Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
Endocrinology ; 149(7): 3643-55, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18403486

ABSTRACT

The human glycoprotein hormone alpha-subunit (alphaGSU) gene is transcriptionally regulated by glucocorticoids in a cell type-specific fashion. In direct contrast to repression of alphaGSU by glucocorticoids in placenta, glucocorticoid receptor (GR) modulation in the pituitary is little understood. We show that glucocorticoids stimulate the alphaGSU promoter in immortalized pituitary gonadotrope-derived LbetaT2 cells, whereas estrogens, androgens, and progestins have no significant effect. Moreover, GR acts in a dose-dependent manner at physiological concentrations of glucocorticoids. Transient transfection of GR with dexamethasone (Dex) treatment further stimulates the alphaGSU promoter, but this induction is severely diminished using a receptor mutated in the DNA-binding domain. Truncation and cis mutations demonstrate that glucocorticoid response element 2 (GRE2) and cAMP-response element 2 (CRE2) within -168 bp of the human alphaGSU promoter are critical for induction. Moreover, dominant-negative CRE-binding protein markedly inhibits basal but also Dex induction of alphaGSU promoter activity. Additionally, GR specifically binds to GRE2 in the human alphaGSU promoter in vitro and to the 5' region of the endogenous mouse alphaGSU gene in vivo. Furthermore, overexpression of the homeobox factor, Distal-less 3 that regulates this gene in placental cells through a site partially overlapping GRE2, blocks Dex induction of alphaGSU in gonadotrope cells, indicating that placenta-specific expression of Dlx3 may interfere with GR, resulting in repression in placental cells vs. induction in gonadotrope cells. These results demonstrate the stimulatory role played by glucocorticoids in alphaGSU gene expression in the pituitary gonadotrope, in contrast to repression in placental cells, and highlight the tissue-specific nature of steroid hormone action.


Subject(s)
Glucocorticoids/pharmacology , Glycoprotein Hormones, alpha Subunit/metabolism , Gonadotrophs/drug effects , Base Sequence , Binding Sites , Cell Line , Chromatin Immunoprecipitation , Dexamethasone/pharmacology , Dose-Response Relationship, Drug , Electrophoretic Mobility Shift Assay , Gene Expression/drug effects , Glycoprotein Hormones, alpha Subunit/genetics , Gonadotrophs/metabolism , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Humans , Luciferases/genetics , Luciferases/metabolism , Mutagenesis, Site-Directed , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Promoter Regions, Genetic/genetics , Protein Binding , Receptors, Glucocorticoid/genetics , Receptors, Glucocorticoid/metabolism , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Sequence Homology, Nucleic Acid , Transcription Factors/genetics , Transcription Factors/metabolism , Transfection
2.
Neuroendocrinology ; 84(6): 353-63, 2006.
Article in English | MEDLINE | ID: mdl-17192702

ABSTRACT

Orexin A, a recently discovered hypothalamic peptide, has been shown to have a stimulatory effect on release of gonadotropin-releasing hormone (GnRH) from rat hypothalamic explants in vitro. However, it is presently unclear whether in vivo this effect is mediated directly at the level of the GnRH neuron, or via multiple afferent neuronal connections. Therefore, in the present study, we investigated the direct action of orexin A on GnRH neurons using the immortalized GnRH-secreting GT1-7 hypothalamic cells. Orexin-1 receptor (OX1R) expression was detected in GT1-7 cells by RT-PCR and Western blot. Results showed that 0.1-1 nM orexin A, when administered in culture media for 4 h, can significantly stimulate GnRH mRNA expression in GT1-7 cells (p < 0.05). Administration of 1 microM OX1R antagonist, SB-334867, completely blocked the observed orexin A responses in these cells, indicating that orexin A stimulation of GnRH neurons is specifically through OX1R. Moreover, 0.1 nM orexin A stimulated GnRH release after 30-45 min. To examine possible signal transduction pathways involved in mediating these effects, a MEK inhibitor (UO-126), PKC inhibitor (calphostin C), and PKA inhibitor (H-89), were used, with each blocking orexin A-induced GnRH transcription and release from immortalized cells. Collectively, our results show that orexin A is capable of directly stimulating GnRH transcription and neuropeptide release from these immortalized hypothalamic neurons, and that the effects of orexin A appear to be mediated via the OX1R, coupled with activation of the PKC-, MAPK- and PKA-signaling pathways. It is suggested that the stimulatory effect of orexin A on GnRH transcription and release may also occur directly at the level of GnRH neurons in vivo.


Subject(s)
Gene Expression Regulation/drug effects , Gonadotropin-Releasing Hormone/metabolism , Hypothalamus/metabolism , Intracellular Signaling Peptides and Proteins/pharmacology , Neuropeptides/pharmacology , Neurotransmitter Agents/pharmacology , Animals , Cells, Cultured , Cyclic AMP-Dependent Protein Kinases/physiology , Gonadotropin-Releasing Hormone/genetics , Humans , Hypothalamus/cytology , Male , Mice , Mice, Inbred C57BL , Mitogen-Activated Protein Kinase Kinases/physiology , Neurons/metabolism , Orexin Receptors , Orexins , Protein Kinase C/physiology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/metabolism , Receptors, Neuropeptide/genetics , Receptors, Neuropeptide/metabolism , Signal Transduction/physiology
3.
Steroids ; 68(10-13): 861-7, 2003 Nov.
Article in English | MEDLINE | ID: mdl-14667978

ABSTRACT

Ovarian cell death is an essential process for the homeostasis of ovarian function in human and other mammalian species. It ensures the selection of the dominant follicle and the demise of excess follicles. In turn, this process minimizes the possibility of multiple embryo development during pregnancy and assures the development of few, but healthy embryos. Degeneration of the old corpora lutea in each estrous/menstrual cycle by programmed cell death is essential to maintain the normal cyclicity of ovarian steroidogenesis. Although there are multiple pathways that can determine cell death or survival, crosstalk among endocrine, paracrine and autocrine factors, as well as among protooncogenes, tumor suppressor genes, survival genes and death genes, plays an important role in determining the fate of ovarian somatic and germ cells. The establishment of immortalized rat and human steroidogenic granulosa cell lines and the investigation of pure populations of primary granulosa cells allows systematic studies of the mechanisms that control steroidogenesis and apoptosis in granulosa cells. We have discovered that during initial stages of granulosa cell apoptosis progesterone production does not decrease. In contrast, we found that it is elevated up to 24h following the onset of the apoptotic stimuli exerted by starvation, cAMP, p53 or TNF-alpha stimulation, before total cell collapse. These observations raise the possibility for an alternative unique apoptotic pathway, one not involving mitochondrial Cyt C release associated with the destruction of mitochondrial structure and steroidogenic function. Using mRNA from apoptotic cells and affymetrix DNA microarray technology we discovered that granzyme B, a protease that normally resides in T cytotoxic lymphocytes and natural killer cells of the immune system is expressed and activated in granulosa cells. Thus, the apoptotic signals could bypass mitochondrial signals for apoptosis, which can preserve their steroidogenic activity until complete cell destruction. This unique apoptotic pathway assures cyclicity of estradiol and progesterone release in the estrous/menstruous cycle even during the initial stages of apoptosis.


Subject(s)
Apoptosis , Ovary/metabolism , Ovary/pathology , Steroids/metabolism , Animals , Cell Death , Cell Line , Cell Survival , Cyclic AMP/metabolism , Female , Granulosa Cells/metabolism , Granulosa Cells/pathology , Granzymes , Humans , Kinetics , Mitochondria/metabolism , Models, Biological , Oligonucleotide Array Sequence Analysis , Progesterone/metabolism , Rats , Serine Endopeptidases/pharmacology , T-Lymphocytes/metabolism , Time Factors , Tumor Necrosis Factor-alpha/metabolism , Tumor Suppressor Protein p53/metabolism
4.
FASEB J ; 17(10): 1256-66, 2003 Jul.
Article in English | MEDLINE | ID: mdl-12832290

ABSTRACT

Follicle-stimulating hormone (FSH) controls the development of follicle-enclosed oocytes in the mammalian ovary by interacting with specific receptors located exclusively on granulosa cells. Its biological activity involves stimulation of intercellular communication, intracellular signaling, and up-regulation of steroidogenesis; the entire spectrum of genes regulated by FSH is not yet fully characterized. We have established monoclonal rat FSH-responsive granulosa cell lines that express FSH receptors at 20-fold higher rates than with primary cells, and thus increased the probability of yielding a distinct spectrum of genes modulated by FSH. Using Affymetrix DNA microarrays, we discovered 11 genes not reported earlier to be up-regulated by FSH and 9 genes not reported earlier to be down-regulated by FSH. Modulation of signal transduction associated with G-protein signaling, phosphorylation of proteins, and intracellular-extracellular ion balance was suggested by up-regulation of decay accelerating factor GPI-form precursor (DAF), membrane interacting protein RGS16, protein tyrosine phosphatase (PTPase), oxidative stress-inducible protein tyrosine phosphatase (OSIPTPase), and down-regulation of rat prostatic acid phosphatase (rPAP), Na+, K+-ATPase, and protein phosphatase 1beta. Elevation in granzyme-like proteins 1 and 3, and natural killer (NK) cell protease 1 (NKP-1) along with reduction in carboxypeptidase E indicates possible FSH-mediated preparation of the cells for apoptosis. Up-regulation of vascular endothelial growth factors indicates the ability of FSH to produce angiogenic factors upon their maturation; whereas, reduction in insulin-like growth factor binding protein (IGFBP3) indicates its increased potential to promote p53-induced apoptosis. Striking similarities in FSH modulation of gene expression were found in primary cultures of human granulosa cells obtained from IVF patients although these cells expressed only 1% of FSH receptor compared with immortalized rat cells, as indicated by microarray technique, which probably is in the normal range of expression of this receptor in nontransformed cells. These findings should increase our understanding of the mechanism of FSH action in stimulating development of the ovarian follicular cells, of intracellular and intercellular communication, and of increasing the potential of ovarian follicular cells to undergo apoptosis during the process of selection of the dominant follicle.


Subject(s)
Follicle Stimulating Hormone/pharmacology , Gene Expression Regulation , Granulosa Cells/metabolism , Animals , Antioxidants/metabolism , Apoptosis , Cell Communication , Cell Line, Transformed , Cells, Cultured , Cytokines/biosynthesis , Cytokines/genetics , Female , Gene Expression Profiling , Gene Expression Regulation/drug effects , Granulosa Cells/drug effects , Granulosa Cells/physiology , Growth Substances/biosynthesis , Growth Substances/genetics , Humans , Nervous System Physiological Phenomena , Peptide Hydrolases/biosynthesis , Peptide Hydrolases/genetics , Phosphoprotein Phosphatases/biosynthesis , Phosphoprotein Phosphatases/genetics , Protein Kinases/biosynthesis , Protein Kinases/genetics , Rats , Signal Transduction , Steroids/biosynthesis
5.
Biochem Pharmacol ; 64(5-6): 843-50, 2002 Sep.
Article in English | MEDLINE | ID: mdl-12213578

ABSTRACT

Glucocorticoids play a major role in attenuation of the inflammatory response. These steroid hormones are able to induce apoptosis in cells of the hematopoietic system such as monocytes, macrophages, and T lymphocytes that are involved in the inflammation reaction. In contrast, it was discovered recently that in glandular cells such as the mammary gland epithelia, hepatocytes, ovarian follicular cells, and in fibroblasts glucocorticoids protect against apoptotic signals evoked by cytokines, cAMP, tumor suppressors, and death genes. The anti-apoptotic effect of glucocorticoids is exerted by modulation of several survival genes such as Bcl-2, Bcl-x(L), and NFkB, in a cell-specific manner. Moreover, upregulation or downregulation of the same gene product can occur in a cell-dependent manner following stimulation by glucocorticoids. This phenomenon is probably due to composite regulatory cross-talk among multiple nuclear coactivators or corepressors, which mediate the transcription regulation of the genes, by their interaction with the glucocorticoid receptor. These observations suggest that the anti-inflammatory action of glucocorticoids is exerted by two complementary mechanisms: on one hand, they induce death of the cells that provoke the inflammation, and on the other hand they protect the resident cells of the inflamed tissue by arresting apoptotic signals. Moreover, the complementary action of glucocorticoids provides a new insight to the therapeutic potential of these hormones.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Apoptosis , Glucocorticoids/pharmacology , Animals , Apoptosis/genetics , Breast/cytology , Breast/drug effects , Female , Fibroblasts/cytology , Fibroblasts/drug effects , Humans , Liver/cytology , Liver/drug effects , Ovary/cytology , Ovary/drug effects
6.
J Clin Endocrinol Metab ; 87(7): 3441-51, 2002 Jul.
Article in English | MEDLINE | ID: mdl-12107264

ABSTRACT

Human granulosa cells obtained from in vitro fertilization patients are highly luteinized, but can still be stimulated by LH/cAMP for production of progesterone. This stimulation involved enhancement of apoptosis. Incubation of the cells with dexamethasone (Dex) reduced the apoptotic incidence compared with nontreated cells and completely abolished the increase in apoptosis stimulated by LH or forskolin, concomitantly with a pronounced increase in progesterone production. Organization of the actin cytoskeleton was dramatically reduced after LH/forskolin stimulation. In contrast, Dex prevented disorganization of the actin filament networks. LH and forskolin also decreased the organization of gap junctions, which could be prevented by Dex. However, the intracellular level of connexin 43 was elevated in the presence of LH, forskolin, and Dex. Endogenous levels of the survival gene protein Bcl-2 were significantly elevated in all cultures treated with Dex compared with either nonstimulated cultures or cultures stimulated with LH and forskolin. Our data suggest that LH/cAMP can stimulate steroidogenesis even during the initial stage of apoptosis of human granulosa cells, whereas Dex, which blocks apoptosis, could further elevate progesterone production. Moreover, the integrity of gap junctions and the actin cytoskeleton as well as elevated levels of Bcl-2 may play an important role in the suppression of apoptosis of human granulosa cells.


Subject(s)
Apoptosis/drug effects , Apoptosis/physiology , Dexamethasone/pharmacology , Fertilization in Vitro , Granulosa Cells/drug effects , Granulosa Cells/physiology , Actins/drug effects , Actins/physiology , Cell Communication/physiology , Cells, Cultured , Colforsin/pharmacology , Connexin 43/metabolism , Female , Gap Junctions/drug effects , Glucocorticoids/pharmacology , Humans , Intracellular Membranes/metabolism , Luteinizing Hormone/pharmacology , Proto-Oncogene Proteins c-bcl-2/metabolism
7.
Mol Cell Endocrinol ; 189(1-2): 1-9, 2002 Mar 28.
Article in English | MEDLINE | ID: mdl-12039060

ABSTRACT

Glucocorticoids play a major role in attenuation of the inflammatory response. These steroid hormones are able to induce apoptosis in cells of the hematopoietic system such as monocytes, macrophages and T-lymphocytes that are involved in the inflammation reaction. In contrast, it was discovered recently that in glandular cells such as the mammary gland epithelia, hepatocytes, ovarian follicular cells and in fibroblasts glucocorticoids protect against apoptotic signals evoked by cytokines, cAMP, tumor suppressors and death genes. The anti-apoptotic effect of glucocorticoids is exerted by modulation of several survival genes such as Bcl-2, Bcl-x(L) and NFkappaB, in a cell type-specific manner. Moreover, up regulation or down regulation of the same gene product can occur in a cell type-dependent manner following stimulation by glucocorticoids. This phenomenon is probably due to composite regulatory cross-talk among multiple nuclear coactivators or corepressors, which mediate the transcriptional regulation of the genes, by their interaction with the glucocorticoid receptor (GR). These observations suggest that the anti-inflammatory action of glucocorticoids is exerted by two complementary mechanisms: on the one hand, they induce death of the cells that provoke the inflammation, and on the other hand, they protect the resident cells of the inflamed tissue by arresting apoptotic signals.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Apoptosis/drug effects , Glucocorticoids/pharmacology , Animals , Apoptosis/genetics , Apoptosis/physiology , Breast/physiology , Cell Line , Female , Fibroblasts/metabolism , Humans , Inflammation/metabolism , Liver/physiology , Models, Biological , Ovary/physiology , Receptors, Glucocorticoid/metabolism
8.
Biochem Biophys Res Commun ; 294(1): 51-9, 2002 May 31.
Article in English | MEDLINE | ID: mdl-12054739

ABSTRACT

Tumor necrosis factor alpha (TNF alpha) plays a role in mammalian ovarian follicular development, steroidogenesis, ovulation, luteolysis, and atresia, but the exact mechanism of TNF alpha action is not completely understood. Induction of apoptosis and suppression of steroidogenesis by TNF alpha in primary preovulatory rat and human granulosa cells, as well as, in human granulosa cells immortalized by mutated p53, were characterized in the present work. Dexamethasone (Dex) and hydrocortisone efficiently suppressed TNF alpha-induced apoptosis in granulosa cells. TNF alpha dramatically reduced intracellular levels of Bcl-2, while Dex abrogated this reduction. TNF alpha reduced considerably intracellular levels of StAR protein, a key regulating factor in steroidogenesis. This reduction can be explained only in part by elimination of cells through apoptosis, since loss of steroidogenic capacity was much higher and faster than the rate and extent of loss of cell viability induced by TNF alpha, suggesting independent mechanisms for TNF alpha-induction of apoptosis and TNF alpha-suppression of steroidogenesis.


Subject(s)
Apoptosis/drug effects , Dexamethasone/pharmacology , Granulosa Cells/drug effects , Hydrocortisone/pharmacology , Proto-Oncogene Proteins c-bcl-2/metabolism , Tumor Necrosis Factor-alpha/pharmacology , Animals , Cells, Cultured , Dose-Response Relationship, Drug , Down-Regulation , Female , Flow Cytometry , Humans , In Situ Nick-End Labeling , Phosphoproteins/metabolism , Progesterone/metabolism , Radioimmunoassay , Rats , Tumor Necrosis Factor-alpha/administration & dosage , Tumor Suppressor Protein p53/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...