 1999 Mar.
Article in English | MEDLINE | ID: mdl-10022892

ABSTRACT

The gene encoding the Na/I symporter (NIS) is expressed at high levels only in thyroid follicular cells, where its expression is regulated by the thyroid-stimulating hormone via the second messenger, cyclic AMP (cAMP). In this study, we demonstrate the presence of an enhancer that is located between nucleotides -2264 and -2495 in the 5'-flanking region of the NIS gene and that recapitulates the most relevant aspects of NIS regulation. When fused to either its own or a heterologous promoter, the NIS upstream enhancer, which we call NUE, stimulates transcription in a thyroid-specific and cAMP-dependent manner. The activity of NUE depends on the four most relevant sites, identified by mutational analysis. The thyroid-specific transcription factor Pax8 binds at two of these sites. Mutations that interfere with Pax8 binding also decrease transcriptional activity of the NUE. Furthermore, expression of Pax8 in nonthyroid cells results in transcriptional activation of NUE, strongly suggesting that the paired-domain protein Pax8 plays an important role in NUE activity. The NUE responds to cAMP in both protein kinase A-dependent and -independent manners, indicating that this enhancer could represent a novel type of cAMP responsive element. Such a cAMP response requires Pax8 but also depends on the integrity of a cAMP responsive element (CRE)-like sequence, thus suggesting a functional interaction between Pax8 and factors binding at the CRE-like site.


Subject(s)
Carrier Proteins/genetics , Cyclic AMP/metabolism , DNA-Binding Proteins/metabolism , Enhancer Elements, Genetic , Membrane Proteins/genetics , Symporters , Thyroid Gland/metabolism , Trans-Activators/metabolism , Transcription, Genetic , Animals , Base Sequence , Binding Sites , Cell Line , DNA-Binding Proteins/genetics , Molecular Sequence Data , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , PAX8 Transcription Factor , Paired Box Transcription Factors , Rats , Regulatory Sequences, Nucleic Acid , Thyroid Gland/cytology , Thyroid Nuclear Factor 1 , Trans-Activators/genetics , Transcription Factors/genetics , Transcription Factors/metabolism , Transcriptional Activation
13.
Cancer Res ; 58(13): 2888-94, 1998 Jul 01.
Article in English | MEDLINE | ID: mdl-9661907

ABSTRACT

Expression of mutated versions of the p53 gene deranged the differentiation program of thyroid cells and resulted in deregulated growth. Specifically, p53 mutants in several residues of the DNA-binding region induced thyrotropin (TSH) -independent growth and inhibition of the expression of thyroid-specific genes. The loss of the differentiated phenotype invariably correlated with the blockage of the expression of the genes coding for the thyroid transcriptional factors PAX-8 and TTF2. Conversely, thyroid cells transfected with a p53 gene mutated at codon 392, located outside the DNA-binding region, stimulated the expression of differentiation genes in the absence of the TSH, and induced TSH-independent growth. cAMP intracellular levels were higher in thyroid cells transfected with the p53 gene mutated at the 392 site than in the untransfected thyroid cells, but lower in the cells transfected with the other mutated p53 genes. Fra-1 and c-jun were induced by p53, resulting in increased AP-1 levels. The results of this study suggest that p53 exerts effects on cAMP transduction pathway in thyroid cells, which are exquisitely sensitive to cAMP.


Subject(s)
Cell Differentiation/genetics , Genes, p53/physiology , Thyroid Gland/cytology , Animals , Binding Sites , Cell Division/genetics , Cells, Cultured , Cyclic AMP/metabolism , Genes, p53/genetics , Mutation , Peroxidases/genetics , Peroxidases/metabolism , Phenotype , Rats , Receptors, Thyrotropin/genetics , Receptors, Thyrotropin/metabolism , Thyroglobulin/genetics , Thyroglobulin/metabolism , Transcription Factor AP-1/metabolism , Transfection , Tumor Suppressor Protein p53/chemistry , Tumor Suppressor Protein p53/genetics
14.
Endocrinology ; 139(6): 2796-802, 1998 Jun.
Article in English | MEDLINE | ID: mdl-9607786

ABSTRACT

Mechanisms responsible for the lack of thyroid-specific differentiation markers in Ha-ras transformed FRTL-5 cells have been investigated. In vivo cell labeling and immunoprecipitation demonstrate that phosphorylation of the thyroid transcription factor-1 (TTF-1) is clearly reduced in thyroid cells transformed with the Ha-ras oncogene. Fingerprinting analysis of phosphotryptic peptides from FRTL-5 and Ha-ras-FRTL-5 cells also reveals a heterogeneous pattern of TTF-1 phosphorylation in the transformed cell line. This heterogeneity is localized in the amino terminal cluster of phosphoserines, as determined by transfection of HeLa cells with TTF-1 mutants in which serine residues have been replaced by alanines. Amplification and nucleotide sequence of the 5'-coding region of the TTF-1 gene in Ha-ras-FRTL-5 cells rule out the possibility that differences in phosphorylation were the consequence of any mutational event affecting residues within the N-terminal protein sequence. Hypophosphorylated TTF-1 is still able to bind its DNA consensus sequence within the thyroglobulin promoter, although a reporter construct whose expression is exclusively dependent on TTF-1 is not transactivated. Transfection of Ha-ras-FRTL-5 cells with an expression vector encoding the cAMP dependent protein kinase A (PKA) catalytic subunit partially reestablishes TTF-1 transcriptional activity. Taken together, these results indicate that the lack of specific thyroid gene expression in Ha-ras-FRTL-5 cells could be a direct consequence of the inability of TTF-1 to promote transcription.


Subject(s)
Genes, ras/physiology , Nuclear Proteins/metabolism , Thyroid Gland/cytology , Thyroid Gland/metabolism , Transcription Factors/metabolism , Amino Acid Sequence , Cell Differentiation/physiology , Cell Line, Transformed , Cyclic AMP-Dependent Protein Kinases/metabolism , HeLa Cells , Humans , Molecular Sequence Data , Mutation/physiology , Nuclear Proteins/genetics , Phosphorylation , Serine/metabolism , Thyroid Nuclear Factor 1 , Transcription Factors/genetics
15.
J Biol Chem ; 273(3): 1477-82, 1998 Jan 16.
Article in English | MEDLINE | ID: mdl-9430685

ABSTRACT

Thyroid transcription factor-1 (TTF-1) is a homeodomain-containing transcription factor that is required for thyroid-specific expression of the thyroglobulin and thyroperoxidase genes as well as for lung-specific expression of the surfactant protein A, B, and C and the CC10 and the HNF-3 alpha genes. TTF-1 is a phosphoprotein, and the phosphorylation of TTF-1 has been studied already. However, the kinase(s) that could be responsible for this phosphorylation have not been known. In this paper we report the identification by in-gel kinase assay of a 56-kDa serine/threonine kinase that is able to phosphorylate TTF-1 in thyroid cells. The cloning of this kinase revealed that we had identified the rat homolog of the human MST2 kinase. The pathway in which human MST2 functions is not known; however, it does not appear to involve either mitogen-activated protein kinases such as Erk1 and Erk2 nor the stress-activated protein kinases such as JNK and p38. We show that the activity responsible for TTF-1 phosphorylation is constitutive in thyroid cells. Furthermore, we demonstrate that TTF-1 is phosphorylated in vivo by rMST2 at the same residues that had been identified previously as the major phosphorylation sites. Thus, TTF-1 represents the first identified target of this class of protein kinases.


Subject(s)
Homeodomain Proteins/metabolism , Mitogen-Activated Protein Kinases , Nuclear Proteins/metabolism , Protein Serine-Threonine Kinases/metabolism , Thyroid Gland/metabolism , Transcription Factors/metabolism , Amino Acid Sequence , Animals , Base Sequence , HeLa Cells , Humans , Mitogen-Activated Protein Kinase 9 , Molecular Sequence Data , Phosphorylation , Protein Kinases/metabolism , Rats , Sequence Homology, Amino Acid , Sequence Homology, Nucleic Acid , Serine-Threonine Kinase 3 , Thyroid Nuclear Factor 1
16.
Cell Growth Differ ; 9(1): 97-103, 1998 Jan.
Article in English | MEDLINE | ID: mdl-9438393

ABSTRACT

The most frequent genetic alterations described thus far in human papillary thyroid carcinomas are somatic rearrangements of the RET proto-oncogene, which generate the chimeric RET/PTC oncogenes. We recently found that the expression of the RET/PTC1 oncogene blocked the expression of the thyroid-differentiated phenotype in rat thyroid epithelial cell line PC CI 3 (PC). Here, we show that this block occurs at a transcriptional level; indeed, the thyroid-specific thyroglobulin and thyroperoxidase gene promoters were inactive in PC-PTC cells. Specific transcription factors, namely, TTF-1 and Pax-8, regulate the expression of differentiated functions in thyroid cells. Here, we show that Pax-8 is expressed at reduced levels in PC-PTC cells and that its adoptive overexpression is unable to restore the activity of target promoters. In contrast, TTF-1 expression is unaltered in PC-PTC cells; however, by using a synthetic promoter that contains its specific target sequence, we demonstrate that TTF-1 is inactive in PC-PTC cells. We conclude that the RET/PTC1 oncogene alters the expression of the thyroid-differentiated phenotype by at least two different mechanisms, ie., down-regulation of Pax-8 protein and mRNA expression and impaired function of TTF-1 and Pax-8, which occurs at a posttranslational level.


Subject(s)
DNA-Binding Proteins/metabolism , Drosophila Proteins , Nuclear Proteins/metabolism , Oncogenes , Proto-Oncogene Proteins/genetics , Receptor Protein-Tyrosine Kinases/genetics , Trans-Activators/metabolism , Transcription Factors/metabolism , Animals , Carcinoma, Papillary/genetics , Cells, Cultured , Down-Regulation , Humans , PAX8 Transcription Factor , Paired Box Transcription Factors , Promoter Regions, Genetic , Proto-Oncogene Mas , Proto-Oncogene Proteins c-ret , Rats , Thyroid Neoplasms/genetics , Thyroid Nuclear Factor 1
17.
Biochim Biophys Acta ; 1354(2): 171-81, 1997 Nov 01.
Article in English | MEDLINE | ID: mdl-9396634

ABSTRACT

The thyroglobulin gene, the substrate for thyroid hormone biosynthesis, is not expressed in the FRT cell line, which, even though it manifests the polarised epithelial phenotype, does not express any of the thyroid functional properties. Two transcription factors, TTF-1 and Pax-8, have been implicated in thyroid specific expression of the thyroglobulin gene. FRT cells contain Pax-8 but they lack TTF-1. In this paper, we show that transfection of TTF-1 expression vectors in FRT cells results in activation of thyroglobulin gene expression. If the expression vector encoded for TTF-1-ER, a fusion gene coding for the entire TTF-1 protein fused to the hormone-binding domain of the steroid receptor, under the control of the RSV promoter, thyroglobulin gene expression was controlled by estrogen. These data provide a direct demonstration that TTF-1 activates the chromosomal thyroglobulin promoter. Since transfection of TTF-1 expression vectors in non-thyroid cell types did not result in thyroglobulin gene expression, it is suggested that Pax-8, in addition, perhaps, to a specific cellular environment, might be required for thyroid specific expression of the thyroglobulin gene.


Subject(s)
Gene Expression Regulation , Nuclear Proteins/genetics , Thyroglobulin/genetics , Thyroid Gland/metabolism , Transcription Factors/genetics , Animals , Blotting, Western , Cell Line , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Estradiol/pharmacology , Fluorescent Antibody Technique , Genes, Reporter , Immunohistochemistry , Nuclear Proteins/physiology , PAX8 Transcription Factor , Paired Box Transcription Factors , Precipitin Tests , Promoter Regions, Genetic , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Recombinant Fusion Proteins/biosynthesis , Respiratory Syncytial Viruses/genetics , Thyroglobulin/biosynthesis , Thyroid Nuclear Factor 1 , Trans-Activators/genetics , Trans-Activators/metabolism , Transcription Factors/physiology , Transfection
18.
J Biol Chem ; 272(37): 23334-9, 1997 Sep 12.
Article in English | MEDLINE | ID: mdl-9287345

ABSTRACT

The hormonal regulation of both thyroglobulin and thyroperoxidase promoter activity in FRTL-5 thyroid cells takes place, at least in part, through a hormone-responsive element to which the thyroid transcription factor TTF-2 binds. The TTF-2 cDNA, encoded by the titf2 locus, has recently been cloned and classified as a member of the forkhead transcription factor family. Here, we demonstrate that TTF-2 mRNA levels become undetectable in FRTL-5 thyroid cells cultured for 4 days in 0.2% serum and in the absent of thyrotropin (TSH) and insulin. Addition of TSH, insulin or insulin-like growth factor I (IGF-I) to the culture medium increases the levels of this transcription factor in a dose- and time- dependent manner and requires ongoing protein synthesis. The TSH effect is greater than that produced by insulin or IGF-I and is similar to the effect produced by the cAMP analog forskolin. The TSH and insulin effects are additive. In all cases, the mRNA levels increase is accompanied by an increase in transcription rate, as demonstrated by run-off assays. These data demonstrate that the TTF-2 mRNA is under tight hormonal control. This is consistent with an important role for TTF-2 as a mediator of the transcriptional activation of thyroid-specific genes (thyroglobulin and thyroperoxidase) by TSH via cAMP and by insulin through the IGF-I receptor.


Subject(s)
DNA-Binding Proteins/genetics , Insulin-Like Growth Factor I/pharmacology , Insulin/pharmacology , Repressor Proteins/genetics , Thyrotropin/pharmacology , Transcription Factors/genetics , Transcription, Genetic/drug effects , Colforsin/pharmacology , Cyclic AMP/analogs & derivatives , DNA-Binding Proteins/biosynthesis , Dose-Response Relationship, Drug , Forkhead Transcription Factors , Gene Expression Regulation , Glyceraldehyde-3-Phosphate Dehydrogenases/biosynthesis , Glyceraldehyde-3-Phosphate Dehydrogenases/genetics , Iodide Peroxidase/biosynthesis , Nuclear Proteins , RNA, Messenger/biosynthesis , Repressor Proteins/biosynthesis , Signal Transduction , Thyroglobulin/biosynthesis , Thyroid Gland/cytology , Transcription Factors/biosynthesis
19.
EMBO J ; 16(11): 3185-97, 1997 Jun 02.
Article in English | MEDLINE | ID: mdl-9214635

ABSTRACT

Expression of thyroglobulin (Tg) and thyroperoxidase (TPO) genes in thyroid follicular cells occurs in the mouse at embryonic day (E)14.5. Two transcription factors, TTF-1 and Pax-8, have been implicated in transcriptional activation of Tg and TPO, even though the onset of their expression is at E9.5, suggesting that additional events are necessary for transcriptional activation of Tg and TPO genes. We report in this paper the cloning of TTF-2, a DNA binding protein that recognizes sites on both Tg and TPO promoters. TTF-2 is a new forkhead domain-containing protein whose expression is restricted to the endodermal lining of the foregut and to the ectoderm that will give rise to the anterior pituitary. TTF-2 shows transient expression in the developing thyroid and anterior pituitary. In the thyroid, TTF-2 expression is down-regulated just before the onset of Tg and TPO gene expression, suggesting that this transcription factor plays the role in development of a negative controller of thyroid-specific gene expression.


Subject(s)
DNA-Binding Proteins/genetics , Gene Expression Regulation, Developmental , Repressor Proteins/genetics , Thyroid Gland/embryology , Amino Acid Sequence , Animals , Base Sequence , Cell Differentiation , Cells, Cultured , Chromosome Mapping , Cloning, Molecular , DNA, Complementary/genetics , DNA-Binding Proteins/biosynthesis , DNA-Binding Proteins/metabolism , Forkhead Transcription Factors , Insulin/pharmacology , Insulin-Like Growth Factor I/pharmacology , Mice , Molecular Sequence Data , Nuclear Proteins/metabolism , PAX8 Transcription Factor , Paired Box Transcription Factors , Peroxidases/biosynthesis , Pituitary Gland, Anterior/embryology , Protein Binding , Repressor Proteins/biosynthesis , Sequence Analysis, DNA , Sequence Homology, Amino Acid , Thyroglobulin/biosynthesis , Thyroid Gland/cytology , Thyroid Gland/drug effects , Thyroid Gland/growth & development , Thyroid Nuclear Factor 1 , Time Factors , Trans-Activators/metabolism , Transcription Factors/metabolism
20.
J Biol Chem ; 271(4): 2249-54, 1996 Jan 26.
Article in English | MEDLINE | ID: mdl-8567686

ABSTRACT

The phosphorylation of thyroid transcription factor-1 (TTF-1), is homeodomain-containing transcription factor that is required for thyroid-specific expression of the thyroglobulin and thyroperoxidase gene promoters, has been studied. Phosphorylation occurs on a maximum of seven serine residues that are distributed in three tryptic peptides. Mutant derivatives of TTF-1, with alanine sites, have been constructed and used to assess the functional relevance of TTF-1 phosphorylation. The DNA binding activity of TTF-1 appears to be phosphorylation-independent, as indicated also by the performance of TTF-1 purified from an overexpressing Escherichia coli strain. Transcriptional activation by TTF-1 could require phosphorylation only in specific cell types since in a co-transfection assay in heterologous cells both wild-type and mutant proteins show a similar transcriptional activity.


Subject(s)
DNA-Binding Proteins/chemistry , Nuclear Proteins/chemistry , Phosphoproteins/chemistry , Transcription Factors/chemistry , Amino Acid Sequence , Animals , Base Sequence , DNA Primers/chemistry , DNA-Binding Proteins/physiology , Gene Expression Regulation , HeLa Cells , Humans , Molecular Sequence Data , Nuclear Proteins/physiology , Peptide Mapping , Phosphorylation , Phosphoserine/chemistry , Rats , Thyroid Nuclear Factor 1 , Transcription Factors/physiology , Transcription, Genetic
SELECTION OF CITATIONS
SEARCH DETAIL
...