Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2009 Nov 1.
Published in final edited form as: Neurotoxicology. 2008 Jul 6;29(6):988–997. doi: 10.1016/j.neuro.2008.06.008

DISRUPTED FEMALE REPRODUCTIVE PHYSIOLOGY FOLLOWING NEONATAL EXPOSURE TO PHYTOESTROGENS OR ESTROGEN SPECIFIC LIGANDS IS ASSOCIATED WITH DECREASED GNRH ACTIVATION AND KISSPEPTIN FIBER DENSITY IN THE HYPOTHALAMUS

Heather L Bateman 1, Heather B Patisaul 1
PMCID: PMC2647326  NIHMSID: NIHMS82313  PMID: 18656497

Abstract

It is well established that estrogen administration during neonatal development can advance pubertal onset and prevent the maintenance of regular estrous cycles in female rats. This treatment paradigm also eliminates the preovulatory rise of gonadotropin releasing hormone (GnRH). It remains unclear, however, through which of the two primary forms of the estrogen receptor (ERα or ERβ) this effect is mediated. It is also unclear whether endocrine disrupting compounds (EDCs) can produce similar effects. Here we compared the effect of neonatal exposure to estradiol benzoate (EB), the ERα specific agonist 1,3,5-tris(4-Hydroxyphenyl)-4-propyl-1H-pyrazole (PPT), the ERβ specific agonist diarylpropionitrile (DPN) and the naturally occurring EDCs genistein (GEN) and equol (EQ) on pubertal onset, estrous cyclicity, GnRH activation, and kisspeptin content in the anteroventral periventricular (AVPV) and arcuate (ARC) nuclei. Vaginal opening was significantly advanced by EB and GEN. By ten weeks post-puberty, irregular estrous cycles were observed in all groups except the control group. GnRH activation, as measured by the percentage of immunopositive GnRH neurons that were also immunopositive for Fos, was significantly lower in all treatment groups except the DPN group compared to the control group. GnRH activation was absent in the PPT group. These data suggest that neonatal exposure to EDCs can suppress GnRH activity in adulthood, and that ERα plays a pivotal role in this process. Kisspeptins (KISS) have recently been characterized to be potent stimulators of GnRH secretion. Therefore we quantified the density of KISS immunolabeled fibers in the AVPV and ARC. In the AVPV, KISS fiber density was significantly lower in the EB and GEN groups compared to the control group but only in the EB and PPT groups in the ARC. The data suggest that decreased stimulation of GnRH neurons by KISS could be a mechanism by which EDCs can impair female reproductive function.

Keywords: genistein, equol, soy, isoflavones, gonadotropin, puberty, development, endocrine disruption, DPN, PPT

1. Introduction

It has been hypothesized that developmental exposure to compounds with estrogen-like action may be a factor in the advancement of puberty and the decrease in human fecundity (Freni-Titulaer et al., 1986; Schoental, 1983). The mechanism by which this may occur remains unclear but both are regulated primarily by the hypothalamic-pituitary-gonadal (HPG) axis suggesting that disruption within this system may underlie both outcomes. Within the HPG axis, reproductive maturation and function is coordinated by the release of gonadotropin releasing hormone (GnRH) (Elkind-Hirsch et al., 1981; Gorski et al., 1975) from a neuronal population surrounding the vascular organ of the lamina terminalis (OVLT) a region just rostral to the anterior ventral periventricular nucleus (AVPV). The neural components of the HPG axis that regulate GnRH secretion are sexually differentiated by endogenous gonadal hormones, primarily estradiol, through a series of pre- and perinatal critical periods (Cooke et al., 1998). Therefore it is possible that disruption of this sex-specific organization by exposure to endocrine disrupting compounds (EDCs) during one or more of these critical periods could affect both the timing of puberty and the maintenance of regular estrus. In rats, it is well established that the administration of steroid hormones during the neonatal critical period can advance puberty and permanently eliminate the ability to generate regular preovulatory GnRH surges (Arai and Gorski, 1968; Kauffman et al., 2007a; Simerly, 2002). Therefore, for the present experiments EDC exposure was confined to the neonatal period. The goals of the present study were to (1) determine if advanced puberty and impaired estrus function in female rats following neonatal exposure to EDCs is associated with impaired GnRH activation within the hypothalamus and (2) explore the estrogen receptor dependent mechanisms by which this may be occurring.

Although GnRH neurons express the beta form of the estrogen receptor (ERβ) throughout development (Herbison and Pape, 2001; Hrabovszky et al., 2000; Hrabovszky et al., 2001) and thus may respond to estrogenic compounds directly, it is generally accepted that hormonal and other environmental signals are largely conveyed to GnRH neurons from other hormone-responsive neurons within the hypothalamus, particularly the AVPV. The AVPV appears to be the primary regulator of GnRH neuronal activity in females as lesions to this region abolish estrogen-induced LH surges and disrupt the estrous cycle (Terasawa et al., 1980; Wiegand et al., 1980). We have previously shown that neonatal exposure to the EDCs genistein (GEN) or Bisphenol-A (BPA) can affect sexually dimorphic gene expression in the AVPV (Patisaul et al., 2006) and AVPV volume (Patisaul et al., 2007) suggesting that this region may be particularly sensitive to disruption by EDCs. Estrogen receptor containing neurons that send axonal projections to GnRH neurons are abundant in the AVPV (Gu and Simerly, 1997; Polston et al., 2004; Polston and Simerly, 2006; Shughrue et al., 1997; Simerly et al., 1990). Of these, a subset also express the KiSS-1 gene, which has recently been found to play a key role in both the timing of puberty and the regulation of the estrous cycle (Kauffman et al., 2007a; Smith et al., 2006a; Smith et al., 2006b). The KiSS-1 gene codes for a family of proteins called kisspeptins (KISS), also known as metastins (Navarro et al., 2004), which act as endogenous ligands for the G protein-coupled receptor GPR54 (Kotani et al., 2001; Lee et al., 1999; Muir et al., 2001). GnRH neurons constitutively express GPR54 (Irwig et al., 2004) and activation of this receptor appears to be vital for triggering the onset of puberty as humans lacking GPR 54 are hypogonadal and fail to enter puberty (de Roux et al., 2003; Seminara et al., 2003). The number of KiSS-1 neurons in the AVPV is markedly sexually dimorphic with females having substantially more than males (Clarkson and Herbison, 2006). Neonatal androgen exposure has recently been shown to result in a male pattern of KiSS-1 gene expression in female rats (Kauffman et al., 2007b). However, this masculinizing effect was likely mediated by estrogens, aromatized in the brain from the administered androgens, because it is well established that estrogens are primarily responsible for masculinizing the hypothalamic nuclei of the rodent HPG axis (Gorski, 1985; Simerly, 2002). Therefore, we hypothesized that neonatal exposure to estrogenic EDCs could interfere with the female-typical organization of KISS neurons resulting in reduced numbers of KISS efferent fibers projecting rostrally from the AVPV towards GnRH neurons.

A second population of KISS neurons resides in the arcuate nucleus (ARC). In contrast to the AVPV, KiSS-1 expression in the ARC does not appear to be sexually dimorphic or affected by neonatal androgen treatment (Kauffman et al., 2007b). In adult rodents, gonadal steroids amplify the expression of KiSS-1 mRNA in the AVPV but inhibit its expression in the ARC (Irwig et al., 2004; Navarro et al., 2004; Smith et al., 2005; Smith et al., 2006b) suggesting that the two populations play distinct roles in the regulation of gonadotropin secretion. It has previously been hypothesized that the AVPV population is critical for steroid positive feedback while the ARC population regulates steroid negative feedback (Kauffman et al., 2007a; Kauffman et al., 2007b; Smith et al., 2006b). To determine if neonatal EDC exposure could affect either population, KISS fiber density was quantified in the ARC as well as the AVPV.

The EDCs selected to test this hypotheses were the isoflavone phytoestrogens equol (EQ) and genistein (GEN). Many EDCs bind to at least one form of the estrogen receptor and affect transcription but the phytoestrogens are particularly potent compared to most synthetic EDCs (Kuiper et al., 1997; Kuiper et al., 1998; Mueller et al., 2003). GEN is commonly present in soy based foods while EQ is a metabolite of the soy isoflavone daidzein (Setchell et al., 2003) and is generated entirely from intestinal microflora. Not all humans are capable of biotransforming daidzein to EQ (Setchell et al., 2002). For example, infants cannot generate EQ, but EQ produced by the mother, as well as GEN and other phytoestrogens, readily cross the placenta (Todaka et al., 2005). Phytoestrogens have previously been shown to disrupt reproductive physiology in both laboratory animals and humans. For example, neonatal exposure to phytoestrogens has been associated with advanced pubertal onset in Puerto Rican girls (Freni- Titulaer et al., 1986; Schoental, 1983) and a retrospective cohort study found that young women fed soy-based infant formula as infants reported longer menstrual bleeding and menstrual discomfort (Strom et al., 2001). Studies in mice have shown that neonatal exposure to GEN can advance puberty, lengthen the reproductive cycle, and alter ovarian morphology (Jefferson et al., 2006a; Jefferson et al., 2005). In contrast, relatively little is known about the consequences of neonatal exposure to EQ in either rodents or humans.

For the present experiments, the synthetic estrogen estradiol benzoate (EB) was used as a positive control because neonatal exposure to EB has previously been shown to advance puberty and suppress ovarian cyclicity (Aihara and Hayashi, 1989; Nagao et al., 1999). We also used the ERα specific agonist 1,3,5-tris(4-Hydroxyphenyl)-4-propyl-1H-pyrazole (PPT) and the ERβ specific agonist diarylpropionitrile (DPN) as additional controls to gain insight as to which form of the ER might mediate the effects of EDCs on HPG organization and function. It is widely hypothesized that ERα and ERβ play distinct roles in estrogen-dependent brain organization. Use of synthetic agonists specific for each ER subtype (Harris et al., 2002) and knockout animals lacking ERα, ERβ, or both (Couse et al., 1995a; Couse et al., 1995b; Rissman et al., 1999) have proven to be very useful for identifying and defining these roles. In terms of behavior in the adult animal, ERα has generally been shown to be essential for the regulation of sexual behavior and reproductive function (Hewitt and Korach, 2003; Ogawa et al., 1998; Rissman et al., 1999), while ERβ plays a more significant role in the modulation of estrogen action on anxiety and stress (Lund et al., 2005) although by no means do they do so exclusively of the other (Choleris et al., 2006). Therefore we hypothesized that the ERα agonist PPT would have the most significant impact on female reproductive physiology, GnRH neuronal activation, and KISS fiber density.

In the present study, animals were exposed to either vehicle, EB, GEN, EQ, DPN or PPT in the first 4 days of life, then monitored for changes in reproductive physiology, GnRH activation and hypothalamic KISS fiber density. In mammals, puberty is manifested by changes in sex-specific physiology and behavior. The primary mechanism for this awakening is still unclear (Sisk and Foster, 2004; Terasawa and Fernandez, 2001) but in rodents vaginal opening is a hallmark of puberty and was therefore used as the physiological marker of pubertal onset. The ability to generate and maintain a regular estrous cycle was assessed using vaginal cytology. The animals were then ovariectomized, hormone replaced and sacrificed at a time point when GnRH activation is maximal. The brains were then collected, sectioned, and stained by immunofluorescent immunohistochemistry. GnRH activation was determined by quantifying the co-expression of GnRH and Fos in the OVLT. KISS content was determined by quantifying the relative density of KISS fibers in the AVPV and ARC using confocal microscopy.

2. Materials and Methods

2.1 Animals and neonatal treatment

Timed pregnant Long Evans rats (n = 10; Charles River, NC) were individually housed in a humidity and temperature controlled room with a 12-h light, 12-h dark cycle (lights on from 10:00 to 22:00) at 23°C and 50% average relative humidity at the Biological Resource Facility at North Carolina State University (NCSU). Because standard lab chows are soy-based and thus contain significant amounts of phytoestrogens (Boettger-Tong et al., 1998; Brown and Setchell, 2001; Thigpen et al., 1999), all of the animals were fed a semi-purified, phytoestrogen-free diet ad libitum for the duration of the experiment (AIN-93G, Test Diet, Richmond, IN). Eight of the 10 dams littered on the same evening, therefore, female pups from these 8 dams were cross fostered and treated within 4–6 hours of birth (6–10 females per dam). Within each litter, only two females that were biologically related to the mother were kept with that litter and the rest were obtained from other litters. All remaining females were culled. Although they were not used for this study, some males were kept with their mothers (a maximum of 5 per dam) to reduce maternal anxiety and the risk of cannibalism. In all cases, the cross fostered litter was smaller than the original biological litter and all of the pups within a cross fostered litter were given the same treatment to avoid cross contamination. There were two vehicle treated litters (6 treatment conditions; 8 total litters) but all litters were cross fostered.

Beginning on the day of birth, females were subcutaneously (sc) injected with sesame oil (0.05ml, Sigma, St. Louis), estradiol benzoate (EB, 50 µg, Sigma), the ERα agonist propyl-pyrazole-triol (PPT; 1 mg/kg bw, Tocris Biosciences, Ellisville, MS), the ERβ agonist diarylpropionitrile (DPN; 1 mg/kg bw, Tocris Biosciences), racemic equol (EQ; 10 mg/kg bw, generously supplied by Mike Adams of Wake Forest University) or genistein (GEN; 10 mg/kg bw, Indofine Chemical Company, Hillsborough, NJ). All compounds were dissolved in ethanol, and then sesame oil at a ratio of 10% EtOH and 90% oil as we have done previously (Patisaul et al., 2006). The vehicle was also prepared with this ratio. We have found this vehicle to cause less skin irritation than the alternative vehicle, DMSO. This dose of GEN is similar to the total amount of soy phytoestrogens consumed daily by children fed soy infant formula (Setchell et al., 1997) and the dose of EQ was chosen to match. The dose of DPN and PPT was approximately equivalent to what has been used in previously published studies examining the effects of these compounds on estrogen mediated behavior and uterine weight in adults (Frasor et al., 2003; Harris et al., 2002; Lund et al., 2005; Rhodes and Frye, 2006; Walf et al., 2004). DPN is an ERβ selective agonist with a 70-fold greater relative binding affinity and 170-fold greater relative potency in transcription assays for ERβ than ERα (Meyers et al., 2001). Additionally, PPT, is selective for ERα, with a 400-fold preference for ERα and minimal binding to ERβ (Stauffer et al., 2000; Sun et al., 2002). The animals (n = 8–16 per group) received injections daily from the day of birth (PND 0) through PND 3 (4 injections total). Not all animals were used for the present study.

All pups were weaned into littermate pairs on PND 22, ear tagged, and maintained on a reverse light schedule (lights off from 10:00 to 22:00). Upon weaning, the animals were checked daily for vaginal opening. Monitoring of the estrous cycle by vaginal lavage commenced approximately two weeks after vaginal opening and all samples were collected in the morning, just after lights off. Because frequent lavage can induce pseudopregnancy, all animals observed to have stopped cycling were resampled 2–3 weeks later to confirm that the irregular cycle was not due to a pseudopregnancy. Sampling proceeded for 10 weeks with an additional 2–3 weeks for validation.

Animal care and maintenance were conducted in accordance with the applicable portions of the Animal Welfare Act and the U.S. Department of Health and Human Services “Guide for the Care and use of Laboratory Animals.” All experimental procedures involving animals were approved by the NCSU Institutional Animal Care and Use Committee.

2.2 Ovariectomy, tissue collection, and immunohistochemistry

Animals were gonadectomized (GNX) under isoflurane anesthesia (completed over 10 days beginning on postnatal day 146) and allowed 6 weeks to recover to provide adequate time for endogenous hormone levels to diminish. The OVX females were then injected sc with 10 µg EB dissolved in sesame oil at 0900 h, followed 48 hours later by a sc injection of 500 µg of progesterone dissolved in the same vehicle. All animals were sacrificed by transcardial perfusion with 4% paraformaldehyde 6–8 hours after the progesterone injection. It has previously been established that this is the point at which GnRH activity following sequential steroid hormone administration is maximal (Finn et al., 1998; Wu et al., 1992). Brains were removed, post-fixed in 20% sucrose paraformaldehyde, cryoprotected overnight in potassium phosphate buffer containing 20% sucrose, rapidly frozen in powdered dry ice, and stored at −80°C. Brains were then sliced into 35 µm coronal sections, and divided into two series of free-floating alternating sections. Tissue was stored in antifreeze solution (20% glycerol, 30% ethylene glycol in KPBS) at −20°C until immunohistochemical processing. One set of tissue was immunolabeled for GnRH and Fos while the other set was immunolabeled for KISS using immunofluorescent immunohistochemistry (IHC) methods described in detail elsewhere (Patisaul et al., 2008). Briefly, the first set of tissue was incubated in a cocktail of primary antibodies directed against GnRH (raised in rabbit, 1:3000, Chemicon LHRH, AB1567) and Fos (raised in goat, 1:250, Santa Cruz, SC-52-6) followed by the secondary antibodies Alexa-Fluor donkey anti-rabbit 568 and Alexa-Fluor donkey anti-goat 488 at 1:200. The second set of tissue was incubated in a primary antibody directed against kisspeptin-10, raised in rabbit and generously gifted by Alain Caraty of Institut National de la Recherche Agronomique/Centre National de la Recherche Scientifique, Université de Tours, at 1:6,000 and the Alexa-Fluor donkey anti-rabbit 568 secondary antibody. After secondary antibody incubation, sections were counterstained with Hoechst 33258 (Invitrogen), rinsed, mounted onto slides (Suprefrost Plus, Fisher, Pittsburgh, PA), and coverslipped using a standard glycerol mountant.

2.3 Quantification of GnRH and Fos immunoreactivity in the OVLT

GnRH/Fos double-immunofluorescent label was visualized in the organum vasculosum of the lamina terminalis (OVLT). Anatomical identification of each section was made by comparing the counterstained sections to a brain atlas (Paxinos and Watson, 1998; Paxinos and Watson, 2004). GnRH and Fos immunostaining was observed to be consistent and evenly distributed throughout the OVLT. Two midlevel sections per animal were selected and analyzed using a Leica 5000DM microscope fitted with 20X, and 40X objective lenses and filter cubes for Cy3 and FITC. To quantify double label, first GnRH immunostaining was observed with the FITC filter and photographed at 20X and 40X with a Retiga 1800 digital camera. Second, Fos immunostaining was observed in the same region with the Cy3 filter and photographed. The images were then merged using the MCID elite Image Analysis (Interfocus Imaging Ltd., Cambridge, England) software package. Cells immunostained for GnRH only, and cells immunlabeled for both GnRH and Fos were then hand counted by an individual blind to the treatment groups and verified by a second independent observer.

2.4 Quantification of KISS fiber density in the AVPV and ARC

KISS immunoreactivity (-ir) has previously been shown to be localized to extended lengths of fibers throughout the AVPV and within cell bodies located in the medial and caudal AVPV (Clarkson and Herbison, 2006). We aimed to quantify the density of AVPV KISS fibers projecting rostrally from the AVPV to the OVLT GnRH neurons, therefore one rostral AVPV section was selected for each animal using a brain atlas (Paxinos and Watson, 1998; Paxinos and Watson, 2004). In the ARC, KISS-ir has been shown to be localized to fibers and cell bodies throughout the ARC so one midlevel section of the ARC was selected for imaging and quantification. Thus, the selected regions are representative of the AVPV and ARC but do not include all KISS fibers present in either. KISS-ir was visualized on a Zeiss LSM 510 Meta confocal microscope fitted with a 63x oil corrective objective lens housed at the CIIT Centers for Health Research at the Hamner Institute (Research Triangle Park, NC). For each scan, a set of serial image planes (z-step distance = 1 µm) was collected through the entire thickness of the section. Individual images were acquired sequentially for light emitted from each fluorophore and parsed into separate stacks of images for analysis using the Image J software package (National Institutes of Health (NIH), Bethesda, MD) as previously described (Patisaul et al., 2008). To control for variations in tissue thickness that would result in unequal numbers of image planes, substacks of consecutive image planes that excluded the rostral and caudal edges of the tissue sections were created for each set of scans. Substacks consisted of 16 sequential image planes for the AVPV and 22 sequential images planes for the ARC. Only data from sections with consistent staining throughout the entire thickness were included in the analysis. Individual images contained within each substack were first binarized and depixelated to minimize the inclusion of background fluorescence. Fibers were then skeletonized to a thickness of one pixel to compensate for differences in individual fiber thickness and brightness. The number of resulting bright pixels in each image plane was then quantified using the Image J Voxel Counter plug-in (NIH). The voxel counts were averaged within the substack to obtain a single measure for each section that was then used as a quantitative representation of average KISS fiber density within the volume sampled (Patisaul et al., 2008).

2.5 Statistical Analysis

Day of vaginal opening was compared across treatment groups by one-way analysis of variance (ANOVA) with treatment as a factor and followed up with Fisher’s Least Significant Difference (LSD) post-hoc test for individual comparisons (SYSTAT) with the significance level set at p < 0.05. The percentage of immunopositive GnRH cells co-labeled with Fos within each group was also compared using a one-way ANOVA followed by LSD post hoc tests. As anticipated (Wray and Gainer, 1987; Wray and Hoffman, 1986) the number of GnRH neurons counted did not significantly differ between groups. Sample sizes for the KISS analysis in the AVPV were small, particularly in the DPN and PPT groups, due to the strict selection criteria used to ensure that each analyzed section was anatomically matched. Therefore, the data was divided into two groups and analyzed separately to address two specific hypotheses: (1) does neonatal exposure to phytoestrogens affect the density of AVPV KISS fibers and (2) through which ER subtype is this effect likely mediated. To address the first hypothesis, the density of voxels containing KISS fibers for the OIL, EB, EQ and GEN groups were compared by one-way ANOVA followed by LSD post hoc tests. To address the second, the OIL, EB, DPN and PPT groups were compared using the same methods. Samples sizes for the KISS analysis in the ARC were sufficient, therefore all groups were compared by one-way ANOVA followed by LSD post hoc tests. In all cases, the significance level was set at p < 0.05.

3. Results

3.1 Age at Vaginal Opening

There was a significant effect of treatment on age at vaginal opening (F(5,53) = 29.401, p ≤ 0.0001). Compared to the controls (n = 12), vaginal opening was significantly advanced by EB (n = 15, p < 0.001) and GEN (n = 8, p < 0.03) but not EQ (n = 10, Fig. 1.A.). Neither the ERα agonist PPT nor the ERβ agonist DPN had a significant effect on the day of vaginal opening. The data suggest that EDC exposure during the neonatal critical period in which the HPG axis is undergoing steroid directed organization advance pubertal onset in females but the relative roles each ER subtype may play in the mediation of this effect remains unclear.

FIG. 1.

FIG. 1

(A) Day of vaginal opening (indicative of pubertal onset) was significantly advanced in animals neonatally treated with EB (n = 15) or GEN (n = 8) but not EQ (n = 10) compared to the oil controls (n = 12). (B) Animals in all groups displayed regular estrous cycles in the first few weeks post-vaginal opening, however some individuals within each treatment groups except the control group (OIL, n = 8) eventually entered into a state of persistent estrus or diestrus. Females in the EB group (n = 10) entered persistent estrus earliest and all animals were in a state of persistent estrus by three weeks after the onset of testing. By ten weeks, 57% of the DPN animals (n = 7), 25% of the EQ animals (n = 8), 29% of the GEN (n = 7) animals and none of the PPT animals (n = 7) displayed a regular 4–5 day estrus cycle. The remainder had entered either a persistent estrus or diestrus. (Means ± s.e.m. *P ≤0.03)

3.2 Estrous Cycle Data

Beginning approximately two weeks after vaginal opening, regularity of the estrous cycle was assessed weekly in a cohort of animals by vaginal lavage. Regular 4 day estrous cycles commenced in all treatment groups. As expected (Aihara and Hayashi, 1989), all of the EB females (n = 10) stopped cycling within three weeks after testing began and entered persistent estrus. By 10 weeks, less than 30% of the EQ (n = 8) and GEN (n = 7) treated females displayed regular estrous cycles (Fig. 1.B.). The remaining 70% were in either persistent estrus or diestrus. Smears from these animals contained a mixture of cornified cells, nucleated epithelial cells and leukocytes. The rate at which the EQ and GEN treated animals became acyclic was also very similar. All of the PPT treated animals were in persistent estrus by 8 weeks of test onset. In contrast, 57% of the DPN treated animals were still displaying regular estrous cycles 10 weeks after monitoring began. Smears from DPN animals that had stopped cycling primarily contained cornified cells but a number of nucleated cells were also present in most cases. The data suggest that neonatal EDC exposure can result in impaired ovarian cyclicity and that both ER subtypes may play a mechanistic role in mediating this effect.

3.3 GnRH Activation

The animals were then hormonally replaced by the sequential administration of EB and progesterone and sacrificed 6 hours after the progesterone injection. This is the point at which Fos expression in GnRH neurons is greatest (Finn et al., 1998; Wu et al., 1992). GnRH activity was determined by quantifying the percentage of OVLT GnRH neurons that co-expressed Fos. There was a significant effect of treatment on the percentage of GnRH neurons immunoreactive for Fos (F(5,25) = 8.458, p ≤ 0.0001). Hormone administration successfully induced Fos expression in the GnRH neurons of the control females (n = 7) (Fig. 2). In contrast, less than 2% of GnRH neurons were colabeled with Fos in the EB females (n = 8, p < 0.001). GnRH activation was 50% lower in the EQ animals (n = 4, p < 0.022) and 70% lower in the GEN (n = 4, p ≤ 0.002) animals compared to the controls. None of the GnRH neurons in the PPT animals contained Fos (p ≤ 0.001) suggesting that agonism of ERα during the neonatal period may be a critical mechanism by which neonatal estrogen or EDC treatment suppresses GnRH activation in response to female gonadal hormones. GnRH activation was also reduced in the DPN group compared to the control group (although not significantly, p ≤ 0.057) suggesting that ERβ may also play a mechanistic role in this process.

FIG 2.

FIG 2

(A–B) Immunolabeling of GnRH (red) and Fos (green) was readily observed. (A) A single labeled GnRH neuron (red) showing no co-expression of Fos and (B) a double labeled neuron immunopositive for GnRH (red) and Fos (green) are depicted here. (C) The percentage of GnRH neurons immunopositive for Fos was lowest in the EB treated animals (n=8). Neonatal EQ treatment (n=4) resulted in a 50% decrease in Fos colabeling and neonatal GEN treatment (n=4) resulted in a 70% decrease compared to controls (OIL). No colabeled cells were observed in any of the PPT (n = 4) animals. GnRH activation was markedly lower in the DPN group but this effect did not quite reach statistical significance (p = 0.057). (Means ± s.e.m. *P ≤0.03, **P ≤0.002, Scale bar = 20 µm.)

3.4 AVPV KISS Fiber Density

The observed depletion of GnRH activation may not necessarily result from disruption at the level of the GnRH neuron itself, but rather from changes to the hormone sensitive neuronal inputs on GnRH neurons including KISS-producing neurons in the AVPV. KISS-ir was localized to extended lengths of fibers throughout the AVPV (Fig. 3A and B) as has been observed previously (Clarkson and Herbison, 2006). Only sections from animals which had consistent staining throughout the entire thickness of the AVPV were selected for the KISS-ir analysis, a constraint that resulted in relatively small sample sizes in some groups. Therefore the data were divided into two sets for analysis. The first contained the OIL, EB, EQ and GEN groups (Fig. 3C) and tested the hypothesis that neonatal phytoestrogen treatment could alter KISS fiber density in the AVPV. There was a significant effect of treatment in this set (F(3,21) = 3.502, p ≤ 0.033). Compared to the control females (n = 8), neonatal treatment with EB (n = 4) decreased average KISS fiber density 64%. (p < 0.006) and GEN reduced KISS content by 60% (n = 5, p < 0.05). Neonatal treatment with EQ reduced KISS fiber density by 25% compared to the controls, an effect that was not statistically significant (n = 8, p < 0.17). The second analysis was conducted to gain insight as to which ER subtype might be mediating the effect of EB and GEN on KISS-ir and contained the OIL, EB, DPN and PPT groups (Fig. 3D). Sample sizes in the DPN and PPT groups were very small and thus diminished the statistical power of this analysis. As a result, there was no significant main effect of treatment on average KISS fiber density in the AVPV within this set (F(3,15) = 2.435, p = 0.105). It has previously been shown that neonatal administration of steroid hormones can reduce the kisspeptin content of the anterior hypothalamus and suppress the GnRH surge associated with ovulation in rodents (Kauffman et al., 2007a). Therefore, our data suggest that decreased KISS expression may be a mechanism by which GnRH activation is impaired in animals neonatally treated with phytoestrogens but do not yield insight as to which ER subtype might mediate this effect.

FIG 3.

FIG 3

Representative confocal images (5 merged consecutive optical planes) depicting the extended lengths of immunolabeled KISS-ir fibers in the AVPV of an (A) Oil (B) EB (C) EQ and (D) GEN treated animal. (E) Compared to the OIL treated control animals KISS immunoreactivity was significantly reduced by neonatal administration of EB and GEN but not EQ. (F) Sample sizes in the DPN and PPT groups were too small to conduct a meaningful analysis but the quantification of KISS immunoreactivity is presented here (F) alongside the data from the OIL and EB groups for illustrative purposes. (Means ± s.e.m. *p ≤0.05, **p ≤ 0.006, Scale bar = 20µm.)

3.5 ARC KISS Fiber Density

KISS-ir was localized to extended lengths of fibers and a small number of cell bodies throughout the ARC which is consistent with what has been observed previously (Clarkson and Herbison, 2006). One midlevel section per animal from animals which had consistent staining throughout the entire ARC was selected for analysis. There was a significant main effect of treatment on KISS fiber density in the ARC (F(5, 37) = 11.644, p ≤ 0.001, Fig. 4). Compared to the control females (n = 10), KISS fiber density was lowest in the EB treated group (n = 8, 72% lower than the controls, p ≤ 0.001) and also significantly lower in the PPT group (n = 6, p ≤ 0.001). Neither EQ (n = 7) nor GEN (n = 5) treatment had a significant effect on KISS fiber density in the ARC nor did DPN (n = 7) suggesting that this measure is primarily sensitive to ERα agonism.

FIG 4.

FIG 4

KISS-ir in the ARC was significantly lower in the animals neonatally treated with EB or the ERα agonist PPT compared to the oil treated controls (OIL). The phytoestrogens GEN and EQ did not significantly affect ARC KISS-ir nor did the ERβ agonist DPN.

4. Discussion

EB and GEN were the only two compounds that significantly affected both the timing of vaginal opening and the ability to maintain a regular estrous cycle. GnRH activation was strongly impaired in these animals, as well as the PPT animals, suggesting that the mechanism of disruption is ERα dependent. Because GnRH neurons do not express ERα, this observation supports the hypothesis that endocrine disruption occurs within ERα expressing neurons that send efferent projections to GnRH neurons, such as KISS neurons. KISS fiber density was significantly lower in both the AVPV and ARC of the EB treated animals but only in the AVPV of the GEN treated animals. The AVPV population of KISS neurons is thought to regulate steroid positive feedback in mammals, including humans (Navarro et al., 2004; Pompolo et al., 2006; Roa et al., 2006; Shibata et al., 2007; Smith et al., 2006b; Tena-Sempere, 2006). Therefore, collectively the observations in the present study suggest that impaired organization of the steroid positive feedback circuitry on GnRH neurons might be the primary mechanism underling the observed changes in reproductive physiology following neonatal exposure to EDCs.

Prior studies have repeatedly demonstrated that developmental exposure to estrogenic compounds, including phytoestrogens and other EDCs, can alter the timing of puberty, disrupt ovarian cycle function, and masculinize the female hypothalamus (Colborn et al., 1993; Jefferson et al., 2006b; Lephart et al., 2005; McLachlan et al., 2006; Patisaul, 2005; Patisaul and Polston, 2007). To our knowledge, this is the first time that similar effects for the isoflavone metabolite EQ have been reported. Advanced puberty following exposure to GEN, or soy protein containing GEN and other isoflavones, has been observed previously (Badger et al., 2001). For example, Leaf et al, found that exposure to four days of 10 mg/kg bw per day of GEN, beginning on gestational day 15, (via administration by sc injection to the pregnant dam) also resulted in accelerated vaginal opening and altered ovarian cycle frequency (Levy et al., 1995). A similar outcome was reported for rats exposed to 40 mg/kg bw GEN per day from birth to weaning (Lewis et al., 2003) but not 4 mg/kg bw GEN per day. This may indicate that the dose of 10 mg/kg bw used in the present study is approaching the minimum level for which an effect of GEN on these endpoints is observed. In the present study, we found no significant effect of EQ on the timing of vaginal opening, but it remains to be seen whether or not there is a dose effect of EQ. One study failed to find advanced pubertal onset in the rat following oral administration of GEN across the first five days of life in doses ranging from 12.5 mg/kg bw to 100 mg/kg bw (Nagao et al., 2001), which may indicate that the route of exposure may also affect outcome. In the present study we opted to administer our compounds by sc injection because a thorough literature search indicated that this was the exposure route most frequently used in other studies using PPT and DPN. Therefore we administered all of the test compounds by sc injection to be consistent and ensure effective delivery of PPT and DPN. A newly published study has also now demonstrated that subcutaneous injection of GEN is a suitable model for oral exposure indicating that route of exposure may not be a serious concern (Jefferson et al., 2007).

Neonatal administration of GEN or EQ also significantly abrogated GnRH neuronal activation following hormone priming in adulthood. This observation is consistent with a prior study which showed that GEN has a defeminizing effect on GnRH secretion (Faber and Hughes, 1993). The specific mechanism by which this occurs is unclear although activity on one or both ER subtype has long been hypothesized to be crucial. ERα and ERβ are differentially distributed throughout the hypothalamus (Li et al., 1997; Shughrue et al., 1996; Shughrue et al., 1997) suggesting that they play distinct roles in GnRH activation and signaling. Therefore, selective disruption through one ER subtype may produce disparate outcomes on GnRH function. Phytoestrogens generally have a higher relative binding affinity for ERβ than ERα in vitro (Kuiper et al., 1998; Mueller et al., 2004). However, in vivo they have been shown to alter both ERα and ERβ dependent gene expression in the brain (Jacob et al., 2001; Patisaul et al., 2002; Patisaul et al., 1999) indicating that they at least have the potential to act through either subtype. The degree to which GnRH activation was suppressed by the phytoestrogens was more similar to the results seen with DPN than with PPT suggesting that the phytoestrogens may be operating through an ERβ mediated mechanism. Alternatively, this observation may also indicate that the phytoestrogens are activating as weak agonists on ERα. This distinction is significant because GnRH neurons express ERβ but not ERα throughout development (Herbison and Pape, 2001; Hrabovszky et al., 2000; Hrabovszky et al., 2001). Therefore, activity through ERβ would suggest that phytoestrogens can act on GnRH neurons directly. Further work is necessary to resolve this critical issue.

In the present study, the ERβ agonist DPN produced a less severe outcome than PPT on the ability to maintain a regular estrous cycle and the capacity to activate GnRH neurons in response to hormone priming. The observation that both PPT and DPN suppressed ovarian cycle function is consistent with effects previously reported following post-natal exposure to two other ER specific agonists (Patchev et al., 2004). In this prior study, agonism of ERβ was found to be more potent on estrous cyclicity, an effect opposite to what was observed here. In the present study, although impairment of GnRH activation by ERβ agonism did not reach statistical significance, it is possible that, within the DPN treated group, GnRH activation was lowest in the individual animals that entered persistent diestrus and higher in the animals that did not. This would then support the hypothesis that EDCs can directly interact with GnRH through ERβ. Unfortunately the sample sizes in the present study were too small to investigate this possibility but future studies will address this issue. The present data suggest that ERα plays a more significant mechanistic role because while the DPN effect did not quite reach statistical significance, ovarian cyclicity and GnRH activation were completely absent in the PPT group. Further studies using a range of DPN and PPT doses are needed to determine if this simply reflects a dose effect and that higher doses of DPN could also eliminate ovarian cyclicity and GnRH activation.

Since GnRH neurons do not express ERα it has been hypothesized that estrogen signaling is conveyed to GnRH neurons by ERα-expressing neurons elsewhere in the hypothalamus, particularly the AVPV (Simerly, 1998; Simerly, 2002). In the female AVPV, estrogen administration markedly increases KiSS-1 mRNA expression and the number of KISS-ir cell bodies (Irwig et al., 2004; Navarro et al., 2004; Smith et al., 2005) but this effect can be eliminated by neonatal androgen treatment (Kauffman et al., 2007b). Here we have shown that neonatal exposure to EB or GEN had a similar effect on AVPV KISS fiber density. AVPV KISS neurons express ERα (Smith et al., 2005; Smith et al., 2006b) so presumably ERα plays a mechanistic role in this effect but unfortunately sample sizes were not sufficient to ascertain if DPN or PPT had an effect on KISS fiber density in the AVPV. In the adult female, it has recently been demonstrated that neuronal ERα is required for the generation of the LH surge (Roa et al., 2008; Wintermantel et al., 2006) but the relative roles ERα and ERβ play in the organization of the neural circuitry required to generate a gonadotropin surge in response to hormone priming are less well understood. The differential distribution of ERα and ERβ in the neonatal rat AVPV have not yet been definitively described making it difficult to conclude that ERα is playing the most substantial role in mediating the organizing effects of estrogens. For example, a pair of studies conducted by the same research group (Orikasa et al., 2002; Orikasa and Sakuma, 2003) have clearly identified both ERβ-ir and ERβ mRNA in the AVPV of male and female rats on PND 7 and beyond (day of birth defined as PND 1 in these studies) but did not look earlier. In contrast a study conducted by a different research group observed no ERβ-ir within the AVPV in the first 2 weeks of life (Perez et al., 2003). Therefore the neonatal distribution of ERα and ERβ in the rat AVPV remains undefined.

We also found that neonatal exposure to EB reduced KISS fiber density in the ARC following hormone priming in adulthood. ERα appears to play a mechanistic role in this process as KISS fiber density was also reduced in the ARC of the PPT but not the DPN treated animals, an effect which is not unexpected given that ERα is abundant in the ARC while ERβ is absent (Shughrue and Merchenthaler, 2001; Simerly et al., 1990). The significant effect of EB was somewhat surprising given that the number of KiSS-1 neurons nor the quantity of KiSS-1 expression in the ARC has not been found to be sexually dimorphic in rats (Kauffman et al., 2007b). Prior studies have shown that KiSS-1 expression and the number of cell bodies immunoreactive for kisspeptin in the ARC are significantly decreased by steroid hormone administration in adults (Kauffman et al., 2007b). Very few KISS-ir cell bodies were present in any of our ARC sections, regardless of treatment, an observation that is consistent with these prior studies. Without an untreated control group, it is difficult to ascertain whether the absolute number of KISS neurons in the ARC, or the AVPV, is reduced by EDC treatment or if their response to hormone priming in adulthood is modified. Experiments designed to distinguish between these two alternative hypotheses are underway. It will also be critical to determine if the KISS fibers quantified in the present study make direct contact with OVLT GnRH neurons.

Finally, it is important to note that it is possible that GEN may produce its effects via mechanisms that are not dependent on ERs. GEN is a potent tyrosine kinase inhibitor so some of the observed effects may be attributable, at least in part, to this mechanism of action. GEN has also been found to alter aromatase production and steroid hormone secretion in vivo (Akingbemi et al., 2004; Brooks and Thompson, 2005; Kishida et al., 2001; Pelissero et al., 1996; Whitehead et al., 2002). Although our findings suggest that ERα plays a mechanistic role, particularly in the suppression of the estrous cycle and GnRH activation, further biochemical and pharmacological experiments are required to conclusively determine the specific mechanisms by which GEN, EQ and other EDCs alter HPG organization. It is also essential to acknowledge that premature vaginal opening and estrous cycle loss could result from disruption anywhere within the HPG axis including the ovary. Disrupted ovarian morphology, including absence of corpora lutea, multi-oocyte follicles and inhibited oocyte nest breakdown (Jefferson et al., 2006a; Jefferson et al., 2002; Jefferson et al., 2006b; Kouki et al., 2003; Nagao et al., 2001) have been reported in both rats and mice following developmental exposure to GEN. By ovariectomizing and hormone replacing our animals prior to sacrifice we aimed to eliminate this potentially confounding effect of ovarian dysfunction.

Unlike rodents, the human hypothalamus is differentiated during gestation, therefore the critical period akin to the one explored in the present study occurs before birth in humans. The GnRH pulse generator is functional by the end of the first trimester (Grumbach, 2002), but it is currently unknown when or how KISS neurons differentiate across gestation and early development in humans. GEN, EQ and most other phytoestrogens readily cross the placenta (Todaka et al., 2005) as do many other EDCs suggesting that human hypothalamic development may be vulnerable to disruption by these compounds however human exposure levels may be too low to produce appreciable effects. Identifying the most sensitive and physiologically relevant critical periods as well as the most salient dose ranges of exposure is essential for environmental risk assessment. Our data support the hypothesis that the time during which the hypothalamus undergoes sexual differentiation is likely one of these critical periods. These findings emphasize the need for further studies aimed at uncovering the mechanisms by which exposure to EDCs, during pre- and postnatal development ultimately impact the health of exposed offspring.

Acknowledgements

We appreciate the assistance of Kenneth H. Pollock for his guidance with the statistical analysis. We also thank Linda Hester and Barbara J. Welker for their assistance with animal husbandry and care and Andrew Crowther for helping to perfuse the animals and for conducting much of the tissue sectioning. We are also grateful to Jillian A. Mickens for her assistance with the IHC and for her critical reading of this manuscript. Finally, we gratefully acknowledge and appreciate Alain Caraty for contributing the kisspeptin antibody, Mike Adams for providing the equol, and the CIIT Centers for Health Research at the Hamner Institute for use of their confocal microscope. This work was supported by NIEHS grant 1R01ES016001-01 to H.B.P.

Footnotes

Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

REFERENCES

  1. Aihara M, Hayashi S. Induction of persistent diestrus followed by persistent estrus is indicative of delayed maturation of tonic gonadotropin-releasing systems in rats. Biol Reprod. 1989;40:96–101. doi: 10.1095/biolreprod40.1.96. [DOI] [PubMed] [Google Scholar]
  2. Akingbemi BT, Sottas CM, Koulova AI, Klinefelter GR, Hardy MP. Inhibition of testicular steroidogenesis by the xenoestrogen bisphenol A is associated with reduced pituitary luteinizing hormone secretion and decreased steroidogenic enzyme gene expression in rat Leydig cells. Endocrinology. 2004;145:592–603. doi: 10.1210/en.2003-1174. [DOI] [PubMed] [Google Scholar]
  3. Arai Y, Gorski RA. Critical exposure time for androgenization of the developing hypothalamus in the female rat. Endocrinology. 1968;82:1010–1014. doi: 10.1210/endo-82-5-1010. [DOI] [PubMed] [Google Scholar]
  4. Badger TM, Ronis MJ, Hakkak R. Developmental effects and health aspects of soy protein isolate, casein, and whey in male and female rats. Int J Toxicol. 2001;20:165–174. doi: 10.1080/109158101317097755. [DOI] [PubMed] [Google Scholar]
  5. Boettger-Tong H, Murthy L, Chiappetta C, Kirkland JL, Goodwin B, Adlercreutz H, Stancel GM, Mäkelä S. A case of a laboratory animal feed with high estrogenic activity and its impact on in vivo responses to exogenously administered estrogens. Environmental Health Perspectives. 1998;106:369–373. doi: 10.1289/ehp.98106369. [DOI] [PMC free article] [PubMed] [Google Scholar]
  6. Brooks JD, Thompson LU. Mammalian lignans and genistein decrease the activities of aromatase and 17beta-hydroxysteroid dehydrogenase in MCF-7 cells. J Steroid Biochem Mol Biol. 2005;94:461–467. doi: 10.1016/j.jsbmb.2005.02.002. [DOI] [PubMed] [Google Scholar]
  7. Brown NM, Setchell KD. Animal models impacted by phytoestrogens in commercial chow: implications for pathways influenced by hormones. Lab Invest. 2001;81:735–747. doi: 10.1038/labinvest.3780282. [DOI] [PubMed] [Google Scholar]
  8. Choleris E, Ogawa S, Kavaliers M, Gustafsson JA, Korach KS, Muglia LJ, Pfaff DW. Involvement of estrogen receptor alpha, beta and oxytocin in social discrimination: A detailed behavioral analysis with knockout female mice. Genes Brain Behav. 2006;5:528–539. doi: 10.1111/j.1601-183X.2006.00203.x. [DOI] [PubMed] [Google Scholar]
  9. Clarkson J, Herbison AE. Postnatal development of kisspeptin neurons in mouse hypothalamus; sexual dimorphism and projections to gonadotropin-releasing hormone neurons. Endocrinology. 2006;147:5817–5825. doi: 10.1210/en.2006-0787. [DOI] [PMC free article] [PubMed] [Google Scholar]
  10. Colborn T, vomSaal FS, Soto AM. Developmental effects of endocrine-disrupting chemicals in wildlife and humans. Environmental Health Perspectives. 1993;101:378–384. doi: 10.1289/ehp.93101378. [DOI] [PMC free article] [PubMed] [Google Scholar]
  11. Cooke B, Hegstrom CD, Villeneuve LS, Breedlove SM. Sexual differentiation of the vertebrate brain:principles and mechanisms. Front Neuroendocrinol. 1998;19:323–362. doi: 10.1006/frne.1998.0171. [DOI] [PubMed] [Google Scholar]
  12. Couse JF, Curtis SW, Washburn TF, Eddy EM, Schomberg DW, Korach KS. Disruption of the mouse oestrogen receptor gene: resulting phenotypes and experimental findings. Biochem Soc Trans. 1995a;23:929–935. doi: 10.1042/bst0230929. [DOI] [PubMed] [Google Scholar]
  13. Couse JF, Curtis SW, Washburn TF, Lindzey J, Golding TS, Lubahn DB, Smithies O, Korach KS. Analysis of transcription and estrogen insensitivity in the female mouse after targeted disruption of the estrogen receptor gene. Mol Endocrinol. 1995b;9:1441–1454. doi: 10.1210/mend.9.11.8584021. [DOI] [PubMed] [Google Scholar]
  14. de Roux N, Genin E, Carel JC, Matsuda F, Chaussain JL, Milgrom E. Hypogonadotropic hypogonadism due to loss of function of the KiSS1-derived peptide receptor GPR54. Proc Natl Acad Sci U S A. 2003;100:10972–10976. doi: 10.1073/pnas.1834399100. [DOI] [PMC free article] [PubMed] [Google Scholar]
  15. Elkind-Hirsch K, King JC, Gerall AA, Arimura AA. The luteinizing hormone-releasing hormone (LHRH) system in normal and estrogenized neonatal rats. Brain Res Bull. 1981;7:645–654. doi: 10.1016/0361-9230(81)90112-x. [DOI] [PubMed] [Google Scholar]
  16. Faber KA, Hughes CL., Jr Dose-response characteristics of neonatal exposure to genistein on pituitary responsiveness to gonadotropin releasing hormone and volume of the sexually dimorphic nucleus of the preoptic area (SDN-POA) in postpubertal castrated female rats. Reprod Toxicol. 1993;7:35–39. doi: 10.1016/0890-6238(93)90007-t. [DOI] [PubMed] [Google Scholar]
  17. Finn PD, Steiner RA, Clifton DK. Temporal patterns of gonadotropin-releasing hormone (GnRH), c-fos,and galanin gene expression in GnRH neurons relative to the luteinizing hormone surge in the rat. J Neurosci. 1998;18:713–719. doi: 10.1523/JNEUROSCI.18-02-00713.1998. [DOI] [PMC free article] [PubMed] [Google Scholar]
  18. Frasor J, Barnett DH, Danes JM, Hess R, Parlow AF, Katzenellenbogen BS. Response-specific and ligand dose-dependent modulation of estrogen receptor (ER) alpha activity by ERbeta in the uterus. Endocrinology. 2003;144:3159–3166. doi: 10.1210/en.2002-0143. [DOI] [PubMed] [Google Scholar]
  19. Freni-Titulaer LW, Cordero JF, Haddock L, Lebron G, Martinez R, Mills JL. Premature thelarche in Puerto Rico. A search for environmental factors. Am J Dis Child. 1986;140:1263–1267. doi: 10.1001/archpedi.1986.02140260065028. [DOI] [PubMed] [Google Scholar]
  20. Gorski RA. Sexual dimorphisms of the brain. J Anim Sci. 1985;61 Suppl 3:38–61. doi: 10.1093/ansci/61.supplement_3.38. [DOI] [PubMed] [Google Scholar]
  21. Gorski RA, Mennin SP, Kubo K. The neural and hormonal bases of the reproductive cycle of the rat. Adv Exp Med Biol. 1975;54:115–153. doi: 10.1007/978-1-4684-8715-2_6. [DOI] [PubMed] [Google Scholar]
  22. Grumbach MM. The neuroendocrinology of human puberty revisited. Horm Res. 2002;57 Suppl 2:2–14. doi: 10.1159/000058094. [DOI] [PubMed] [Google Scholar]
  23. Gu GB, Simerly RB. Projections of the sexually dimorphic anteroventral periventricular nucleus in the female rat. J Comp Neurol. 1997;384:142–164. [PubMed] [Google Scholar]
  24. Harris HA, Katzenellenbogen JA, Katzenellenbogen BS. Characterization of the biological roles of the estrogen receptors, ERalpha and ERbeta, in estrogen target tissues in vivo through the use of an ERalpha-selective ligand. Endocrinology. 2002;143:4172–4177. doi: 10.1210/en.2002-220403. [DOI] [PubMed] [Google Scholar]
  25. Herbison AE, Pape JR. New evidence for estrogen receptors in gonadotropin-releasing hormone neurons. Front Neuroendocrinol. 2001;22:292–308. doi: 10.1006/frne.2001.0219. [DOI] [PubMed] [Google Scholar]
  26. Hewitt SC, Korach KS. Oestrogen receptor knockout mice: roles for oestrogen receptors alpha and beta in reproductive tissues. Reproduction. 2003;125:143–149. doi: 10.1530/rep.0.1250143. [DOI] [PubMed] [Google Scholar]
  27. Hrabovszky E, Shughrue PJ, Merchenthaler If, Hajszan T, Carpenter CD, Liposits Z, Petersen SL. Detection of estrogen receptor-beta messenger ribonucleic acid and 125I-estrogen binding sites in luteinizing hormone-releasing hormone neurons of the rat brain. Endocrinology. 2000;141:3506–3509. doi: 10.1210/endo.141.9.7788. [DOI] [PubMed] [Google Scholar]
  28. Hrabovszky E, Steinhauser A, Barabas K, Shughrue PJ, Petersen SL, Merchenthaler I, Liposits Z. Estrogen receptor-beta immunoreactivity in luteinizing hormone-releasing hormone neurons of the rat brain. Endocrinology. 2001;142:3261–3264. doi: 10.1210/endo.142.7.8176. [DOI] [PubMed] [Google Scholar]
  29. Irwig MS, Fraley GS, Smith JT, Acohido BV, Popa SM, Cunningham MJ, Gottsch ML, Clifton DK, Steiner RA. Kisspeptin activation of gonadotropin releasing hormone neurons and regulation of KiSS-1 mRNA in the male rat. Neuroendocrinology. 2004;80:264–272. doi: 10.1159/000083140. [DOI] [PubMed] [Google Scholar]
  30. Jacob DA, Temple JL, Patisaul HB, Young LJ, Rissman EF. Coumestrol antagonizes neuroendocrine actions of estrogen via the estrogen receptorα. Proceedings of the Society for Experimental Biology and Medicine. 2001;226:301–306. doi: 10.1177/153537020122600406. [DOI] [PubMed] [Google Scholar]
  31. Jefferson W, Newbold R, Padilla-Banks E, Pepling M. Neonatal genistein treatment alters ovarian differentiation in the mouse: inhibition of oocyte nest breakdown and increased oocyte survival. Biol Reprod. 2006a;74:161–168. doi: 10.1095/biolreprod.105.045724. [DOI] [PubMed] [Google Scholar]
  32. Jefferson WN, Couse JF, Padilla-Banks E, Korach KS, Newbold RR. Neonatal exposure to genistein induces estrogen receptor (ER)alpha expression and multioocyte follicles in the maturing mouse ovary: evidence for ERbeta-mediated and nonestrogenic actions. Biol Reprod. 2002;67:1285–1296. doi: 10.1095/biolreprod67.4.1285. [DOI] [PubMed] [Google Scholar]
  33. Jefferson WN, Padilla-Banks E, Newbold RR. Adverse effects on female development and reproduction in CD-1 mice following neonatal exposure to the phytoestrogen genistein at environmentally relevant doses. Biol Reprod. 2005;73:798–806. doi: 10.1095/biolreprod.105.041277. [DOI] [PubMed] [Google Scholar]
  34. Jefferson WN, Padilla-Banks E, Newbold RR. Studies of the effects of neonatal exposure to genistein on the developing female reproductive system. J AOAC Int. 2006b;89:1189–1196. [PubMed] [Google Scholar]
  35. Jefferson WN, Padilla-Banks E, Newbold RR. Disruption of the female reproductive system by the phytoestrogen genistein. Reprod Toxicol. 2007;23:308–316. doi: 10.1016/j.reprotox.2006.11.012. [DOI] [PubMed] [Google Scholar]
  36. Kauffman AS, Clifton DK, Steiner RA. Emerging ideas about kisspeptin- GPR54 signaling in the neuroendocrine regulation of reproduction. Trends Neurosci. 2007a;30:504–511. doi: 10.1016/j.tins.2007.08.001. [DOI] [PubMed] [Google Scholar]
  37. Kauffman AS, Gottsch ML, Roa J, Byquist AC, Crown A, Clifton DK, Hoffman GE, Steiner RA, Tena-Sempere M. Sexual differentiation of Kiss1 gene expression in the brain of the rat. Endocrinology. 2007b;148:1774–1783. doi: 10.1210/en.2006-1540. [DOI] [PubMed] [Google Scholar]
  38. Kishida M, McLellan M, Miranda JA, Callard GV. Estrogen and xenoestrogens upregulate the brain aromatase isoform (P450aromB) and perturb markers of early development in zebrafish (Danio rerio) Comp Biochem Physiol B Biochem Mol Biol. 2001;129:261–268. doi: 10.1016/s1096-4959(01)00319-0. [DOI] [PubMed] [Google Scholar]
  39. Kotani M, Detheux M, Vandenbogaerde A, Communi D, Vanderwinden JM, Le Poul E, Brezillon S, Tyldesley R, Suarez-Huerta N, Vandeput F, Blanpain C, Schiffmann SN, Vassart G, Parmentier M. The metastasis suppressor gene KiSS-1 encodes kisspeptins, the natural ligands of the orphan G protein-coupled receptor GPR54. J Biol Chem. 2001;276:34631–34636. doi: 10.1074/jbc.M104847200. [DOI] [PubMed] [Google Scholar]
  40. Kouki T, Kishitake M, Okamoto M, Oosuka I, Takebe M, Yamanouchi K. Effects of neonatal treatment with phytoestrogens, genistein and daidzein, on sex difference in female rat brain function: estrous cycle and lordosis. Horm Behav. 2003;44:140–145. doi: 10.1016/s0018-506x(03)00122-3. [DOI] [PubMed] [Google Scholar]
  41. Kuiper GGJM, Carlsson B, Grandien K, Enmark E, Häggblad J, Hilsson S, Gustafsson JA. Comparison of the ligand binding specificity and transcript tissue distribution of estrogen receptors αandβ. Endocrinology. 1997;138:863–870. doi: 10.1210/endo.138.3.4979. [DOI] [PubMed] [Google Scholar]
  42. Kuiper GGJM, Lemmen JG, Carlsson B, Corton JC, Safe SH, Van Der Saag PT, Van Der Berg B, Gustafsson JA. Interaction of estrogenic chemicals and phytoestrogens with estrogen receptor β. Endocrinology. 1998;139:4252–4263. doi: 10.1210/endo.139.10.6216. [DOI] [PubMed] [Google Scholar]
  43. Lee DK, Nguyen T, O'Neill GP, Cheng R, Liu Y, Howard AD, Coulombe N, Tan CP, Tang-Nguyen AT, George SR, O'Dowd BF. Discovery of a receptor related to the galanin receptors. FEBS Lett. 1999;446:103–107. doi: 10.1016/s0014-5793(99)00009-5. [DOI] [PubMed] [Google Scholar]
  44. Lephart ED, Setchell KD, Lund TD. Phytoestrogens: hormonal action and brain plasticity. Brain Res Bull. 2005;65:193–198. doi: 10.1016/j.brainresbull.2004.11.022. [DOI] [PubMed] [Google Scholar]
  45. Levy JR, Faber KA, Ayyash L, Hughes CL., Jr The effect of prenatal exposure to the phytoestrogen genistein on sexual differentiation in rats. Proceedings of the Society for Experimental Biology and Medicine. 1995;208:60–66. doi: 10.3181/00379727-208-43832. [DOI] [PubMed] [Google Scholar]
  46. Lewis RW, Brooks N, Milburn GM, Soames A, Stone S, Hall M, Ashby J. The effects of the phytoestrogen genistein on the postnatal development of the rat. Toxicol Sci. 2003;71:74–83. doi: 10.1093/toxsci/71.1.74. [DOI] [PubMed] [Google Scholar]
  47. Li X, Schwartz PE, Rissman EF. Distribution of estrogen receptor-β -like immunoreactivity in rat forebrain. Neuroendocrinology. 1997;66:63–67. doi: 10.1159/000127221. [DOI] [PubMed] [Google Scholar]
  48. Lund TD, Rovis T, Chung WC, Handa RJ. Novel actions of estrogen receptor-beta on anxiety-related behaviors. Endocrinology. 2005;146:797–807. doi: 10.1210/en.2004-1158. [DOI] [PubMed] [Google Scholar]
  49. McLachlan JA, Simpson E, Martin M. Endocrine disrupters and female reproductive health. Best Pract Res Clin Endocrinol Metab. 2006;20:63–75. doi: 10.1016/j.beem.2005.09.009. [DOI] [PubMed] [Google Scholar]
  50. Meyers MJ, Sun J, Carlson KE, Marriner GA, Katzenellenbogen BS, Katzenellenbogen JA. Estrogen receptor-beta potency-selective ligands: structure-activity relationship studies of diarylpropionitriles and their acetylene and polar analogues. J Med Chem. 2001;44:4230–4251. doi: 10.1021/jm010254a. [DOI] [PubMed] [Google Scholar]
  51. Mueller SO, Kling M, Arifin Firzani P, Mecky A, Duranti E, Shields-Botella J, Delansorne R, Broschard T, Kramer PJ. Activation of estrogen receptor alpha and ERbeta by 4-methylbenzylidene-camphor in human and rat cells: comparison with phyto- and xenoestrogens. Toxicol Lett. 2003;142:89–101. doi: 10.1016/s0378-4274(03)00016-x. [DOI] [PubMed] [Google Scholar]
  52. Mueller SO, Simon S, Chae K, Metzler M, Korach KS. Phytoestrogens and their human metabolites show distinct agonistic and antagonistic properties on estrogen receptor alpha (ERalpha) and ERbeta in human cells. Toxicol Sci. 2004;80:14–25. doi: 10.1093/toxsci/kfh147. [DOI] [PubMed] [Google Scholar]
  53. Muir AI, Chamberlain L, Elshourbagy NA, Michalovich D, Moore DJ, Calamari A, Szekeres PG, Sarau HM, Chambers JK, Murdock P, Steplewski K, Shabon U, Miller JE, Middleton SE, Darker JG, Larminie CG, Wilson S, Bergsma DJ, Emson P, Faull R, Philpott KL, Harrison DC. AXOR12, a novel human G protein-coupled receptor, activated by the peptide KiSS-1. J Biol Chem. 2001;276:28969–28975. doi: 10.1074/jbc.M102743200. [DOI] [PubMed] [Google Scholar]
  54. Nagao T, Saito Y, Usumi K, Kuwagata M, Imai K. Reproductive function in rats exposed neonatally to bisphenol A and estradiol benzoate. Reprod Toxicol. 1999;13:303–311. doi: 10.1016/s0890-6238(99)00017-9. [DOI] [PubMed] [Google Scholar]
  55. Nagao T, Yoshimura S, Saito Y, Nakagomi M, Usumi K, Ono H. Reproductive effects in male and female rats of neonatal exposure to genistein. Reprod Toxicol. 2001;15:399–411. doi: 10.1016/s0890-6238(01)00141-1. [DOI] [PubMed] [Google Scholar]
  56. Navarro VM, Castellano JM, Fernandez-Fernandez R, Barreiro ML, Roa J, Sanchez-Criado JE, Aguilar E, Dieguez C, Pinilla L, Tena-Sempere M. Developmental and hormonally regulated messenger ribonucleic acid expression of KiSS-1 and its putative receptor, GPR54, in rat hypothalamus and potent luteinizing hormone-releasing activity of KiSS-1 peptide. Endocrinology. 2004;145:4565–4574. doi: 10.1210/en.2004-0413. [DOI] [PubMed] [Google Scholar]
  57. Ogawa S, Eng V, Taylor J, Lubahn DB, Korach KS, Pfaff DW. Roles of estrogen receptor-alpha gene expression in reproduction- related behaviors in female mice. Endocrinology. 1998;139:5070–5081. doi: 10.1210/endo.139.12.6357. [DOI] [PubMed] [Google Scholar]
  58. Orikasa C, Kondo Y, Hayashi S, McEwen BS, Sakuma Y. Sexually dimorphic expression of estrogen receptor beta in the anteroventral periventricular nucleus of the rat preoptic area: implication in luteinizing hormone surge. Proc Natl Acad Sci U S A. 2002;99:3306–3311. doi: 10.1073/pnas.052707299. [DOI] [PMC free article] [PubMed] [Google Scholar]
  59. Orikasa C, Sakuma Y. Possible involvement of preoptic estrogen receptor beta positive cells in luteinizing hormone surge in the rat. Domest Anim Endocrinol. 2003;25:83–92. doi: 10.1016/s0739-7240(03)00047-x. [DOI] [PubMed] [Google Scholar]
  60. Patchev AV, Gotz F, Rohde W. Differential role of estrogen receptor isoforms in sex-specific brain organization. Faseb J. 2004;18:1568–1570. doi: 10.1096/fj.04-1959fje. [DOI] [PubMed] [Google Scholar]
  61. Patisaul HB. Phytoestrogen action in the adult and developing brain. J Neuroendocrinol. 2005;17:57–64. doi: 10.1111/j.1365-2826.2005.01268.x. [DOI] [PubMed] [Google Scholar]
  62. Patisaul HB, Fortino AE, Polston EK. Neonatal genistein or bisphenol-A exposure alters sexual differentiation of the AVPV. Neurotoxicol Teratol. 2006;28:111–118. doi: 10.1016/j.ntt.2005.11.004. [DOI] [PubMed] [Google Scholar]
  63. Patisaul HB, Fortino AE, Polston EK. Differential disruption of nuclear volume and neuronal phenotype in the preoptic area by neonatal exposure to genistein and bisphenol-A. Neurotoxicology. 2007;28:1–12. doi: 10.1016/j.neuro.2006.10.001. [DOI] [PubMed] [Google Scholar]
  64. Patisaul HB, Fortino AE, Polston EK. Sex differences in serotonergic but not gamma-aminobutyric acidergic (GABA) projections to the rat ventromedial nucleus of the hypothalamus. Endocrinology. 2008;149:397–408. doi: 10.1210/en.2007-0666. [DOI] [PubMed] [Google Scholar]
  65. Patisaul HB, Melby M, Whitten PL, Young LJ. Genistein affects ERβ- but not ERα-dependent gene expression in the hypothalamus. Endocrinology. 2002;143:2189–2197. doi: 10.1210/endo.143.6.8843. [DOI] [PubMed] [Google Scholar]
  66. Patisaul HB, Polston EK. Influence of endocrine active compounds on the developing rodent brain. Brain Res Rev. 2007 doi: 10.1016/j.brainresrev.2007.06.008. [DOI] [PubMed] [Google Scholar]
  67. Patisaul HB, Whitten PL, Young L. Regulation of estrogen receptor beta mRNA in the brain: opposite effects of 17β-estradiol and the phytoestrogen, coumestrol. Brain Research. Molecular Brain Research. 1999;67:165–171. doi: 10.1016/s0169-328x(99)00058-3. [DOI] [PubMed] [Google Scholar]
  68. Paxinos G, Watson C. The rat brain in stereotaxic coordinates. vol. San Diego: Academic Press; 1998. [Google Scholar]
  69. Paxinos G, Watson C. The rat brain in stereotaxic coordinates : [the new coronal set] vol. London: Elsevier Academic; 2004. [Google Scholar]
  70. Pelissero C, Lenczowski M, Chinzi D, Davail-Cuisset B, Sumpter J, Fostier A. Effects of flavonoids on aromatase activity, and in vitro study. Journal of Steroid Biochemistry and Molecular Biology. 1996;57:215–223. doi: 10.1016/0960-0760(95)00261-8. [DOI] [PubMed] [Google Scholar]
  71. Perez SE, Chen EY, Mufson EJ. Distribution of estrogen receptor alpha and beta immunoreactive profiles in the postnatal rat brain. Brain Res Dev Brain Res. 2003;145:117–139. doi: 10.1016/s0165-3806(03)00223-2. [DOI] [PubMed] [Google Scholar]
  72. Polston EK, Gu G, Simerly RB. Neurons in the principal nucleus of the bed nuclei of the stria terminalis provide a sexually dimorphic GABAergic input to the anteroventral periventricular nucleus of the hypothalamus. Neuroscience. 2004;123:793–803. doi: 10.1016/j.neuroscience.2003.09.034. [DOI] [PubMed] [Google Scholar]
  73. Polston EK, Simerly RB. Ontogeny of the projections from the anteroventral periventricular nucleus of the hypothalamus in the female rat. J Comp Neurol. 2006;495:122–132. doi: 10.1002/cne.20874. [DOI] [PubMed] [Google Scholar]
  74. Pompolo S, Pereira A, Estrada KM, Clarke IJ. Colocalization of kisspeptin and gonadotropin-releasing hormone in the ovine brain. Endocrinology. 2006;147:804–810. doi: 10.1210/en.2005-1123. [DOI] [PubMed] [Google Scholar]
  75. Rhodes ME, Frye CA. ERbeta-selective SERMs produce mnemonic-enhancing effects in the inhibitory avoidance and water maze tasks. Neurobiol Learn Mem. 2006;85:183–191. doi: 10.1016/j.nlm.2005.10.003. [DOI] [PubMed] [Google Scholar]
  76. Rissman EF, Wersinger SR, Fugger HN, Foster TC. Sex with knockout models: behavioral studies of estrogen receptor alpha. Brain Res. 1999;835:80–90. doi: 10.1016/s0006-8993(99)01452-3. [DOI] [PubMed] [Google Scholar]
  77. Rao J, Vigo E, Castellano JM, Gaytan F, Navarro VM, Aguilar E, Dijcks FA, Ederveen AG, Pinilla L, van Noort PI, Tena-Sempere M. Opposite Roles of Estrogen Receptor (ER) {alpha} and ER{beta} in the Modulation of Luteinizing Hormone Responses to Kisspeptin in the Female Rat: Implications for the Generation of the Preovulatory Surge. Endocrinology. 2008 doi: 10.1210/en.2007-1540. [DOI] [PubMed] [Google Scholar]
  78. Roa J, Vigo E, Castellano JM, Navarro VM, Fernandez-Fernandez R, Casanueva FF, Dieguez C, Aguilar E, Pinilla L, Tena-Sempere M. Hypothalamic expression of KiSS-1 system and gonadotropin-releasing effects of kisspeptin in different reproductive states of the female Rat. Endocrinology. 2006;147:2864–2878. doi: 10.1210/en.2005-1463. [DOI] [PubMed] [Google Scholar]
  79. Schoental R. Precocious sexual development in Puerto Rico and oestrogenic mycotoxins (zearalenone) Lancet. 1983;1:537. doi: 10.1016/s0140-6736(83)92229-8. [DOI] [PubMed] [Google Scholar]
  80. Seminara SB, Messager S, Chatzidaki EE, Thresher RR, Acierno JS, Jr, Shagoury JK, Bo-Abbas Y, Kuohung W, Schwinof KM, Hendrick AG, Zahn D, Dixon J, Kaiser UB, Slaugenhaupt SA, Gusella JF, O'Rahilly S, Carlton MB, Crowley WF, Jr, Aparicio SA, Colledge WH. The GPR54 gene as a regulator of puberty. N Engl J Med. 2003;349:1614–1627. doi: 10.1056/NEJMoa035322. [DOI] [PubMed] [Google Scholar]
  81. Setchell KD, Brown NM, Desai PB, Zimmer-Nechimias L, Wolfe B, Jakate AS, Creutzinger V, Heubi JE. Bioavailability, disposition, and dose-response effects of soy isoflavones when consumed by healthy women at physiologically typical dietary intakes. J Nutr. 2003;133:1027–1035. doi: 10.1093/jn/133.4.1027. [DOI] [PubMed] [Google Scholar]
  82. Setchell KD, Brown NM, Lydeking-Olsen E. The clinical importance of the metabolite equol-a clue to the effectiveness of soy and its isoflavones. J Nutr. 2002;132:3577–3584. doi: 10.1093/jn/132.12.3577. [DOI] [PubMed] [Google Scholar]
  83. Setchell KDR, Zimmer-Nechemias L, Cai J, Heubi JE. Exposure of infants to phyto-oestrogens from soy-based infant formula. Lancet. 1997;350:23–27. doi: 10.1016/S0140-6736(96)09480-9. [DOI] [PubMed] [Google Scholar]
  84. Shibata M, Friedman RL, Ramaswamy S, Plant TM. Evidence that down regulation of hypothalamic KiSS-1 expression is involved in the negative feedback action of testosterone to regulate luteinising hormone secretion in the adult male rhesus monkey (Macaca mulatta) J Neuroendocrinol. 2007;19:432–438. doi: 10.1111/j.1365-2826.2007.01549.x. [DOI] [PubMed] [Google Scholar]
  85. Shughrue P, Merchenthaler I. Distribution of estrogen receptor beta immunoreactivity in the rat central nervous system. J Comp Neurol. 2001;436:64–81. [PubMed] [Google Scholar]
  86. Shughrue PJ, Komm B, Merchenthaler I. The distribution of estrogen receptor-β mRNA in the rat hypothalamus. Steroids. 1996;61:678–681. doi: 10.1016/s0039-128x(96)00222-x. [DOI] [PubMed] [Google Scholar]
  87. Shughrue PL, Lane MV, Merchenthaler I. Comparative distribution of estrogen receptor αand βmRNA in the rat central nervous system. Journal of Comparative Neurology. 1997;388:507–525. doi: 10.1002/(sici)1096-9861(19971201)388:4<507::aid-cne1>3.0.co;2-6. [DOI] [PubMed] [Google Scholar]
  88. Simerly RB. Organization and regulation of sexually dimorphic neuroendocrine pathways. Behav Brain Res. 1998;92:195–203. doi: 10.1016/s0166-4328(97)00191-5. [DOI] [PubMed] [Google Scholar]
  89. Simerly RB. Wired for reproduction: organization and development of sexually dimorphic circuits in the mammalian forebrain. Annu Rev Neurosci. 2002;25:507–536. doi: 10.1146/annurev.neuro.25.112701.142745. [DOI] [PubMed] [Google Scholar]
  90. Simerly RB, Chang C, Muramatsu M, Swanson LW. Distribution of androgen and estrogen receptor mRNA-containing cells in the rat brain: an in situ hybridization study. J Comp Neurol. 1990;294:76–95. doi: 10.1002/cne.902940107. [DOI] [PubMed] [Google Scholar]
  91. Sisk CL, Foster DL. The neural basis of puberty and adolescence. Nat Neurosci. 2004;7:1040–1047. doi: 10.1038/nn1326. [DOI] [PubMed] [Google Scholar]
  92. Smith JT, Clifton DK, Steiner RA. Regulation of the neuroendocrine reproductive axis by kisspeptin-GPR54 signaling. Reproduction. 2006a;131:623–630. doi: 10.1530/rep.1.00368. [DOI] [PubMed] [Google Scholar]
  93. Smith JT, Cunningham MJ, Rissman EF, Clifton DK, Steiner RA. Regulation of Kiss1 gene expression in the brain of the female mouse. Endocrinology. 2005;146:3686–3692. doi: 10.1210/en.2005-0488. [DOI] [PubMed] [Google Scholar]
  94. Smith JT, Popa SM, Clifton DK, Hoffman GE, Steiner RA. Kiss1 neurons in the forebrain as central processors for generating the preovulatory luteinizing hormone surge. J Neurosci. 2006b;26:6687–6694. doi: 10.1523/JNEUROSCI.1618-06.2006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  95. Stauffer SR, Coletta CJ, Tedesco R, Nishiguchi G, Carlson K, Sun J, Katzenellenbogen BS, Katzenellenbogen JA. Pyrazole ligands: structure-affinity/activity relationships and estrogen receptor-alpha-selective agonists. J Med Chem. 2000;43:4934–4947. doi: 10.1021/jm000170m. [DOI] [PubMed] [Google Scholar]
  96. Strom BL, Schinnar R, Ziegler EE, Barnhart KT, Sammel MD, Macones GA, Stallings VA, Drulis JM, Nelson SE, Hanson SA. Exposure to soy-based formula in infancy and endocrinological and reproductive outcomes in young adulthood. Jama. 2001;286:807–814. doi: 10.1001/jama.286.7.807. [DOI] [PubMed] [Google Scholar]
  97. Sun J, Huang YR, Harrington WR, Sheng S, Katzenellenbogen JA, Katzenellenbogen BS. Antagonists selective for estrogen receptor alpha. Endocrinology. 2002;143:941–947. doi: 10.1210/endo.143.3.8704. [DOI] [PubMed] [Google Scholar]
  98. Tena-Sempere M. KiSS-1 and reproduction: focus on its role in the metabolic regulation of fertility. Neuroendocrinology. 2006;83:275–281. doi: 10.1159/000095549. [DOI] [PubMed] [Google Scholar]
  99. Terasawa E, Fernandez DL. Neurobiological mechanisms of the onset of puberty in primates. Endocr Rev. 2001;22:111–151. doi: 10.1210/edrv.22.1.0418. [DOI] [PubMed] [Google Scholar]
  100. Terasawa E, Wiegand SJ, Bridson WE. A role for medial preoptic nucleus on afternoon of proestrus in female rats. Am J Physiol. 1980;238:E533–E539. doi: 10.1152/ajpendo.1980.238.6.E533. [DOI] [PubMed] [Google Scholar]
  101. Thigpen JE, Setchell KDR, Ahlmark KB, Locklear J, Spahr T, Caviness GF, Goelz MF, Haseman JK, Newbold RR, Forsythe DB. Phytoestrogen content of purified open- and closed-formula laboratory animal diets. Laboratory Animal Sciences. 1999;49:530–536. [PubMed] [Google Scholar]
  102. Todaka E, Sakurai K, Fukata H, Miyagawa H, Uzuki M, Omori M, Osada H, Ikezuki Y, Tsutsumi O, Iguchi T, Mori C. Fetal exposure to phytoestrogens--the difference in phytoestrogen status between mother and fetus. Environ Res. 2005;99:195–203. doi: 10.1016/j.envres.2004.11.006. [DOI] [PubMed] [Google Scholar]
  103. Walf AA, Rhodes ME, Frye CA. Antidepressant effects of ERbeta-selective estrogen receptor modulators in the forced swim test. Pharmacol Biochem Behav. 2004;78:523–529. doi: 10.1016/j.pbb.2004.03.023. [DOI] [PubMed] [Google Scholar]
  104. Whitehead SA, Cross JE, Burden C, Lacey M. Acute and chronic effects of genistein, tyrphostin and lavendustin A on steroid synthesis in luteinized human granulosa cells. Hum Reprod. 2002;17:589–594. doi: 10.1093/humrep/17.3.589. [DOI] [PubMed] [Google Scholar]
  105. Wiegand SJ, Terasawa E, Bridson WE, Goy RW. Effects of discrete lesions of preoptic and suprachiasmatic structures in the female rat. Alterations in the feedback regulation of gonadotropin secretion. Neuroendocrinology. 1980;31:147–157. doi: 10.1159/000123066. [DOI] [PubMed] [Google Scholar]
  106. Wintermantel TM, Campbell RE, Porteous R, Bock D, Grone HJ, Todman MG, Korach KS, Greiner E, Perez CA, Schutz G, Herbison AE. Definition of estrogen receptor pathway critical for estrogen positive feedback to gonadotropin-releasing hormone neurons and fertility. Neuron. 2006;52:271–280. doi: 10.1016/j.neuron.2006.07.023. [DOI] [PMC free article] [PubMed] [Google Scholar]
  107. Wray S, Gainer H. Effect of neonatal gonadectomy on the postnatal development of LHRH cell subtypes in male and female rats. Neuroendocrinology. 1987;45:413–419. doi: 10.1159/000124767. [DOI] [PubMed] [Google Scholar]
  108. Wray S, Hoffman G. A developmental study of the quantitative distribution of LHRH neurons within the central nervous system of postnatal male and female rats. J Comp Neurol. 1986;252:522–531. doi: 10.1002/cne.902520408. [DOI] [PubMed] [Google Scholar]
  109. Wu TJ, Segal AZ, Miller GM, Gibson MJ, Silverman AJ. FOS expression in gonadotropin-releasing hormone neurons: enhancement by steroid treatment and mating. Endocrinology. 1992;131:2045–2050. doi: 10.1210/endo.131.5.1425409. [DOI] [PubMed] [Google Scholar]

RESOURCES