Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 66
Filter
1.
Mol Pharmacol ; 79(6): 1053-60, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21415305

ABSTRACT

The xenobiotic receptors, constitutive androstane receptor (CAR), and pregnane X receptor (PXR) regulate and alter the metabolism of xenobiotic substrates. Among the 19 functional UDP-glucuronosyltransferases (UGTs) in humans, UGT2B7 is involved in the metabolism of many structurally diverse xenobiotics and plays an important role in the clearance and detoxification of many therapeutic drugs. To examine whether this gene is regulated by CAR and PXR in vivo, transgenic mice expressing the entire UGT2B7 gene (TgUGT2B7) were created. Gene expression profiles revealed that UGT2B7 is differentially expressed in liver, kidney, adipocytes, brain, and estrogen-sensitive tissues, such as ovary and uterus. Liver UGT2B7 expression levels were decreased when TgUGT2B7 mice were treated with the CAR ligand 1,4-b-s-[2-(3,5,-dichloropyridyloxy)] (TCPOBOP) but not the PXR ligand pregnenolone 16α-carbonitrile. Although TCPOBOP decreased the levels of UGT2B7 mRNA in TgUGT2B7 mice, it had no affect on Tg(UGT2B7)Car(-/-) mice, adding support for a CAR-dependent mechanism contributing toward UGT2B7 gene suppression. Expression of promoter constructs in HepG2 cells showed the CAR-dependent inhibition was linked to hepatocyte nuclear factor-4α (HNF4α)-mediated transactivation of the UGT2B7 promoter. The inhibitory effect of CAR on UGT2B7 gene expression was validated in chromatin immunoprecipitation assays in which TCPOBOP treatment blocked HNF4α binding to the UGT2B7 promoter. These results suggest that HNF4α plays an important role in the constitutive expression of hepatic UGT2B7, and CAR acts as a negative regulator by interfering with HNF4α binding activity.


Subject(s)
Glucuronosyltransferase/antagonists & inhibitors , Receptors, Cytoplasmic and Nuclear/physiology , Animals , Base Sequence , Constitutive Androstane Receptor , DNA Primers , Gene Expression Profiling , Humans , Mice , Mice, Inbred C57BL , Mice, Transgenic
2.
Endocrinology ; 142(6): 2284-95, 2001 Jun.
Article in English | MEDLINE | ID: mdl-11356674

ABSTRACT

FSH is secreted by gonadotropes of the anterior pituitary and plays a crucial role in mammalian reproduction. However, little is known about FSH gene regulation due to the lack of a gonadotrope cell line that synthesizes FSH. The LbetaT2 mouse pituitary cell line, isolated by targeted tumorigenesis in transgenic mice, has the characteristics of a mature gonadotrope, including expression of GnRH receptor, steroidogenic factor 1, and both the alpha- and beta-subunits of LH, but was thought not to express FSH. Using RT-PCR, we show that these cells synthesize FSH beta- subunit messenger RNA, which is induced by activin and inhibited by follistatin. Furthermore, in transient transfections an ovine FSHbeta 5'-regulatory region (5.5 kb) confers LbetaT2 cell-specific expression to a reporter gene compared with other pituitary and nonpituitary cell lines. This FSHbeta regulatory region responds to activin specifically in LbetaT2 cells, an effect that is blocked by follistatin. The LHbeta, alpha-subunit, and GnRH receptor regulatory regions are induced by activin and blocked by follistatin. Furthermore, LbetaT2 cells express the components of the activin system, and addition of follistatin alone reduces FSHbeta gene expression, demonstrating that an endogenous activin autocrine loop regulates FSH in these cells. In addition, GnRH stimulates both the FSHbeta and LHbeta regulatory regions, specifically in LbetaT2 cells. Surprisingly, GnRH induction is reduced by follistatin, suggesting its dependence on endogenous activin. As the mouse GnRH receptor promoter is inhibited by follistatin, reduction of GnRH receptor levels might be one mechanism by which follistatin interferes with GnRH induction of gonadotropin genes. In summary, LbetaT2 cells exhibit the characteristics of fully differentiated gonadotropes, including the expression of LH, FSH, GnRH receptor, and components of the activin/follistatin system, as well as display the appropriate responses to activin and GNRH:


Subject(s)
Follicle Stimulating Hormone/genetics , Gene Expression Regulation/drug effects , Gonadotropin-Releasing Hormone/pharmacology , Inhibins/pharmacology , Pituitary Gland, Anterior/metabolism , Activin Receptors , Activins , Animals , Blotting, Southern , Cell Line , Follicle Stimulating Hormone, beta Subunit , Follistatin , Gene Expression , Glycoprotein Hormones, alpha Subunit/genetics , Glycoproteins/genetics , Glycoproteins/pharmacology , Humans , Inhibins/genetics , Luteinizing Hormone/genetics , Mice , Mice, Transgenic , RNA, Messenger/analysis , Receptors, Growth Factor/genetics , Regulatory Sequences, Nucleic Acid , Reverse Transcriptase Polymerase Chain Reaction , Sheep , Transcription, Genetic/drug effects , Transfection
3.
Mol Endocrinol ; 14(11): 1811-9, 2000 Nov.
Article in English | MEDLINE | ID: mdl-11075814

ABSTRACT

The neuropeptide GnRH is a central regulator of mammalian reproductive function produced by a dispersed population of hypothalamic neurosecretory neurons. The principal action of GnRH is to regulate release of the gonadotropins, LH and FSH, by the gonadotrope cells of the anterior pituitary. Using a cultured cell model of mouse pituitary gonadotrope cells, alphaT3-1 cells, we present evidence that GnRH stimulation of alphaT3-1 cells results in an increase in cap-dependent mRNA translation. GnRH receptor activation results in increased protein synthesis through a regulator of mRNA translation initiation, eukaryotic translation initiation factor 4E-binding protein, known as 4EBP or PHAS (protein, heat, and acid stable). Although the GnRH receptor is a member of the rhodopsin-like family of G protein-linked receptors, we show that activation of translation proceeds through a signaling pathway previously described for receptor tyrosine kinases. Stimulation of translation by GnRH is protein kinase C and Ras dependent and sensitive to rapamycin. Furthermore, GnRH may also regulate the cell cycle in alphaT3-1 cells. The activation of a signaling pathway that regulates both protein synthesis and cell cycle suggests that GnRH may have a significant role in the maintenance of the pituitary gonadotrope population in addition to directing the release of gonadotropins.


Subject(s)
Carrier Proteins , Gonadotropin-Releasing Hormone/metabolism , Pituitary Gland/cytology , Pituitary Gland/physiology , Protein Biosynthesis , Protein Kinases , 5' Untranslated Regions , Adaptor Proteins, Signal Transducing , Animals , Cell Cycle Proteins , Cells, Cultured , Encephalomyocarditis virus/genetics , Eukaryotic Initiation Factors , Genes, ras , Gonadotropin-Releasing Hormone/pharmacology , Insulin/pharmacology , Mice , Phosphoproteins/metabolism , Phosphorylation , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Pituitary Gland/drug effects , Protein Kinase C/metabolism , Protein Subunits , RNA Caps/genetics , RNA, Messenger/genetics , Receptors, LHRH/biosynthesis , Receptors, LHRH/genetics , Sirolimus/pharmacology , TOR Serine-Threonine Kinases
4.
Lab Invest ; 80(11): 1657-70, 2000 Nov.
Article in English | MEDLINE | ID: mdl-11092526

ABSTRACT

A line of transgenic mice that develops submandibular gland adenocarcinoma of intercalated duct origin was established. In these mice, the oncogene SV40 T antigen (Tag) is expressed from the neonatal submandibular gland secretory protein b (Smgb) gene promoter. This hybrid gene directs expression of the oncoprotein to neonatal submandibular gland proacinar and terminal tubule cells and to intercalated ducts of the adult gland. Transgene expression resulted in duct luminal cell hyperplasia as early as 20 to 30 days postnatally, which progressed to dysplasia by 3 to 4 months of age. Marked dysplasia and in situ carcinoma were evident at 4 to 6 months of age. All histologic changes were more pronounced in males. Submandibular gland adenocarcinoma developed stochastically in more than half of the adult male mice by 12 months of age (average age: 10.8 months, range: 6 to 13.5 months). Tag expression persisted in in situ carcinoma and all tumors. Using a combination of immunocytochemical and ultrastructural criteria, submandibular gland dysplasia and tumors were found to originate from intercalated ducts. The dysplastic ducts and adenocarcinoma in Smgb-Tag mice were morphologically similar to previously reported Tag-induced dysplasias of striated ducts and granular convoluted tubules and a Tag-induced adenocarcinoma of striated duct origin. These findings demonstrate that salivary gland dysplasias and tumors of similar histologic appearance can arise from distinct differentiated cell types. Analysis of the molecular changes accompanying tumor formation in Smgb-Tag mice could increase knowledge of human salivary gland tumorigenesis.


Subject(s)
Adenocarcinoma/pathology , Salivary Gland Neoplasms/pathology , Submandibular Gland/pathology , Animals , Base Sequence , DNA Primers , Immunohistochemistry , Male , Mice , Mice, Transgenic , Reverse Transcriptase Polymerase Chain Reaction
5.
Mol Endocrinol ; 14(9): 1509-22, 2000 Sep.
Article in English | MEDLINE | ID: mdl-10976927

ABSTRACT

Neuroendocrine control of the reproductive cascade is mediated by GnRH, which in mammals is produced by a subset of neurons scattered throughout the hypothalamus and forebrain. Utilizing a cultured cell model of GnRH neurons (GT1-7 cells), two regulatory regions in the rat GnRH 5' flanking DNA were identified as essential for cell-type specificity: a 300-bp enhancer and a 173-bp conserved proximal promoter. Using transient transfections to compare expression in GT1-7 cells to a non-GnRH-expressing cell type (NIH 3T3), we show that the GnRH enhancer and the proximal promoter each play roles in conferring this specificity. Deletion of footprint 2 (FP2; -26 to -76) from the promoter when coupled to the GnRH enhancer diminishes reporter activity in GT1-7 cells more strongly than in NIH 3T3 cells. Furthermore, deletion of FP2 from the promoter when coupled to the heterologous Rous sarcoma virus 5'-long terminal repeat promoter abolishes the difference in reporter activity between GT1-7 and NIH 3T3 cells, suggesting that FP2 of the GnRH promoter is necessary for cell-specific expression. In addition, FP2 alone is sufficient to confer cell-specific expression and can interact with the GnRH enhancer to augment reporter gene expression specifically in GT1-7 cells. Finally, a 31-bp sequence from within FP2 (-63 to -33) synergistically activates transcription when coupled with the GnRH enhancer in GT1-7 cells but not in NIH 3T3 cells. Thus, this 31-bp region contains elements necessary for interaction between the GnRH enhancer and promoter. We show that two of five protein complexes that bind to the -63 to -33 region are GT1-7 cell specific, and both of them appear to be homeodomain proteins. The identification of a cell-specific element in the GnRH proximal promoter significantly advances our understanding of the transcriptional basis for neuron-specific GnRH gene expression.


Subject(s)
Enhancer Elements, Genetic , Gene Expression Regulation , Gonadotropin-Releasing Hormone/genetics , Neurons/physiology , Promoter Regions, Genetic , 3T3 Cells , Animals , Base Sequence , Cell Line , Genes, Reporter , Luciferases/genetics , Mice , Mutagenesis, Site-Directed , Neurons/cytology , Rats , Transfection , beta-Galactosidase/genetics
6.
Mol Endocrinol ; 14(8): 1246-56, 2000 Aug.
Article in English | MEDLINE | ID: mdl-10935548

ABSTRACT

The GnRH gene is expressed exclusively in a highly restricted population of approximately 800 neurons in the mediobasal hypothalamus in the mouse. The Otx2 homeoprotein has been shown to colocalize with GnRH in embryonic mouse brain. We have identified a highly conserved bicoid-related Otx target sequence within the proximal promoter region of the GnRH gene from several species. This element from the rat GnRH promoter binds baculovirus-expressed Otx2 protein and Otx2 protein in nuclear extracts of a hypothalamic GnRH-expressing neuronal cell line, GT1-7. Transient transfection assays indicate that the GnRH promoter Otx/bicoid site is required for specific expression of the GnRH gene in GT1-7 cells and that it can confer specificity to a neutral Rous sarcoma virus (RSV) promoter in GT1-7 cells but not in NIH3T3 cells. Overexpression of mouse Otx2 in GT1-7 cells induces expression of a GnRH promoter plasmid, an effect that is dependent upon the Otx binding site. Thus, the GnRH proximal promoter is regulated by the Otx2 homeoprotein. Finally, we have now demonstrated the presence of Otx2 protein in the GnRH neurons of the adult mouse hypothalamus. These data suggest that Otx2 is important in the development of the GnRH neuron and/or in the maintenance of GnRH expression in the adult mouse hypothalamus.


Subject(s)
Gonadotropin-Releasing Hormone/genetics , Homeodomain Proteins/metabolism , Nerve Tissue Proteins/metabolism , Trans-Activators/metabolism , 3T3 Cells , Animals , Avian Sarcoma Viruses/genetics , Base Sequence , Binding Sites , Cell Line , Conserved Sequence , Gene Expression Regulation , Gonadotropin-Releasing Hormone/metabolism , Homeodomain Proteins/genetics , Hypothalamus/metabolism , Male , Mice , Nerve Tissue Proteins/genetics , Neurons/metabolism , Organ Specificity , Otx Transcription Factors , Promoter Regions, Genetic , Rats , Trans-Activators/genetics , Transcription, Genetic
7.
Mol Endocrinol ; 14(2): 212-28, 2000 Feb.
Article in English | MEDLINE | ID: mdl-10674395

ABSTRACT

The physiological actions of nitric oxide (NO) as a signaling molecule in endothelial and brain cells and as a toxic molecule used by activated immune cells have been the focus of a wide range of studies. Nevertheless, the downstream effector molecules of this important neuromodulator are not well understood. We have previously demonstrated that expression of the gene for the reproductive neuropeptide, GnRH, is repressed by the glutamate/NO/cyclic GMP (cGMP) signal transduction pathway through cGMP-dependent protein kinase in the hypothalamic GnRH-secreting neuronal cell line GT1-7. This repression localized within a previously characterized 300-bp neuron-specific enhancer. Here, we find that mutation of either of two adjacent elements within the enhancer eliminates repression by this pathway. An AT-rich sequence located at -1695 has homology to the octamer motif known to bind POU-homeodomain proteins, while the adjacent element at -1676 has homology to the C/EBP (CCAAT/enhancer-binding protein) protein family consensus sequence. Antibody supershift assays reveal that one of the proteins bound at the -1695 sequence is Oct-1, and one of the proteins bound to the element at -1676 is C/EBPbeta. These two proteins can bind simultaneously to the adjacent -1695 and -1676 binding sites in vitro. In nuclear extracts of GT1-7 cells treated with an NO donor, the intensity of the Oct-1 complex is increased. However, although Western blot analysis indicates that neither Oct-1 nor C/EBPbeta protein levels are increased, the relative binding affinity of Oct-1 is increased. Dephosphorylation of the nuclear extracts decreases binding of the Oct-1 complex to the -1695 site only in NO donor-treated extracts. Thus, we conclude that Oct-1 and C/EBPbeta are both downstream transcriptional regulators involved in the repression of GnRH gene expression by the glutamate/NO/ cGMP signal transduction pathway.


Subject(s)
Cyclic GMP/metabolism , DNA-Binding Proteins/metabolism , Gonadotropin-Releasing Hormone/genetics , Nuclear Proteins/metabolism , Repressor Proteins/metabolism , Transcription Factors/metabolism , Animals , Binding Sites , CCAAT-Enhancer-Binding Proteins , Cell Line/drug effects , Cyclic GMP/pharmacology , Enhancer Elements, Genetic , Gene Expression Regulation , Glutamic Acid/metabolism , Gonadotropin-Releasing Hormone/drug effects , Gonadotropin-Releasing Hormone/metabolism , Host Cell Factor C1 , Hypothalamus/cytology , Mice , N-Methylaspartate/pharmacology , Neurons/drug effects , Neurons/metabolism , Nitric Oxide/metabolism , Nitroprusside/pharmacology , Octamer Transcription Factor-1 , Rats , Signal Transduction
8.
J Clin Endocrinol Metab ; 85(1): 390-7, 2000 Jan.
Article in English | MEDLINE | ID: mdl-10634415

ABSTRACT

The Prophet of Pit-1 gene (PROP1) encodes a paired-like homeodomain protein, which is expressed early in pituitary gland development. When mutated, it is responsible for combined pituitary hormone deficiency (CPHD) in humans, as well as in Ames dwarf mice (df/df). Several independent mutations in the homeodomain of PROP1 have been identified as causative for the human CPHD phenotype, which has been characterized, thus far, as absence or low levels of GH, PRL, TSH, LH, and FSH. Here, we report 10 CPHD cases, 9 of which were born to consanguineous marriages occurring in a large family living in an isolated area in the Southeast of Brazil. All affected patients present complete absence of puberty and low GH, PRL, TSH, LH, and FSH associated with severe hypoplasia of the pituitary gland, as seen by MRI. All 3 exons of the PROP1 genes of these patients were sequenced. The 301-302delAG frameshift mutation was found in both alleles of each affected case. Surprisingly, we observed ACTH/cortisol insufficiency associated with the PROP1 phenotype. The patients' ages varied between 8 and 67 yr, and cortisol response impairment was identified in 5 of 6 of the older patients and in an 11-yr-old patient. Previous studies have not fully characterized patients at advanced ages, leading us to conclude that the phenotype of this PROP1 mutation includes late-onset adrenal insufficiency. We present an extensive clinical analysis of all of these patients. The presence of ACTH/cortisol deficiency in this family bearing the PROP1 301-302delAG mutation indicates the importance of a complete endocrine characterization and of life-long monitoring of PROP1 patients.


Subject(s)
Adrenocorticotropic Hormone/deficiency , Homeodomain Proteins/genetics , Pituitary Hormones/deficiency , Pituitary-Adrenal System/physiopathology , Sequence Deletion/genetics , Transcription Factors/genetics , Adult , Aged , DNA/analysis , DNA/genetics , Female , Gonadotropin-Releasing Hormone , Human Growth Hormone/blood , Human Growth Hormone/deficiency , Humans , Hydrocortisone/deficiency , Hypoglycemic Agents , Insulin , Insulin-Like Growth Factor I/metabolism , Luteinizing Hormone/blood , Luteinizing Hormone/deficiency , Male , Middle Aged , Pedigree , Pituitary Gland/pathology , Pituitary Hormones/blood , Pituitary-Adrenal Function Tests , Sexual Maturation/physiology
9.
Nucleic Acids Res ; 28(4): 1036-43, 2000 Feb 15.
Article in English | MEDLINE | ID: mdl-10648798

ABSTRACT

The AP-2 family of transcriptional regulator proteins has three members, alpha, beta and gamma. AP-2alpha and gamma are expressed in placenta and in the human trophoblast cell line JEG-3. AP-2 has been shown to regulate expression of the placental human chorionic gonado-tropin (hCG) alpha- and beta-subunit genes, however, previous work did not distinguish between the family members. Tryptic peptides of the AP-2 protein complexes purified from JEG-3 cells by oligo-affinity chromatography using the hCGalpha AP-2 site match the amino acid sequence of AP-2gamma. The fact that AP-2gamma is present at significant levels and binds the hCGalpha trophoblast-specific element suggests that AP-2gamma is at least part of the binding complex in vivo and plays a role in regulating hCG expression. We show that mutation of each of four AP-2 binding sites within the hCGbeta promoter decreases expression in transfection assays, demonstrating that all four sites are required for maximal expression in JEG-3 cells. Furthermore, we find differences in regulation of the family members: AP-2alpha mRNA levels increase in response to cAMP while AP-2gamma mRNA levels do not. The demonstrated importance of the AP-2 sites in controlling hCGalpha and beta expression and the likely involvement of more than one family member suggest that a balance in AP-2 proteins is involved in coordinate regulation of these genes. Moreover, many placenta-restricted genes are regulated by AP-2 proteins, thus members of this family may play an important overall role in placenta-specific expression.


Subject(s)
Chorionic Gonadotropin/genetics , Cyclic AMP/physiology , DNA-Binding Proteins/physiology , Gene Expression Regulation/physiology , Transcription Factors/physiology , Base Sequence , DNA Primers , DNA-Binding Proteins/chemistry , Humans , Molecular Sequence Data , Peptide Mapping , Placenta/metabolism , Transcription Factor AP-2 , Transcription Factors/chemistry , Tumor Cells, Cultured
10.
Neuroendocrinology ; 70(5): 353-9, 1999 Nov.
Article in English | MEDLINE | ID: mdl-10567861

ABSTRACT

Evidence suggests that insulin-like growth factors (IGFs; IGF-I and IGF-II) are involved in the regulation of reproductive function including the development of the gonadotropin-releasing hormone (GnRH) neuronal system and the modulation of GnRH secretory activities. To further characterize the regulatory role of the IGF system on GnRH neuronal function, we have examined the gene expression of IGF-I, IGF-II, IGF-I receptor (IGF-IR), and IGF-binding proteins (IGFBPs) in a GnRH neuronal cell line (GT1-7 cells). The relative effects of IGFs and insulin on GnRH secretion by these cells was also investigated. RT-PCR analysis demonstrated IGF-I, IGF-II and IGF-IR mRNAs in GT1-7 cells. The mRNAs for IGFBP-2, -3, -4, -5 and -6 but not IGFBP-1 were also detected. Immunoreactive protein bands for IGFBP-2, -4 and -5 but not for other IGFBPs were demonstrated by Western blot with IGFBP-5 appearing to be the most abundant IGFBP secreted by GT1-7 cells. IGFBP-5 production by GT1-7 cells was stimulated by both IGF-I and IGF-II in a dose-dependent manner with approximately equal potency, whereas insulin caused no significant effect. GnRH secretion by GT1-7 cells treated with IGF-I or IGF-II but not insulin showed an increase (80-100%) at 2 h of treatment followed by a decrease (46%) at 6 h that continued up to 24 h. We conclude that the expression of IGFs, IGF-IR and IGFBPs and their interactions in the regulation of GnRH secretion by GT1-7 cells as demonstrated by our study provide a basis for an autocrine regulatory role for the IGF system in GnRH neuronal secretory activities.


Subject(s)
Gonadotropin-Releasing Hormone/genetics , Neurons/physiology , Somatomedins/genetics , Animals , Blotting, Western , Cells, Cultured , DNA Primers , Gene Expression/physiology , Gonadotropin-Releasing Hormone/analysis , Hypoglycemic Agents/pharmacology , Hypothalamus/cytology , Insulin/pharmacology , Insulin-Like Growth Factor Binding Proteins/analysis , Insulin-Like Growth Factor Binding Proteins/genetics , Insulin-Like Growth Factor I/pharmacology , Insulin-Like Growth Factor II/pharmacology , Neurons/chemistry , Neurons/cytology , RNA, Messenger/analysis , Receptor, IGF Type 1/analysis , Receptor, IGF Type 1/genetics , Somatomedins/analysis
11.
Neuroendocrinology ; 70(4): 246-54, 1999 Oct.
Article in English | MEDLINE | ID: mdl-10529619

ABSTRACT

Activin is essential for the regulation of normal mammalian reproductive function at both the pituitary and gonadal levels. However, its central actions in the control of the hypothalamic-pituitary-gonadal axis remain largely unexplored. The present study aims to determine whether activin could regulate the reproductive axis at the level of the hypothalamus, through control of the GnRH neuroendocrine system. Using the GnRH-secreting GT1-7 neuronal cell line as a model system, we demonstrate expression of mRNAs encoding activin receptor types I, IB, and II. We examined the effects of activin A on GnRH protein secretion and mRNA levels in GT1-7 cells. Treatment with rh-activin A regulated both GnRH protein secretion and GnRH mRNA expression in the GT1-7 cells in a time-dependent fashion. Using transient transfection assays, we explored a potential transcriptional basis for these changes. Activin A increased reporter gene activity driven by minimal GnRH enhancer and promoter elements, suggesting that activin may regulate GnRH gene expression at the level of transcription. Lastly, activin A treatment of male rat hypothalami, in vitro, increased GnRH protein secretion. Collectively, molecular and physiological evidence support the presence of an activin system which might act at a hypothalamic site to regulate mammalian reproduction via activation of GnRH synthesis and release.


Subject(s)
Gonadotropin-Releasing Hormone/genetics , Gonadotropin-Releasing Hormone/metabolism , Hypothalamus/cytology , Inhibins/pharmacology , Activin Receptors , Activins , Animals , Cell Line, Transformed , Gene Expression/drug effects , In Vitro Techniques , Male , Neurons/cytology , Neurons/drug effects , Neurons/metabolism , Neurosecretory Systems/cytology , RNA, Messenger/analysis , Rats , Rats, Sprague-Dawley , Receptors, Growth Factor/metabolism , Transcriptional Activation/drug effects
12.
Cell Tissue Res ; 298(1): 55-61, 1999 Oct.
Article in English | MEDLINE | ID: mdl-10555539

ABSTRACT

The pituitary homeobox1 gene (Ptx1) was initially identified as encoding a pituitary-restricted transcription factor for the proopiomelanocortin (POMC) gene. In order to elucidate the expression pattern of the Ptx1 protein, we investigated the localization of the protein in adult rat pituitary gland and in various pituitary cell lines. We produced an antibody specific for Ptxl protein, and confirmed its specificity by Western blot analysis. Immunohistochemically, many nuclei in the anterior pituitary cells as well as in the intermediate cells were positive for Ptxl staining with this specific antibody. Immunohistochemical double staining revealed the presence of Ptx1 not only in all types of hormone-secreting cells but also in some folliculo-stellate (FS) cells. Furthermore, the expression of Ptx1 mRNA was confirmed in various pituitary cell lines and in the FS cell line by using the reverse transcriptase-polymerase chain reaction (RT-PCR) method. Our studies indicated that Ptxl may not only play a role as a basic transcriptional factor for production of various hormones, but may also play some important role(s) in FS cells. Possible synergistic actions with other factors remain to be investigated. The novel finding of Ptx1 in FS cells is of particular interest, and may suggest that FS cells and hormone-secreting cells are derived from a common cellular ancestor.


Subject(s)
Homeodomain Proteins/genetics , Pituitary Gland/metabolism , Transcription Factors/genetics , Animals , Base Sequence , Cell Line , DNA Primers/genetics , Female , Gene Expression , Genes, Homeobox , Immunohistochemistry , Male , Paired Box Transcription Factors , Pituitary Gland/cytology , Pituitary Hormones/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Rats, Wistar , Reverse Transcriptase Polymerase Chain Reaction
13.
Endocrinology ; 139(11): 4476-82, 1998 Nov.
Article in English | MEDLINE | ID: mdl-9794455

ABSTRACT

TSH is expressed in two populations of thyrotropes in the pituitary: one in the pars distalis and a second in the pars tuberalis. Pars distalis thyrotropes exhibit classical endocrine inhibition of TSH by thyroid hormone, whereas pars tuberalis thyrotropes do not. The majority of our understanding of TSH subunit gene regulation has come from studies conducted in dispersed pituitary, dispersed thyrotropic tumors, or the GH3 somatolactotrope cell line. However, the dispersed pituitary model is limited because of its inherent heterogeneity, thyrotropic tumors are difficult to grow and maintain, and the GH3 cells lack endogenous TSH expression. The recent derivation of a clonal thyrotrope cell line, T alphaT1, that expresses thyrotrope-specific markers, overcomes these limitations. However, because it was not possible to distinguish whether the tumor from which the T alphaT1 cells are derived originated in the pars distalis or the pars tuberalis, it was necessary to define their cellular origin and thereby establish their status as representative thyrotrope cells for future molecular studies. In this study, we demonstrate that the T alphaT1 cells express thyroid hormone receptors (beta1 and beta2) and their heterodimeric partner, retinoid X receptor-gamma. Treatment with T3 causes a dose- and time-dependent decrease in the expression of the TSH beta-subunit messenger RNA. In contrast to previous reports in rat pituitary cultures, T3 does not alter TSH beta-subunit messenger RNA stability in the T alphaT1 cells. Based on these data and the presence of thyrotrope-specific isoforms of the transcription factor Pit-1, we conclude that the T alphaT1 cells represent differentiated thyrotropes of the pars distalis and will be a useful model system for future analysis of the cis- and trans-acting factors necessary for thyrotrope-specific and thyroid hormone-regulated TSH gene expression.


Subject(s)
Gene Expression Regulation/drug effects , Thyroid Gland/metabolism , Thyroid Hormones/pharmacology , Thyrotropin/genetics , Animals , Blotting, Northern , Cell Line , DNA-Binding Proteins/biosynthesis , DNA-Binding Proteins/genetics , Depression, Chemical , Homeodomain Proteins/biosynthesis , Homeodomain Proteins/genetics , Mice , Rats , Reverse Transcriptase Polymerase Chain Reaction , Thyroid Gland/cytology , Transcription Factor Pit-1 , Transcription Factors/biosynthesis , Transcription Factors/genetics , Transcription, Genetic
14.
Mol Cell Endocrinol ; 140(1-2): 25-30, 1998 May 25.
Article in English | MEDLINE | ID: mdl-9722164

ABSTRACT

Targeted expression of oncogenes in transgenic mice can immortalize specific cell types to serve as valuable cultured model systems. Utilizing promoter regions from pituitary genes activated in the gonadotrope/thyrotrope lineage at discrete stages of development, we have demonstrated that targeted oncogene expression in transgenic mice can produce cell lines representing sequential stages of differentiation. Each cell line expresses a specific subset of the genes denoting differentiated function such as the subunits of the glycoprotein hormones, hormone receptors, and transcriptional regulatory proteins. These model systems have allowed detailed investigations into molecular and cellular mechanisms otherwise inaccessible in vivo or in complex primary pituitary cell cultures.


Subject(s)
Pituitary Gland, Anterior/cytology , Animals , Cell Differentiation , Cell Line, Transformed , Cell Lineage , Gene Expression Regulation , Mice , Mice, Transgenic , Oncogenes/genetics , Pituitary Gland, Anterior/growth & development , Pituitary Hormones/genetics , Pituitary Neoplasms/pathology , Promoter Regions, Genetic/genetics , Tumor Cells, Cultured
15.
Mol Cell Endocrinol ; 140(1-2): 151-5, 1998 May 25.
Article in English | MEDLINE | ID: mdl-9722183

ABSTRACT

Gonadotropin-releasing hormone (GnRH) is essential for normal reproductive maturation and function. We present a review of the known mechanisms of hypothalamic GnRH transcriptional control through the conserved GnRH promoter. Understanding this promoter region will allow us to comprehend better the complexities of the hypothalamic pituitary-gonadal axis.


Subject(s)
Gonadotropin-Releasing Hormone/genetics , Hormones/metabolism , Promoter Regions, Genetic/genetics , Signal Transduction/genetics , Transcription Factors/metabolism , Animals , DNA-Binding Proteins/metabolism , Gene Expression Regulation , Humans , Octamer Transcription Factor-6
16.
Mol Cell Endocrinol ; 140(1-2): 157-61, 1998 May 25.
Article in English | MEDLINE | ID: mdl-9722184

ABSTRACT

The hypothalamic gonadotropin-releasing hormone (GnRH) neurons are important regulators of reproductive function. During development, these cells arise in the olfactory placode and migrate to the central nervous system, where they form a diffuse population of neurosecretory cells that mediate central nervous system control of reproduction. Little is known of the mechanisms regulating the differentiation of these cells. Studies of the transcriptional regulation of the GnRH gene have demonstrated the importance of the GATA family of zinc-finger transcription factors in gene expression. Although GATA factors are not highly expressed in mature GnRH-secreting neurons, we report that GATA-4 is highly expressed in migrating GnRH neurons in the developing mouse. We also report that a second DNA binding activity regulating GnRH gene expression at the site of GATA-factor action persists in mature hypothalamus and may also play a role in gene expression in the differentiated GnRH neuron.


Subject(s)
Cell Movement , DNA-Binding Proteins/genetics , Embryo, Mammalian/metabolism , Gonadotropin-Releasing Hormone/genetics , Neurons/metabolism , Transcription Factors/genetics , Animals , DNA-Binding Proteins/analysis , Embryonic and Fetal Development , Enhancer Elements, Genetic/genetics , GATA4 Transcription Factor , Gene Expression Regulation , Gonadotropin-Releasing Hormone/analysis , Hypothalamus/metabolism , Immunohistochemistry , Mice , Neurons/cytology , Transcription Factors/analysis
17.
Mol Endocrinol ; 12(5): 714-26, 1998 May.
Article in English | MEDLINE | ID: mdl-9605934

ABSTRACT

Tissue-specific expression of the mammalian FTZ-F1 gene is essential for adrenal and gonadal development and sexual differentiation. The FTZ-F1 gene encodes an orphan nuclear receptor, termed SF-1 (steroidogenic factor-1) or Ad4BP, which is a primary transcriptional regulator of several hormone and steroidogenic enzyme genes that are critical for normal physiological function of the hypothalamic-pituitary-gonadal axis in reproduction. The objective of the current study is to understand the molecular mechanisms underlying transcriptional regulation of SF-1 gene expression in the pituitary. We have studied a series of deletion and point mutations in the SF-1 promoter region for transcriptional activity in alphaT3-1 and L/betaT2 (pituitary gonadotrope), CV-1, JEG-3, and Y1 (adrenocortical) cell lines. Our results indicate that maximal expression of the SF-1 promoter in all cell types requires an E box element at -82/-77. This E box sequence (CACGTG) is identical to the binding element for USF (upstream stimulatory factor), a member of the helix-loop-helix family of transcription factors. Studies of the SF-1 gene E box element using gel mobility shift and antibody supershift assays indicate that USF may be a key transcriptional regulator of SF-1 gene expression.


Subject(s)
DNA-Binding Proteins/genetics , Gene Expression Regulation/physiology , Helix-Loop-Helix Motifs/physiology , Leucine Zippers/physiology , Pituitary Gland, Anterior/physiology , Transcription Factors/genetics , Transcription Factors/physiology , Animals , Binding Sites/genetics , Biological Transport/genetics , Cell Line , DNA-Binding Proteins/metabolism , Fushi Tarazu Transcription Factors , Haplorhini , Homeodomain Proteins , Humans , Introns , Macromolecular Substances , Mice , Mutagenesis, Site-Directed , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Pituitary Gland, Anterior/cytology , Pituitary Gland, Anterior/metabolism , Promoter Regions, Genetic/genetics , Rats , Receptors, Cytoplasmic and Nuclear , Regulatory Sequences, Nucleic Acid/genetics , Steroidogenic Factor 1 , Transcription, Genetic , Tumor Cells, Cultured , Upstream Stimulatory Factors
18.
Mol Endocrinol ; 12(4): 469-81, 1998 Apr.
Article in English | MEDLINE | ID: mdl-9544983

ABSTRACT

The GnRH gene is exclusively expressed in a discrete population of neurons in the hypothalamus. The promoter-proximal 173 bp of the rat GnRH gene are highly conserved through evolution and are bound by multiple nuclear proteins found in the neuronal cell line, GT1-7, a model for the GnRH-expressing hypothalamic neuron. To explore the protein-DNA interactions that occur within this promoter and the role of these interactions in targeting GnRH gene expression, we have mutagenized individual binding sites in this region. Deoxyribonuclease I protection experiments reveal that footprint 2, a 51-bp sequence that confers a 20-fold induction of the GnRH gene, is comprised of at least three independent protein-binding sites. Transfections of the GnRH promoter-reporter plasmid containing a series of block mutations of footprint 2 into GT1-7 neurons indicate that each of the three putative component sites contributes to transcriptional activity. Mutations in footprint 4 also decrease GnRH gene expression. Footprint 4 and the promoter-proximal site in footprint 2 contain octamer-like motifs, an element that is also present in the neuron-specific enhancer of the rat GnRH gene located approximately 1.6 kb upstream of the promoter. Previous studies in our laboratory have demonstrated that two enhancer octamer sites are bound by the POU-homeodomain transcription factor Oct-1 in GT1-7 cells. We now show that Oct-1 binds the octamer motifs within footprints 2 and 4. Thus, Oct-1 plays a critical role in the regulation of GnRH transcription, binding functional elements in both the distal enhancer and the promoter-proximal conserved region.


Subject(s)
DNA-Binding Proteins/metabolism , Gonadotropin-Releasing Hormone/genetics , Promoter Regions, Genetic/physiology , Transcription Factors/metabolism , Transcription, Genetic , Animals , Base Sequence , Binding Sites/genetics , Cell Line , DNA Footprinting , Host Cell Factor C1 , Mice , Molecular Sequence Data , Mutagenesis, Site-Directed , Octamer Transcription Factor-1 , Promoter Regions, Genetic/genetics , Protein Binding/genetics , Rats
19.
Mol Endocrinol ; 12(3): 364-77, 1998 Mar.
Article in English | MEDLINE | ID: mdl-9514154

ABSTRACT

Neuron-specific expression of the GnRH gene is dependent on an upstream multicomponent enhancer. This enhancer is functional in a small population of GnRH-producing hypothalamic neurons which, through the secretion of GnRH, mediates central nervous system control of reproductive function. GnRH enhancer function requires activation by the GATA family of transcription factors that act through tandem consensus GATA-binding motifs, GATA-A and GATA-B. Here we show that two newly identified DNA-binding factors, termed GBF-A1/A2 and GBF-B1, bind the GnRH enhancer at sites overlapping the GATA factor-binding motifs. In vitro bindings of GATA, GBF-A1/A2, and GBF-B1 to the GnRH enhancer sequences are independent. Specific mutation of either the consensus GATA motif or the GBF-B1 site of GATA-B does not alter binding of the overlapping factor in vitro. Utilizing a GnRH-expressing neuronal cell line as a model system, we show by transient transfection that GBF-B1 is necessary for enhancer activity and independently activates the GnRH promoter. Transactivation of the GnRH enhancer in GT1 cells and in NIH 3T3 cells by GATA-4 is modulated by GBF-B1 binding, suggesting GBF-B1 interferes with GATA factor binding through a steric mechanism.


Subject(s)
Enhancer Elements, Genetic , Gonadotropin-Releasing Hormone/genetics , Gonadotropin-Releasing Hormone/metabolism , Transcription Factors/metabolism , Animals , Base Sequence , Binding Sites , Binding, Competitive , Cell Line , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Deoxyribonuclease I/metabolism , G-Box Binding Factors , GATA4 Transcription Factor , Gene Expression Regulation , Humans , Mice , Molecular Sequence Data , Mutation , Neurons/physiology , Promoter Regions, Genetic , Transcription Factors/genetics , Transcription, Genetic
20.
Development ; 122(10): 3319-29, 1996 Oct.
Article in English | MEDLINE | ID: mdl-8898243

ABSTRACT

Targeted expression of oncogenes in transgenic mice can immortalize specific cell types to serve as valuable cultured model systems. Utilizing promoter regions from a set of genes expressed at specific stages of differentiation in a given cell lineage, we demonstrate that targeted oncogenesis can produce cell lines representing sequential stages of development, in essence allowing both spatial and temporal immortalization. Our strategy was based on our production of a committed but immature pituitary gonadotrope cell line by directing expression of the oncogene SV40 T antigen using a gonadotrope-specific region of the human glycoprotein hormone alpha-subunit gene in transgenic mice. These cells synthesize alpha-subunit and gonadotropin-releasing hormone (GnRH) receptor, yet are not fully differentiated in that they do not synthesize the beta-subunits of luteinizing hormone (LH) or follicle-stimulating hormone (FSH). This observation lead to the hypothesis that targeting oncogenesis with promoters that are activated earlier or later in development might immortalize cells that were more primitive or more differentiated, respectively. To test this hypothesis, we used an LHbeta promoter to immortalize a cell that represents a subsequent stage of gonadotrope differentiation (expression of alpha-subunit, GnRH receptor, and LH beta-subunit but not FSH beta-subunit). Conversely, targeting oncogenesis with a longer fragment of the human alpha-subunit gene (which is activated earlier in development) resulted in the immortalization of a progenitor cell that is more primitive, expressing only the alpha-subunit gene. Interestingly, this transgene also immortalized cells of the thyrotrope lineage that express both alpha- and beta-subunits of thyroid-stimulating hormone and the transcription factor GHF-1 (Pit-1). Thus, targeted tumorigenesis immortalizes mammalian cells at specific stages of differentiation and allows the production of a series of cultured cell lines representing sequential stages of differentiation in a given cell lineage.


Subject(s)
Pituitary Gland/cytology , Pituitary Gland/metabolism , Animals , Antigens, Polyomavirus Transforming/genetics , Carcinogenicity Tests , Cell Differentiation , Female , Gene Expression , Gonadotropin-Releasing Hormone/genetics , Gonadotropin-Releasing Hormone/metabolism , Homeodomain Proteins/metabolism , Humans , LIM-Homeodomain Proteins , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Inbred ICR , Mice, Transgenic , Oncogenes/genetics , Pituitary Hormones, Anterior/genetics , Pituitary Hormones, Anterior/metabolism , Pituitary Neoplasms/pathology , Transcription Factors/metabolism , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL
...