Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
Endocrinology ; 163(2)2022 02 01.
Article in English | MEDLINE | ID: mdl-34967898

ABSTRACT

Leydig cells produce androgens that are essential for male sex differentiation and reproductive function. Leydig cell function is regulated by several hormones and signaling molecules, including growth hormone (GH). Although GH is known to upregulate Star gene expression in Leydig cells, its molecular mechanism of action remains unknown. The STAT5B transcription factor is a downstream effector of GH signaling in other systems. While STAT5B is present in both primary and Leydig cell lines, its function in these cells has yet to be ascertained. Here we report that treatment of MA-10 Leydig cells with GH or overexpression of STAT5B induces Star messenger RNA levels and increases steroid hormone output. The mouse Star promoter contains a consensus STAT5B element (TTCnnnGAA) at -756 bp to which STAT5B binds in vitro (electrophoretic mobility shift assay and supershift) and in vivo (chromatin immunoprecipitation) in a GH-induced manner. In functional promoter assays, STAT5B was found to activate a -980 bp mouse Star reporter. Mutating the -756 bp element prevented STAT5B binding but did not abrogate STAT5B-responsiveness. STAT5B was found to functionally cooperate with DNA-bound cJUN. The STAT5B/cJUN cooperation was only observed in Leydig cells and not in Sertoli or fibroblast cells, indicating that additional Leydig cell-enriched transcription factors are required. The STAT5B/cJUN cooperation was lost only when both STAT5B and cJUN elements were mutated. In addition to identifying the Star gene as a novel target for STAT5B in Leydig cells, our data provide important new insights into the mechanism of GH and STAT5B action in the regulation of Leydig cell function.


Subject(s)
Growth Hormone/pharmacology , Leydig Cells/metabolism , Phosphoproteins/genetics , Proto-Oncogene Proteins c-jun/physiology , STAT5 Transcription Factor/genetics , Animals , Base Sequence , Binding Sites , Cell Line , DNA/chemistry , DNA/metabolism , Gene Expression/drug effects , Leydig Cells/classification , Male , Mice , Phosphoproteins/analysis , Phosphoproteins/physiology , Promoter Regions, Genetic , RNA, Messenger/analysis , STAT5 Transcription Factor/analysis , STAT5 Transcription Factor/physiology , Up-Regulation/drug effects
2.
Endocrinology ; 158(6): 1886-1897, 2017 06 01.
Article in English | MEDLINE | ID: mdl-28379539

ABSTRACT

The three FOXA transcription factors are mainly known for their roles in the liver. However, Foxa3-deficient mice become progressively sub/infertile due to germ cell loss. Because no data were available regarding the localization of the FOXA3 protein in the testis, immunohistochemistry was performed on mouse testis sections. In the fetal testis, a weak but consistent staining for FOXA3 is detected in the nucleus of Sertoli cells. In prepubertal and adult life, FOXA3 remains present in Sertoli cells of some but not all seminiferous tubules. FOXA3 is also detected in the nucleus of some peritubular cells. From postnatal day 20 onward, FOXA3 is strongly expressed in the nucleus of Leydig cells. To identify FOXA3 target genes in Leydig cells, MLTC-1 Leydig cells were transfected with a series of Leydig cell gene reporters in the presence of a FOXA3 expression vector. The platelet-derived growth factor receptor α (Pdgfra) promoter was significantly activated by FOXA3. The Pdgfra promoter contains three potential FOX elements and progressive 5' deletions and site-directed mutagenesis revealed that the most proximal element at -78 bp was sufficient to confer FOXA3 responsiveness. FOXA3 from Leydig cells could bind to this element in vitro (electrophoretic mobility shift assay) and was recruited to the proximal Pdgfra promoter in vivo (chromatin immunoprecipitation). Finally, endogenous Pdgfra messenger RNA levels were reduced in FOXA3-deficient MLTC-1 Leydig cells. Taken together, our data identify FOXA3 as a marker of the Sertoli cell lineage and of the adult Leydig cell population, and as a regulator of Pdgfra transcription in Leydig cells.


Subject(s)
Cell Lineage/genetics , Hepatocyte Nuclear Factor 3-gamma/genetics , Leydig Cells/metabolism , Receptor, Platelet-Derived Growth Factor alpha/genetics , Testis/cytology , Animals , Cell Line , Gene Expression Regulation , Male , Mice , Rats , Receptor, Platelet-Derived Growth Factor alpha/metabolism , Testis/metabolism
3.
Endocrinology ; 156(12): 4695-706, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26393304

ABSTRACT

Testosterone is essential for spermatogenesis and the development of male sexual characteristics. However, steroidogenesis produces a significant amount of reactive oxygen species (ROS), which can disrupt testosterone production. The myocyte enhancer factor 2 (MEF2) is an important regulator of organogenesis and cell differentiation in various tissues. In the testis, MEF2 is present in Sertoli and Leydig cells throughout fetal and adult life. MEF2-deficient MA-10 Leydig cells exhibit a significant decrease in steroidogenesis concomitant with a reduction in glutathione S-transferase (GST) activity and in the expression of the 4 Gsta members (GST) that encode ROS inactivating enzymes. Here, we report a novel role for MEF2 in ROS detoxification by directly regulating Gsta expression in Leydig cells. Endogenous Gsta1-4 mRNA levels were decreased in MEF2-deficient MA-10 Leydig cells. Conversely, overexpression of MEF2 increased endogenous Gsta1 levels. MEF2 recruitment to the proximal Gsta1 promoter and direct binding on the -506-bp MEF2 element were confirmed by chromatin immunoprecipitation and DNA precipitation assays. In MA-10 Leydig cells, MEF2 activates the Gsta1 promoter and cooperates with Ca(2+)/calmodulin-dependent kinases I to further enhance Gsta1 promoter activity. These effects were lost when the -506-bp MEF2 element was mutated or when a MEF2-Engrailed dominant negative protein was used. Similar results were obtained on the Gsta2, Gsta3, and Gsta4 promoters, suggesting a global role for MEF2 factors in the regulation of all 4 Gsta genes. Altogether, our results identify a novel role for MEF2 in the expression of genes involved in ROS detoxification, a process essential for adequate testosterone production in Leydig cells.


Subject(s)
Gene Expression Regulation, Developmental , Glutathione Transferase/genetics , MEF2 Transcription Factors/genetics , RNA, Messenger/metabolism , Reactive Oxygen Species/metabolism , Testosterone/biosynthesis , Animals , Calcium-Calmodulin-Dependent Protein Kinase Type 1/metabolism , Cell Line , Chromatin Immunoprecipitation , Gene Knockout Techniques , Glutathione Transferase/metabolism , Isoenzymes/genetics , Isoenzymes/metabolism , Leydig Cells , MEF2 Transcription Factors/metabolism , Male , Mice , Promoter Regions, Genetic
4.
Biol Reprod ; 91(1): 26, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24899578

ABSTRACT

Testosterone production is dependent on cholesterol transport within the mitochondrial matrix, an essential step mediated by a protein complex containing the steroidogenic acute regulatory (STAR) protein. In steroidogenic Leydig cells, Star expression is hormonally regulated and involves several transcription factors. NR2F2 (COUP-TFII) is an orphan nuclear receptor that plays critical roles in cell differentiation and lineage determination. Conditional NR2F2 knockout prior to puberty leads to male infertility due to insufficient testosterone production, suggesting that NR2F2 could positively regulate steroidogenesis and Star expression. In this study we found that NR2F2 is expressed in the nucleus of some peritubular myoid cells and in interstitial cells, mainly in steroidogenically active adult Leydig cells. In MA-10 and MLTC-1 Leydig cells, small interfering RNA (siRNA)-mediated NR2F2 knockdown reduces basal steroid production without affecting hormone responsiveness. Consistent with this, we found that STAR mRNA and protein levels were reduced in NR2F2-depleted MA-10 and MLTC-1 cells. Transient transfections of Leydig cells revealed that a -986 bp mouse Star promoter construct was activated 3-fold by NR2F2. Using 5' progressive deletion constructs, we mapped the NR2F2-responsive element between -131 and -95 bp. This proximal promoter region contains a previously uncharacterized direct repeat 1 (DR1)-like element to which NR2F2 is recruited and directly binds. Mutations in the DR1-like element that prevent NR2F2 binding severely blunted NR2F2-mediated Star promoter activation. These data identify an essential role for the nuclear receptor NR2F2 as a direct activator of Star gene expression in Leydig cells, and thus in the control of steroid hormone biosynthesis.


Subject(s)
COUP Transcription Factor II/metabolism , Gonadal Steroid Hormones/biosynthesis , Leydig Cells/metabolism , Phosphoproteins/metabolism , Animals , COUP Transcription Factor II/genetics , Cell Line , Male , Mice , Phosphoproteins/genetics , Promoter Regions, Genetic , RNA, Small Interfering
5.
J Mol Endocrinol ; 53(1): 43-55, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24780841

ABSTRACT

Insulin-like 3 (INSL3), a hormone produced by Leydig cells, regulates testicular descent during foetal life and bone metabolism in adults. Despite its importance, little is known about the molecular mechanisms controlling INSL3 expression. Reduced Insl3 mRNA levels were reported in the testis of mice deficient for chicken ovalbumin upstream promoter-transcription factor II (COUP-TFII), an orphan nuclear receptor known to play critical roles in cell differentiation and lineage determination in several tissues. Although COUP-TFII-deficient mice had Leydig cell dysfunction and impaired fertility, it remained unknown whether Insl3 expression was directly regulated by COUP-TFII. In this study, we observed a significant decrease in Insl3 mRNA levels in MA-10 Leydig cells depleted of COUP-TFII. Furthermore, a -1087 bp mouse Insl3 promoter was activated fourfold by COUP-TFII in MA-10 Leydig cells. Using 5' progressive deletions, the COUP-TFII-responsive element was located between -186 and -79 bp, a region containing previously uncharacterised direct repeat 0-like (DR0-like) and DR3 elements. The recruitment and direct binding of COUP-TFII to the DR0-like element were confirmed by chromatin immunoprecipitation and DNA precipitation assay respectively. Mutation of the DR0-like element, which prevented COUP-TFII binding, significantly decreased COUP-TFII-mediated activation of the -1087 bp Insl3 reporter in CV-1 fibroblast cells but not in MA-10 Leydig cells. Finally, we found that COUP-TFII cooperates with the nuclear receptor steroidogenic factor 1 (SF1) to further enhance Insl3 promoter activity. Our results identify Insl3 as a target for COUP-TFII in Leydig cells and revealed that COUP-TFII acts through protein-protein interactions with other DNA-bound transcription factors, including SF1, to activate Insl3 transcription in these cells.


Subject(s)
COUP Transcription Factor II/metabolism , Insulin/genetics , Leydig Cells/metabolism , Proteins/genetics , Animals , COUP Transcription Factor II/antagonists & inhibitors , COUP Transcription Factor II/genetics , Cell Line , Gene Expression Regulation , Gene Knockdown Techniques , Male , Mice , Promoter Regions, Genetic , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Small Interfering/genetics , Sequence Deletion , Steroidogenic Factor 1/metabolism
6.
Mol Endocrinol ; 28(6): 886-98, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24694307

ABSTRACT

Leydig cell steroidogenesis is controlled by the pituitary gonadotropin LH that activates several signaling pathways, including the Ca(2+)/calmodulin kinase I (CAMKI) pathway. In other tissues, CAMKI regulates the activity of the myocyte enhancer factor 2 (MEF2) transcription factors. MEF2 factors are essential regulators of cell differentiation and organogenesis in numerous tissues but their expression and role in the mammalian gonad had not been explored. Here we show that MEF2 factors are expressed in a sexually dimorphic pattern in the mouse gonad. MEF2 factors are present in the testis throughout development and into adulthood but absent from the ovary. In the testis, MEF2 was localized mainly in the nucleus of both somatic lineages, the supporting Sertoli cells and the steroidogenic Leydig cells. In Leydig cells, MEF2 was found to activate the expression of Nr4a1, a nuclear receptor important for hormone-induced steroidogenesis. In these cells MEF2 also cooperates with forskolin and CAMKI to enhance Nr4a1 promoter activity via two MEF2 elements (-318 and -284 bp). EMSA confirmed direct binding of MEF2 to these elements whereas chromatin immunoprecipitation revealed that MEF2 recruitment to the proximal Nr4a1 promoter was increased following hormonal stimulation. Modulation of endogenous MEF2 protein level (small interfering RNA-mediated knockdown) or MEF2 activity (MEF2-Engrailed active dominant negative) led to a significant decrease in Nr4a1 mRNA levels in Leydig cells. All together, our results identify MEF2 as a novel testis-specific transcription factor, supporting a role for this factor in male sex differentiation and function. MEF2 was also positioned upstream of NR4A1 in a regulatory cascade controlling Leydig cell gene expression.


Subject(s)
Gene Expression Regulation , Leydig Cells/metabolism , MEF2 Transcription Factors/metabolism , Nuclear Receptor Subfamily 4, Group A, Member 1/genetics , Animals , Base Sequence , Calcium-Calmodulin-Dependent Protein Kinase Type 1/genetics , Calcium-Calmodulin-Dependent Protein Kinase Type 1/metabolism , Cell Line , Consensus Sequence , Female , Gene Expression , Humans , Male , Mice , Nuclear Receptor Subfamily 4, Group A, Member 1/metabolism , Organ Specificity , Ovary/metabolism , Promoter Regions, Genetic , Protein Binding , Rats
7.
Can J Physiol Pharmacol ; 87(9): 694-701, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19794520

ABSTRACT

The expression of tissue factor (TF) in tumors reportedly exacerbates the aggressiveness of several types of cancers. The shedding of TF-containing membrane particles is believed to influence the ability of tumors to expand and metastasize, and these microparticles may also be harmful in the onset of disseminated intravascular coagulation in specific cancers. Furthermore, the intracellular signaling that is elicited after the formation of the TF / coagulation factor VIIa complex at the cell membrane modulates the activity of adhesion molecules and mitogen-activated protein (MAP) kinases. To evaluate whether TF overexpression in tumor cells modulates its shedding and neighboring stromal cells by its catalytic or intracellular activity, TF-GFP (green fluorescent protein) and a tailless form (TFDeltaC-GFP) were stably expressed in the rat Morris hepatoma and human HT1080 fibrosarcoma cell lines. Both TF proteins were efficiently produced by tumor cells and functionally active, and their clotting activity could be blocked by the active site-inhibited factor VIIa (ASIS). TF-expressing tumorigenic cells produced a soluble factor that increased the migration of arterial smooth muscle cells in vitro. This effect was abrogated by ASIS and the PAR-1 receptor antagonist ATAP-2, showing that it is dependent on the proteolytic activity of the TF ligand factor VIIa and the thrombin-activated cell membrane receptor. We propose that TF-containing microparticles that are released in the culture medium by tumor cells influence the migratory behavior of neighboring stromal cells, thus aiding the cancer cell's tumorigenic potential.


Subject(s)
Cell Movement/physiology , Myocytes, Smooth Muscle/cytology , Neoplasms/metabolism , Thromboplastin/biosynthesis , Animals , Blood Coagulation , Blotting, Western , Catalysis , Cell Line, Tumor , Cell Membrane/metabolism , Cell Migration Assays , Cell Proliferation , Coculture Techniques , Green Fluorescent Proteins/genetics , Humans , Neoplasms/pathology , Neovascularization, Pathologic , Rabbits , Rats , Recombinant Fusion Proteins/genetics , Thromboplastin/genetics , Transfection
8.
Cancer Lett ; 275(1): 127-38, 2009 Mar 08.
Article in English | MEDLINE | ID: mdl-19022561

ABSTRACT

Excessive exposure to estradiol represents the main risk factor for endometrial cancer. The abnormally high estradiol levels in the endometrium of women with endometrial cancer are most likely due to overproduction by the tumour itself. Endometrial cancer cells express the genes encoding the steroidogenic enzymes involved in estradiol synthesis. Here we used RT-PCR and Western blot to show that the nuclear receptors SF1 and LRH1, two well-known regulators of steroidogenic gene expression in gonadal and adrenal cells, are also expressed in endometrial cancer cell lines. By transient transfections, we found that SF1 and LRH1, but not the related nuclear receptor NUR77, can activate the promoters of three human steroidogenic genes: STAR, HSD3B2, and CYP19A1 PII. Similarly, forskolin but not PMA, could activate all three promoters. In addition, we found that both SF1 and LRH1 can transcriptionally cooperate with the AP-1 family members c-JUN and c-FOS, known to be associated with enhanced proliferation of endometrial carcinoma cells, to further enhance activation of the STAR, HSD3B2, and CYP19A1 PII promoters. All together, our data provide novel insights into the mechanisms of steroidogenic gene expression in endometrial cancer cells and thus in the regulation of estradiol biosynthesis by tumour cells.


Subject(s)
Endometrial Neoplasms/metabolism , Gene Expression Regulation, Neoplastic , Promoter Regions, Genetic , Receptors, Cytoplasmic and Nuclear/metabolism , Steroidogenic Factor 1/metabolism , Steroids/metabolism , Transcription Factor AP-1/metabolism , Transcription, Genetic , Aromatase/metabolism , Colforsin/pharmacology , Female , Humans , Nuclear Receptor Subfamily 4, Group A, Member 1 , Phosphoproteins/metabolism , Progesterone Reductase/metabolism , Receptors, Steroid/metabolism
9.
Mol Endocrinol ; 22(9): 2021-37, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18599618

ABSTRACT

Cholesterol transport in the mitochondrial membrane, an essential step of steroid biosynthesis, is mediated by a protein complex containing the steroidogenic acute regulatory (StAR) protein. The importance of this transporter is underscored by mutations in the human StAR gene that cause lipoid congenital adrenal hyperplasia, male pseudohermaphroditism, and adrenal insufficiency. StAR transcription in steroidogenic cells is hormonally regulated and involves several transcription factors. The nuclear receptor NUR77 is present in steroidogenic cells, and its expression is induced by hormones known to activate StAR expression. We have now established that StAR transcription in cAMP-stimulated Leydig cells requires de novo protein synthesis and involves NUR77. We found that cAMP-induced NUR77 expression precedes that of StAR both at the mRNA and protein levels in Leydig cells. In these cells, small interfering RNA-mediated NUR77 knockdown reduces cAMP-induced StAR expression. Chromatin immunoprecipitation assays revealed a cAMP-dependent increase in NUR77 recruitment to the proximal StAR promoter, whereas transient transfections in MA-10 Leydig cells confirmed that NUR77 can activate the StAR promoter and that this requires an element located at -95 bp. cAMP-induced StAR and NUR77 expression in Leydig cells was found to require a Ca2+/calmodulin-dependent protein kinase (CaMK)-dependent signaling pathway. Consistent with this, we show that within the testis, CaMKI is specifically expressed in Leydig cells. Finally, we report that CaMKI transcriptionally cooperates with NUR77, but not steroidogenic factor 1, to further enhance StAR promoter activity in Leydig cells. All together, our results implicate NUR77 as a mediator of cAMP action on StAR transcription in steroidogenic Leydig cells and identify a role for CaMKI in this process.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinase Type 1/genetics , Calcium-Calmodulin-Dependent Protein Kinase Type 1/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Leydig Cells/metabolism , Phosphoproteins/genetics , Receptors, Steroid/genetics , Receptors, Steroid/metabolism , Animals , Base Sequence , Cell Line , Cyclic AMP/metabolism , Cyclic AMP/pharmacology , Cyclic AMP-Dependent Protein Kinases/metabolism , DNA/genetics , Humans , Leydig Cells/drug effects , Male , Mice , Nuclear Receptor Subfamily 4, Group A, Member 1 , Promoter Regions, Genetic/drug effects , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Small Interfering/genetics , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Signal Transduction , Transcription, Genetic , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL