Introduction
Ovulation is a complex endocrine and molecular process, leading to the release of the oocyte from the dominant follicle. The process begins with a pre-ovulatory LH and follicle FSH surge that stimulates the expression of a number of genes that trigger various physiological, events such as cumulus expansion, granulosa cell luteinization and follicular rupture (
Espey, Richards, 2002Temporal and spatial patterns of ovarian gene transcription following an ovulatory dose of gonadotrophin in the rat.
,
Richards et al, 2002- Richards J.S.
- Russell D.L.
- Ochsner S.
- Hsieh M.
- Doyle K.H.
- Falender A.E.
- Lo Y.K.
- Sharma S.C.
Novel signaling pathways that control ovarian follicular development, ovulation and luteinization.
,
Stouffer, 2003Progesterone as a mediator of gonadotrophin action in the corpus luteum: beyond steroidogenesis.
). The binding of progesterone to the progesterone receptor is an essential factor in ovulation. This finding is supported by numerous studies in rodents and a primate showing that administration of anti-progestins, such as mifepristone, inhibits ovulation (
Loutradis et al, 1991- Loutradis D.
- Bletsa R.
- Aravantinos L.
- Kallianidis K.
- Michalas S.
- Psychoyos A.
Preovulatory effects of the progesterone antagonist mifepristone (RU486) in mice.
,
Richards, 2005Ovulation: new factors that prepare the oocyte for fertilization.
). Treatment of progesterone-receptor-knockout mice with exogenous gonadotropins results in normal follicular development, but follicular rupture does not occur despite the luteinization of the granulosa cells. This finding indicates that progesterone receptor is essential for LH-dependent follicular rupture, but is not involved in granulosa and luteal cell differentiation for progesterone biosynthesis (
Lydon et al, 1995- Lydon J.P.
- DeMayo F.J.
- Funk C.R.
- Mani S.K.
- Hughes A.R.
- Montgomery C.A.
- Shyamala G.
- Conneely O.M.
- O'Malley B.W.
Mice lacking progesterone receptor exhibit pleiotropic reproductive abnormalities.
,
Robker et al, 2009- Robker R.L.
- Akison L.K.
- Russell D.L.
Control of oocyte release by progesterone receptor-regulated gene expression.
). Progesterone receptor mediates the expression of several matrix metalloproteinases (MMPs), including metalloproteinases 3 and 10 (MMP3/10) and members of the desintegrin and metalloproteinase with thrombospondin motifs family (ADAMTS-1), which remodel the extracellular matrix of the follicle wall, ultimately resulting in rupture. All of these MMPs are present in the ovarian granulosa cells of rodents, sheep and humans after the LH surge (
Espey et al, 2000- Espey L.L.
- Yoshioka S.
- Russell D.L.
- Robker R.L.
- Fujii S.
- Richards J.S.
Ovarian expression of a disintegrin and metalloproteinase with thrombospondin motifs during ovulation in the gonadotropin-primed immature rat.
,
McCord et al, 2012- McCord L.A.
- Li F.
- Rosewell K.L.
- Brännström M.
- Curry T.E.
Ovarian expression and regulation of the stromelysins during the periovulatory period in the human and the rat.
,
Robker et al, 2000- Robker R.L.
- Russell D.L.
- Espey L.L.
- Lydon J.P.
- O'Malley B.W.
- Richards J.S.
Progesterone regulated genes in the ovulation process: ADAMTS-1 and cathepsin L proteases.
,
Yung et al, 2010- Yung Y.
- Maman E.
- Konopnicki S.
- Cohen B.
- Brengauz M.
- Lojkin I.
- Dal Canto M.
- Fadini R.
- Dor J.
- Hourvitz A.
ADAMTS-1: a new human ovulatory gene and a cumulus marker for fertilization capacity.
). In mice, hypoxia-inducible factor 1-alpha (HIF1A) mRNA and protein expression are also induced through the progesterone receptor. Inhibiting HIF1A transcriptional activity with echinomycin suppresses ovulation by preventing follicular rupture, in part via effects on ADAMTS-1 expression (
Kim et al, 2009- Kim J.
- Bagchi I.C.
- Bagchi M.K.
Signaling by hypoxia-inducible factors is critical for ovulation in mice.
).
Previous reports from our laboratory provided evidence of transient progesterone receptor expression in human granulosa cells during the peri-ovulatory period, disappearing after the LH surge (
García et al, 2012- García V.
- Kohen P.
- Maldonado C.
- Sierralta W.
- Muñoz A.
- Villarroel C.
- Strauss 3rd., J.F.
- Devoto L.
Transient expression of progesterone receptor and cathepsin-l in human granulosa cells during the periovulatory period.
). In addition, in-vitro studies on human granulosa cells showed that progesterone receptor mRNA expression significantly increases 6 h after HCG stimulation. This increase is blocked by a PKA inhibitor (
García et al, 2012- García V.
- Kohen P.
- Maldonado C.
- Sierralta W.
- Muñoz A.
- Villarroel C.
- Strauss 3rd., J.F.
- Devoto L.
Transient expression of progesterone receptor and cathepsin-l in human granulosa cells during the periovulatory period.
). Interestingly, in mice, LH can increase progesterone receptor expression through other signalling pathways, such as PI3K, PKC and ERK1/2 (
Robker et al, 2009- Robker R.L.
- Akison L.K.
- Russell D.L.
Control of oocyte release by progesterone receptor-regulated gene expression.
). These findings have not been confirmed in human granulosa cells.
Few data are available on the various gonadotrophin-activated (LH, HCG and FSH) signalling pathways involved in triggering the rupture of the dominant follicle. On the basis of our previous in-vivo findings of transient progesterone receptor expression in pre-ovulatory granulosa cells, the present study was carried out in order to determine progesterone receptor expression and localization in primary granuloa cell cultures treated with HCG, LH and FSH; (2) evaluate the roles of the various HCG-LH-FSH-activated signalling pathways (PKA, PKC, ERK1/2, AKT, or both) in progesterone receptor expression; (3) determine ADAMTS-1 and MMP3/10 expression in granuluosa cells in the pre-ovulatory follicle after HCG administration in IVF cycles; and (4) to assess the gonadotrophin-progesterone-receptor-dependent role of HIF1A in the expression of ADAMTS-1 and MMP3/10 in granulosa-cell cultures.
Materials and methods
Participants
Follicular aspirates from 22 healthy women (25–32 years old) undergoing ovarian stimulation for IVF for treatment of male factor infertility, were aspirated using a KITAZATO G -21 needle 36 h after HCG administration (10,000 IU). The aspiration used a pump setting of 150 mm Hg. This procedure setting removes granulosa cells with minimal aspiration of theca and endothelial cells. The granulosa cells were obtained after centrifugation of follicular fluid at 400 x g for 5 min. The pellet was suspended, and red blood cells and detritus were removed using a Histopaque (Sigma-Aldrich) gradient. Macrophages were eliminated by pre-plating for 30 min at 37°C, and cells were cultured as previously described (
Olivero et al, 2008- Olivero P.
- Leiva-Salcedo E.
- Devoto L.
- Stutzin A.
Activation of Cl- channels by human chorionic gonadotrophin in luteinized granulosa cells of the human ovary modulates progesterone biosynthesis.
). The purity of the granulosa cell cultures was assessed by immunodetection of the steroidogenic acute regulatory protein. This protein is expressed in granulosa cell after the LH peak in normal cycles or after HCG administration in ovarian stimulation cycles (
Henríquez et al, 2016- Henríquez S.
- Kohen P.
- Xu X.
- Veenstra T.D.
- Muñoz A.
- Palomino W.A.
- Strauss 3rd., J.F.
- Devoto L.
Estrogen metabolites in human corpus luteum physiology: differential effects on angiogenic activity.
). A portion of the granulosa cells obtained 36 h after HCG administration was used immediately to determine MMPs expression by Western blotting.
The Institutional Review Board of School of Medicine, University of Chile, approved the study on 8 September 2015, and signed informed consent was obtained from all women's participating in the study.
Granulosa-lutein cell cultures
Granulosa cells were cultured for 72 h in growth medium (M199) supplemented with 10% fetal bovine serum (FBS) followed by 24 h in serum-free medium (M199, Sigma-Aldrich, Missouri, USA). Progesterone receptor was detected in granulosa cells using temporal course of 6, 8, 12, 18 and 24 h under basal conditions and in the presence and absence of HCG (10 IU/ml, Sigma-Aldrich, Missouri, USA), LH (10 IU/ml, Luveris, Merck-Serono, Darmstadt, Germany), FSH (10 IU/ml, GONAL-f, Merck-Serono), H-89 (10 µM, Calbiochem, Merck millipore, Darmstadt, Germany), AKT inhibitor (LY 294002) (20 µM, Calbiochem, Merck millipore, Darmstadt, Germany), UO126 (10 µM, Calbiochem), bisindolylmaleimide I (BIM) (2,5 µM, Calbiochem, Merck millipore, Darmstadt, Germany). The concentrations of gonadotrophins and inhibitors were selected for maximal stimulation (gonadotrophins) or minimum inhibitory concentrations (inhibitors) based on dose-response curves from our previous studies and published data (
García et al, 2012- García V.
- Kohen P.
- Maldonado C.
- Sierralta W.
- Muñoz A.
- Villarroel C.
- Strauss 3rd., J.F.
- Devoto L.
Transient expression of progesterone receptor and cathepsin-l in human granulosa cells during the periovulatory period.
,
Kong et al, 2005- Kong D.
- Park E.J.
- Stephen A.G.
- Calvani M.
- Cardellina J.H.
- Monks A.
- Fisher R.J.
- Shoemaker R.H.
- Melillo G.
Echinomycin, a small-molecule inhibitor of hypoxia-inducible factor-1 DNA-binding activity.
). Under these culture conditions, the granulosa cells do not express progesterone receptor (basal state), but they do recover the ability to express progesterone receptor and MMPs after gonadotrophin stimulus.
Protein extracts from granulosa cells obtained immediately after follicular aspiration after HCG administration were used to determine MMP expression ex vivo.
Gonadotrophin-dependent signalling pathways in granulosa cells were determined immediately after 24 h FBS deprivation. The granulosa cells were cultured for 5, 15 and 30 min under basal conditions and in the presence of HCG (10 IU/ml), LH (10 IU/ml) or FSH (10 IU/ml) to determine the levels of ERK1/2, AKT and CREB (protein phosphorylated by PKA). MARCKS (a protein phosphorylated by PKC) levels were determined only in the presence of HCG (10 IU/ml) and FSH (10 IU/ml). ADAMTS-1 and MMP3/10 levels were quantified in extracts of granulosa cells cultured for 24 h under basal conditions and in the presence or absence of HCG (10 IU/ml), LH (10 IU/ml) and FSH (10 IU/ml), RU 486 (100 nM, Sigma-Aldrich, Missouri, USA) or echinomycin (0.5 uM, Sigma-Aldrich, Missouri, USA).
The cellular viability was evaluated at the end of the culture with MTS assay, a colorimetric method for determining the number of cell viable, described by the manufacturer (The CellTiter 96® AQueous One Solution Cell Proliferation Assay, Promega Corp, Wisconsin, USA).
Immunofluorescence
Progesteron receptor was detected by immunofluorescence in granulosa cells (50,000 cells per cover slip) cultured in the conditions described above. The coverslips were methanol-fixed and blocked with 3% bovine serum albumin in phosphate-buffered saline for 1 h at room temperature, followed by overnight incubation at 4°C with a primary monoclonal antibody capable of recognizing both progesterone receptor isoforms (progesterone receptor A/B antibody, Cell Signaling Technology, Massachusetts, USA, diluted 1:100). Subsequently, cells were incubated with anti-mouse secondary antibody conjugated with FITC (Sigma-Aldrich, Missouri, USA) for 1 h at 37°C, diluted 1:1000. The cellular localization of HIF1A in granulosa cells was carried out by time courses at 6, 12, 18 and 24 h under basal conditions, and in the presence and absence of HCG (10 IU/ml) and mifepristone (100 nM RU486). HIF1A was detected by overnight incubation at 4°C with anti-HIF 1A, rabbit polyclonal antibody (Santa Cruz Biotechnology, Texas, USA, diluted 1:100) and incubated with anti-rabbit secondary antibody conjugated with Alexa Fluor 488 (Molecular Probes, Inc., Oregon, USA) for 1 h at 37°C, diluted 1:1000. The primary antibody was omitted in the negative control. The cells were embedded and mounted for microscopy with ProLong Gold mounting medium with DAPI, a DNA-intercalating agent 4′,6-diamidino-2-phenylindol dihydrochloride, for detection of nuclear DNA (Molecular Probes, Inc., Oregon, USA). Immunofluorescence was documented with an epifluorescence microscope (Olympus BX-51TF; Olympus Optical Co. Ltd, Tokyo, Japan) and CoolSNAP-Pro camera (Media Cybernetics, Maryland, USA).
Western blotting
The levels of ERK1/2, AKT, CREB (proteins phosphorylated by PKA) and MARCKS (a protein phosphorylated by protein kinase C) were measured using Western blotting in homogenates of granulosa cells. Gel electrophoresis was carried out in 10% SDS-PAGE. Total protein (30 µg), determined by the dye-binding assay (Bio-Rad Lab. Inc., California, USA), was loaded in each lane and were transferred onto nitrocellulose membranes (Thermo Fisher Scientific, Massachusetts, USA) as previously reported (
Kohen et al, 2013- Kohen P.
- Henríquez S.
- Rojas C.
- Gerk P.M.
- Palomino W.A.
- Strauss 3rd., J.F.
- Devoto L.
2-Methoxyestradiol in the human corpus luteum throughout the luteal phase and its influence on lutein cell steroidogenesis and angiogenic activity.
). After the transfer, blots were blocked in 3% bovine serum albumn in T-TBS (20 mM Tris, 500 mM NaCl and 0.01% Tween 20) for 1 h and incubated overnight at 4°C with primary antibody: (1) anti-phospho or total ERK1/2 (Mouse monoclonal antibody, Santa Cruz Biotechnology, Texas, USA, diluted 1:3000); (2) anti-phospho or total AKT (Rabbit polyclonal antibody, Cell Signaling Technology, Massachusetts, USA, diluted 1:1000); (3) anti-phospho or total CREB (Rabbit polyclonal antibody, Cell Signaling Technology, diluted 1:500); (4) anti-phosphor or total MARCKS (Rabbit polyclonal antibody, Cell Signaling Technology, Massachusetts, USA, diluted 1:1000); (5) anti-ADAMTS-1 (H-60) rabbit polyclonal antibody for the mature 85 kDa isoform (Santa Cruz Biotechnology, Inc, Texas, USA, diluted 1:500); (6) anti-MMP3/10 (F-10) mouse monoclonal antibody (Santa Cruz Biotechnology, Inc, Texas, USA, diluted 1:300); or (7) anti-β-actin (Mouse monoclonal antibody, Sigma-Aldrich, Missouri, USA, diluted 1:10000). Horseradish peroxidase-conjugated secondary antibody: Peroxidase-conjugated affiniPure Goat Anti-Rabbit IgG (Jackson Immuno Research, Laboratories, Inc., Pennsylvania, USA, diluted 1:5000) and Peroxidase-conjugated affiniPure Goat Anti-Mouse IgG (Jackson Immuno Research, Laboratories, Inc. diluted 1:5000) were incubated for 1 h at 37 C and detected by chemiluminescence (Western Lightning Plus-ECL, Perking Elmer, Inc., Pennsylvania, USA) using an Ultra Quant 6.2 Image Reader and analyzed by Image Quant 5.2 software (Molecular Dynamic Inc., Sunnyvale, CA, USA). Densitometric analyses of immunoreactive bands were normalized to beta-actin or by total protein when referring to the phosphorylated protein.
Statistical analysis
The statistical analysis of the densitometric data from Western blotting (n = 4) was carried out using a non-parametric method: Kruskal–Wallis (one-way analysis of variance) followed by a Dunn's Multiple Comparison post-hoc test with GraphPad Prism software (GraphPad Software Inc., San Diego, CA, USA). Previously, the data were checked for normality with the Shapiro–Wilk test, P < 0.05, indicating that our data do not have normal distribution. Significance was defined as P < 0.05. Data are presented as means ± SEM.
Discussion
Ovulation is an orchestrated process largely synchronized by a mid-cycle surge of pituitary gonadotropins (LH and FSH). Physiologically, the LH and FSH surges occur simultaneously (
Richards et al, 2002- Richards J.S.
- Russell D.L.
- Ochsner S.
- Hsieh M.
- Doyle K.H.
- Falender A.E.
- Lo Y.K.
- Sharma S.C.
Novel signaling pathways that control ovarian follicular development, ovulation and luteinization.
). These gonadotrophin surges initiate a series of biochemical events in the pre-ovulatory follicle, resulting in expansion of the cumulus oocyte complex, rupture of the dominant follicle and ultimately expulsion of the oocyte. The gonadotrophin cascade initiates expression of a number of specific transcription factors, steroids receptors, cytokines and metalloproteinases and induces progesterone synthesis. During induced ovulatory cycles, HCG administration is used as a substitute for the LH surge, resulting in follicular rupture and stimulating the final stages of ovum maturation (
Espey, Richards, 2002Temporal and spatial patterns of ovarian gene transcription following an ovulatory dose of gonadotrophin in the rat.
,
Zafeiriou et al, 2000- Zafeiriou S.
- Loutradis D.
- Michalas S.
The role of gonadotropins in follicular development and their use in ovulation induction protocols for assisted reproduction.
). It has been postulated that proteinases digest the connective tissue matrix to allow for follicular rupture. The present study was designed to examine the various LH-, FSH-, and HCG-driven signalling pathways involved in progesterone receptor and MMP expression in primary cultures of human granulosa cells.
A previous study from our laboratory found that progesterone receptor mRNA was transiently up-regulated in human granulosa cell culture 6 h after HCG administration. This stimulation was achieved through the LH and HCG, cAMP, and PKA pathways (
García et al, 2012- García V.
- Kohen P.
- Maldonado C.
- Sierralta W.
- Muñoz A.
- Villarroel C.
- Strauss 3rd., J.F.
- Devoto L.
Transient expression of progesterone receptor and cathepsin-l in human granulosa cells during the periovulatory period.
). The present findings indicate that progesterone receptor expression in human granulosa cell culture becomes detectable with immunofluorescence after 12 h of treatment with LH, HCG and FSH. This stimulation occurs through the PKA, PKC and ERK1/2-signalling pathways. These results confirm the report by
Robker et al, 2009- Robker R.L.
- Akison L.K.
- Russell D.L.
Control of oocyte release by progesterone receptor-regulated gene expression.
that, in the mouse and the rat, LH and HCG activation of the PKA, PKC or MAPK signalling pathways is required to stimulate progesterone receptor mRNA expression in granulosa cells. It is important to note that our experiments were conducted on granulosa cells obtained 36 h after HCG administration, when progesterone receptors are not expressed. Interestingly, these cells become sensitive to gonadotrophins after 72 h of culture followed by 24 h in serum-free medium, expressing progesterone receptor after 12 h gonadotrophin stimulation.
During the pre-ovulatory period, remodelling of the follicle wall extracellular matrix facilitates rupture and expulsion of the oocyte. These biological events depend on the induction and activation of various transcription factors and MMPs. The present study establishes that HCG operates through progesterone receptor to regulate HIF1A transcription factor expression along with ADAMTS-1 and MMP3/10 expression in human granulosa cell cultures. The antiprogestin RU486 abolished this regulation. These findings corroborate in-vivo studies showing that HIF1A is critical for follicular rupture in mice (
Kim et al, 2009- Kim J.
- Bagchi I.C.
- Bagchi M.K.
Signaling by hypoxia-inducible factors is critical for ovulation in mice.
). Furthermore, these results are consistent with findings of hypoxic stress within the periovulatory follicle in mice, resulting in high levels of HIF1A associated with follicular growth and rupture (
Basini et al, 2004- Basini G.
- Bianco F.
- Grasselli F.
- Tirelli M.
- Bussolati S.
- Tamanini C.
The effects of reduced oxygen tension on swine granulosa cell.
,
Zhang et al, 2012- Zhang J.
- Zhang Z.
- Wu Y.
- Chen L.
- Luo Q.
- Chen J.
- Huang X.
- Cheng Y.
- Wang Z.
Regulatory effect of hypoxia-inducible factor-1α on hCG-stimulated endothelin-2 expression in granulosa cells from the PMSG-treated rat ovary.
). Recent studies in human fibroblast culture indicate that MMP3 expression is exclusively regulated by HIF1A in response to inflammation that suggests that metalloproteinases expression is involved in follicular rupture (
Ahn et al, 2008- Ahn J.K.
- Koh E.M.
- Cha H.S.
- Lee Y.S.
- Kim J.
- Bae E.K.
- Ahn K.S.
Role of hypoxia-inducible factor-1alpha in hypoxia-induced expressions of IL-8, MMP-1 and MMP-3 in rheumatoid fibroblast-like synoviocytes.
). It is well known that inflammation is a key component of the ovulation process (
,
Espey et al, 1994- Espey L.L.
- Lipner H.
- Knobil E.
- Neill J.D.
Ovulation. The Physiology of Reproduction.
).
In human granulosa cells, MMP3/10 expression is dependent of the progesterone receptor. It is tempting, however, to speculate that different mechanisms are involved in MMP3/10 expression, as the presence of the specific inhibitor of HIF1A, echinomycin, abolished the progesterone-receptor-dependent increase in MMP3/10. In contrast, progesterone-receptor-dependent induction of ADAMTS-1 was evidently not mediated through HIF1A. Importantly, MMP3/10 and ADAMTS-1 are expressed before follicular rupture, as they were detected in granulosa cells immediately after collection from the pre-ovulatory follicle. These results suggest the possibility of a differential and newly identified progesterone-receptor-dependent, HIF1A-mediated regulation mechanism involved in human follicular rupture.
Clinicians have recently been using GnRH agonist for triggering of ovulation (through an endogenous LH and FSH surge). This study shows that LH and FSH significantly stimulate the expression of both proteases in a progesterone receptor-dependent manner, but the combined effects of LH and FSH together are similar to the effects of LH and FSH separately.
In conclusion, HCG, LH, and FSH activate intracellular signalling pathways involving PI3K, PKC, ERK 1/2 and PKA. Only PKC, ERK 1/2 and PKA, however, stimulate progesterone receptor expression in human granulosa cells. progesterone receptor activation regulates proteases involved in the ovulatory process, such as ADAMTS-1 and MMP3/10. Furthermore, progesterone receptor expression up-regulates HIF-1A, which increases MMP3/10 levels, but not ADAMTS-1. These observations provide a better understanding of the mechanisms by which LH and proesterone receptor signalling pathways mediate the ovulation process in humans.
Article info
Publication history
Published online: June 22, 2017
Accepted:
June 2,
2017
Received in revised form:
May 31,
2017
Received:
November 24,
2016
Declaration: The authors report no financial or commercial conflicts of interest.
Copyright
© 2017 Reproductive Healthcare Ltd. Published by Elsevier Ltd. All rights reserved.