Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
2.
Mol Cell Endocrinol ; 411: 49-57, 2015 Aug 15.
Article in English | MEDLINE | ID: mdl-25917455

ABSTRACT

A disintegrin and metalloprotease-17 (ADAM17) is thought to play a key role in the release of soluble and active epiregulin (EREG) and amphiregulin (AREG) in ovarian follicles but its transcriptional regulation in follicular cells remains largely unknown. The objectives of this study were to characterize the regulation of ADAM17 transcripts in bovine follicles prior to ovulation and to investigate its transcriptional control in bovine granulosa cells. To study the regulation of ADAM17 transcripts, RT-PCR analyses were performed using total RNA extracted from bovine follicles collected between 0 h and 24 h post-hCG. Results showed that levels of ADAM17 mRNA were low prior to hCG (0 h), markedly and transiently increased 6-12 h post-hCG (P <0.05), and returned to low baseline levels at 24 h post-hCG in granulosa and theca interna cells of preovulatory follicles. To determine the transcriptional control of ADAM17 expression, primary cultures of bovine granulosa cells were used. Forskolin (FSK) stimulation induced a pattern of ADAM17 mRNA up-regulation in vitro similar to that observed by hCG in vivo. 5'-Deletion mutagenesis studies identified a minimal region of the bovine ADAM17 promoter containing basal and FSK-inducible activities, which were dependent on the presence of a consensus AP1 cis-element. Electrophoretic mobility shift assays revealed an interaction between AP1 and the trans-acting factor Fra2. Chromatin immunoprecipitation assays confirmed an endogenous interaction between Fra2 and the ADAM17 promoter in granulosa cell cultures. FSK-inducible ADAM17 promoter activity and mRNA expression were suppressed by PKA and ERK1/2 inhibitors but not by a p38MAPK inhibitor, pointing to the importance of PKA and ERK1/2 signaling pathways in the up-regulation of bovine ADAM17 mRNA. Collectively, these findings describe the gonadotropin/FSK-dependent up-regulation of ADAM17 transcripts in bovine preovulatory follicles and unravel for the first time some of the molecular mechanisms involved in ADAM17 gene expression in granulosa cells of a monoovulatory species.


Subject(s)
ADAM Proteins/metabolism , Granulosa Cells/metabolism , Ovarian Follicle/metabolism , ADAM Proteins/genetics , ADAM17 Protein , Animals , Cattle , Chorionic Gonadotropin/pharmacology , Colforsin/pharmacology , Female , Gene Expression Regulation/drug effects , Granulosa Cells/drug effects , Ovarian Follicle/drug effects , Ovulation/genetics , Promoter Regions, Genetic/drug effects , Up-Regulation/drug effects
3.
Biol Reprod ; 91(6): 139, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25339105

ABSTRACT

The luteinizing hormone preovulatory surge stimulates several signal pathways essential for ovulation, and the regulator of G-protein signaling protein-2 (RGS2) is thought to be involved in this process. The objectives of this study were to characterize the regulation of RGS2 transcripts in equine and bovine follicles prior to ovulation and to determine its transcriptional control in bovine granulosa cells. To assess the regulation of equine RGS2 prior to ovulation, RT-PCR was performed using total RNA extracted from equine follicles collected at various times after human chorionic gonadotropin (hCG) injection. Results showed that RGS2 mRNA levels were very low at 0 h but markedly increased 12-39 h post-hCG (P < 0.05). In the bovine species, results revealed that RGS2 mRNA levels were low in small and dominant follicles and in ovulatory follicles obtained at 0 h, but markedly increased in ovulatory follicles 6-24 h post-hCG (P < 0.05). To study the molecular control of RGS2 expression, primary cultures of bovine granulosa cells were used. Stimulation with forskolin induced an up-regulation of RGS2 mRNA in vitro. Studies using 5'-deletion mutants identified a minimal region containing full-length basal and forskolin-inducible RGS2 promoter activities. Site-directed mutagenesis indicated that these activities were dependent on CRE and ETS1 cis-elements. Electrophoretic mobility shift assays confirmed the involvement of these elements and revealed their interactions with CREB1 and ETS1 proteins. Chromatin immunoprecipitation assays confirmed endogenous interactions of these proteins with the RGS2 promoter in granulosa cells. Forskolin-inducible RGS2 promoter activity and mRNA expression were markedly decreased by PKA and ERK1/2 inhibitors, and treatment with an antagonist of PGR (RU486) and inhibitors of PTGS2 (NS398) and EGFR (PD153035) blocked the forskolin-dependent RGS2 transcript expression, suggesting the importance of RGS2 in ovulation. Collectively, this study reports for the first time the gonadotropin-dependent up-regulation of RGS2 in equine and bovine preovulatory follicles and presents some of the regulatory controls involved in RGS2 gene expression in granulosa cells.


Subject(s)
Cattle/genetics , Granulosa Cells/metabolism , Horses/genetics , Ovarian Follicle/metabolism , Ovulation/genetics , RGS Proteins/genetics , Animals , Cattle/physiology , Cells, Cultured , Female , Follicular Phase/drug effects , Follicular Phase/genetics , Follicular Phase/metabolism , Gene Expression Regulation/drug effects , Gonadotropins/pharmacology , Granulosa Cells/drug effects , Horses/physiology , Ovarian Follicle/drug effects , Ovulation/drug effects , Ovulation/metabolism , RGS Proteins/metabolism , Transcriptional Activation/drug effects
4.
Mol Cell Endocrinol ; 391(1-2): 10-21, 2014 Jun 25.
Article in English | MEDLINE | ID: mdl-24784705

ABSTRACT

The interaction between stromal cell-derived factor-1 (SDF1) and chemokine CXC motif receptor 4 (CXCR4) has been implicated in leukocyte attraction, tissue remodeling and angiogenesis. The objective of the present study was to characterize the expression and regulation of SDF1 and CXCR4 in equine follicles during the ovulatory process. Equine preovulatory follicles were isolated during estrus 0-39h after hCG treatment. Follicle wall preparations (theca interna with attached granulosa cells) and isolated preparations of granulosa cells and theca interna were obtained, and total RNA extracts were analyzed by RT-PCR/Southern blot. Results showed that levels of CXCR4 transcripts were induced by hCG in follicles at 36 h post-hCG (P<0.05 vs 0 h), with the induction observed in both granulosa and theca cells. Immunoblotting and immunohistochemical analyses confirmed an increase in CXCR4 protein in follicles after hCG treatment. In contrast, levels of SDF1 transcripts were very low in granulosa cells but high in theca interna cells throughout most of the ovulatory period. Studies in vivo performed with bovine preovulatory follicles collected 0-24h post-hCG revealed a marked and significant up-regulation of CXCR4 transcripts after hCG (P<0.05), as observed in equine follicles. A similar pattern of CXCR4 mRNA up-regulation was observed in cultures of bovine granulosa cells treated with forskolin (P<0.05). This forskolin-dependent induction of CXCR4 mRNA was suppressed by co-treatment with inhibitors of PKA, ERK1/2 and EGFR, and by the progesterone receptor antagonist RU486 (P<0.05), underscoring the contribution of multiple signaling pathways. In complementary studies, treatment of bovine granulosa cells with EGF or the hypoxia mimetic cobalt chloride significantly increased CXCR4 transcript levels, whereas co-treatment with forskolin and a CXCR4 antagonist repressed the expression of several ovulation-related genes. Collectively, this study describes for the first time the gonadotropin-dependent up-regulation of CXCR4 transcript in ovarian follicles of large monoovulatory species, provides some insights into the regulation of CXCR4 gene expression in granulosa cells, and identifies a potential link between follicular SDF1/CXCR4 activation and the regulation of ovulation-related genes.


Subject(s)
Chemokine CXCL12/genetics , Granulosa Cells/metabolism , Ovulation/physiology , RNA, Messenger/genetics , Receptors, CXCR4/genetics , Theca Cells/metabolism , Amino Acid Sequence , Animals , Cattle , Cells, Cultured , Chemokine CXCL12/metabolism , Chorionic Gonadotropin/administration & dosage , Cobalt/pharmacology , Colforsin/pharmacology , Epidermal Growth Factor/pharmacology , Estrus/physiology , Female , Gene Expression Regulation , Granulosa Cells/cytology , Granulosa Cells/drug effects , Horses , Humans , Mifepristone/pharmacology , Molecular Sequence Data , Protein Kinase Inhibitors/pharmacology , RNA, Messenger/agonists , RNA, Messenger/antagonists & inhibitors , RNA, Messenger/metabolism , Receptors, CXCR4/agonists , Receptors, CXCR4/antagonists & inhibitors , Receptors, CXCR4/metabolism , Signal Transduction , Theca Cells/cytology , Theca Cells/drug effects
5.
Biol Reprod ; 89(4): 98, 2013 Oct.
Article in English | MEDLINE | ID: mdl-24006283

ABSTRACT

Vanin-2 (VNN2) is known to be involved in inflammation and leukocyte migration, but its regulation in follicles remains unknown. The objectives of this work were to study the regulation of VNN2 transcripts in bovine follicles prior to ovulation and to characterize the control of its expression in bovine granulosa cells. VNN2 expression was studied using total RNA extracted from granulosa cells of small follicles (2-4 mm in diameter), dominant follicles obtained on Day 5 of the estrous cycle, ovulatory follicles obtained 0-24 h after human chorionic gonadotropin (hCG), and corpora lutea on Day 5 of the cycle. The results from RT-PCR analyses showed that levels of VNN2 mRNA were high in ovulatory follicles 24 h post-hCG but low in the other tissues. In ovulatory follicles, levels of VNN2 mRNA were low at 0 h but significantly up-regulated 12-24 h post-hCG. To determine factors controlling VNN2 gene expression, established primary cultures of granulosa cells isolated from bovine dominant follicles were used. Treatment with forskolin elevated VNN2 mRNA expression as observed in vivo. Mutation studies identified the minimal region conferring basal and forskolin-stimulated VNN2 promoter activities, which were dependent on chicken ovalbumin upstream promoter-transcription factor (COUP-TF), GATA, and Ebox cis-elements. Electrophoretic mobility shift assays identified COUP-TF, GATA4, and upstream stimulating factor proteins as key factors interacting with these elements. Chromatin immunoprecipitation assays confirmed basal and forskolin-induced interactions between these proteins and the VNN2 promoter in bovine granulosa cell cultures. VNN2 promoter activity and mRNA expression were markedly stimulated by forskolin and overexpression of the catalytic subunit of PKA, but inhibited by PKA and ERK1/2 inhibitors. Collectively, the findings from this study describe for the first time the gonadotropin/forskolin-dependent up-regulation of VNN2 transcripts in granulosa cells of preovulatory follicles and provide insights into some of the molecular bases of VNN2 gene expression in follicular cells.


Subject(s)
Amidohydrolases/metabolism , Cell Adhesion Molecules/metabolism , Ovarian Follicle/metabolism , Proestrus , Promoter Regions, Genetic , Transcription, Genetic , Up-Regulation , Amidohydrolases/biosynthesis , Amidohydrolases/genetics , Animals , Cattle , Cell Adhesion Molecules/biosynthesis , Cell Adhesion Molecules/genetics , Cells, Cultured , Chorionic Gonadotropin/pharmacology , Colforsin/pharmacology , Corpus Luteum/cytology , Corpus Luteum/drug effects , Corpus Luteum/metabolism , Enzyme Activators/pharmacology , Female , Fertility Agents, Female/pharmacology , GPI-Linked Proteins/biosynthesis , GPI-Linked Proteins/genetics , GPI-Linked Proteins/metabolism , Granulosa Cells/cytology , Granulosa Cells/drug effects , Granulosa Cells/metabolism , Humans , MAP Kinase Signaling System/drug effects , Mutation , Ovarian Follicle/cytology , Ovarian Follicle/drug effects , Proestrus/drug effects , Promoter Regions, Genetic/drug effects , Recombinant Proteins/biosynthesis , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Response Elements/drug effects , Transcription, Genetic/drug effects , Up-Regulation/drug effects
6.
Endocrinology ; 154(8): 2857-69, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23751874

ABSTRACT

The ovulatory process involves a complex remodeling of the extracellular matrix during which a desintegrin and metalloproteinase with thrombospondin motif 1 (ADAMTS1) is thought to play a key role, but its transcriptional regulation in bovine follicles remains largely unknown. The objectives of this study were to characterize the regulation of ADAMTS1 in bovine follicles before ovulation and to determine its transcriptional control in bovine granulosa cells. Regulation of ADAMTS1 was assessed using total RNA isolated from bovine preovulatory follicles obtained at various times after human chorionic gonadotropin treatment. Results from RT-PCR analyses showed that levels of ADAMTS1 mRNA were very low at 0 hours but increased at 6 to 24 hours after human chorionic gonadotropin in granulosa cells. To determine the regulatory mechanisms controlling ADAMTS1 gene expression in vitro, primary cultures of bovine granulosa cells were established, and treatment with forskolin up-regulated ADAMTS1 mRNA levels. Promoter activity assays, 5'-deletion, and site-directed mutagenesis identified a minimal region conferring full-length basal and forskolin-stimulated ADAMTS1 promoter activities, with both being dependent on Ebox cis-acting elements. EMSAs revealed upstream stimulating factor (USF) proteins as key trans-activating factors interacting with Ebox. Chromatin immunoprecipitation assays confirmed such interactions between USF and Ebox in vivo, and USF binding to Ebox elements was increased by forskolin treatment. ADAMTS1 promoter activity and mRNA expression were increased by forskolin and overexpression of the catalytic subunit of protein kinase A, but not by cotreatment with inhibitors of protein kinase A, ERK1/2, and epidermal growth factor receptor signaling pathways. Furthermore, treatment with a soluble epidermal growth factor induced ADAMTS1 mRNA expression in granulosa cells. Collectively, results from this study describe the gonadotropin/forskolin-dependent up-regulation of ADAMTS1 mRNA in granulosa cells of bovine preovulatory follicles in vivo and in vitro and identify for the first time some of the molecular mechanisms responsible for ADAMTS1 promoter activation in follicular cells of a large monoovulatory species.


Subject(s)
ADAM Proteins/genetics , Gene Expression Regulation , Granulosa Cells/metabolism , Ovarian Follicle/metabolism , Promoter Regions, Genetic/genetics , ADAM Proteins/metabolism , Animals , Binding Sites/genetics , Cattle , Cells, Cultured , Chorionic Gonadotropin/pharmacology , Colforsin/pharmacology , Female , Humans , Luciferases/genetics , Luciferases/metabolism , Mutation , Ovulation/genetics , Protein Binding , Reverse Transcriptase Polymerase Chain Reaction , Time Factors , Transcription, Genetic/drug effects , Up-Regulation/drug effects , Upstream Stimulatory Factors/genetics , Upstream Stimulatory Factors/metabolism
7.
Gen Comp Endocrinol ; 180: 39-47, 2013 Jan 01.
Article in English | MEDLINE | ID: mdl-23178756

ABSTRACT

Little is known about the expression and regulation of epiregulin (EREG) and amphiregulin (AREG) in ovarian follicles of large monoovulatory animal species. To characterize the gonadotropin-dependent regulation of EREG and AREG mRNAs in equine follicles prior to ovulation, extracts were prepared from equine follicles collected during estrus between 0 and 39h post-hCG and corpora lutea obtained on day 8 of the estrous cycle (day 0=day of ovulation). Results from RT-PCR/Southern blot analyses showed that levels of EREG and AREG mRNAs were very low in follicles obtained at 0h but increased thereafter (P<0.05), with maximal levels observed 33-39h post-hCG. This significant increase was observed in both granulosa and theca cells. Immunohistochemistry and immunoblot analyses confirmed the hCG-dependent induction of EREG protein in both cell types. RT-PCR/Southern blot analyses of ADAM17, which encodes an enzyme that cleaves and releases soluble bioactive EREG and AREG, showed that levels of its transcript were high and remained constant throughout the period studied. Studies on the hCG-dependent regulation of EREG and AREG in bovine preovulatory follicles in vivo showed that the induction of both transcripts was transient, observed predominantly at 6h post-hCG and localized only in granulosa cells. To characterize the effect of epidermal growth factor receptor (EGFR) activation on the expression of ovulation-related genes in granulosa cells of a large monoovulatory animal species, primary cultures of bovine granulosa cells were established. Results from RT-PCR analyses revealed that EREG and AREG mRNAs were induced by forskolin treatment in vitro; but the EGFR inhibitor PD153035 suppressed the forskolin-dependent induction of several ovulation-related transcripts, including PTGS2, PTGER2, TNFAIP6, PGR, MMP1, VEGFA, and CTSL2 mRNAs. Moreover, these transcripts were induced in granulosa cell cultures by EGF, an analog of EREG and AREG. Collectively, this study identifies differences in the temporal and cellular localization of EREG and AREG expression in equine and bovine preovulatory follicles, and underscores the potential role of follicular EGFR activation in the regulation of ovulation-regulated genes in large monoovulatory species.


Subject(s)
Chorionic Gonadotropin/pharmacology , Epidermal Growth Factor/metabolism , Glycoproteins/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Ovarian Follicle/drug effects , Ovarian Follicle/metabolism , Amphiregulin , Animals , Cattle , Cells, Cultured , EGF Family of Proteins , Epiregulin , Female , Horses , Humans , Ovulation/drug effects , Ovulation/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...