IN THE OVARY, folliculogenesis begins with the recruitment

Size: px
Start display at page:

Download "IN THE OVARY, folliculogenesis begins with the recruitment"

Transcription

1 /05/$15.00/0 Endocrinology 146(8): Printed in U.S.A. Copyright 2005 by The Endocrine Society doi: /en Estrogen Receptor- Is Critical to Granulosa Cell Differentiation and the Ovulatory Response to Gonadotropins John F. Couse, Mariana M. Yates, Bonnie J. Deroo, and Kenneth S. Korach Receptor Biology Section, Laboratory of Reproductive and Developmental Toxicology, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina The process of granulosa cell differentiation that occurs in preovulatory follicles is dependent on FSH but requires augmentation by estradiol. To determine which estrogen receptor (ER) form mediates the effects of estradiol during gonadotropin-induced follicle growth, differentiation, and rupture, we characterized the response of ER - and ER -null mice to gonadotropin-induced ovulation. Immature mice were treated with an ovulatory regimen of exogenous gonadotropins and tissues were collected at distinct time points for morphological, biochemical, gene expression, and immunohistochemical analyses. Granulosa cells of ER knockout (ERKO) preovulatory follicles exhibited an attenuated response to FSH-induced differentiation, as evident by reduced aromatase activity and estradiol synthesis, and insufficient expression of LH receptor. As a result, ERKO ovaries were unable to fully respond to an ovulatory bolus of gonadotropin, IN THE OVARY, folliculogenesis begins with the recruitment of a cohort of primordial follicles from the existing pool. Once enlisted, the selected follicles embark on an irreversible course of oocyte growth, granulosa cell proliferation, and thecal cell differentiation. The resulting preantral follicles become highly responsive to FSH from the anterior pituitary and are induced to differentiate to the preovulatory stage. Fully differentiated preovulatory follicles are characterized by an increased capacity to synthesize large amounts of estradiol via aromatization of thecal-derived androgens, formation of a large fluid-filled antrum, and the acquisition of LH-receptor and responsiveness. The synchronized assembly of mature follicles in the ovary provides for increasing levels of circulating estradiol, which ultimately peaks and acts on the hypothalamic-pituitary axis to elicit secretion of a massive bolus of LH. The acquisition of LH receptor by healthy preovulatory follicles allows them to selectively respond to the LH surge, leading to ovulation (rupture and expulsion of the oocyte) and terminal differentiation of the First Published Online April 14, 2005 Abbreviations: AR, Androgen receptor; COC, cumulus-oocyte complex; CREB, camp response element binding protein; ER, estrogen receptor; ERE, estrogen response element; ERKO, mice null for ER ; ERKO, mice null for ER ; hcg, human choriogonadotropin; PKA, protein kinase A; PMSG, pregnant mare serum gonadotropin; PR, progesterone receptor; SF, steroidogenic factor; SQ-RT-PCR, semiquantitative RT-PCR. Endocrinology is published monthly by The Endocrine Society ( the foremost professional society serving the endocrine community. leading to a reduced rate of follicle rupture; insufficient induction of prostaglandin-synthase 2 and progesterone receptor; an aberrant increase in aromatase activity and plasma estradiol; and incomplete expansion of the cumulus-oocyte complex. Parallel characterization of ERKO females indicated a minimal role for ER in granulosa cell differentiation, ovulation, and the concomitant changes in gene expression, although some abnormalities were revealed. These studies demonstrate that ER -mediated estradiol actions are vital to FSH-induced granulosa cell differentiation; and in the absence of ER, preovulatory follicles are deficient in the necessary cellular organization (i.e. antrum and cumulus oocyte complex), enzymatic activity (i.e. capacity to convert androgen precursor to estradiol), and receptor signaling pathways (i.e. LH receptor) to respond to a gonadotropin surge and expel a healthy oocyte. (Endocrinology 146: , 2005) somatic cells to form a corpus luteum (1). Therefore, follicle growth and ovulation are remarkably synchronous processes that depend on the functions of multiple ovarian cell types and a myriad of endocrine signaling pathways. This complexity of follicular growth and ovulation in the mammalian ovary is illustrated by the growing number of genetargeted mouse models that exhibit disruptions in one or both ovarian processes and hence have reduced fecundity or are infertile. For example, FSH-mediated follicular growth and differentiation are greatly impacted in mice null for IGF-I (Igf1) (2), growth differentiation factor (Gdf)-9 (3), cyclin D2 (Ccnd2) (4), -inhibin (Inha) (5), FSH- (Fshb) (6), FSH receptor (Fshr) (7), or phosphodiesterase-4d (Pde4d) (8). In turn, mice null for connexin-37 (Gja4) (9), prostaglandin-synthase-2 (Ptgs2) (10), CCAAT/enhancer binding protein- (Cebpb) (11), or progesterone receptor (Pgr) (12) possess ovaries that exhibit fully developed preovulatory follicles that fail to rupture after LH stimulation. We have previously reported that mice null for estrogen receptor (ER)- (Esr1) or estrogen receptor- (Esr2) also exhibit distinct ovarian defects that lead to compromised fertility (13). Mice null for ER ( ERKO) are anovulatory and invariably exhibit ovaries that possess multiple hemorrhagic/cystic follicles and elevated steroid synthesis, both of which are principally due to hyperstimulation of the ovary by elevated basal levels of circulating LH (14, 15). However, immature ERKO females do successfully ovulate when treated with exogenous gonadotropins if administered before onset of the overt hypergonadotropic phenotypes in the 3247

2 3248 Endocrinology, August 2005, 146(8): Couse et al. Superovulation of ER-Null Mice ovary (14). In contrast, ERKO mice exhibit ovaries that are grossly normal but are oligoovulatory and fail to exhibit efficient ovulation even when treated with exogenous gonadotropins (13). Therefore, the ovarian phenotypes in ERKO mice suggest that ER plays a more critical intraovarian role than ER. This hypothesis is supported by a plethora of evidence that ER is the predominant ER form expressed in the granulosa cells of growing and mature follicles in the rodent ovary (16 18). It has been long recognized that estradiol augments the actions of FSH on granulosa cells and both hormones are necessary to establish a fully differentiated and healthy preovulatory follicle, but the mechanisms involved are less clear (19). For example, estradiol alone has little effect on rodent granulosa cells but is required for maximum FSH stimulation of the following: 1) aromatase (Cyp19) expression (20) and estradiol synthesis (21, 22), 2) LH receptor (Lhcgr) expression (23) and LH responsiveness (24 26), 3) antrum formation (27), 4) gap-junction formation (28, 29), and 5) prevention of atresia (30). Indeed, immature rats treated with an aromatase inhibitor during gonadotropininduced ovulation exhibit a severely reduced number of healthy preovulatory follicles and ovulation rates (31). Therefore, the following studies were undertaken to further characterize the ovulatory defects in ERKO mice with the aim of determining the contribution of each ER form to follicle growth and rupture. Immature control, ERKO, and ERKO females were induced to ovulate via treatment with exogenous pregnant mare serum gonadotropin (PMSG) followed 48 h later by human choriogonadotropin (hcg). Ovaries were collected at four specific points during the course of treatment for biochemical, immunohistochemical, and histomorphological analyses. Immature ERKO females yielded a reduced number of ovulated oocytes, compared with similarly treated control and ERKO females. Gene expression analyses demonstrated that ERKO and ERKO ovaries show relatively normal expression patterns for most genes during follicle growth and ovulation. However, PMSG-induced granulosa cell differentiation was clearly deficient in ERKO ovaries as indicated by reduced estradiol synthesis and minimal Cyp19 and Lhcgr expression; hence, the response to subsequent hcg stimulation was severely compromised in ERKO ovaries. Overall, these data indicate that ER but not ER plays an integral role in the processes of gonadotropin-induced granulosa cell differentiation that are necessary for a full response to LH-induced ovulation. Materials and Methods Animals and treatments All procedures were performed in accordance with the National Institutes of Health Guide for the Care and Use of Laboratory Animals and were preapproved by the National Institute of Environmental Health Sciences Institutional Animal Care and Use Committee. The generation of mice null for the Esr1 (ER ) oresr2 (ER ) genes has been described previously (13). All animals were maintained in plastic cages under a 12-h light, 12-h dark schedule in a temperature-controlled room (21 22 C), fed National Institutes of Health 31 mouse chow and fresh water ad libitum. Immature ERKO (Esr1 / ) female mice were generated via heterozygous (Esr1 / ) breeding pairs. Immature ERKO (Esr2 / ) female mice were generated via breeding homozygous (Esr2 / ) males with heterozygous (Esr2 / ) females. All females were weaned at 21 d of age, genotyped as previously described (15), and subjected to gonadotropin-induced ovulation while between 23 and 30 d of age. The average animal weights at the time of necropsy were as follows: wild-type, 13.8 g; heterozygous, 14.3 g; ERKO, 13.2 g; and ERKO, 14.8 g. Gonadotropin-induced ovulation consisted of treating animals with 3.25 IU PMSG (Sigma Chemical Co., St. Louis, MO) between 1300 and 1500 h followed 48 h later with hcg (Sigma) of doses varying from 1.1 to 17.6 IU. Both drugs were dissolved in 0.85% saline solution and injected sc in a total volume of 0.1 ml. At necropsy, animals were killed by CO 2 -asphyxiation; whole blood was immediately collected via the descending vena cava; and the ovaries were dissected, trimmed of oviduct, and surrounding tissue and snap frozen, fixed in cold 10% buffered formalin, or processed for granulosa cell isolation. Oocyte counts after induced ovulation Animals were killed h after hcg treatment and the ovaries and oviduct removed to M-2 medium (Specialty Media, Lavallette, NJ). The cumulus-oocyte complexes were surgically extracted from each oviduct, pooled into a single droplet of M-2 medium supplemented with 0.3% hyaluronidase (Sigma), and allowed to undergo enzymatic disassociation at 37 C for approximately 15 min. The number of oocytes per animal was then determined by manually counting using a MZ6 stereoscope (Leica, Québec, Canada). The data presented are the combination of multiple induced ovulation experiments carried out over a period of time. Granulosa cell isolation Ovaries were removed and immediately transferred to a 100-mm cell culture dish containing 15 ml ice-cold M199 medium supplemented with 1 mg/ml BSA, 2.5 g/ml Fungizone, and 50 g/ml gentamicin (all reagents from Invitrogen, Carlsbad, CA). Ovaries were pooled according to genotype and treatment. Once all ovaries were collected, the granulosa cells from each were expressed by manual puncture with 25-gauge needles followed by slight pressure applied with a sterile spatula. Follicular debris was then removed manually and the granulosa cell suspension filtered through a 150- m Nitex nylon membrane (Sefar America Inc., Depew, NY) mounted in Swinnex filters (Millipore, Billerica, MA). The granulosa cells were then pelleted by centrifugation at 250 g for 5 min at 4 C, followed by two washes in DMEM/F-12 medium. The final cell pellet was frozen at 80 C until RNA extraction. RNA isolation and semiquantitative RT-PCR (SQ-RT-PCR) gene expression assays Total RNA was isolated from frozen ovaries using TRIZOL reagent (Invitrogen) according to the manufacturer s protocol. All RNA samples were rid of contaminating DNA using the DNA-free reagents (Ambion, Austin, TX) according to the manufacturer s protocol, followed by normalization to a concentration of 0.2 g/ml in RNase-free water. A SQ-RT-PCR approach was used to assess gene expression levels as previously described (15). For each individual ovarian RNA sample, 1 g of RNA was used in a 25- l cdna reaction using random hexamers and the Superscript cdna synthesis system (Invitrogen) according to the manufacturer s protocol. PCRs were prepared using the equivalent of 1 l cdna per 15- l reaction for each respective primer set (Table 1) using PCR reagents and Platinum Taq polymerase (Invitrogen) as previously described (15). PCR was carried out in a Thermo Hybaid multiblock system (Franklin, MA) at 95 C for 30 sec, 58 C for 45 sec, and 72 C for 30 sec for cycles, depending on the level of gene expression. Primers for ribosomal protein L7 (Rpl7) were included in all reactions as an internal positive control and for normalization. All samples were electrophoresed on an agarose gel (2% NuSieve/0.7% SeaKem, BMA Bioproducts, Rockland, ME) in 1 Tris-borate EDTA followed by immobilization to BrightStar nylon membrane (Ambion) using the Royal Genie blotter (Idea Scientific, Minneapolis, MN). All blots were probed with a nested oligo specific to sequences internal to the PCR primers (Table 1) that was 5 -radiolabeled with [ 33 P- ATP] (Amersham Pharmacia Biotech, Buckinghamshire, UK) using T 4 polynucleotide kinase (New England Biolabs, Beverly, MA). Hybridization of blots was carried out in Rapid-Hyb buffer (Amersham Pharmacia Biotech) with more than cpm of probe per milliliter overnight in a rotisserie hybridization oven (Thermo Hybaid) at 42 C, followed by washing according to the manufacturer s protocol. Final SQ-RT-PCR blots were exposed to a Phos-

3 Couse et al. Superovulation of ER-Null Mice Endocrinology, August 2005, 146(8): TABLE 1. Genes evaluated over the course of gonadotropin-induced ovulation in immature ERKO mice Gene Accession no. Amplimer Probe Results a Adamts1 NM Fig. 7 Amh X No change throughout Amhr2 M No change throughout Ar M No change throughout Areg NM Fig. 5 Bak1 Y Fold at 10 h post hcg Bax L Fold at 10 h post hcg Bcl2 M No change throughout Btc NM Fig. 5 Casp2 Y No change throughout Casp3 U No change throughout Casp8 AF No change throughout Casp9 AB No change throughout Ccnd2 M No change throughout Ccne1 NM No change throughout Cdkn1a U Fold at 10 h post hcg Cdkn1b U Fold at 20 h post hcg Cyp19 D Fig. 4 Ereg NM Fig. 5 Fshr NM Fig. 4 Gata4 U No change throughout Gata6 S No change throughout Has2 NM Fold at 4 h post hcg Hsd17b3 U Detectable in ERKO only Hsd17b7 NM Fig. 5 Lhcgr NM Fig. 4 Mmp2 M Fold at 20 h post hcg Mmp19 AF Figs. 7 and 8 Nra51 AB No change throughout Nr5a2 NM No change throughout Pgr M Figs. 7 and 8 Plat J Fold at 4 h post hcg Prkar2b AI No change throughout Ptgs2 M Fig. 6 Rgs2 NM No change throughout Timp BC Fig. 5 Timp3 L Fold at 20 h post hcg Tnfaip6 NM Fig. 5 a No change throughout, change of less than 2-fold control-vehicle; all fold changes refer to fold control-vehicle. phorimager screen and the data analyzed with a Storm 860 and accompanying ImageQuant Software (Molecular Dynamics, Sunnyvale, CA). RNA isolation and Northern blot analysis of gene expression TRIZOL reagent (Invitrogen) was used according to the manufacturer s protocol to extract total RNA from individual pairs of frozen ovaries or isolated granulosa cells from pooled ovaries. Northern blots were generated using 20 g total RNA from whole ovaries or 5 g total RNA from isolated granulosa cells using NorthernMax (Ambion) reagents according to the manufacturer s protocol. RNAs were fractionated on a denaturing 1.2% agarose gel and immobilized to Brightstar nylon membrane (Ambion) by capillary action according to the manufacturer s protocol. All membranes were prehybridized and hybridized in NorthernMax Pre/Hyb buffer (Ambion) in a hybridization oven (Thermo Hybaid) at 65 C. All blots were probed with strippable, radiolabeled riboprobes generated using Strip-EZ riboprobe transcription (Ambion) reagents and [ 32 P]- UTP (Amersham). Blots were probed with cpm of purified probe per milliliter of hybridization buffer at 65 C overnight and washed according to the membrane manufacturer s protocol. Blots were then exposed to a PhosphorImager screen and analyzed with a Storm 860 and accompanying ImageQuant Software (Molecular Dynamics). Once exposed, the blots were immediately stripped using the Strip-EZ (Ambion) reagents and prepared for reprobing. All probes used herein have been previously described: -actin (Actb) (15), Cyp19 (15), Lhcgr (32), Ptgs2, and Pgr (14). Histological and immunohistochemical evaluation of ovaries Formalin-fixed ovaries were embedded in paraffin, sectioned, and stained with hematoxylin and eosin according to standard laboratory protocols or left unstained and stored at 4 C. For immunohistochemical detection of Ptgs2, sections were subjected to deparaffinization/antigen retrieval using 1 Reveal (Biocare Medical, Walnut Creek, CA) in a decloaking chamber (Biocare Medical) according to the manufacturer s protocol, followed by multiple washes in water and then 1 automation buffer (Biomeda, Foster City, CA). Endogenous peroxidase activity was then quenched by incubating the sections in 3% hydrogen peroxide for 10 min. Sections were then blocked for endogenous avidin and biotin using an avidin/biotin blocking reagents kit (Vector Laboratories, Burlingame, CA) and then placed under a general blocking reagent of 10% BSA in 1 automation buffer for 30 min. Sections were then incubated with goat-raised antirat Ptgs2 (M-19) (Santa Cruz Biotechnology, Santa Cruz, CA) at a dilution of 1:5000, followed by a 1:500 dilution of biotinylated antigoat IgG (Vector Laboratories), followed by a 1:50 dilution of extraavidin peroxidase (Sigma). All dilutions and subsequent washings were done in 1 automation buffer. Specific immunoreactivity was detected by applying NovaRED (Vector Laboratories) to sections for 5 min. Immunohistochemical detection of progesterone receptor (PR) followed a similar protocol except sections were first deparaffinized via xylene washes followed by antigen retrieval using 1 nuclear decloaker (Biocare Medical) in a decloaking chamber (Biocare Medical) according to the manufacturer s protocol. Sections were then incubated with rabbit-raised antimouse PR (a gift from Dr. Dean P. Edwards, University of

4 3250 Endocrinology, August 2005, 146(8): Couse et al. Superovulation of ER-Null Mice Colorado Health Sciences Center, Denver, CO) at a dilution of 1:100; followed by a 1:500 dilution of biotinylated antirabbit IgG (Amersham Pharmacia Biotech). All photomicrographs were collected using a BX-50 microscope and OLY-750 video camera (Olympus, Tokyo, Japan). Western blot analysis Cytosolic protein fractions were isolated from individual pairs of frozen ovaries using the NE-PER (Pierce Biotechnology, Rockford, IL) reagents according to the manufacturer s protocol. Final protein preparations were quantified using the BCA-200 Protein Assay Reagents (Pierce Biotechnology) and BSA as a standard. All blots were generated using 30 g of cytosolic protein per sample prepared in NuPage LDS sample buffer with reducing reagent (Invitrogen), fractionated on a 4 12% Tris-Bis acrylamide gel in 3[N-morpholino]propanesulfonic acid buffer (Invitrogen), and immobilized to nitrocellulose membrane (Invitrogen) according to the manufacturer s protocol. For detection of Ptgs2, blots were probed with rabbit-raised antimouse cyclooxygenase-2 (Cayman Chemical Co., Ann Arbor, MI) at a dilution of 1:1,000, followed by a 1:20,000 dilution of horseradish peroxidase-conjugated antirabbit IgG (Cell Signaling Technology, Beverly, MA). For detection of PR, blots were probed with rabbit-raised antimouse PR (a gift from Dr. Dean P. Edwards) at 10 g/ml, followed by a 1:20,000 dilution of horseradish peroxidase-conjugated antirabbit IgG (Cell Signaling Technology). Blocking was carried out in a solution of 5% nonfat dry milk in 1 Tris-buffered saline with Tween 20, and all postantibody washes were carried out in 1 Tris-buffered saline with Tween 20 alone. Final detection of immunoreactivity was carried out using the SuperSignal West- Dura (Pierce Biotechnology) reagents according to the manufacturer s protocol and captured via exposure to x-ray film or using an EpiChemi 3 Darkroom (UVP Bioimaging Systems, Upland, CA). IU) and hcg (2.2 IU) indicated no difference between immature wild-type (Esr1 / or Esr2 / ) and heterozygous (Esr2 / ) females, allowing the results from all three genotypes to be combined into a single group that was defined thereafter as controls. As shown in Fig. 1A, the average oocyte yield following standard induced ovulation in immature ERKO females ( oocyte/female) was significantly reduced (P 0.05), compared with immature control ( ) and ERKO ( ) females. The rate of ovulation in ERKO females observed herein is consistent with our original description of the ERKO in which assays were conducted on a much smaller number of animals (13). All three genotypes exhibited a broad range of oocyte yields, characteristic of gonadotropin-induced ovulation in mice. However, seven of 31 (22.5%) treated ERKO females failed to ovulate even a single recoverable oocyte, whereas the lowest individual yield among control and ERKO females was six and seven oocytes, respectively. To determine whether an increased dose of hcg might improve the ovulatory response in immature ERKO females, animals were treated with PMSG (3.25 IU) followed 48 h later with a dose of hcg ranging from 1.1 to 17.6 IU (Fig. 1B). As expected, 2.2 IU hcg proved to be the optimum dose in controls as the average yield after 1.1 IU hcg was signif- Hormone RIAs Upon necropsy, whole blood was collected from the inferior vena cava and mixed with 10 l heparin (60 mg/ml) on ice. Plasma was then collected and stored at 70 C until assay. All hormone assays were carried out on plasma collected from individual animals. Plasma progesterone and estradiol were assayed in singlicate per animal on 25- and 200- l aliquots, respectively, using the active progesterone RIA and ultrasensitive estradiol double-antibody RIA kits (Diagnostic Systems Laboratories, Webster, TX) according to the manufacturer s protocol. All final assays were quantified using a Packard Multi-Prias 2 -counter (PerkinElmer, Boston, MA). The following parameters applied for each RIA: estradiol (one assay; least detectable concentration 2.2 pg/ml; average intraassay coefficient of variation 18%; average interassay coefficient of variation not applicable); and progesterone (two assays; least detectable concentration 0.3 ng/ml; and average intraassay coefficient of variation 14%, and average interassay coefficient of variation 13%). Statistical analysis All data were analyzed for statistical significance (P 0.05) using JMP software (SAS Institute, Cary, NC). Statistical analysis of oocyte counts indicated some difference between experimental trials but within the expected range given the variability of ovulation as an experimental end point. Therefore, our statistical analysis did not warrant the omission of any one experimental trial and allowed the data of all trials to be combined into a single set. Because the oocyte yield data were skewed, the Kruskal-Wallis ANOVA was used followed by the Mann-Whitney U test to determine where significant differences existed. Plasma steroid levels and gene expression data sets were first tested for homoscedasticity of variance using the Levene s test and, if failed, were log transformed before further statistical analysis. All data sets were then evaluated using a one-way ANOVA followed by the Tukey-Kramer honestly significant difference post hoc test when applicable. Results ER -null females exhibit reduced oocyte yield after induced ovulation Statistical comparison of oocyte yields after treatment with a previously determined optimum regimen of PMSG (3.25 FIG. 1. Oocyte yield after induced ovulation in immature control and ERKO mice. Immature mice were treated with PMSG (3.25 IU) followed 48 h later with hcg at the dose indicated. Oviducts were then collected h after hcg treatment and the oocytes extracted and counted. A, Results after treatment with gonadotropin doses previously determined to be optimum for induced ovulation. Each dot represents the oocyte yield of an individual animal within their respective genotype; vertical bars represent the average oocyte yield within each genotype. B, hcg dose-response for control and ERKO females showing the average oocyte yield for each genotype after being treated with PMSG (3.25 IU) followed by increasing doses of hcg 48 h later. a, P 0.05 vs. control with 2.2 IU hcg; b, P 0.05 vs. oocyte yield with 2.2 IU hcg within the same genotype.

5 Couse et al. Superovulation of ER-Null Mice Endocrinology, August 2005, 146(8): icantly reduced (P 0.05), and no significant improvement was observed among the three doses greater than 2.2 IU. Simply doubling the hcg dose to 4.4 IU did improve the ovulatory response in immature ERKO females, but no further improvement was observed with 8.8 IU or 17.6 IU hcg. All three higher doses of hcg led to an improved frequency of recoverable oocytes from the oviducts of ERKO females, with the lowest yield being three oocytes. Nonetheless, even at 17.6 IU hcg, 8-fold the optimum, the average oocyte yield among immature ERKO females remained below that of controls at all doses. These data confirm our previous findings that ERKO females exhibit an attenuated response to gonadotropin-induced ovulation in terms of oocyte yield and further demonstrates this deficit cannot be overcome with increased doses of hcg. ER-null females exhibit aberrant plasma steroid levels during induced ovulation The plasma levels of estradiol and progesterone fluctuate in dramatic but predictable patterns during folliculogenesis and ovulation and can therefore be used as parameters for evaluating the ovarian response to gonadotropin-induced ovulation. As shown in Fig. 2A, plasma estradiol levels in control females increased 6-fold (from 6.8 to 40.5 pg/ml) 48 h after a single PMSG treatment but dropped precipitously after hcg treatment to a nadir just before ovulation ( h after hcg). Plasma progesterone levels in control females were relatively unaffected by PMSG but rose rapidly within 4 h of hcg treatment and peaked at 10-fold the basal level and then gradually decreased thereafter (Fig. 2A). These data are congruent with the temporal changes in plasma steroid levels documented to occur during the rodent ovarian cycle (33). Immature ERKO females exhibited a control-like pattern of plasma estradiol over the course of induced ovulation (Fig. 2B). However, plasma estradiol levels 48 h after PMSG and 4 h after hcg were much higher in ERKO vs. controls and proved to be statistically significant (P 0.05) at the latter time point. By h after hcg treatment, plasma estradiol levels in ERKO females were similar to that of control. The pattern and level of plasma progesterone among ERKO females mirrored that of control, differing only at the final time point (20 h after hcg), at which ERKO levels were approximately half that of control (9.2 vs. 20 ng/ml) (Fig. 2B). In contrast, immature ERKO females exhibited a strikingly abnormal pattern of estradiol secretion such that plasma levels were abnormally low 48 h after PMSG treatment (P 0.05 vs. control) but rose rapidly to a peak of more than 40 pg/ml within 4 h after hcg treatment (Fig. 2C), a significant divergence from the precipitous drop in plasma estradiol that followed hcg in control females (compare with Fig. 2A). Estradiol levels did eventually decline in ERKO females, but the expected nadir at 10 h after hcg remained more than 3-fold higher (P 0.05) than controls. The pattern and level of progesterone secretion in immature ERKO females was similar to control, rapidly peaking 4 h after hcg and decreasing thereafter. Therefore, ERKO females exhibit a normal pattern but somewhat aberrant levels of sex steroid secretion over the course of induced ovulation, whereas FIG. 2. Plasma levels of ovarian steroids during gonadotropin-induced ovulation. Immature mice were treated with PMSG (3.25 IU) followed 48 h later with hcg (2.2 IU). Animals were killed at the indicated times over the course of gonadotropin-induced ovulation. Plasma was prepared from whole blood and assayed for estradiol and progesterone content by RIA. Shown are the average ( SEM) plasma levels of estradiol and progesterone at each time point for control (A), ERKO (B), and ERKO females (C). a, P 0.05 vs. average control estradiol level at the same time point; b, P 0.05 vs. average control progesterone at the same time point. ERKO females exhibit an abnormal pattern, marked by an aberrant increase in estradiol after hcg treatment that results in a simultaneous peak in estradiol and progesterone synthesis just hours before ovulation. Ovarian histology during induced ovulation Ovaries from immature untreated control, ERKO, and ERKO females were relatively indistinguishable because all exhibited a sparse number of growing follicles, most of which possessed multiple indications of atresia (data not shown). Some small antral follicles were observed in untreated ERKO ovaries (data not shown), congruent with previous descriptions of premature stimulation by rising endogenous

6 3252 Endocrinology, August 2005, 146(8): Couse et al. Superovulation of ER-Null Mice gonadotropins in this model (14, 32). PMSG treatment induced the appearance of multiple growing antral follicles in the ovaries of all three genotypes within 48 h. Compared with follicles in control and ERKO ovaries, maturing follicles in ERKO ovaries tended to exhibit slightly undersized antra (data not shown), but this was variable and difficult to quantify. Almost all ERKO ovaries exhibited at least one large hemorrhagic and cystic follicle 48 h after PMSG treatment, similar to the cystic follicles described by others to occur spontaneously in older ERKO females (32). None of the PMSG-stimulated control or ERKO ovaries exhibited ERKO-like hemorrhagic follicles. By 10 h after hcg treatment, control and ERKO ovaries exhibited multiple preovulatory follicles undergoing expansion of the cumulus-oocyte complex (COC) (Fig. 3, A E) or already beginning corpus luteum formation (data not shown). In contrast, visible COC expansion among ovulatory follicles in ERKO ovaries 12 h after hcg treatment (Fig. 3, C and F) was noticeably variable because observance of an ovulatory follicle undergoing COC expansion juxtaposed to another that was not in ERKO ovaries was common. SQ-RT-PCR for gene expression during induced ovulation The mouse ovary exhibits a complex but synchronous pattern of gene expression during folliculogenesis and ovulation. Because the ERs are ligand-inducible transcription factors, the insufficient response of ERKO follicles to gonadotropin-induced maturation and ovulation may be due to the loss of ER -mediated expression of genes that are critical to these processes. To address this issue, we evaluated gene expression in the ovaries of immature control, ERKO, and ERKO females over the course of standard gonadotropininduced ovulation with 3.25 IU PMSG followed 48 h later with 2.2 IU hcg. Ovaries were collected from animals of each genotype at five distinct time points during induced ovulation: 1) no treatment, 2) 48 h after PMSG, 3) 4 h after hcg, 4) 10 h after hcg, and 5) h after hcg. The finite amount of RNA available from the ovaries of individual animals limited the number of genes that could be evaluated by traditional means. We therefore developed a SQ-RT-PCR FIG. 3. ERKO follicles fail to exhibit cumulus-oocyte expansion 10 h after hcg-induced ovulation. Immature mice were treated with PMSG (3.25 IU) followed 48 h later with hcg (2.2 IU). Animals were killed 10 h after hcg treatment, just before the time of follicle rupture. Shown are representative cross-sections of control (A and D), ERKO (B and E) and ERKO (C and F) follicles. There is obvious expansion of the COC in the control and ERKO examples, whereas in both ERKO examples, the cumulus oophorus cells remain relatively packed around the oocyte and still retain some attachment to the mural granulosa cells. method that allowed for evaluation of the expression of more than 35 different genes in the ovaries of individual animals. The SQ-RT-PCR method also permitted the sample numbers in this initial screen of gene expression to be more than eight per genotype per time point, making the assay more robust, allowing for proper statistical analysis of the data. Table 1 lists all of the genes evaluated in this study, including those that exhibited little change in expression over the course of induced ovulation or in which changes occurred but no genotypic differences were observed. Genes that exhibited more dramatic changes in expression over the course of induced ovulation are discussed in greater detail below. Certain genes that are known to play an important role in folliculogenesis and ovulation were found to be aberrantly expressed in ERKO ovaries by SQ-RT-PCR and were therefore evaluated further by: 1) Northern blot on RNA prepared from whole ovaries and isolated granulosa cells, 2) Western blot analysis of whole ovarian protein extracts, and 3) immunohistochemistry in whole ovaries at the most relevant time point. In general, Northern blot analyses confirmed the results initially generated by SQ-RT-PCR, thereby validating this method as an acceptable semi-high-throughput screen for changes and abnormalities in gene expression. ER -null granulosa cells exhibit insufficient preovulatory differentiation The process of granulosa cell differentiation that occurs during progression from a preantral to preovulatory follicle is dependent on sufficient FSH stimulation (6, 7, 34) and is marked by the acquisition of increased aromatase activity and LH receptor. Because estradiol is known to augment the actions of FSH (19), we were specifically interested in evaluating the expression pattern of the genes encoding Fshr, Lhcgr, and Cyp19 in control and ERKO ovaries 48 h after PMSG (an FSH analog) treatment. As expected, Fshr expression was increased 3-fold in PMSG-treated vs. untreated control ovaries (Fig. 4A). Similar PMSG induction of Fshr expression was observed in ERKO and ERKO ovaries, indicating this effect of PMSG is independent of ER actions (Fig. 4A). Within 4 h after hcg treatment, Fshr expression returned to basal levels in all three genotypes, although post-hcg levels in ERKO ovaries were elevated vs. control (Fig. 4A). Acquisition of LH receptor by preovulatory granulosa cells is a distinct marker of differentiation and is critical to their ability to respond to the LH surge and initiate ovulation. As expected, Lhcgr expression was increased 5-fold 48 h after PMSG treatment in control ovaries and remained relatively unchanged after hcg treatment when evaluated by SQ-RT- PCR in whole ovaries (Fig. 4A). Immature ERKO ovaries exhibited a similar but exaggerated increase ( 5-fold control levels) in Lhcgr expression 48 h after PMSG treatment, congruent with our previous descriptions of elevated Lhcgr expression in the ovaries and granulosa cells of adult ERKO ovaries (14, 32). ERKO ovaries also exhibited a relatively normal PMSG induction of Lhcgr expression when evaluated by SQ-RT-PCR in whole ovaries (Fig. 4A). However, whereas hcg had little effect on Lhcgr expression in control and ERKO ovaries, it led to a further doubling in ERKO ovaries after 4 h. Similar expression data for Lhcgr were generated when whole ovary RNA prep-

7 Couse et al. Superovulation of ER-Null Mice Endocrinology, August 2005, 146(8): FIG. 4. Ovarian expression of Fshr, Lhcgr, and Cyp19 48 h after PMSG treatment in control and ERKO ovaries. Immature mice were treated with PMSG (3.25 IU) followed 48 h later with hcg (2.2 IU). Animals were killed at the indicated times over the course of gonadotropin-induced ovulation, whole ovaries collected, and total RNA extracted for gene expression analysis. A, Quantitative data from SQ-RT-PCR for Fshr, Lhcgr, and Cyp19. All data are expressed as the average fold control-none (untreated) SEM. B, Northern blot analysis for Cyp19 and Lhcgr expression in whole ovaries or granulosa cells isolated from control and ERKO females during the indicated treatments and time points. a, P 0.05 vs. control at the same time point. arations were evaluated by Northern blot analysis, although some reduction in the levels of Lhcgr transcripts after PMSG treatment was apparent in ERKO ovaries (Fig. 4B). Because Lhcgr expression is constitutive in thecal cells, it is difficult to accurately assess granulosa cell-specific expression when evaluated in whole ovarian RNA preparations. Therefore, total RNA was prepared from enriched granulosa cell samples collected from control and ERKO ovaries 48 h after PMSG and 4 h after hcg (which followed 48 h PMSG treatment). As shown in Fig. 4B, granulosa cell-specific levels of Lhcgr transcripts in ERKO ovaries were dramatically reduced, compared with control ovaries after PMSG and PMSG/hCG treatment. Interestingly, the Lhcgr probe used for Northern blot analysis indicated two prominent transcripts in whole ovary RNA but only one in enriched granulosa cells (Fig. 4B). The specificity of the Lhcgr probe used herein has been previously documented (14, 32). The reduced level Lhcgr expression in PMSG-stimulated ERKO granulosa cells was also confirmed by Taq-man quantitative RT-PCR (data not shown). These data strongly indicate that granulosa cells of preovulatory follicles in ERKO ovaries fail to develop sufficient LH receptor levels. Cyp19 is necessary for the final stages of estradiol synthesis and is specifically induced by FSH in the granulosa cells of preovulatory follicles (19). As expected, PMSG treatment led to a doubling of Cyp19 expression that was transiently reduced after hcg in immature control ovaries (Fig. 4A), a pattern that closely mimics the plasma estradiol levels shown in Fig. 2A. Northern blot analysis of RNA from whole ovary or isolated granulosa cells confirmed the induction of Cyp19 expression by PMSG in control ovaries (Fig. 4B). ERKO ovaries exhibited a similar, control-like expression pattern of Cyp19 except that levels were much higher after PMSG treatment, strongly correlating with the above normal plasma estradiol levels observed at this time point in ERKO females (Fig. 2B). In contrast to control and ERKO females, immature ERKO females exhibited an aberrant Cyp19 expression pattern such that PMSG treatment had little effect but hcg induced an almost 10-fold increase within 4 h (Fig. 4A). The minimal induction of Cyp19 expression by PMSG but aberrant increase after hcg in ERKO ovaries was confirmed by Northern blot analysis (Fig. 4B) and Taq-man quantitative RT-PCR (data not shown) of whole ovary and isolated granulosa cell RNAs. Furthermore, plasma estradiol levels in immature ERKO females (Fig. 2C) mirrored the observed Cyp19 expression pattern, including a striking increase 4 h after hcg treatment. These data indicate that preovulatory granulosa cells do not properly differentiate and develop sufficient aromatase activity in response to PMSG but then respond inappropriately to subsequent hcg treatment by dramatically increasing Cyp19 expression, as opposed to the hcg-elicited decrease observed in control and ERKO ovaries. ER -null granulosa cells exhibit an attenuated response to hcg Perhaps the most dynamic changes in ovarian gene expression occur shortly after the LH surge and can be mimicked by hcg administration in PMSG-primed mice. Several genes evaluated in this study exhibited robust induction 4 h after hcg exposure in control ovaries, only to return to near baseline levels within h; these included amphiregulin (Areg), betacellulin (Btc), Cebpb, epiregulin (Ereg), hyaluronan synthase-2 (Has2), Pgr, Ptgs2, tissue plasminogen activator (Plat), and TNF- induced protein 6 (Tnfaip6) (Table 1 and Figs. 5 7). Such rapid and transient up-regulation in the ovary after hcg treatment or just before ovulation has been previously reported for each of these genes (1, 35). Several other genes exhibited equally impressive induction in control ovaries that was delayed until 10 h after hcg treatment and more proximal to the time of follicular rupture, including A disintegrin and metalloproteinase with thrombospondinlike repeats-1 (Adamts1), cyclin-dependent kinase inhibitor 1A (Cdkn1a), matrix metalloproteinase (Mmp) 19, and tissueinhibitor of metalloproteinase (Timp) (Table 1 and Fig. 5). Finally, several genes exhibited peak expression in the postovulatory ovary ( 20 h after hcg treatment), including 17 -hydroxysteroid dehydrogenase type VII (Hsd17b7), Mmp2, and Timp3 (Table 1 and Fig. 5). Once again, the hcg induction and expression patterns observed herein for each of the above genes has been previously reported (36 38). In general, immature ERKO ovaries exhibited a control-like expression pattern for all of the genes evaluated (Table 1 and Fig. 5). Some minor differences in the level of peak expression were observed in ERKO vs. control ovaries, such as an abovenormal increase in Areg and Tnfaip6 expression at 4 h post hcg

8 3254 Endocrinology, August 2005, 146(8): Couse et al. Superovulation of ER-Null Mice FIG. 5. Ovarian gene expression over the course of gonadotropin-induced ovulation in control and ERKO ovaries. Immature mice were treated with PMSG (3.25 IU) followed 48 h later with hcg (2.2 IU). Animals were killed at the indicated times over the course of gonadotropin-induced ovulation, whole ovaries collected, and total RNA extracted for gene expression analysis. Shown is the quantitative data from SQ-RT-PCR for Areg, Btc, Cebpb, Ereg, Hsd17b7, Mmp19, Timp, and Tnfaip6. All data are expressed as the average fold control-none (untreated) SEM. a, P 0.05 vs. control at the same time point. and Timp expression at 10 h after hcg and a slightly lower increase in Mmp19 expression at 20 h after hcg (Fig. 5). One striking abnormality observed in ERKO ovaries was premature induction of Hsd17b7 expression with PMSG alone such that levels were increased more than 15-fold vs. approximately 3-fold in control ovaries. The Hsd17b7 gene encodes the enzyme 17 -HSD VII, which was recently described as specific to luteal cells and necessary for the final step in estradiol synthesis in the corpus luteum (38). Immature control ovaries exhibited a slight increase in Hsd17b7 expression after PMSG treatment but a much greater response after hcg, which elicited a steady induction that peaked at more than 12-fold 20 h after hcg (Fig. 5). Immature ERKO ovaries also expressed considerable levels of Hsd17b3 (Table 1); which encodes a Leydig cell-specific enzyme involved in testosterone synthesis and is previously described to be uniquely expressed in ERKO ovaries (15). Immature ERKO ovaries exhibited appropriate hcg induction of several genes, including Areg, Cebpb, Ereg, and Has2. However, hcg failed to fully induce expression of several other genes in ERKO ovaries relative to control, including Adamts1, Btc, Mmp19, Pgr, Ptgs2, and Timp (Figs. 5 7). Interestingly, Timp and Mmp19 expression in ERKO ovaries did eventually increase to control-like levels but not until 20 h after hcg, a considerable delay, compared with the peak at 4 h after hcg observed in control ovaries (Fig. 5). Ptgs2 and PR are both obligatory to follicular rupture (10, 12), and each exhibited a severely blunted response to hcg in ERKO ovaries. Therefore, the expression pattern of Ptgs2 and Pgr in control and ERKO ovaries after hcg treatment was evaluated in greater detail. SQ-RT-PCR analysis indicated that Ptgs2 mrna levels were induced in ERKO ovaries 4 h after hcg treatment but that peak levels were more than 60% lower (P 0.05) than that observed in control and ERKO ovaries (Fig. 6A). These data were confirmed by Northern blot analysis, which demonstrated that Ptgs2 transcripts were barely detectable in ERKO ovaries and isolated granulosa cells collected 4 h after hcg treatment of PMSG-primed mice vs. the extraordinary induction observed in similarly treated control ovaries (Fig. 6B). Western blot analysis of cytosolic protein preparations from control, ERKO, and ERKO ovaries further illustrated that the levels of immunoreactive Ptgs2 (cyclooxygenase-2) after hcg induction were barely detectable in ERKO ovaries but easily observed in control and ERKO ovaries (Fig. 6C). Immunohistochemical detection of Ptgs2 in control and ERKO ovaries collected at 4 and 10 h after hcg treatment indicated that expression was localized to the granulosa cells of ovulatory and ovulating follicles in both genotypes (Fig. 6D). Ptgs2 immunoreactivity was especially strong among granulosa cells lining the antrum and forming the cumulus oophorus (Fig. 6D), in agreement with previous descriptions of ovarian Ptgs2 expression during induced ovulation in mice (39). In contrast, a minimal number of granulosa cells of ERKO preovulatory follicles 4 and 10 h after hcg treatment exhibited Ptgs2 immunoreactivity, and these cells were sparse and exhibited no apparent pattern (Fig. 6D). Although a discernible range of Ptgs2 expression was apparent when comparing ERKO preovula-

9 Couse et al. Superovulation of ER-Null Mice Endocrinology, August 2005, 146(8): FIG. 6. ERKO ovaries exhibit an attenuated induction of the Ptgs2 gene after hcg treatment during gonadotropin-induced ovulation. Immature mice were treated with PMSG (3.25 IU) followed 48 h later with hcg (2.2 IU). Animals were killed at the indicated time points over the course of gonadotropin-induced ovulation, whole ovaries collected for RNA or protein extraction, and immunohistochemistry. A, Quantitative data from SQ-RT-PCR for Ptgs2 gene expression showing the rapid rise in expression 4 h after hcg treatment that is attenuated in ERKO ovaries. Data are expressed as the average fold control-none (untreated) SEM. B, Northern blot analysis for Ptgs2 gene expression in whole ovaries or granulosa cells isolated from control and ERKO females during the indicated treatments and time points. Ptgs2 expression peaks at 4 h after hcg and decreases precipitously thereafter; Ptgs2 transcripts are essentially undetectable in ERKO ovaries 4 h after hcg treatment. C, Western blot analysis for Ptgs2 protein in control, ERKO, and ERKO ovaries 48 h after PMSG treatment and 4 h after subsequent hcg treatment. D, Immunohistochemistry for Ptgs2 in control, ERKO, and ERKO ovaries collected 4 and 10 h after hcg treatment. Shown are representative photomicrographs of low- and high-power magnification for each genotype. Ptgs2 immunoreactivity is clearly localized to the granulosa cells of preovulatory follicles in control and ERKO ovaries and peaks at 4 h after hcg but remains 10 h after hcg treatment. The representative ERKO ovaries illustrate the severely attenuated induction of Ptgs2 in the granulosa cells of preovulatory follicles. Some Ptgs2 immunoreactivity is detectable in ERKO preovulatory follicles, but this is generally restricted to the cumulus-oophorus. a, P 0.05 vs. control at the same time point. tory follicles, even within the same ovary, none exhibited control-like levels of immunoreactivity. Furthermore, the preovulatory rise in Tnfaip6 expression (Fig. 5), which is reported to be partially dependent on prerequisite increases in Ptgs2 function within the ovulating follicle (40), was blunted in ERKO ovaries vs. control ovaries at 10 h post-hcg treatment, suggesting insufficient Ptgs2 action in the former (Fig. 5). Assays for Pgr expression over the course of induced ovulation in mice indicated an extraordinary increase in Pgr mrna levels at 4 h post-hcg treatment in PMSG-primed mice, followed by a return to barely detectable levels within 6 h (Fig. 7). This dramatic and transient induction of Pgr expression after hcg has been well described (1, 19) and is critical to follicular rupture (12). In turn, although Pgr mrna levels were increased in ERKO ovaries 4 h after hcg treatment, the level of peak induction was more than 70% less (P 0.05) than that observed in control ovaries (Fig. 6A) when assessed by SQ-RT-PCR. Northern blot analysis of RNA from whole ovaries or isolated granulosa cells of control mice confirmed the induction of Pgr expression 4 h after hcg treatment in control females but only barely detectable levels in similarly treated ERKO ovaries and granulosa cells (Fig. 7B). Immunohistochemistry for PR immunoreactivity during induced ovulation indicated a pattern that was strikingly similar to that of Ptgs2 (Fig. 7C). Control and

10 3256 Endocrinology, August 2005, 146(8): Couse et al. Superovulation of ER-Null Mice FIG. 7. ERKO ovaries exhibit an attenuated induction of the Pgr gene after hcg treatment during gonadotropin-induced ovulation. Immature mice were treated with PMSG (3.25 IU) followed 48 h later with hcg (2.2 IU). Animals were killed at the indicated time points over the course of gonadotropin-induced ovulation, whole ovaries collected for RNA or protein extraction, and immunohistochemistry. A, Quantitative data from SQ-RT-PCR for Pgr gene expression showing the rapid rise in expression 4 h after hcg treatment that is attenuated in ERKO ovaries. Data are expressed as the average fold control-none (untreated) SEM. B, Northern blot analysis for Pgr gene expression in whole ovaries or granulosa cells isolated from control and ERKO females during the indicated treatments and time points. Pgr expression peaks at 4 h after hcg and is undetectable at all other time points. Pgr transcripts are essentially undetectable in ERKO ovaries 4 h after hcg treatment. C, Immunohistochemistry for PR in control, ERKO, and ERKO ovaries collected 4 h after hcg treatment. Shown are representative photomicrographs of low- and high-power magnification for each genotype. PR immunoreactivity is clearly localized to the granulosa cells of preovulatory follicles in control and ERKO ovaries. The representative ERKO ovaries illustrate the severely attenuated induction of PR in the granulosa cells of preovulatory follicles. D, Quantitative data from SQ-RT-PCR for Adamts1 gene expression, a PR-regulated gene in the ovulatory follicle, showing the lack of increased expression 12 h after hcg treatment in ERKO ovaries. Data are expressed as the average fold control-none (untreated) SEM. ERKO ovaries collected 4 h after hcg treatment possessed multiple growing follicles; however, only preovulatory follicles exhibited strong PR immunoreactivity (Fig. 7C). PR expression was evenly distributed throughout the mural granulosa cells but notably absent from the cumulus oophorus in both genotypes, in agreement with previous descriptions of hcg-induced PR expression in preovulatory follicles (41). In contrast, the multiple preovulatory follicles present in immature ERKO ovaries collected 4 h after hcg exposure exhibited minimal PR immunoreactivity (Fig. 7C). Because expression of the Adamts1 gene is reported to depend on increased PR functions in the preovulatory follicle (36), we were interested in determining whether expression of this gene in ERKO preovulatory follicles was altered as well. Interestingly, ERKO ovaries exhibited a normal initial rise in Adamts1 mrna levels after hcg exposure but failed to exhibit any further increase thereafter vs. a more than 4-fold rise observed in control ovaries 10 h after hcg (Fig. 7D). Recent studies have indicated a bimodal mechanism of Adamts1 induction after hcg exposure or the LH surge such that the acute increase in expression (within 4 h) is LH dependent, but the more dramatic increase that provides for peak expression at 10 h after hcg is PR dependent (36). These data support our findings that PR functions in ERKO ovaries after hcg treatment are insufficient. PR expression is reduced in PMSG-treated ER -null ovaries To date, most investigations of ovarian PR expression during folliculogenesis have focused on the dramatic rise that occurs in granulosa cells shortly after the LH surge or hcg treatment. However, we observed a small but measurable (2-fold) increase in Pgr mrna levels in control ovaries 48 h after PMSG treatment (Fig. 8A). Furthermore, Western blot analyses indicated that both PR-A and PR-B were detectable in whole ovarian homogenates after PMSG treatment (Fig. 8B). However, ERKO ovaries exhibited reduced basal Pgr mrna levels relative to control ovaries and no change after PMSG treatment when assessed by SQ-RT-PCR (Fig. 8A). Western blot analysis confirmed that PR-A and PR-B were barely detectable before and after PMSG treatment in ERKO

11 Couse et al. Superovulation of ER-Null Mice Endocrinology, August 2005, 146(8): on gonadotropin-induced ovulation and the concomitant changes in gene expression, although some abnormalities were revealed. Therefore, we conclude that ER is more important in mediating the intraovarian actions of estradiol that are critical to maximizing the follicular response to gonadotropins. FIG. 8. ERKO ovaries exhibit reduced Pgr expression 48 h after PMSG treatment. Immature mice were treated with PMSG (3.25 IU) followed 48 h later with hcg (2.2 IU). Animals were killed at the indicated time points over the course of gonadotropin-induced ovulation and whole ovaries collected for RNA and protein extraction. A, Quantitative data from SQ-RT-PCR for Pgr gene expression showing a 2-fold induction after PMSG treatment in control ovaries but lower basal levels and no increase in similarly treated ERKO ovaries. Data are expressed as the average fold control-none (untreated) SEM. B, Western blot analysis for PR protein in control, ERKO, and ERKO ovaries 48 h after PMSG treatment illustrating that PR-A and PR-B are undetectable in ERKO ovaries but clearly present in control and ERKO ovaries after PMSG treatment. A nonspecific (n.s.) immunoreactive band migrates just above the PR-B band and appears at equal amounts in all samples, thereby acting as an internal loading control. Ut, Wild-type mouse uterus to serve as a positive control for PR. a, P 0.05 vs. control at the same time point. ovaries, whereas both forms were easily observed in preparations of PMSG-treated control ovaries (Fig. 8B). ERKO ovaries exhibited slightly higher basal levels of Pgr mrna and minimal induction after PMSG treatment, but levels of PR protein in ERKO ovaries were comparable with control before and after PMSG treatment (Fig. 8B). Due to high background, it was difficult to localize the basal and PMSGinduced PR expression by immunohistochemistry in ovaries from any one of the three genotypes (data not shown). Still, these data are the first to demonstrate that constitutive PR expression in the murine ovary is ER dependent. Discussion The primary cause of reduced fecundity in ER -null female mice is infrequent and inefficient ovulation (13, 42). In the current study, we provide evidence that the ovulatory phenotype in ERKO females is due to the loss of intraovarian ER functions that facilitate gonadotropin-induced differentiation of preovulatory follicles. Two classic indices of preovulatory granulosa cell differentiation, increased Cyp19 expression and aromatase activity and acquired Lhcgr expression and LH responsiveness, were markedly deficient in ERKO ovaries after PMSG treatment. As a result, the response of PMSG-primed ERKO ovaries to an ovulatory dose of hcg was severely attenuated, as evident by a reduced rate of ovulation, insufficient induction of Ptgs2 and PR functions, an aberrant increase in Cyp19 expression and plasma estradiol, and incomplete expansion of the COC. Our parallel characterization of ER -null females indicates that loss of intraovarian ER function had minimal impact ER -null follicles fail to differentiate after PMSG treatment FSH is undoubtedly the primary stimulus for differentiation of follicles from the preantral to preovulatory stage. Female mice null for FSH signaling due to the loss of hormone (6, 43) or receptor (7, 44) exhibit follicles that fail to differentiate beyond the preantral stage and produce minimal estradiol and hence are infertile. However, FSH alone does not provide for complete follicle and granulosa cell differentiation in rodents. Instead, the synergistic actions of estradiol are required to maximize FSH induction of antrum formation (27), Cyp19 expression and aromatase activity (20 22), and LH responsiveness (25, 26, 45). Because ER is the more prominent ER form present in rodent granulosa cells (16 18) and exhibits an expression pattern similar to FSH receptor, it is the most plausible candidate to mediate the cooperative actions of estradiol with FSH. Our thorough analysis of induced ovulation in immature ERKO ovaries supports this role of ER in amplifying the granulosa cell response to FSH. Substantial Cyp19 expression and estradiol synthesis is a hallmark of healthy preovulatory follicles in the mammalian ovary (46). FSH stimulation of the Cyp19 gene is augmented by estradiol (20 22) and several findings suggest this to be an ER -mediated effect, including the highly correlative expression pattern of ER and Cyp19 in granulosa cells (47 49); the preservation of synergism in isolated granulosa cells, ruling out a paracrine type mechanism; and the inhibition of FSH-induced aromatase activity in granulosa cells by ER antagonists (50). Our data provide the most definitive evidence to date that ER mediates this effect of estradiol and is required for maximum FSH stimulation of aromatase activity by demonstrating that PMSG-stimulated ERKO ovaries and granulosa cells exhibit reduced Cyp19 expression and that ERKO females fail to present the expected preovulatory rise in plasma estradiol. This conclusion is further supported by our recent findings that individual ERKO follicles maintained in vitro secrete significantly less estradiol relative to wild-type follicles when stimulated with FSH (51). Furthermore, ERKO ovaries (Fig. 4) (15) and individual ERKO follicles (51) exhibit elevated Cyp19 expression and estradiol synthesis, excluding a direct role for ER. Direct estradiol/er regulation of the Cyp19 gene is unlikely because there are no known EREs in the human or rat Cyp19 promoter II and estradiol alone has no stimulatory effect (20). In turn, FSH stimulation of aromatase activity in granulosa cells involves the binding of camp/protein kinase A (PKA)-activated camp response element binding protein (CREB) and steroidogenic factor (SF)-1 to their cognate response elements within the Cyp19 promoter II (19, 52 54). LRH-1, which may substitute for SF-1 in granulosa cells (55), and GATA-4 (56) have also been postulated to be involved. We found that SF-1 (Nr5a1), LRH-1 (Nr5a2) and Gata4 expression were not overtly altered in immature ERKO ovaries during gonadotropin-induced ovu-

REPRODUCTIVE CYCLE OF FEMALE MAMMAL

REPRODUCTIVE CYCLE OF FEMALE MAMMAL REPRODUCTIVE CYCLE OF FEMALE MAMMAL Fig. 8-12 Secondary follicles growing follicles increase in number of layers of granulosa cells Tertiary follicles maturing follicles antrum formation fluid filled space

More information

OVARY The surface of the ovary is covered with surface epithelium

OVARY The surface of the ovary is covered with surface epithelium OVARY Cow The ovary, or female gonad, is: 1. an exocrine gland, producing oocytes 2. an endocrine gland, secreting hormones, i.e., estrogen and progesterone OVARY OVARY The surface of the ovary is covered

More information

The intra-follicular molecular biology mandating advancement of egg retrieval in some women

The intra-follicular molecular biology mandating advancement of egg retrieval in some women The intra-follicular molecular biology mandating advancement of egg retrieval in some women David H. Barad, USA Director of Assisted Reproductive Technology, The Center for Human Reproduction New York

More information

CASE 41. What is the pathophysiologic cause of her amenorrhea? Which cells in the ovary secrete estrogen?

CASE 41. What is the pathophysiologic cause of her amenorrhea? Which cells in the ovary secrete estrogen? CASE 41 A 19-year-old woman presents to her gynecologist with complaints of not having had a period for 6 months. She reports having normal periods since menarche at age 12. She denies sexual activity,

More information

LH (Bovine) ELISA Kit

LH (Bovine) ELISA Kit LH (Bovine) ELISA Kit Catalog Number KA2280 96 assays Version: 05 Intended for research use only www.abnova.com Table of Contents Introduction... 3 Intended Use... 3 Background... 3 Principle of the Assay...

More information

Raoul Orvieto. The Chaim Sheba Medical Center Tel Hashomer, Israel. Declared no potential conflict of interest

Raoul Orvieto. The Chaim Sheba Medical Center Tel Hashomer, Israel. Declared no potential conflict of interest Raoul Orvieto The Chaim Sheba Medical Center Tel Hashomer, Israel Declared no potential conflict of interest LH in antagonist cycles; is the story really written? Raoul Orvieto M.D. Israel Overview Role

More information

Chapter 27 The Reproductive System. MDufilho

Chapter 27 The Reproductive System. MDufilho Chapter 27 The Reproductive System 1 Figure 27.19 Events of oogenesis. Before birth Meiotic events 2n Oogonium (stem cell) Mitosis Follicle development in ovary Follicle cells Oocyte 2n Primary oocyte

More information

Superovulation of Beef Heifers with Follicle Stimulating Hormone or Human Menopausal Gonadotropin: Acute Effects on Hormone Secretion

Superovulation of Beef Heifers with Follicle Stimulating Hormone or Human Menopausal Gonadotropin: Acute Effects on Hormone Secretion Superovulation of Beef Heifers with Follicle Stimulating Hormone or Human Menopausal Gonadotropin: Acute Effects on Hormone Secretion A.S. Leaflet R1362 Acacia A. Alcivar, graduate research assistant,

More information

Expression of acid base transporters in the kidney collecting duct in Slc2a7 -/-

Expression of acid base transporters in the kidney collecting duct in Slc2a7 -/- Supplemental Material Results. Expression of acid base transporters in the kidney collecting duct in Slc2a7 -/- and Slc2a7 -/- mice. The expression of AE1 in the kidney was examined in Slc26a7 KO mice.

More information

Female Reproductive Physiology. Dr Raelia Lew CREI, FRANZCOG, PhD, MMed, MBBS Fertility Specialist, Melbourne IVF

Female Reproductive Physiology. Dr Raelia Lew CREI, FRANZCOG, PhD, MMed, MBBS Fertility Specialist, Melbourne IVF Female Reproductive Physiology Dr Raelia Lew CREI, FRANZCOG, PhD, MMed, MBBS Fertility Specialist, Melbourne IVF REFERENCE Lew, R, Natural History of ovarian function including assessment of ovarian reserve

More information

Milder is better? Advantages and disadvantages of "mild" ovarian stimulation for human in vitro fertilization

Milder is better? Advantages and disadvantages of mild ovarian stimulation for human in vitro fertilization Milder is better? Advantages and disadvantages of "mild" ovarian stimulation for human in vitro fertilization Revelli et al. Reproductive Biology and Endocrinology 2011, 9:25 Presenter: R2 孫怡虹 Background

More information

Superovulation of Beef Heifers with Follicle Stimulating Hormone or Human Menopausal Gonadotropin: Acute Effects on Hormone Secretion

Superovulation of Beef Heifers with Follicle Stimulating Hormone or Human Menopausal Gonadotropin: Acute Effects on Hormone Secretion Beef Research Report, 1996 Animal Science Research Reports 1997 Superovulation of Beef Heifers with Follicle Stimulating Hormone or Human Menopausal Gonadotropin: Acute Effects on Hormone Secretion Acacia

More information

Islet viability assay and Glucose Stimulated Insulin Secretion assay RT-PCR and Western Blot

Islet viability assay and Glucose Stimulated Insulin Secretion assay RT-PCR and Western Blot Islet viability assay and Glucose Stimulated Insulin Secretion assay Islet cell viability was determined by colorimetric (3-(4,5-dimethylthiazol-2-yl)-2,5- diphenyltetrazolium bromide assay using CellTiter

More information

ABSTRACT. Key words: ovulation, ovary, human, follicle, collagen, MMP and TIMP. ISBN-10: ISBN-13:

ABSTRACT. Key words: ovulation, ovary, human, follicle, collagen, MMP and TIMP. ISBN-10: ISBN-13: HUMAN OVULATION Studies on collagens, gelatinases and tissue inhibitors of metalloproteinases Anna Karin Lind Department of Obstetrics and Gynecology Institute of Clinical Sciences Sahlgrenska University

More information

SUPPLEMENTARY MATERIAL

SUPPLEMENTARY MATERIAL SUPPLEMENTARY MATERIAL Table S1. Primers and fluorescent probes used for qrt-pcr analysis of relative expression levels of PPP family phosphatases. gene name forward primer, 5-3 probe, 5-3 reverse primer,

More information

Ovarian Characteristics, Serum Hormone Concentrations, and Fertility in Lactating Dairy Cows in Response to Equine Chorionic Gonadotropin

Ovarian Characteristics, Serum Hormone Concentrations, and Fertility in Lactating Dairy Cows in Response to Equine Chorionic Gonadotropin Ovarian Characteristics, Serum Hormone Concentrations, and Fertility in Lactating Dairy Cows in Response to quine Chorionic Gonadotropin S. L. Pulley, L. D. Wallace, H. I. Mellieon, and J. S. Stevenson

More information

CLARITY reveals dynamics of ovarian follicular architecture and vasculature in three-dimensions

CLARITY reveals dynamics of ovarian follicular architecture and vasculature in three-dimensions CLARITY reveals dynamics of ovarian follicular architecture and vasculature in three-dimensions Yi Feng, Peng Cui, Xiaowei Lu, Brian Hsueh, Fredrik Möller Billig, Livia Zarnescu Yanez, Raju Tomer, Derek

More information

different ratios of PMSG and HCG on the occurrence of follicular haemorrhage THE induction of ovulation with PMSG and HCG in the rat has been studied

different ratios of PMSG and HCG on the occurrence of follicular haemorrhage THE induction of ovulation with PMSG and HCG in the rat has been studied Q. Jl exp. Physiol. (1968) 53, 129-135 THE INDUCTION OF OVULATION IN IMMATURE RATS TREATED WITH PREGNANT MARE'S SERUM GONADOTROPHIN AND HUMAN CHORIONIC GONADOTROPHIN. By S. F. LUNN and E. T. BELL. From

More information

Endocrinology of the Female Reproductive Axis

Endocrinology of the Female Reproductive Axis Endocrinology of the Female Reproductive Axis girlontheriver.com Geralyn Lambert-Messerlian, PhD, FACB Professor Women and Infants Hospital Alpert Medical School at Brown University Women & Infants BROWN

More information

GENERAL SUMMARY Corpus luteum is a transient endocrine structure formed from the ruptured ovarian follicle. Its main function is to secrete P 4, a pro

GENERAL SUMMARY Corpus luteum is a transient endocrine structure formed from the ruptured ovarian follicle. Its main function is to secrete P 4, a pro Corpus luteum is a transient endocrine structure formed from the ruptured ovarian follicle. Its main function is to secrete P 4, a pro-gestational hormone, essential for establishment and maintenance of

More information

Two important cells in female are the theca cells and the granulose cells. Granulosa cells are affected by the two gonadotropin hormones; FSH and LH.

Two important cells in female are the theca cells and the granulose cells. Granulosa cells are affected by the two gonadotropin hormones; FSH and LH. 1 UGS physiology sheet #13 lecture 3 Dr.Saleem Khresha. Now we will start discussing the female reproductive system Ovarian Steroids Two important cells in female are the theca cells and the granulose

More information

Chapter 14 Reproduction Review Assignment

Chapter 14 Reproduction Review Assignment Date: Mark: _/45 Chapter 14 Reproduction Review Assignment Multiple Choice Identify the choice that best completes the statement or answers the question. 1. Use the diagram above to answer the next question.

More information

Endocrine control of female reproductive function

Endocrine control of female reproductive function Medicine School of Women s & Children s Health Discipline of Obstetrics & Gynaecology Endocrine control of female reproductive function Kirsty Walters, PhD Fertility Research Centre, School of Women s

More information

(A) PCR primers (arrows) designed to distinguish wild type (P1+P2), targeted (P1+P2) and excised (P1+P3)14-

(A) PCR primers (arrows) designed to distinguish wild type (P1+P2), targeted (P1+P2) and excised (P1+P3)14- 1 Supplemental Figure Legends Figure S1. Mammary tumors of ErbB2 KI mice with 14-3-3σ ablation have elevated ErbB2 transcript levels and cell proliferation (A) PCR primers (arrows) designed to distinguish

More information

Daily blood hormone levels related to the luteinizing hormone surge in anovulatory cycles

Daily blood hormone levels related to the luteinizing hormone surge in anovulatory cycles FRTILITY AND STRILITY Copyright 1983 The American Fertility Society Printed in U.8A. Daily blood hormone levels related to the luteinizing hormone surge in anovulatory cycles Chung H. Wu, M.D. * F. Susan

More information

Characterization of Luteinizing Hormone and Luteinizing Hormone Receptor and Their Indispensable Role in the Ovulatory Process of the Medaka

Characterization of Luteinizing Hormone and Luteinizing Hormone Receptor and Their Indispensable Role in the Ovulatory Process of the Medaka Characterization of Luteinizing Hormone and Luteinizing Hormone Receptor and Their Indispensable Role in the Ovulatory Process of the Medaka Katsueki Ogiwara 1, Chika Fujimori 1, Sanath Rajapakse 2, Takayuki

More information

THE MENSTRUAL CYCLE INA S. IRABON, MD, FPOGS, FPSRM, FPSGE OBSTETRICS AND GYNECOLOGY REPRODUCTIVE ENDOCRINOLOGY AND INFERTILITY

THE MENSTRUAL CYCLE INA S. IRABON, MD, FPOGS, FPSRM, FPSGE OBSTETRICS AND GYNECOLOGY REPRODUCTIVE ENDOCRINOLOGY AND INFERTILITY THE MENSTRUAL CYCLE INA S. IRABON, MD, FPOGS, FPSRM, FPSGE OBSTETRICS AND GYNECOLOGY REPRODUCTIVE ENDOCRINOLOGY AND INFERTILITY REFERENCE Comprehensive Gynecology 7 th edition, 2017 (Lobo RA, Gershenson

More information

REPRODUCCIÓN. La idea fija. Copyright 2004 Pearson Education, Inc., publishing as Benjamin Cummings

REPRODUCCIÓN. La idea fija. Copyright 2004 Pearson Education, Inc., publishing as Benjamin Cummings REPRODUCCIÓN La idea fija How male and female reproductive systems differentiate The reproductive organs and how they work How gametes are produced and fertilized Pregnancy, stages of development, birth

More information

MicroRNA sponges: competitive inhibitors of small RNAs in mammalian cells

MicroRNA sponges: competitive inhibitors of small RNAs in mammalian cells MicroRNA sponges: competitive inhibitors of small RNAs in mammalian cells Margaret S Ebert, Joel R Neilson & Phillip A Sharp Supplementary figures and text: Supplementary Figure 1. Effect of sponges on

More information

Animal Science 434! Tonic and Preovulatory Surge of GnRH! Tonic and Preovulatory Surge of GnRH! Lecture 11: The Follicular Phase of the Estrous Cycle!

Animal Science 434! Tonic and Preovulatory Surge of GnRH! Tonic and Preovulatory Surge of GnRH! Lecture 11: The Follicular Phase of the Estrous Cycle! Tonic and Preovulatory Surge of GnRH! Animal Science 434! Lecture 11: The Follicular Phase of the Estrous Cycle!! (-)! Hypothalamus! GnRH! Estradiol! (-)! Tonic and Preovulatory Surge of GnRH! Anterior!

More information

The reproductive lifespan

The reproductive lifespan The reproductive lifespan Reproductive potential Ovarian cycles Pregnancy Lactation Male Female Puberty Menopause Age Menstruation is an external indicator of ovarian events controlled by the hypothalamicpituitary

More information

emphasized both the need for an adequate amount of fsh and an adequate COMPARISON OF SUPEROVULATION IN THE IMMATURE MOUSE AND RAT

emphasized both the need for an adequate amount of fsh and an adequate COMPARISON OF SUPEROVULATION IN THE IMMATURE MOUSE AND RAT COMPARISON OF SUPEROVULATION IN THE IMMATURE MOUSE AND RAT EVERETT D. WILSON* and M. X. ZARROW Department of Biological Sciences, Purdue University, Lafayette, Indiana, U.S.A. (Received 26th May 1961)

More information

Obesity, Metabolic Hormone Signaling, and Granulosa Cell Gene Expression

Obesity, Metabolic Hormone Signaling, and Granulosa Cell Gene Expression University of Nebraska - Lincoln DigitalCommons@University of Nebraska - Lincoln Theses and Dissertations in Animal Science Animal Science Department 12-2010 Obesity, Metabolic Hormone Signaling, and Granulosa

More information

/06/$15.00/0 Molecular Endocrinology 20(9): Copyright 2006 by The Endocrine Society doi: /me

/06/$15.00/0 Molecular Endocrinology 20(9): Copyright 2006 by The Endocrine Society doi: /me 0888-8809/06/$15.00/0 Molecular Endocrinology 20(9):2062 2079 Printed in U.S.A. Copyright 2006 by The Endocrine Society doi: 10.1210/me.2005-0316 Androgens, Progestins, and Glucocorticoids Induce Follicle-Stimulating

More information

TFEB-mediated increase in peripheral lysosomes regulates. Store Operated Calcium Entry

TFEB-mediated increase in peripheral lysosomes regulates. Store Operated Calcium Entry TFEB-mediated increase in peripheral lysosomes regulates Store Operated Calcium Entry Luigi Sbano, Massimo Bonora, Saverio Marchi, Federica Baldassari, Diego L. Medina, Andrea Ballabio, Carlotta Giorgi

More information

10.7 The Reproductive Hormones

10.7 The Reproductive Hormones 10.7 The Reproductive Hormones December 10, 2013. Website survey?? QUESTION: Who is more complicated: men or women? The Female Reproductive System ovaries: produce gametes (eggs) produce estrogen (steroid

More information

LH (Rodent) ELISA Kit

LH (Rodent) ELISA Kit LH (Rodent) ELISA Kit Catalog Number KA2332 96 assays Version: 05 Intended for research use only www.abnova.com Table of Contents Introduction... 3 Intended Use... 3 Background... 3 Principle of the Assay...

More information

General Laboratory methods Plasma analysis: Gene Expression Analysis: Immunoblot analysis: Immunohistochemistry:

General Laboratory methods Plasma analysis: Gene Expression Analysis: Immunoblot analysis: Immunohistochemistry: General Laboratory methods Plasma analysis: Plasma insulin (Mercodia, Sweden), leptin (duoset, R&D Systems Europe, Abingdon, United Kingdom), IL-6, TNFα and adiponectin levels (Quantikine kits, R&D Systems

More information

GONADOTROPHIN (LUTEINISING)- RELEASING HORMONE AND ANALOGUES (GnRH OR LHRH)

GONADOTROPHIN (LUTEINISING)- RELEASING HORMONE AND ANALOGUES (GnRH OR LHRH) GONADOTROPHIN (LUTEINISING)- RELEASING HORMONE AND ANALOGUES (GnRH OR LHRH) Naturally occurring hormone, produced by the hypothalamus and transferred to the anterior pituitary gland in the hypophyseal

More information

Supplementary data Supplementary Figure 1 Supplementary Figure 2

Supplementary data Supplementary Figure 1 Supplementary Figure 2 Supplementary data Supplementary Figure 1 SPHK1 sirna increases RANKL-induced osteoclastogenesis in RAW264.7 cell culture. (A) RAW264.7 cells were transfected with oligocassettes containing SPHK1 sirna

More information

Ovarian follicular development in cattle

Ovarian follicular development in cattle Ovarian follicular development in cattle John P Kastelic Professor of Theriogenology Head, Department of Production Animal Health University of Calgary Calgary, Alberta, Canada Overview Prenatal development

More information

SISTEMA REPRODUCTOR (LA IDEA FIJA) Copyright 2004 Pearson Education, Inc., publishing as Benjamin Cummings

SISTEMA REPRODUCTOR (LA IDEA FIJA) Copyright 2004 Pearson Education, Inc., publishing as Benjamin Cummings SISTEMA REPRODUCTOR (LA IDEA FIJA) How male and female reproductive systems differentiate The reproductive organs and how they work How gametes are produced and fertilized Pregnancy, stages of development,

More information

Ovarian Follicular Development in the Untreated and

Ovarian Follicular Development in the Untreated and Ovarian Follicular Development in the Untreated and PMSG-treated Cyclic Rat Hajime MIYAMOTO, Goro KATSUURA and Takehiko ISHIBASHI Department of Animal Science, College of Agriculture, Kyoto University,

More information

MICROWELL ELISA LUTEINIZING HORMONE (LH) ENZYMEIMMUNOASSAY TEST KIT LH ELISA. Cat # 4225Z

MICROWELL ELISA LUTEINIZING HORMONE (LH) ENZYMEIMMUNOASSAY TEST KIT LH ELISA. Cat # 4225Z DIAGNOSTIC AUTOMATION, INC. 23961 Craftsman Road, Suite D/E/F, Calabasas, CA 91302 Tel: (818) 591-3030 Fax: (818) 591-8383 onestep@rapidtest.com technicalsupport@rapidtest.com www.rapidtest.com See external

More information

Development Supplementary information. Supplementary Figures * * +/+ +/- -/- +/+ +/- -/-

Development Supplementary information. Supplementary Figures * * +/+ +/- -/- +/+ +/- -/- Development 144: doi:1.1242/dev.1473: Supplementary information Supplementary Figures A (f) FRT LoxP 2 3 4 B All Males Females I Ovary 1 (+) 77 bps (f) 78 bps (-) >13 bps (-) 2 4 (-) 424 bps M +/f +/-

More information

Supplementary Figure 1: Neuregulin 1 increases the growth of mammary organoids compared to EGF. (a) Mammary epithelial cells were freshly isolated,

Supplementary Figure 1: Neuregulin 1 increases the growth of mammary organoids compared to EGF. (a) Mammary epithelial cells were freshly isolated, 1 2 3 4 5 6 7 8 9 10 Supplementary Figure 1: Neuregulin 1 increases the growth of mammary organoids compared to EGF. (a) Mammary epithelial cells were freshly isolated, embedded in matrigel and exposed

More information

Neil Goodman, MD, FACE

Neil Goodman, MD, FACE Initial Workup of Infertile Couple: Female Neil Goodman, MD, FACE Professor of Medicine Voluntary Faculty University of Miami Miller School of Medicine Scope of Infertility in the United States Affects

More information

Abstracts for the KSAR and JSAR Joint Symposium. Fertility control in female domestic animals: From basic understanding to application

Abstracts for the KSAR and JSAR Joint Symposium. Fertility control in female domestic animals: From basic understanding to application Abstracts for the KSAR and JSAR Joint Symposium Fertility control in female domestic animals: From basic understanding to application Current Research Orientation in Livestock Reproduction in Korea Choong-Saeng

More information

Interval between PMSG Priming and hcg Injection in Superovulation of the Mongolian Gerbil

Interval between PMSG Priming and hcg Injection in Superovulation of the Mongolian Gerbil J. Mamm. Ova Res. Vol. 21, 105 109, 2004 105 Original Interval between PMSG Priming and hcg Injection in Superovulation of the Mongolian Gerbil Yuichi Kameyama 1 *, Kaori Arai 1 and Yoshiro Ishijima 1

More information

Is it the seed or the soil? Arthur Leader, MD, FRCSC

Is it the seed or the soil? Arthur Leader, MD, FRCSC The Physiological Limits of Ovarian Stimulation Is it the seed or the soil? Arthur Leader, MD, FRCSC Objectives 1. To consider how ovarian stimulation protocols work in IVF 2. To review the key events

More information

Timing of Ovulation Induction Matters in Women with Low Functional Ovarian Reserve

Timing of Ovulation Induction Matters in Women with Low Functional Ovarian Reserve Timing of Ovulation Induction Matters in Women with Low Functional Ovarian Reserve Norbert Gleicher, MD Medical Director And C hief Scientist, C enter For Human Reproduction, New York, NY President, Foundation

More information

Abraxis Progesterone (bovine) ELISA Kit

Abraxis Progesterone (bovine) ELISA Kit Abraxis Progesterone (bovine) ELISA Kit Enzyme immunoassay for the quantitative determination of progesterone in bovine milk/serum/plasma samples PN5081M 96 Tests For Research Use Only. Not for use in

More information

In Vitro Fertilization in Clomiphene-Resistant Women with Polycystic Ovary Syndrome

In Vitro Fertilization in Clomiphene-Resistant Women with Polycystic Ovary Syndrome Original Article Effect of Laparoscopic Ovarian Drilling on Outcomes of In Vitro Fertilization in Clomiphene-Resistant Women with Polycystic Ovary Syndrome Maryam Eftekhar, M.D. 1, Razieh Deghani Firoozabadi,

More information

Impact of High-Fat Environment on Ovarian Androgen Synthesis in Rats and the Associated Pathophysiological Changes

Impact of High-Fat Environment on Ovarian Androgen Synthesis in Rats and the Associated Pathophysiological Changes Research in Obstetrics and Gynecology 2015, 3(1): 8-12 DOI: 10.5923/j.rog.20150301.03 Impact of High-Fat Environment on Ovarian Androgen Synthesis in Rats and the Associated Pathophysiological Changes

More information

REPRODUCTION & GENETICS. Hormones

REPRODUCTION & GENETICS. Hormones REPRODUCTION & GENETICS Hormones http://www.youtube.com/watch?v=np0wfu_mgzo Objectives 2 Define what hormones are; Compare and contrast the male and female hormones; Explain what each hormone in the mail

More information

Soy phytoestrogen effects on progesterone receptor and ovalbumin synthesis in the chick oviduct.

Soy phytoestrogen effects on progesterone receptor and ovalbumin synthesis in the chick oviduct. Soy phytoestrogen effects on progesterone receptor and ovalbumin synthesis in the chick oviduct. L.M. STEVENSON*, S.H. OATES, A.L. DOERNTE, J.B. HESS and W.D. BERRY Poultry Science Department, Auburn University,

More information

Gene expression of follicle-stimulating hormone receptors in granulosa cells in poor ovarian responders

Gene expression of follicle-stimulating hormone receptors in granulosa cells in poor ovarian responders Journal of Physics: Conference Series PAPER OPEN ACCESS Gene expression of follicle-stimulating hormone receptors in granulosa cells in poor ovarian responders To cite this article: P A Iffanolida et al

More information

FREE-ROAMING HORSE AND BURRO FERTILITY CONTROL WORKSHOP Albuquerque, NM November 8, 2018

FREE-ROAMING HORSE AND BURRO FERTILITY CONTROL WORKSHOP Albuquerque, NM November 8, 2018 FREE-ROAMING HORSE AND BURRO FERTILITY CONTROL WORKSHOP Albuquerque, NM November 8, 2018 Current Contraceptive Use pzp GonaCon Porcine Zona Pellucida Antibodies to ZP3 Cons: Requires boosters Continuous

More information

Female Reproductive System. Lesson 10

Female Reproductive System. Lesson 10 Female Reproductive System Lesson 10 Learning Goals 1. What are the five hormones involved in the female reproductive system? 2. Understand the four phases of the menstrual cycle. Human Reproductive System

More information

Endocrinology laboratory Department of Zoology Kalyani University Kalyani, West Bengal India

Endocrinology laboratory Department of Zoology Kalyani University Kalyani, West Bengal India Epidermal growth factor (EGF) promotes ovarian steroidogenesis and epidermal growth factor receptor (EGFR) signaling is required for gonadotropin-induced steroid production in common carp Cyprinus carpio

More information

Reproductive Hormones

Reproductive Hormones Reproductive Hormones Male gonads: testes produce male sex cells! sperm Female gonads: ovaries produce female sex cells! ovum The union of male and female sex cells during fertilization produces a zygote

More information

Investigation: The Human Menstrual Cycle Research Question: How do hormones control the menstrual cycle?

Investigation: The Human Menstrual Cycle Research Question: How do hormones control the menstrual cycle? Investigation: The Human Menstrual Cycle Research Question: How do hormones control the menstrual cycle? Introduction: The menstrual cycle (changes within the uterus) is an approximately 28-day cycle that

More information

A COH protocol with human recombinant FSH (Gonal-F, Merck Serono S.A., Madrid,

A COH protocol with human recombinant FSH (Gonal-F, Merck Serono S.A., Madrid, Supplemental Methods Detailed protocol for controlled ovarian stimulation A COH protocol with human recombinant FSH (Gonal-F, Merck Serono S.A., Madrid, Spain) was initiated on menstrual cycle day 2 at

More information

BMP6 treatment compensates for the molecular defect and ameliorates hemochromatosis in Hfe knockout mice

BMP6 treatment compensates for the molecular defect and ameliorates hemochromatosis in Hfe knockout mice SUPPLEMENTARY MATERIALS BMP6 treatment compensates for the molecular defect and ameliorates hemochromatosis in Hfe knockout mice Elena Corradini, Paul J. Schmidt, Delphine Meynard, Cinzia Garuti, Giuliana

More information

Human Immunodeficiency Virus type 1 (HIV-1) p24 / Capsid Protein p24 ELISA Pair Set

Human Immunodeficiency Virus type 1 (HIV-1) p24 / Capsid Protein p24 ELISA Pair Set Human Immunodeficiency Virus type 1 (HIV-1) p24 / Capsid Protein p24 ELISA Pair Set Catalog Number : SEK11695 To achieve the best assay results, this manual must be read carefully before using this product

More information

LH (Canine) ELISA Kit

LH (Canine) ELISA Kit LH (Canine) ELISA Kit Catalog Number KA2292 96 assays Version: 05 Intended for research use only www.abnova.com Table of Contents Introduction... 3 Intended Use... 3 Background... 3 Principle of the Assay...

More information

Reproduction. AMH Anti-Müllerian Hormone. Analyte Information

Reproduction. AMH Anti-Müllerian Hormone. Analyte Information Reproduction AMH Anti-Müllerian Hormone Analyte Information - 1-2011-01-11 AMH Anti-Müllerian Hormone Introduction Anti-Müllerian Hormone (AMH) is a glycoprotein dimer composed of two 72 kda monomers 1.

More information

FSH (Rodent) ELISA Kit

FSH (Rodent) ELISA Kit FSH (Rodent) ELISA Kit Catalog Number KA2330 96 assays Version: 06 Intended for research use only www.abnova.com Table of Contents Introduction... 3 Intended Use... 3 Background... 3 Principle of the Assay...

More information

Animal and Veterinary Science Department University of Idaho. REGULATION OF REPRODUCTION AVS 222 (Instructor: Dr. Amin Ahmadzadeh) Chapter 5

Animal and Veterinary Science Department University of Idaho. REGULATION OF REPRODUCTION AVS 222 (Instructor: Dr. Amin Ahmadzadeh) Chapter 5 Animal and Veterinary Science Department University of Idaho REGULATION OF REPRODUCTION AVS 222 (Instructor: Dr. Amin Ahmadzadeh) Chapter 5 I. DEFINITIONS A. Endocrine Gland B. Hormone Chemical messenger

More information

A Tale of Three Hormones: hcg, Progesterone and AMH

A Tale of Three Hormones: hcg, Progesterone and AMH A Tale of Three Hormones: hcg, Progesterone and AMH Download the Ferring AR ipad/iphone app from the Apple Store: http://bit.ly/1okk74m Human Ovarian Steroidogenesis and Gonadotrophin Stimulation Johan

More information

LH activity administration during the

LH activity administration during the LH activity administration during the luteal-follicular transition Richard Fleming On behalf of the Luveris Pre-treatment group University of Glasgow Scotland Androgens have a Paracrine action in the Early

More information

Fish follicle-stimulating hormone, FSH ELISA Kit

Fish follicle-stimulating hormone, FSH ELISA Kit Fish follicle-stimulating hormone, FSH ELISA Kit Catalog No: E0830f 96 Tests Operating instruction www.eiaab.com FOR RESEARCH USE ONLY; NOT FOR THERAPEUTIC OR DIAGNOSTIC APPLICATIONS! PLEASE READ THROUGH

More information

CARD HyperOva (Superovulation Reagent for mouse)

CARD HyperOva (Superovulation Reagent for mouse) Product manual (Superovulation Reagent for mouse) Cat. No. KYD-010-EX -X5 Size: 5 1 ML Origin Serum of goat, Horse-derived villus gonatropin. Composition 1. Inhibin antiserum (Goat). 2. Equine chorionic

More information

SCHOOL OF MEDICINE AND HEALTH SCIENCES DIVISION OF BASIC MEDICAL SCIENCES DISCIPLINE OF BIOCHEMISTRY & MOLECULAR BIOLOGY

SCHOOL OF MEDICINE AND HEALTH SCIENCES DIVISION OF BASIC MEDICAL SCIENCES DISCIPLINE OF BIOCHEMISTRY & MOLECULAR BIOLOGY 1 SCHOOL OF MEDICINE AND HEALTH SCIENCES DIVISION OF BASIC MEDICAL SCIENCES DISCIPLINE OF BIOCHEMISTRY & MOLECULAR BIOLOGY PBL SEMINAR: SEX HORMONES PART 1 An Overview What are steroid hormones? Steroid

More information

Reproductive Health and Pituitary Disease

Reproductive Health and Pituitary Disease Reproductive Health and Pituitary Disease Janet F. McLaren, MD Assistant Professor Division of Reproductive Endocrinology and Infertility Department of Obstetrics and Gynecology jmclaren@uabmc.edu Objectives

More information

Supporting Information

Supporting Information Supporting Information Pang et al. 10.1073/pnas.1322009111 SI Materials and Methods ELISAs. These assays were performed as previously described (1). ELISA plates (MaxiSorp Nunc; Thermo Fisher Scientific)

More information

Biology of Reproduction- Zool 346 Exam 2

Biology of Reproduction- Zool 346 Exam 2 Biology of Reproduction- Zool 346 Exam 2 ANSWER ALL THE QUESTIONS ON THE ANSWER SHEET. THE ANSWER ON THE ANSWER SHEET IS YOUR OFFICIAL ANSWER. Some critical words are boldfaced. This exam is 7 pages long.

More information

ENDOCRINE CHARACTERISTICS OF ART CYCLES

ENDOCRINE CHARACTERISTICS OF ART CYCLES ENDOCRINE CHARACTERISTICS OF ART CYCLES DOÇ. DR. SEBİHA ÖZDEMİR ÖZKAN KOCAELI UNIVERSITY, SCHOOL OF MEDICINE, DEPARTMENT OF OBSTETRICS AND GYNECOLOGY, IVF UNIT 30.04.2014, ANTALYA INTRODUCTION The endocrine

More information

Effect of Resistin on Granulosa and Theca Cell Function in Cattle

Effect of Resistin on Granulosa and Theca Cell Function in Cattle 1 Effect of Resistin on Granulosa and Theca Cell Function in Cattle D.V. Lagaly, P.Y. Aad, L.B. Hulsey, J.A. Grado-Ahuir and L.J. Spicer Story in Brief Resistin is an adipokine that has not been extensively

More information

A comparison of the effects of estrus cow. nuclear maturation of bovine oocytes

A comparison of the effects of estrus cow. nuclear maturation of bovine oocytes A comparison of the effects of estrus cow serum and fetal calf serum on in vitro nuclear maturation of bovine oocytes J Spiropoulos, SE Long University of Bristol, School of Veterinary Science, Department

More information

SUPPLEMENTARY INFORMATION

SUPPLEMENTARY INFORMATION SUPPLEMENTARY INFORMATION FOR Liver X Receptor α mediates hepatic triglyceride accumulation through upregulation of G0/G1 Switch Gene 2 (G0S2) expression I: SUPPLEMENTARY METHODS II: SUPPLEMENTARY FIGURES

More information

Central Progesterone Involvement in Estrogen- Induced Prolactin and Luteinizing Hormone Secretion Surges in Female Rats

Central Progesterone Involvement in Estrogen- Induced Prolactin and Luteinizing Hormone Secretion Surges in Female Rats Southern Illinois University Carbondale OpenSIUC Honors Theses University Honors Program 5-10-2014 Central Progesterone Involvement in Estrogen- Induced Prolactin and Luteinizing Hormone Secretion Surges

More information

Concept : effects by treatment

Concept : effects by treatment QuickTime and a TIFF (LZW) decompressor are needed to see this picture. ESHRE CAMPUS 2010 MARIBOR INFLUENCE of stimulation on oocyte quality? Johan Smitz, MD, PhD UZBrussel Vrije Universiteit Brussel Brussels,

More information

Supplementary Materials and Methods

Supplementary Materials and Methods Supplementary Materials and Methods Whole Mount X-Gal Staining Whole tissues were collected, rinsed with PBS and fixed with 4% PFA. Tissues were then rinsed in rinse buffer (100 mm Sodium Phosphate ph

More information

Supplementary Material

Supplementary Material 10.1071/RD13007_AC CSIRO 2014 Supplementary Material: Reproduction, Fertility and Development, 2014, 26(2), 337-345. Supplementary Material Table S1. Details of primers used for quantitative reverse transcription-polymerase

More information

Endocrine Glands. Endocrine glands

Endocrine Glands. Endocrine glands ENDOCRINOLOGY Endocrine Glands Endocrine glands Produce substances called hormones. Ductless glands, i.e., they release hormones directly into the bloodstream Hormones only act at their target tissue where

More information

Reproductive Endocrinology. Isabel Hwang Department of Physiology Faculty of Medicine University of Hong Kong Hong Kong May2007

Reproductive Endocrinology. Isabel Hwang Department of Physiology Faculty of Medicine University of Hong Kong Hong Kong May2007 Reproductive Endocrinology Isabel Hwang Department of Physiology Faculty of Medicine University of Hong Kong Hong Kong May2007 isabelss@hkucc.hku.hk A 3-hormone chain of command controls reproduction with

More information

Hormonal Control of Human Reproduction

Hormonal Control of Human Reproduction Hormonal Control of Human Reproduction Bởi: OpenStaxCollege The human male and female reproductive cycles are controlled by the interaction of hormones from the hypothalamus and anterior pituitary with

More information

1. During the follicular phase of the ovarian cycle, the hypothalamus releases GnRH.

1. During the follicular phase of the ovarian cycle, the hypothalamus releases GnRH. 1. During the follicular phase of the ovarian cycle, the hypothalamus releases GnRH. 2. This causes the anterior pituitary to secrete small quantities of FSH and LH. 3. At this time, the follicles in the

More information

Supplementary Figure 1: Hsp60 / IEC mice are embryonically lethal (A) Light microscopic pictures show mouse embryos at developmental stage E12.

Supplementary Figure 1: Hsp60 / IEC mice are embryonically lethal (A) Light microscopic pictures show mouse embryos at developmental stage E12. Supplementary Figure 1: Hsp60 / IEC mice are embryonically lethal (A) Light microscopic pictures show mouse embryos at developmental stage E12.5 and E13.5 prepared from uteri of dams and subsequently genotyped.

More information

9.4 Regulating the Reproductive System

9.4 Regulating the Reproductive System 9.4 Regulating the Reproductive System The Reproductive System to unite a single reproductive cell from a female with a single reproductive cell from a male Both male and female reproductive systems include

More information

ABSTRACT. The purpose of this research was to investigate the role of TGF-β in the luteinizing

ABSTRACT. The purpose of this research was to investigate the role of TGF-β in the luteinizing ABSTRACT SRIPERUMBUDUR, RAJAGOPAL. The Role of TGF-β in Luteinization in the Pig. (Under the direction of Dr. John E. Gadsby). The purpose of this research was to investigate the role of TGF-β in the luteinizing

More information

Analysis of small RNAs from Drosophila Schneider cells using the Small RNA assay on the Agilent 2100 bioanalyzer. Application Note

Analysis of small RNAs from Drosophila Schneider cells using the Small RNA assay on the Agilent 2100 bioanalyzer. Application Note Analysis of small RNAs from Drosophila Schneider cells using the Small RNA assay on the Agilent 2100 bioanalyzer Application Note Odile Sismeiro, Jean-Yves Coppée, Christophe Antoniewski, and Hélène Thomassin

More information

Effects of Label-Dose Permethrin Administration on Reproductive Function and Embryo Quality on Superovulated Beef Heifers

Effects of Label-Dose Permethrin Administration on Reproductive Function and Embryo Quality on Superovulated Beef Heifers Animal Industry Report AS 662 ASL R3050 2016 Effects of Label-Dose Permethrin Administration on Reproductive Function and Embryo Quality on Superovulated Beef Heifers Tyler M. Dohlman Iowa State University,

More information

Mohammad. Renad zakaria ---

Mohammad. Renad zakaria --- 13 Mohammad Renad zakaria --- Before we start: - I didn t follow the record order, for organizing purposes. - I added extra information from our text box which is Guyton 12 th edition, pages 987-997, actually

More information

National Institute for Research in Reproductive Health (NIRRH), Mumbai

National Institute for Research in Reproductive Health (NIRRH), Mumbai National Institute for Research in Reproductive Health (NIRRH), Mumbai Fertility Assessment Kits Product/Process: Fertility assessment kits and their assaysa. Indulsa - a kit for estimation of estrone

More information

I. Endocrine System & Hormones Figure 1: Human Endocrine System

I. Endocrine System & Hormones Figure 1: Human Endocrine System I. Endocrine System & Hormones Figure 1: Human Endocrine System Endocrine System: a) Endocrine glands are ductless since they lack specific vessels for the transport of hormones throughout the body. Instead,

More information

The role of growth factors in regulating cellular events during ovarian follicular development Leon J. Spicer

The role of growth factors in regulating cellular events during ovarian follicular development Leon J. Spicer The role of growth factors in regulating cellular events during ovarian follicular development Leon J. Spicer Department of Animal Science, Oklahoma State University, Stillwater, OK USA SESSION #54 EAAP

More information

Effect of Bovine Follicular Fluid Added to the Maturation Medium on Sperm Penetration in Pig Oocytes Matured In Vitro

Effect of Bovine Follicular Fluid Added to the Maturation Medium on Sperm Penetration in Pig Oocytes Matured In Vitro Article Effect of Bovine Follicular Fluid Added to the Maturation Medium on Sperm Penetration in Pig Oocytes Matured In Vitro Abstract Naoki ISOBE Research Associate Graduate School for International Development

More information