You are on page 1of 11

Hormones and Behavior 98 (2018) 77–87

Contents lists available at ScienceDirect

Hormones and Behavior


journal homepage: www.elsevier.com/locate/yhbeh

Lordosis facilitated by GPER-1 receptor activation involves GnRH-1, T


progestin and estrogen receptors in estrogen-primed rats
Domínguez-Ordóñez R.a, Garcia-Juárez M.a, Lima-Hernández F.J.a, Gómora-Arrati P.a,

Domínguez-Salazar E.b, Blaustein J.D.c, Etgen A.M.d, González-Flores O.a,b,
a
Centro de Investigación en Reproducción Animal, Universidad Autónoma de Tlaxcala-CINVESTAV, México
b
Area de Neurosciencias, Departamento de Biología de la Reproducción, Universidad Autónoma Metropolitana, México
c
Department of Psychological and Brain Sciences, University of Massachusetts, Amherst, MA 01003, USA
d
Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY 10461, USA

A R T I C L E I N F O A B S T R A C T

Keywords: The present study assessed the participation of membrane G-protein coupled estrogen receptor 1 (GPER-1) and
Lordosis gonadotropin releasing hormone 1 (GnRH-1) receptor in the display of lordosis induced by in-
Estradiol tracerebroventricular (icv) administration of G1, a GPER-1 agonist, and by unesterified 17β-estradiol (free E2).
GnRH In addition, we assessed the participation of both estrogen and progestin receptors in the lordosis behavior
G1
induced by G1 in ovariectomized (OVX), E2-benzoate (EB)-primed rats. In Experiment 1, icv injection of G1
G-protein coupled estrogen receptor 1
G15
induced lordosis behavior at 120 and 240 min. In Experiment 2, icv injection of the GPER-1 antagonist G15
Antide significantly reduced lordosis behavior induced by either G1 or free E2. In addition, Antide, a GnRH-1 receptor
RU486 antagonist, significantly depressed G1 facilitation of lordosis behavior in OVX, EB-primed rats. Similarly, icv
Tamoxifen injection of Antide blocked the stimulatory effect of E2 on lordosis behavior. In Experiment 3, systemic injection
of either tamoxifen or RU486 significantly reduced lordosis behavior induced by icv administration of G1 in
OVX, EB-primed rats. The results suggest that GnRH release activates both estrogen and progestin receptors and
that this activation is important in the chain of events leading to the display of lordosis behavior in response to
activation of GPER-1 in estrogen-primed rats.

1. Introduction et al., 2005; Vivacqua et al., 2006) and by 5 mg of RU486, a classical


progestin receptor (PR) antagonist, when administered sc 1 h before E2
Although lordosis in ovariectomized (OVX) rats typically requires (Domínguez-Ordóñez et al., 2015). This implicates the participation of
sequential treatment with 17β-estradiol (E2) and progesterone (P), it both steroid receptors in the short latency behavioral response to E2. It
can also be induced by high or repeated doses of E2 alone (Beyer et al., is important to note that TMX has brain region-specific effects on lor-
1971; Boling and Blandau, 1939; Davidson et al., 1968; Zemlan and dosis (Howard et al., 1984). For example, TMX reduced lordosis be-
Adler, 1977). We and others recently also showed that unesterified 17β- havior induced by P when injected into the ventromedial hypotha-
estradiol (free E2) administered subcutaneously (sc; Domínguez- lamus, but not when administered into the preoptic area or
Ordóñez et al., 2015) or intracerebroventricularly (icv; Domínguez- interpeduncular region (Howard et al., 1984). Moreover, Long et al.
Ordóñez et al., 2015; Long et al., 2014) facilitates lordosis behavior in (2017) showed that TMX and ICI 182,780 (ICI) infused into the arcuate
OVX-E2 benzoate (EB)-primed rats in the absence of P. nucleus facilitate lordosis within 30 min in EB-primed animals. These
The cellular mechanism by which E2 enhances this behavior is un- effects are blocked by pretreatment with the GPER-1 antagonist G15,
clear. However, lordosis behavior induced 39.5 h after EB priming by indicating that they are mediated by GPER-1. Furthermore, RU486
acute sc and intracerebral E2 administration is reduced by tamoxifen administered sc at 48 h after EB-priming facilitated female sexual be-
(TMX), a selective estrogen receptor modulator that has both antagonist havior in Long-Evans rats (Pleim et al., 1990). However, when ad-
and agonist actions on estrogen receptor α and β (ERα/β) and agonist ministered 1 h before P, the PR antagonist reduced P facilitation of
effects on G-protein coupled estrogen receptor 1 (GPER-1; Filardo et al., lordosis, suggesting that RU486 has a dual effect; it may act as an an-
2000; Filardo et al., 2002; MacGregor and Jordan, 1998; Revankar tagonist in the presence of P and as an agonist in its absence.


Corresponding author at: Centro de Investigación en Reproducción Animal, Universidad Autónoma de Tlaxcala-CINVESTAV, México.
E-mail address: oglezflo@gmail.com (O. González-Flores).

https://doi.org/10.1016/j.yhbeh.2017.12.005
Received 28 June 2017; Received in revised form 8 November 2017; Accepted 15 December 2017
0018-506X/ © 2018 Elsevier Inc. All rights reserved.
R. Domínguez-Ordóñez et al. Hormones and Behavior 98 (2018) 77–87

Most of the biological effects of estrogens are mediated by the in our colony at Centro de Investigación en Reproducción Animal-
classical ERs, ERα and ERβ, which up- or down- regulate the expression Panotla. Animals were housed in a reversed light–dark cycle (14 h light,
of their target genes by binding to site-specific DNA sequences (es- 10 h dark, lights on at 2300 h) and a controlled temperature
trogen response elements) and/or specific co-regulatory proteins, in- (23 ± 2 °C) environment. They were fed Purina Rodent Laboratory
cluding co-activators and co-repressors. The role of each ER subtype in Chow 5001 and water ad libitum. Several males were used during
the expression of lordosis behavior has been explored using ER sexual behavior testing (see below).
knockout female mice, antisense oligonucleotides, and ER subtype-
specific agonists and antagonists (Dewing et al., 2007; Kudwa and 2.2. Surgical procedures
Rissman, 2003; Mazzucco et al., 2008; Ogawa et al., 1999; Ogawa et al.,
1998; Rissman et al., 1999). We recently reported that ERα and ERβ Female rats were bilaterally OVX under anesthesia with xylazine
agonists each induce lordosis behavior in estrogen-primed rats, and (4 mg/kg) and ketamine (80 mg/kg) and group housed (4/cage). One
both ER subtype-specific antagonists reduce lordosis behavior induced week later, they were anesthetized with xylazine (4 mg/kg) and keta-
by free E2 (Domínguez-Ordóñez et al., 2016). mine (80 mg/kg) and placed in a Kopf stereotaxic instrument (Tujunga,
More recently, E2 has been found to act on non-classical receptors CA) for implantation of a stainless steel cannula (22 gauge, 17-mm
localized within cell membranes or the endoplasmic reticulum length) into the right lateral ventricle following the Paxinos and Watson
(Domínguez and Micevych, 2010; Filardo and Thomas, 2012; Gaudet (2006) atlas coordinates: anteroposterior + 0.80 mm, mediolateral
et al., 2015; Revankar et al., 2005). One example of this is GPER-1, also − 1.5 mm, dorsoventral −3.5 mm with respect to bregma. A stainless
known as GPR30, which plays an important role in mediating the rapid steel screw was fixed to the skull, and both the cannula and screw were
effects of E2 (Filardo et al., 2000, 2002; Filardo and Thomas, 2012; attached to the bone with dental cement. A dummy cannula (30 gauge)
Gaudet et al., 2015). For example, this receptor regulates the release of provided with a cap was introduced into the guide cannula to prevent
gonadotropin releasing hormone (GnRH) from hypothalamic neurons clogging and contamination. Immediately after each surgical proce-
(Noel et al., 2009; Qiu et al., 2008; Rudolf and Kadokawa, 2013). Sti- dure, rats were injected with penicillin (165,000 IU/kg of procaine
mulation of GnRH neurons with G1, a selective agonist (Bologa et al., benzyl penicillin and 55,000 IU/kg of crystalline benzyl penicillin), and
2006), stimulates GnRH release, and treatment with G15 (GPER-1 an- this continued for 3 days after surgery. After surgery, rats were housed
tagonist), blocks E2-induced GnRH release (Kenealy and Terasawa, individually in plastic cages with food and water available ad libitum
2012). Furthermore, icv administration of either E2 or G1 significantly for recovery until the test day. During this time, animals did not show
increases lordosis 30 min after administration to EB-primed rats, while any apparent discomfort due to isolation.
pretreatment with G15 blocks E2 and G1 facilitation of sexual re- All of the experiments were performed under the guidelines of the
ceptivity (Long et al., 2014). Thus, GPER-1 participates in the acute E2 Mexican Law of Animal Protection (NOM-062-ZOO-1999) under the
facilitation of lordosis behavior estrogen-primed rats. GPER-1 is highly approval and supervision of the Institutional Committee for the Use and
expressed in areas involved in the expression of lordosis, such as the Care of Laboratory Animals of Centro de Investigación y de Estudios
arcuate nucleus, medial preoptic nucleus and ventromedial hypotha- Avanzados.
lamus (Brailoiu et al., 2007; Hazell et al., 2009; Long et al., 2014; Qiu
et al., 2008; Rudolf and Kadokawa, 2013). 2.3. Behavioral testing
GnRH also facilitates lordosis behavior in E2-primed rodents that
have been either OVX or OVX and adrenalectomized (Moss and Tests for sexual behavior were conducted by placing females in a
McCann, 1973; Pfaff, 1973). This effect of GnRH on female sexual be- circular Plexiglas arena (53 cm diameter) with a male after drug ad-
havior is mediated by the GnRH-1 receptor, because Antide, a GnRH-1 ministration as discussed below. The lordosis quotient [LQ = (number
receptor antagonist, blocks lordosis behavior in OVX, E2-primed rats of lordosis / 10 mounts) × 100] and lordosis score (LS) were used to
induced by several agents including GnRH, ring A-reduced progestins, assess receptive behavior in response to the first 10 mounts. LS refers to
and vaginocervical stimulation (Gómora-Arrati et al., 2008). PRs also the intensity of lordosis, which is quantified according to Hardy and
participate in lordosis behavior induced by GnRH, because RU486 in- DeBold (1971). This scale ranges from 0 to 3 for each individual re-
hibits GnRH-induced lordosis and proceptive behaviors (Beyer et al., sponse and consequently, from 0 to 30 for each female that received 10
1997). mounts. In all experiments, the rats were tested at 30, 120, and 240 min
The present study assessed the participation of GPER-1 and GnRH-1 after E2 or G1 infusion by an experimenter blind to treatment groups.
receptors in the display of lordosis induced by icv administration of G1,
a GPER-1 agonist, and by free E2 in OVX, estrogen-primed rats. 2.4. Chemicals
Experiment 1 was designed to confirm prior reports that GPER-1 fa-
cilitates lordosis in OVX, estrogen-primed rats through the adminis- E2 benzoate (EB), unesterified 17β-E2 (E2) and the GnRH-1 an-
tration of G1 and by the icv infusion of the antagonist, G15. Because the tagonist, Antide (acetyl-D-Ala(2-naphthyl)-D-Phe(4-Cl)-D-Ala(3-pyridyl)-
cellular mechanism by which GPER-1 induces lordosis behavior has not Ser-Lys(Nε-nicotinoyl)-D-Lys(Nε-nicotinoyl)-Leu-Lys(Nε-isopr-opyl)-
been well clarified, and because GPER-1 promotes GnRH release Pro-D-Ala-NH2) were purchased from Sigma Chemicals (St. Louis, MO).
(Terasawa and Kenealy, 2012), Experiment 2 tested the ability of the The GPER-1 agonist G1 ( ± )-1-[(3aR*,4S*,9bS*)-4-(6-Bromo-1,3-ben-
GnRH antagonist, Antide, to interfere with G1 and E2 facilitation of zodioxol-5-yl)-3a,4,5,9b-tetrahydro-3H-cyclopenta[c]quinolin-8-yl]-
lordosis. Because G1 may induce lordosis behavior through GnRH re- ethanone and the GPER-1 antagonist G15 (3aS*,4R*,9bR*)-4-(6-Bromo-
lease and subsequent activation of PRs and perhaps ERs, in Experiment 1,3-benzodioxol-5-yl)-3a,4,5,9b-3H-cyclopenta[c]quinoline were pur-
3, we tested the ability of the selective ER modulator, TMX, and the PR chased from Tocris Cookson (St. Louis, MO). PR antagonist, RU486, and
antagonist, RU486, to block lordosis facilitated by G1 in OVX, estrogen- the ER antagonist, TMX, were also purchased from Sigma Chemicals
primed rats. (St. Louis, MO).
EB, E2 and TMX were dissolved in sesame oil vehicle. G1, G15 and
2. Methods Antide were dissolved in 10% DMSO, and RU486 was dissolved in se-
same oil:benzyl benzoate:benzylic alcohol (80:15:5). All drugs and ve-
2.1. Animals hicles infused icv were administered in a volume of 1 μl. To avoid un-
necessary duplication and thus minimize the numbers of animals killed,
A total of 126 female rats were used in this study. Animals were important animal welfare concerns, the same groups of vehicle (DMSO)
sexually inexperienced, female Sprague Dawley rats (240–280 g) bred control animals were used throughout Experiments 1 and 2.

78
R. Domínguez-Ordóñez et al. Hormones and Behavior 98 (2018) 77–87

2.5. Experiment 1 halothane, and 1% methylene blue was administered through the
cannula. The brain was removed and sectioned in the transverse plane
2.5.1. Involvement of GPER-1 in lordosis behavior in OVX, EB-primed rats to confirm the cannula position in the right lateral ventricle. Animals
The objective of this experiment was to characterize the effect of the whose cannula was not in the ventricle (n = 7) were discarded from the
GPER-1 agonist, G1, on lordosis behavior and to establish a dose re- data analysis.
sponse curve of that compound in OVX, estrogen-primed rats. One week
after cannula implantation, rats were primed with a sc injection of 5 μg 2.9. Statistical analysis
(13.28 nmol) of EB in 0.1 ml of oil followed 40 h later by icv infusion of
G1 or 10% DMSO vehicle. G1 was infused at the following doses in To assess the effects of icv administration of G1 or E2, we first used
independent groups: DMSO (control, n = 11); G1, 3.5 (8.5 nmol, the Kruskall–Wallis test (significance level p < 0.05) for each of the
n = 8), 7.5 (18.2 nmol, n = 8), 15 (36.4 nmol, n = 9) or 30 μg three times tested. Because LQ and LS values in some groups were not
(72.8 nmol, n = 9). Those doses were chosen based on the dose of normally distributed, the results of two independent groups at 30, 120,
30 μg used by Lebesgue et al. (2009), where it was effective in inducing and 240 min were compared using the Wilcoxon Mann–Whitney U test.
prolactin secretion. This is an acceptable alternative to the t-test with a power efficiency of
95.5% of the parametric test (Siegel and Castellan, 1995).
2.6. Experiment 2 The effects of G15 and Antide on the behavioral action of G1 and E2
and the effects of TMX and RU486 on the behavioral action of G1 were
Effect of icv administration of GPER-1 and GnRH-1 receptor an- assessed by comparing the LQ obtained with G1 or E2 alone with those
tagonists on G1 induced lordosis behavior in OVX, EB-primed rats. obtained when different antagonists were added. The Wilcoxon
To test the hypothesis that lordosis behavior induced by G1 involves Mann–Whitney U test was used in this case as well. We also calculated
GPER-1 or GnRH-1 receptors, one week after stereotaxic surgery, the effect sizes (Berben et al., 2012) for differences in mean LQ scores be-
animals were injected sc with 5 μg of EB and 39.5 h later received an icv tween females treated with G1 or E2 and those who received those
injection of either G15 or Antide followed by G1 at 40 h as follows: agonists plus different inhibitors. We report Cohen's d for interpreting
25 μg (67.5 nmol) of G15 plus 15 μg of G1; 1 μg (2.4 nmol) of Antide effect sizes for the standardized mean differences.
plus 15 μg of G1 (n = 12). G15 and Antide were administered 30 min
before G1. The dose of G1 was selected based on the results of 3. Results
Experiment 1. The dose of G15 was selected from Long et al. (2014)
while the dose of Antide was selected based on previous work from our 3.1. Experiment 1
laboratory (Gomora-Arrati, et al. 2008).
3.1.1. Icv injections of G1 facilitate lordosis behavior in OVX, estrogen-
2.6.1. Effect of icv administration of GPER-1 and GnRH-1 receptor primed rats
antagonists on E2 induced lordosis behavior in OVX, EB-primed rats Table 1 indicates probability and Mann Whitney U values and effect
One week after stereotaxic surgery, the animals were injected sc sizes of different doses of G1. The control animals infused with DMSO
with 5 μg of EB and 40 h later received an icv injection of 2 ng of E2 expressed low levels of LQ at all times tested. Low LQs were observed
(n = 11). To test the hypothesis that lordosis behavior induced by E2 with most doses of G1 at 30 min post-infusion (Fig. 1; see Table 1 for
involves GPER-1 or GnRH-1 receptors, we used either G15 or Antide statistical values); only animals injected with 3.5 μg were significantly
combined with the E2 as follows: 25 μg of G15 plus E2 (n = 9); 1 μg of different from the control group receiving DMSO (p < 0.05). Com-
Antide plus E2 (n = 10). G15 and Antide were administered 30 min pared to DMSO controls, G1 at 15 (p < 0.01) and 30 (p < 0.01) μg
before E2. The dose of E2 was selected based on previous work from our induced statistically significant increases in lordosis at 120 min whereas
laboratory (Domínguez-Ordóñez et al., 2015). 3.5 (p = 0.08) and 7.5 μg (p = 0.1) were without effect. The highest
level of receptivity (LQ and LS) was observed at 240 min with all four
2.7. Experiment 3 doses (Fig. 1, Table 1). Similar results were obtained for LS (Fig. 1); the
doses of 3.5 and 15 μg (both p < 0.05 vs DMSO) induced increases at
2.7.1. Participation of ER and PR in lordosis behavior induced by G1 in 30 min, but 7.5 and 30 μg had no effect at this time. At 120 min the
OVX, EB-primed rats
This experiment was designed to determine whether lordosis be- Table 1
havior induced by G1 is mediated through PRs and/or ERs. A total of 30 Statistical values associated with lordosis quotients in rats given icv infusion of G1 when
estrogen-primed (5 μg of EB) rats were used. At 39 h after EB injection, compared to control DMSO-treated rats (Experiment 1).
9 females were injected sc with 5 mg of the progestin antagonist,
Treatment Statistical value at each test time
RU486, and at 39.5 h, 10 females received a sc injection of 5 mg of
TMX. Forty hours after EB, all rats received an icv injection of 15 μg of 30 min 120 min 240 min
G1. At 39 h after EB injection, 6 control females received a sc injection
of RU486 vehicle and at 39.5 h, 5 additional control animals received DMSO – – –
G1 3.5 μg d 1.6 1.6 1.6
TMX vehicle, followed by an icv injection of 15 μg of G1. The doses of (n = 8) p< 0.05 NS 0.05
RU486 and TMX were based on previous studies in our laboratory U 16 23 14.5
(Beyer et al., 1997; Domínguez-Ordóñez et al., 2015). Because we have G1 7.5 μg d 0.005 1.4 1
shown in multiple experiments that oil and oil:benzyl benzoate: (n = 8) p< NS NS 0.05
U 43.5 26.5 20
benzylic alcohol, vehicles of TMX and RU486, respectively, do not af-
G1 15 μg d 0.2 2.7 1
fect the expression of lordosis behavior in estrogen-primed rats (Beyer (n = 9) p< NS 0.01 0.05
et al., 1995, 1997; González-Mariscal et al., 1989), these groups were U 27 9 20.5
not included. G1 30 μg d 0.4 1.8 1.6
(n = 9) p< NS 0.01 0.01
U 48 15 13
2.8. Histological examination of cannula placement
Time (min) is relative to infusion of G1 or vehicle. DMSO, vehicle; G1, GPER-1 agonist;
One day after completion of the experiments, rats implanted with Cohen's d, effect size value; p < , probability; U, Mann Whitney U test value; NS, not
icv cannula were euthanized with an overdose of the anesthetic significant.

79
R. Domínguez-Ordóñez et al. Hormones and Behavior 98 (2018) 77–87

Fig. 1. Intracerebral infusion of 3.5 (n = 8), 7.5 (n = 8), 15 (n = 9) and 30 μg (n = 9) of G1 to OVX, EB-primed rats on LQ (A) and LS (B). Only the 3.5 μg dose of G1 induced significant
increases in lordosis behavior at 30 min post injection. At 120 and 240 min after injection of G1 all doses used significantly elevated LQ and LS compared with animal infused only with
vehicle (DMSO, n = 11; black bar). *p < 0.01, + p < 0.05 vs DMSO.

80
R. Domínguez-Ordóñez et al. Hormones and Behavior 98 (2018) 77–87

Fig. 2. The facilitation of LQ (A) and LS (B) with in-


tracerebral infusion of 15 μg of G1 (n = 9) in OVX,
EB-primed rats was significantly antagonized by icv
infusion of 25 μg of the GPER-1, antagonist G15
(67.5 mM; n = 9) or by icv infusion of 1 μg (628 μM)
of Antide, a GnRH-1 antagonist (n = 12) at 30 and
120 min after G1 injection. LQ and LS induced with G1
at 240 min was significantly reduced by Antide and
not by G15. + p < 0.05; *p < 0.01; vs G1. Data for
15 μg of G1 alone are from Fig. 1.

doses of 15 and 30 μg (both p < 0.01) increased LS, but no significant reduced at 240 min. Table 2 indicates Mann Whitney values, p values
effect was found with 3.5and 7.5 μg. However, at 240 min all doses and effect sizes for G15 and Antide combined with G1. Effect sizes
except 7.5 μg increased LS (p < 0.05 vs DMSO). (Table 2) confirm that G15 was most effective at inhibiting G1 facil-
itation of lordosis at 30 (d = 0.6) and 120 (d = 0.8) min compared to
240 min (d = 0.42). Administration of Antide also significantly pre-
3.2. Experiment 2 vented the stimulatory effect of G1 on LQ (Fig. 2A) in estrogen-primed
rats at 30, 120 (both p < 0.01) and 240 min (p < 0.05). Moreover,
3.2.1. Effect of G15 and Antide on lordosis behavior induced by G1 Antide significantly reduced the LS (Fig. 2B) at 30, 120 (both
Injection of G15 significantly decreased LQ induced by G1 at 30 and p < 0.01) and 240 min (p < 0.05). Antide had similar effect sizes as
120 min (both p < 0.01; Fig. 2A). G15 also significantly decreased the those obtained with G15 at 30, 120 and 240 min (see Table 2 for d
LS at 30 and 120 min (both p < 0.01) (Fig. 2B). The inhibitory effect values).
of G15 was transitory, because LQ and LS were not significantly

81
R. Domínguez-Ordóñez et al. Hormones and Behavior 98 (2018) 77–87

Table 2 These demonstrate the large magnitude of the inhibitory effect of those
Statistical values associated with lordosis quotients in rats given icv infusion of the agents on lordosis behavior (Lipsey and Wilson, 2000).
agonists G1 or E2 when compared to agonist plus the antagonists G15 or Antide
In Experiment 1, G1 induced lordosis with a relatively long latency.
(Experiment 2).
At 2 h after infusion, only the highest doses (15 and 30 μg) induced a
Treatment Statistical value at each time clear response, whereas at 4 h post-injection all doses induced intense
and similar levels of lordosis. In a previous report, the administration of
30 min 120 min 240 min G1 into the third ventricle induced lordosis with a very short latency of
DMSO – – – 30 min (Long et al., 2014). That study used OVX Long-Evans rats
G15 + G1 d 0.6 0.8 0.42 primed with 2 μg EB sc 4 times at four day intervals. It is possible that
(n = 9) p< 0.01 0.01 NS the use of different strains of rats (Sprague-Dawley vs Long-Evans) or
U 14 5.5 19.5 the use of repeated doses of EB by Long et al. (2014) is responsible for
Antide + G1 d 0.58 0.76 0.49
the different latencies of G1 action. It should also be noted that com-
(n = 12) p< 0.01 0.01 0.05
U 20 9 25.5 pounds administered by an icv route diffuse into many brain areas. The
G15 + E2 d 0.02 0.76 0.73 compounds move from the lateral ventricle, via bulk flow to the third
(n = 9) p< NS 0.01 0.01 ventricle, then to the fourth ventricle, then over the entire surface of the
U 44 9.5 10
brain. Thus, after icv administration of small molecules, their con-
Antide + E2 d 0.4 0.77 0.62
(n = 10) p< NS 0.01 0.05 centration decreases logarithmically in the brain with each millimeter
U 33 6.5 12 of distance (Pardridge, 2012). In order to compensate for this possible
dilution limitation, G1 (Experiment 1) was administered at different
Time (min) is relative to infusion of G1, E2 or vehicle. DMSO, vehicle; G1, GPER-1 ago- doses, in all cases obtaining the greatest responses at 120 and 240 min
nist; G15, GPER-1 antagonist; Antide, GnRH-1 receptor antagonist; E2 estradiol; Cohen's
post-injection.
d, effect size value; p < , probability; U, Mann Whitney U test value; NS, not significant.
The cellular mechanism through which G1 facilitates lordosis be-
havior is not clear. However, in Experiment 2, G15 reduced the facil-
3.2.2. Effect of G15 and Antide on lordosis behavior induced by free E2
itating effects of G1 and E2 in OVX, EB-primed rats, suggesting the
E2 infused icv induced intense lordosis behavior in EB-primed rats at
participation of GPER-1. This is in agreement with previous results in
120 and 240 min (both p < 0.01). The elevation of LQ and LS by E2
rats as well as mice (Anchan et al., 2014; Long et al., 2014) because
was significantly reduced by G15 at 120 and 240 min (both p < 0.01)
pretreatment with G15 inhibited G1 and E2 facilitation of lordosis. This
(Fig. 3); however, there was no effect on LQ or LS at 30 min. Table 2
corroborates the idea that E2 facilitates lordosis in part through the
indicates Mann Whitney U values, p values and effect sizes for G15 and
activation of GPER-1. GPER-1 has been found at a variety of in-
Antide combined with E2. G15 had a substantial effect size (Table 2) on
tracellular locations in neurons as well as in the plasma membrane in
the inhibition of lordosis (LQ) induced by E2 at 120 (d = 0.76) and 240
brain and anterior pituitary (Brailoiu et al., 2007; Filardo et al., 2007;
(d = 0.73) min, but a small effect size at 30 min (d = 0.02). Antide
Funakoshi et al., 2006). GPER-1 immunoreactivity is detected in the
significantly prevented the stimulatory effect of E2 on LQ in estrogen-
endoplasmic reticulum and plasma membrane of neuronal perikarya
primed rats at 120 (p < 0.01) and 240 min (p < 0.05), but not at
and dendritic shafts as well as in spines and in clusters of vesicles in
30 min. Moreover, Antide significantly reduced the elevation of LS in-
axon terminals of the hippocampus (Waters et al., 2015). GPER-1 im-
duced by E2 (Fig. 3B) only at 120 min (p < 0.01) post-injection. The
munostaining has also been reported in the Golgi apparatus in mag-
greatest effect sizes for Antide inhibition of LS induced by E2 were
nocellular oxytocin neurons (Sakamoto et al., 2007). Activation of
found at 120 and 240 min (see Table 2 for d values).
GPER-1can induce non-genomic signaling, such as activation of the α
subunit of the G protein, which in turn activates adenylyl cyclase.
3.3. Experiment 3 Because Antide inhibited lordosis induced by G1 and E2, we spec-
ulate that lordosis behavior was induced by the release of GnRH and the
3.3.1. Effect of TMX and RU486 on lordosis behavior induced by G1 activation of its GnRH-1 receptor. Previous findings support this in-
Systemic administration of TMX reduced LQ (Fig. 4A) and LS terpretation. a) In primates, GPER-1 is located in hypothalamic GnRH
(Fig. 4B) induced by G1 at 120 (p < 0.001) and 240 min (p < 0.01) neurons where it mediates the rapid actions of E2 on GnRH release
but not at 30 min. Table 3 indicates Mann Whitney U values, p values (Jacobi et al., 2007; Noel et al., 2009). Both E2 and G1 increase in-
and effect sizes for TMX and RU486 combined with G1. Effect sizes tracellular Ca+ and stimulate GnRH release, and these effects are
(Table 3) for TMX were large at 120 (d = 0.8) and 240 (d = 0.72) min blocked by G15 (Noel et al., 2009; Kenealy and Terasawa, 2012;
but small at 30 min (d = 0.21). LQ and LS induced by G1 at 120 and Terasawa and Kenealy, 2012). b) Lordosis behavior induced by GnRH, P
240 min were almost completely suppressed by the sc injection of and some of its ring A-reduced metabolites, leptin or vaginocervical
RU486 (both p < 0.001 vs G1 alone). However, there was no effect at stimulation is reduced by icv administration of Antide (García-Juárez
30 min for LQ or LS. Effect sizes confirm that RU486 was highly ef- et al., 2011; Gómora-Arrati et al., 2008), supporting the idea that GnRH
fective at inhibiting G1-induced lordosis at 120 and 240 min (see release is an obligatory step in the facilitation of lordosis by each of
Table 3 for d values). these.
Interestingly, Experiment 3 demonstrated the involvement of ER
4. Discussion and PR in the regulation of lordosis facilitation by GPER-1, because
both TMX and RU486, respectively, blocked lordosis behavior induced
The present study tested the hypothesis that lordosis behavior in- by G1. In previous studies, TMX injected before estrogen priming in
duced by GPER-1 activation in EB-primed rats involves GnRH-1 re- OVX rats, either into the ventromedial hypothalamus or systemically,
ceptors, PRs and ERs. The results suggest that lordosis behavior induced inhibited estrogen priming of lordosis behavior (Etgen, 1979; Etgen and
through the activation of GPER-1 by E2 or G1 may depend on the re- Shamamian, 1986; Howard et al., 1984). In addition, concurrent icv
lease of GnRH, because icv administration of Antide blocked the facil- infusion of TMX with GnRH, leptin, progestins, or PGE2 significantly
itatory actions of both compounds. TMX and RU486 also inhibited the reduced lordosis induced by these compounds in estrogen-primed rats
facilitation of lordosis by G1, suggesting the participation of both ER (Lima-Hernández et al., 2014), suggesting that ERs also participate in
and PR in this process. We believe that the roles of GnRH-1 receptor, ER activation of lordosis behavior many h after the priming administration
and PR are biologically meaningful because of the effect size estimates of E2.
of the impact of G15, TMX and RU486 treatment (Tables 2 and 3). The agonist and antagonist effects of TMX may vary according to

82
R. Domínguez-Ordóñez et al. Hormones and Behavior 98 (2018) 77–87

Fig. 3. The facilitation of LQ (A) and LS (B) with in-


tracerebral infusion of 2 ng of E2 (n = 11) in OVX, EB-
primed rats was antagonized significantly both by icv
infusion of 25 μg of the GPER-1 antagonist G15
(n = 9) or by icv infusion of 1 μg of Antide (GnRH-1
antagonist, n = 10) at 30, 120 and 240 min after E2
infusion. *p < 0.01; + p < 0.05 vs E2.

the behavior measured and the cellular process; for example, TMX E2 in the arcuate nucleus, effects associated with the blockade of the
blocks E2 induction of PR in the pituitary, but in the absence of E2, it preovulatory-like LH surge and with suppression of the E2-induced
induces PR synthesis (Roy et al., 1979; Sánchez-Criado et al., 2002). prolactin surge (Aquino et al., 2016). On the other hand, nonsteroidal
Thus, like RU486 and its action on the PR (Pleim et al., 1990), TMX antiestrogens, like TMX, ICI or nafoxidine, mimic the effects of E2 on
may act as an antagonist in the presence of E2 or other agonists and as body weight and food intake (Wade and Blaustein, 1978; Wade and
an agonist in the absence of E2. TMX appears to have primarily an- Gray, 1979; Wade and Heller, 1993; Wade et al., 1993). Another clear
tagonist effects on selected E2-dependent reproductive processes, be- agonist effect of TMX is to induce GnRH self-priming (defined as an
cause TMX prevents the E2-induced increase in cFos expression in the enhanced LH secretory response to the second of two pulses of identical
anteroventral periventricular nucleus and also prevents PR induction by concentration of GnRH), probably through binding ERα (Sánchez-

83
R. Domínguez-Ordóñez et al. Hormones and Behavior 98 (2018) 77–87

Fig. 4. LQ (A) and LS (B) were induced in OVX, EB-


primed rats by icv administration of 15 μg of G1
(n = 11). The lordosis behavior induced at 30 min (LQ
and LS) was not significantly reduced by sc adminis-
tration of 5 mg of TMX (ER antagonist, n = 10) or
5 mg of antiprogestin RU486 (RU; n = 9). Lordosis
behavior induced by G1 at 120 and 240 min post-in-
jection was significantly inhibited by sc administration
of TMX or RU486. &p < 0.001; *p < 0.01 vs G1.

Criado et al., 2002, 2005, 2006). that study and the present one that could account for these differing
Recent work by Long et al. (2017) explored time-dependent effects results. First, the rat strains used differed between studies (we used
of TMX and ICI on lordosis expression in EB-primed rats when the drugs Sprague Dawley, whereas the Long study used the Long Evans strain).
were infused via different routes. They showed that systemic adminis- Secondly, the manner in which lordosis was induced differed im-
tration of TMX or icv infusion of ICI facilitated lordosis 4 h later in OVX, portantly between the two studies. In the Long study, lordosis was in-
EB-primed rats. However, when either ER antagonist was directly in- duced in EB-primed rats by the administration of TMX or ICI alone; in
fused in the arcuate nucleus, they induced more intense lordosis be- the present study, lordosis was induced in EB-primed rats by E2 or G1,
havior and with a shorter latency (30 min). This effect was mediated and in this case, TMX inhibited G1-dependent lordosis induction. We
through the activation of GPER-1, because G15 reduced the lordosis did not test for potential agonist effects of TMX alone. Moreover, the
induced by those compounds. strongest, most rapid facilitatory effect of TMX on lordosis behavior was
In contrast to the study by Long et al. (2017), we observed that observed when TMX was infused directly into the arcuate nucleus in the
TMX, administered icv, inhibited G1 facilitation of lordosis behavior in Long study. In the present study, TMX was administered icv only, and
EB-primed rats. There are some methodological differences between relatively rapid inhibition of G1-induced lordosis was observed. As

84
R. Domínguez-Ordóñez et al. Hormones and Behavior 98 (2018) 77–87

Table 3 Ordóñez et al., 2015; García-Juárez et al., 2011; Lima-Hernández et al.,


Statistical values associated with lordosis quotients in rats given icv infusion of G1 2014; Mani and Blaustein, 2012; Mani and Portillo, 2010). The fact that
compared to the antagonists TMX and RU486 (Experiment 3).
RU486 reduced lordosis behavior induced by G1 in the present study
Treatment Statistical value at each time suggests that G1 activates PRs, perhaps indirectly via GnRH release.
This idea is supported by the ability of RU486 to block GnRH self-
30 min 120 min 240 min priming, which requires cross-communication between GnRH receptor-
activated protein kinase A and the PR (Turgeon and Waring, 1994;
DMSO – – –
TMX + G1 d 0.21 0.8 0.72 Waring and Turgeon, 1992). Overall, the results suggest that GPER-1 is
(n = 10) p< NS 0.001 0.01 involved in the stimulation of lordosis behavior in estrogen-primed
U 24 2 7 female rats. Thus, we propose that E2 and G1 bind directly to GPER-1
RU486 + G1 d 0.15 0.84 0.88 (upper panel, Fig. 5), which is present in GnRH neurons in the preoptic
(n = 9) p< NS 0.001 0.001
area and which stimulates an increase in intracellular calcium, inducing
U 28 1.5 0.5
the release of GnRH (Naor et al., 2000; Weiner and Charles, 2001: Kraus
Time (min) is relative to infusion of G1 or vehicle. G1, GPER-1 agonist; TMX, tamoxifen, et al., 2003: Navratil et al., 2003; Higa-Nakamine et al., 2015). GnRH
selective ER modulator; RU486, PR antagonist; Cohen's d, effect size value; p < , prob- neurons project to other regions of the hypothalamus (e.g., para-
ability; U, Mann Whitney U test value; NS, not significant. ventricular nucleus) containing cells that express the GnRH-1 receptor
(Silverman et al., 1987; Wierman et al., 2011; Schauer et al., 2015). The
noted previously, this suggests that TMX may act as an antagonist in the released GnRH may then activate its membrane receptor (GnRH-1) lo-
presence of E2 or other agonists and as an agonist in the absence of E2. calized in postsynaptic neurons, which contain PRs and ERs. The GnRH-
At the level of neural mechanisms, the different effects of TMX on 1 receptor-dependent increase in second messenger production and
lordosis behavior between these two studies could also be due to the kinase activation may then modulate lordosis behavior by activating
fact that TMX, in some experimental conditions, inhibits GnRH release. both ERs and PRs in a ligand-independent manner (Beyer et al., 2003;
For example, TMX effectively prevented the positive, but not negative, Demay et al., 2001; Mani and Blaustein, 2012; Mani and Portillo,
feedback effects of E2 (Donath and Nishino, 1998; Gao et al., 2002; 2010).
Aquino et al., 2016). An alternative intracellular mechanism by which GnRH-1 receptors
The inhibitory effect of RU486 on the actions of G1 also agrees with could induce lordosis behavior is by activating the mitogen-activated
previous reports showing that this PR antagonist reduces lordosis be- protein kinase (MAPK) pathway through the Src/ER/PR/MAPK com-
havior induced by a variety of compounds (Beyer et al., 1995; Beyer plex in hypothalamic neurons (lower panel, Fig. 5; Higa-Nakamine
et al., 1997; Domínguez-Ordóñez et al., 2015; Etgen and Shamamian, et al., 2015; Kraus et al., 2003; Navratil et al., 2003; Naor et al., 2000).
1986; García-Juárez et al., 2011; González-Mariscal et al., 1989; Mani PRs bind the SH3 domain of Src through its proline-rich domain (re-
and Portillo, 2010; Mani and Blaustein, 2012; Vathy et al., 1987). On sidues 421 and 428 located in the N-terminus), while ERs interact di-
the other hand, in certain experimental conditions, RU486 exerts ago- rectly with the SH2 domains through phosphotyrosine 537 located also
nist effects on lordosis behavior (Pleim et al., 1990). However, an at the N-terminus (Ballaré et al., 2003; Boonyaratanakornkit et al.,
agonist effect of the antiprogestin was only observed in the absence of 2001; Faivre and Lange, 2007; Migliaccio et al., 1998). Therefore, PR
P. Several hormone antagonists, like RU486 and TMX, induce partial and ER could heterodimerize through the ERIDI and II (ER-interaction
agonist effects in the absence of natural hormone by a mechanism that domain I and II) in the N-terminal domain of the PR and the ligand-
is not well understood. binding domain of ER. Indeed others (Migliaccio et al., 1998;
Some studies suggest that PRs are common molecular effectors for Boonyaratanakornkit et al., 2001) showed that activation of Src/MAPK
compounds with diverse chemical structures that induce lordosis be- through the progestin agonist, R5020, was blocked by administration of
havior in OVX, estrogen-primed rats (Beyer et al., 2003; Domínguez-

Fig. 5. Proposed cellular mechanisms involved in the facilitation


of lordosis behavior by GPER-1 activation. Upper panel: E2 and
G1 bind to GPER-1 receptor located in the membrane of GnRH
neurons in the preoptic area, activating a G protein (GP) to sti-
mulate the adenylyl cyclase-cAMP-protein kinase A (AC-cAMP-
PKA) system. This increases intracellular calcium (Ca++) con-
centration, which induces GnRH release. Lower panel: GnRH
interaction with the membrane GnRH-1 receptor, located in
postsynaptic hypothalamic neurons. GnRH-1 receptors interact
with a GP to activate the Src-ER-PR-MAPK-complex. PR binds to
the SH3 domain, whereas the ER binds to the Sh2 domain of Src
(see Discussion for references). These interactions activate
MAPK. In turn, MAPK phosphorylates the PR, leading to the
facilitation of lordosis behavior. TMX and RU486 bind to ER and
PR, respectively, where they destabilize the Src complex, pre-
venting the activation of MAPK and the expression of lordosis.
Abbreviations: GPER-1, G-protein coupled estrogen receptor 1;
G1, GPER-1 agonist; E2 17β-estradiol; PR, progestin receptor;
ER, estrogen receptor; GnRH, gonadotropin releasing hormone;
MAPK, mitogen-activated protein kinase.

85
R. Domínguez-Ordóñez et al. Hormones and Behavior 98 (2018) 77–87

TMX, suggesting the participation of both the ER and PR. Dewing, P., Boulware, M.I., Sinchak, K., Christensen, A., Mermelstein, P.G., Micevych, P.,
We propose that the ER and PR may be participating as anchor 2007. Membrane estrogen receptor-alpha interactions with metabotropic glutamate
receptor 1a modulate female sexual receptivity in rats. J. Neurosci. 27, 9294–9300.
proteins forming the Src/ER/PR complex and subsequently activating Domínguez, R., Micevych, P.E., 2010. Estradiol rapidly regulates membrane estrogen
MAPK. In turn, MAPK would be the component that activates the ER receptor α levels in hypothalamic neurons. J. Neurosci. 30, 12589–12596.
(Jiang et al., 2007; Kato et al., 1995; Law et al., 2009) and PR (Hagan Domínguez-Ordóñez, R., García-Juárez, M., Lima-Hernández, F.J., Gómora-Arrati, P.,
Blaustein, J.D., González-Flores, O., 2015. Sexual receptivity facilitated by un-
et al., 2012; Lange, 2004; Shen et al., 2001). This cellular mechanism esterified estradiol: dependence on estrogen and progestin receptors and priming
could participate in the regulation of lordosis behavior induced by dose of estradiol benzoate. Behav. Neurosci. 129, 777–788.
several compounds in OVX, estrogen-primed rats (González-Flores Domínguez-Ordóñez, R., García-Juárez, M., Lima-Hernández, F.J., Gómora-Arrati, P.,
Blaustein, J.D., Etgen, A.M., González-Flores, O., 2016. Estrogen receptor α and β are
et al., 2010). In support of this proposal, we showed that icv adminis- involved in the activation of lordosis behavior in estradiol-primed rats. Horm. Behav.
tration of both PD98059 (inhibitor of MAPK) or PP2 (inhibitor of Src) 1, 1–7.
reduced lordosis behavior induced by icv administration of GnRH Donath, J., Nishino, Y., 1998. Effects of partial versus pure antiestrogens on ovulation and
the pituitary-ovarian axis in the rat. J. Steroid Biochem. Mol. Biol. 66, 247–254.
(González-Flores et al., 2009; Lima-Hernández et al., 2012). Moreover,
Etgen, A.M., 1979. Antiestrogens: effects of tamoxifen, nafoxidine, and CI-628 on sexual
in vitro studies showed that GnRH activation of its GnRH-1 receptor behavior, cytoplasmic receptors, and nuclear binding of estrogen. Horm. Behav. 13,
activates the Src–MAPK pathway (Higa-Nakamine et al., 2015; Kraus 97–112.
et al., 2003). Based on these data, the fact that lordosis behavior in- Etgen, A.M., Shamamian, P., 1986. Regulation of estrogen-stimulated lordosis behavior
and hypothalamic progestin receptor induction by antiestrogens in female rats.
duced by E2 or G1 was blocked with either RU486 or TMX supports the Horm. Behav. 20, 166–180.
participation of Src/PR/ER/MAPK complex in the regulation of lor- Faivre, E.J., Lange, C.A., 2007. Progesterone receptors upregulate Wnt-1 to induce epi-
dosis. In T47D cells in vitro, the progestin R5020 activates this dermal growth factor receptor transactivation and c-Src-dependent sustained acti-
vation of Erk1/2 mitogen-activated protein kinase in breast cancer cells. Mol. Cell.
pathway. Moreover, both RU486 and TMX inhibit progestin stimulation Biol. 27, 466–480.
of c-Src and the MAPK, Erk-2 (Migliaccio et al., 1998). In addition, Filardo, E.J., Thomas, P., 2012. Minireview: G protein-coupled estrogen receptor-1,
because Antide also reduced lordosis behavior induced by E2 and G1, GPER-1: its mechanism of action and role in female reproductive cancer, renal and
vascular physiology. Endocrinology 153, 2953–2962.
we suggest that these compounds may facilitate lordosis by promoting Filardo, E.J., Quinn, J.A., Bland, K.I., Frackelton Jr., A.R., 2000. Estrogen-induced acti-
GnRH release from the hypothalamus, which acts on the GnRH-1 re- vation of Erk-1 and Erk-2 requires the G protein-coupled receptor homolog, GPR30,
ceptor to activate the Src-MAPK pathway. Nonetheless, this is only a and occurs via trans-activation of the epidermal growth factor receptor through re-
lease of HB-EGF. Mol. Endocrinol. 14, 1649–1660.
hypothetical model meant to stimulate further experimentation. Filardo, E.J., Quinn, J.A., Frackelton Jr., A.R., Bland, K.I., 2002. Estrogen action via the G
protein-coupled receptor, GPR30: stimulation of adenylyl cyclase and cAMP-medi-
Acknowledgement ated attenuation of the epidermal growth factor receptor-to-MAPK signaling axis.
Mol. Endocrinol. 16, 70–84.
Filardo, E., Quinn, J., Pang, Y., Graeber, C., Shaw, S., Dong, J., Thomas, P., 2007.
This work was supported by Consejo Nacional de Ciencia y Activation of the novel estrogen receptor G protein-coupled receptor 30 (GPR30) at
Tecnología (CONACYT) Grant Number 134291 and by PRODEP Grant the plasma membrane. Endocrinology 148, 3236–3245.
Number DSA/103.5/16/12094. Funakoshi, T., Yanai, A., Shinoda, K., Kawano, M.M., Mizukami, Y., 2006. G protein-
coupled receptor 30 is an estrogen receptor in the plasma membrane. Biochem.
Biophys. Res. Commun. 346, 904–910.
References Gao, X., Petroff, B.K., Oluola, O., Georg, G., Terranova, P.F., Rozman, K.K., 2002.
Endocrine disruption by indole-3-carbinol and tamoxifen: blockage of ovulation.
Toxicol. Appl. Pharmacol. 183, 179–188.
Anchan, D., Gafur, A., Sano, K., Ogawa, S., Vasudevan, N., 2014. Activation of the GPR30 García-Juárez, M., Beyer, C., Soto-Sánchez, A., Domínguez-Ordóñez, R., Gómora-Arrati,
receptor promotes lordosis in female mice. Neuroendocrinology 100, 71–80. P., Lima-Hernández, F.J., Eguibar, J.R., Etgen, A.M., González-Flores, O., 2011.
Aquino, N.S., Araujo-Lopes, R., Batista, I.A., Henriques, P.C., Poletini, M.O., Franci, C.R., Leptin facilitates lordosis behavior through GnRH-1 and progestin receptors in es-
Reis, A.M., Szawka, R.E., 2016. Hypothalamic effects of tamoxifen on oestrogen trogen-primed rats. Neuropeptides 45, 63–67.
regulation of luteinising hormone and prolactin secretion in female rats. J. Gaudet, H.M., Cheng, S.B., Christensen, E.M., Filardo, E.J., 2015. The G-protein coupled
Neuroendocrinol. 28, 1–13. estrogen receptor, GPER: the inside and inside-out story. Mol. Cell. Endocrinol. 418,
Ballaré, C., Uhrig, M., Bechtold, T., Sancho, E., Di Domenico, M., Migliaccio, A., 207–219.
Auricchio, F., Beato, M., 2003. Two domains of the progesterone receptor interact Gómora-Arrati, P., Beyer, C., Lima-Hernández, F.J., Gracia, M.E., Etgen, A.M., González-
with the estrogen receptor and are required for progesterone activation of the c-Src/ Flores, O., 2008. GnRH mediates estrous behavior induced by ring A reduced pro-
Erk pathway in mammalian cells. Mol. Cell. Biol. 23, 1994–2008. gestins and vaginocervical stimulation. Behav. Brain Res. 187, 1–8.
Berben, L., Sereika, S.M., Engberg, S., 2012. Effect size estimation: methods and ex- González-Flores, O., Gómora-Arrati, P., García-Juárez, M., Gómez-Camarillo, M.A., Lima-
amples. Int. J. Nurs. Stud. 49, 1039–1047. Hernández, F.J., Beyer, C., Etgen, A.M., 2009. Nitric oxide and ERK/MAPK mediation
Beyer, C., Moralí, G., Vargas, R., 1971. Effect of diverse estrogens on estrous behavior and of estrous behavior induced by GnRH, PGE2 and db-cAMP in rats. Physiol. Behav. 23
genital tract development in ovariectomized rats. Horm. Behav. 2, 273–277. (96), 606–612.
Beyer, C., González-Flores, O., González-Mariscal, G., 1995. Ring A reduced progestins González-Flores, O., Beyer, C., Gómora-Arrati, P., García-Juárez, M., Lima-Hernández,
potently stimulate estrous behavior in rats: paradoxical effect through the proges- F.J., Soto-Sánchez, A., Etgen, A.M., 2010. A role for Src kinase in progestin facilita-
terone receptor. Physiol. Behav. 58, 985–993. tion of estrous behavior in estradiol-primed female rats. Horm. Behav. 58, 223–229.
Beyer, C., González-Flores, O., González-Mariscal, G., 1997. Progesterone receptor par- González-Mariscal, G., González-Flores, O., Beyer, C., 1989. Intrahypothalamic injection
ticipates in the stimulatory effect of LHRH, prostaglandin E2, and cyclic AMP on of RU486 antagonizes the lordosis induced by ring A-reduced progestins. Physiol.
lordosis and proceptive behaviours in rats. J. Neuroendocrinol. 9, 609–614. Behav. 46, 435–438.
Beyer, C., González-Flores, O., García-Juárez, M., González-Mariscal, G., 2003. Non-li- Hagan, C.R., Daniel, A.R., Dressing, G.E., Lange, C.A., 2012. Role of phosphorylation in
gand activation of estrous behavior in rodents: cross-talk at the progesterone re- progesterone receptor signaling and specificity. Mol. Cell. Endocrinol. 357, 43–49.
ceptor. Scand. J. Psychol. 44, 221–229. Hardy, D.F., DeBold, J.F., 1971. The relationship between levels of exogenous hormones
Boling, J.M., Blandau, R., 1939. The estrogen-progesterone induction of mating response and the display of lordosis by the female rat. Horm. Behav. 2, 287–297.
in the spayed female rat. Endocrinology 25, 359–364. Hazell, G.G., Yao, S.T., Roper, J.A., Prossnitz, E.R., O'Carroll, A.M., Lolait, S.J., 2009.
Bologa, C.G., Revankar, C.M., Young, S.M., Edwards, B.S., Arterburn, J.B., Kiselyov, A.S., Localization of GPR30, a novel G protein-coupled estrogen receptor, suggests mul-
Parker, M.A., Tkachenko, S.E., Savchuck, N.P., Sklar, L.A., Oprea, T.I., Prossnitz, E.R., tiple functions in rodent brain and peripheral tissues. J. Endocrinol. 202, 223–236.
2006. Virtual and biomolecular screening converge on a selective agonist for GPR30. Higa-Nakamine, S., Maeda, N., Toku, S., Yamamoto, H., 2015. Involvement of protein
Nat. Chem. Biol. 2, 207–212. kinase D1 in signal transduction from the protein kinase C pathway to the tyrosine
Boonyaratanakornkit, V., Scott, M., Ribon, V., Sherman, L., Anderson, S., Maller, J., kinase pathway in response to gonadotropin-releasing hormone. J. Biol. Chem. 290,
Miller, W., Edwards, D.A., 2001. Progesterone receptor contains a proline-rich motif 25974–25985.
that directly interacts with SH3 domains and activates c-Src family tyrosine kinase. Howard, S.B., Etgen, A.M., Barfield, R.J., 1984. Antagonism of central estrogen action by
Mol. Cell 8, 269–280. intracerebral implants of tamoxifen. Horm. Behav. 18, 256–266.
Brailoiu, E., Dun, S.L., Brailoiu, G.C., Mizuo, K., Sklar, L.A., Oprea, T.I., Prossnitz, E.R., Jacobi, J.S., Martin, C., Nava, G., Jeziorski, M.C., Clapp, C., Martínez de la Escalera, G.,
Dun, N.J., 2007. Distribution and characterization of estrogen receptor G protein- 2007. 17-Beta estradiol directly regulates the expression of adrenergic receptors and
coupled receptor 30 in the rat central nervous system. J. Endocrinol. 193, 311–321. kisspeptin/GPR54 system in GT1-7 GnRH neurons. Neuroendocrinoly 86, 260–269.
Davidson, J., Smith, E., Rodgers, C., Bloch, G., 1968. Relative thresholds of behavioral Jiang, J., Sarwar, N., Peston, D., Kulinskaya, E., Shousha, S., Coombes, R.C., Ali, S., 2007.
and somatic responses to estrogen. Physiol. Behav. 3, 227–229. Phosphorylation of estrogen receptor-alpha at Ser167 is indicative of longer disease-
Demay, F., De Monti, M., Tiffoche, C., Vaillant, C., Thieulant, M.L., 2001. Steroid-in- free and overall survival in breast cancer patients. Clin. Cancer Res. 13, 5769–5776.
dependent activation of ER by GnRH in gonadotrope pituitary cells. Endocrinology Kato, S., Endoh, H., Masuhiro, Y., Kitamoto, T., Uchiyama, S., Sasaki, H., Masushige, S.,
142, 3340–3347. Gotoh, Y., Nishida, E., Kawashima, H., Metzger, D., Chambon, P., 1995. Activation of

86
R. Domínguez-Ordóñez et al. Hormones and Behavior 98 (2018) 77–87

the estrogen receptor through phosphorylation by mitogen-activated protein kinase. 985–991.


Science 270, 1491–1494. Revankar, C.M., Cimino, D.F., Sklar, L.A., Arterburn, J.B., Prossnitz, E.R., 2005. A
Kenealy, B.P., Terasawa, E., 2012. Rapid direct action of estradiol in GnRH neurons: transmembrane intracellular estrogen receptor mediates rapid cell signaling. Science
findings and implications. Front. Endocrinol. 2, 106. 307, 1625–1630.
Kraus, S., Benard, O., Naor, Z., Seger, R., 2003. c-Src is activated by the epidermal growth Rissman, E.F., Wersinger, S.R., Fugger, H.N., Foster, T.C., 1999. Sex with knockout
factor receptor in a pathway that mediates JNK and ERK activation by gonadotropin- models: behavioral studies of estrogen receptor alpha. Brain Res. 835, 80–90.
releasing hormone in COS7 cells. J. Biol. Chem. 278, 32618–32630. Roy, E.J., MacLusky, N.J., McEwen, B.S., 1979. Antiestrogen inhibits the induction of
Kudwa, A.E., Rissman, E.F., 2003. Double oestrogen receptor alpha and beta knockout progestin receptor by estradiol in the hypothalamus-preoptic area and pituitary.
mice reveal differences in neural oestrogen-mediated progestin receptor induction Endocrinology 104, 1333–1336.
and female sexual behaviour. J. Neuroendocrinol. 15, 978–983. Rudolf, F.O., Kadokawa, H., 2013. Expression of estradiol receptor, GPR30, in bovine
Lange, C.A., 2004. Making sense of cross-talk between steroid hormone receptors and anterior pituitary and effects of GPR30 agonist on GnRH-induced LH secretion. Anim.
intracellular signaling pathways: who will have the last word? Mol. Endocrinol. 2, Reprod. Sci. 139, 9–17.
269–278. Sakamoto, H., Matsuda, K., Hosokawa, K., Nishi, M., Morris, J.F., Prossnitz, E.R., Kawata,
Law, W.S., Chan, R.Y., Guo, D.A., Wong, M.S., 2009. Mitogen-activated protein kinase M., 2007. Expression of G protein-coupled receptor-30, a G protein-coupled mem-
(MAPK) pathway mediates the oestrogen-like activities of ginsenoside Rg1 in human brane estrogen receptor, in oxytocin neurons of the rat paraventricular and su-
breast cancer (MCF-7) cells. Br. J. Pharmacol. 156, 1136–1146. praoptic nuclei. Endocrinology 148 (2007), 5842–5850.
Lebesgue, D., Reyna-Neyra, A., Huang, X., Etgen, A.M., 2009. GPR30 differentially reg- Sánchez-Criado, J.E., Guelmes, P., Bellido, C., Gónzalez, M., Hernández, G., Aguilar, R.,
ulates short latency responses of luteinising hormone and prolactin secretion to Garrido-Gracia, J.C., Bello, A.R., Alonso, R., 2002. Tamoxifen but not other selective
oestradiol. J. Neuroendocrinol. 21, 743–752. estrogen receptor modulators antagonizes estrogen actions on luteinizing hormone
Lima-Hernández, F.J., Beyer, C., Gómora-Arrati, P., García-Juárez, M., Encarnación- secretion while inducing gonadotropin-releasing hormone self-priming in the rat.
Sánchez, J.L., Etgen, A.M., González-Flores, O., 2012. Src kinase signaling mediates Neuroendocrinology 76, 203–213.
estrous behavior induced by 5β-reduced progestins, GnRH, prostaglandin E2 and Sánchez-Criado, J.E., de Las Mulas, J.M., Bellido, C., Aguilar, R., Garrido-Gracia, J.C.,
vaginocervical stimulation in estrogen-primed rats. Horm. Behav. 62, 579–584. 2005. Gonadotrope oestrogen receptor-alpha and -beta and progesterone receptor
Lima-Hernández, F.J., Gómora-Arrati, P., García-Juárez, M., Blaustein, J.D., Etgen, A.M., immunoreactivity after ovariectomy and exposure to oestradiol benzoate, tamoxifen
Beyer, C., González-Flores, O., 2014. Estrogen receptors regulate the estrous behavior or raloxifene in the rat: correlation with LH secretion. J. Endocrinol. 184, 59–68.
induced by progestins, peptides, and prostaglandin E2. Horm. Behav. 66, 361–368. Sánchez-Criado, J.E., Garrido-Gracia, J.C., Bellido, C., Aguilar, R., Guelmes, P., Abreu, P.,
Lipsey, M.W., Wilson, M.D., 2000. Practical Meta-analysis, 1st ed. Sage Publications, Alonso, R., Barranco, I., Millán, Y., de Las Mulas, J.M., 2006. Oestradiol-17beta in-
Thousand Oaks. New Delhi, London. hibits tamoxifen-induced LHRH self-priming blocking hormone-dependent and li-
Long, N., Serey, C., Sinchak, K., 2014. 17β-estradiol rapidly facilitates lordosis through G gand-independent activation of the gonadotrope progesterone receptor in the rat. J.
protein-coupled estrogen receptor 1 (GPER) via deactivation of medial preoptic nu- Endocrinol. 190, 73–84.
cleus μ-opioid receptors in estradiol primed female rats. Horm. Behav. 66, 663–666. Schauer, C., Tong, T., Petitjean, H., Blum, T., Peron, S., Mai, O., Schmitz, F., Boehm, U.,
Long, N., Long, B., Mana, A., Le, D., Nguyen, L., Chokr, S., Sinchak, K., 2017. Tamoxifen Leinders-Zufall, T., 2015. Hypothalamic gonadotropin-releasing hormone (GnRH)
and ICI 182,780 activate hypothalamic G protein-coupled estrogen receptor 1 to receptor neurons fire in synchrony with the female reproductive cycle. J.
rapidly facilitate lordosis in female rats. Horm. Behav. 89, 98–103. Neurophysiol. 114 (2), 1008–1021 (2015 Aug).
MacGregor, J.I., Jordan, V.C., 1998. Basic guide to the mechanisms of antiestrogen ac- Shen, T., Horwitz, K.B., Lange, C.A., 2001. Transcriptional hyperactivity of human pro-
tion. Pharmacol. Rev. 50, 151–196. gesterone receptors is coupled to their ligand-dependent down-regulation by mitogen
Mani, S.K., Blaustein, J.D., 2012. Neural progestin receptors and female sexual behavior. activated protein kinase-dependent phosphorylation of serine 294. Mol. Cell. Biol. 21,
Neuroendocrinology 96, 152–161. 6122–6131.
Mani, S., Portillo, W., 2010. Activation of progestin receptors in female reproductive Siegel, S., Castellan, N.J., 1995. Estadística no paramétrica aplicada a las ciencias de la
behavior: interactions with neurotransmitters. Front. Neuroendocrinol. 31, 157–171. conducta, 4th ed. Trillas, México.
Mazzucco, C.A., Walker, H.A., Pawluski, J.L., Lieblich, S.E., Galea, L.A., 2008. ERalpha, Silverman, A.J., Jhamandas, J., Renaud, L.P., 1987. Localization of luteinizing hormone-
but not ERbeta, mediates the expression of sexual behavior in the female rat. Behav. releasing hormone (LHRH) neurons that project to the median eminence. J. Neurosci.
Brain Res. 191, 111–117. 8, 2312–2319.
Migliaccio, A., Piccolo, D., Castoria, G., DiDomenico, M., Bilancio, A., Lombardi, M., Terasawa, E., Kenealy, B.P., 2012. Neuroestrogen, rapid action of estradiol, and GnRH
Gong, W., Beato, M., Auricchio, F., 1998. Activation of the Src/p21ras/Erk pathway neurons. Front. Neuroendocrinol. 33, 364–375.
by progesterone receptor via cross-talk with estrogen receptor. EMBO J. 17, Turgeon, J.L., Waring, D.W., 1994. Activation of the progesterone receptor by the go-
2008–2018. nadotropin-releasing hormone self-priming signaling pathway. Mol. Endocrinol. 8,
Moss, R.L., McCann, S.M., 1973. Induction of mating behavior in rats by luteinizing 860–869.
hormone-releasing factor. Science 181, 177–179. Vathy, I.U., Etgen, A.M., Barfield, R.J., 1987. Actions of progestins on estrous behavior in
Naor, Z., Benard, O., Seger, R., 2000. Activation of MAPK cascades by G-protein-coupled female rats. Physiol. Behav. 40, 591–595.
receptors: the case of gonadotropin-releasing hormone receptor. Trends Endocrinol. Vivacqua, A., Bonofiglio, D., Albanito, L., Madeo, A., Rago, V., Carpino, A., Musti, A.M.,
Metab. 11, 91–99. Picard, D., Andò, S., Maggiolini, M., 2006. 17beta-estradiol, genistein, and 4-hy-
Navratil, A.M., Bliss, S.P., Berghorn, K.A., Haughian, J.M., Farmerie, T.A., Graham, J.K., droxytamoxifen induce the proliferation of thyroid cancer cells through the g protein-
Clay, C.M., Roberson, M.S., 2003. Constitutive localization of the gonadotropin-re- coupled receptor GPR30. Mol. Pharmacol. 70, 1414–1423.
leasing hormone (GnRH) receptor to low density membrane microdomains is ne- Wade, G.N., Blaustein, J.D., 1978. Effects of an anti-estrogen on neural estradiol binding
cessary for GnRH signaling to ERK. J. Biol. Chem. 278, 31593–31602. and on behaviors in female rats. Endocrinology 102, 245–251.
Noel, S.D., Keen, K.L., Baumann, D.I., Filardo, E.J., Terasawa, E., 2009. Involvement of G- Wade, G.N., Gray, J.M., 1979. Gonadal effects on food intake and adiposity: a metabolic
protein coupled receptor 30 (GPR30) in rapid action of estrogen in primate LHRH hypothesis. Physiol. Behav. 22, 583–593.
neurons. Mol. Endocrinol. 23, 349–359. Wade, G.N., Heller, H.W., 1993. Tamoxifen mimics the effects of estradiol on food intake,
Ogawa, S., Eng, V., Taylor, J., Lubahn, D.B., Korach, K.S., Pfaff, D.W., 1998. Roles of body weight, and body composition in rats. Am. J. Phys. 264, R1219–R1223.
estrogen receptor-alpha gene expression in reproduction-related behaviors in female Wade, G.N., Powers, J.B., Blaustein, J.D., Green, D.E., 1993. ICI 182,780 antagonizes the
mice. Endocrinology 139, 5070–5081. effects of estradiol on estrous behavior and energy balance in Syrian hamsters. Am. J.
Ogawa, S., Chan, J., Chester, A.E., Gustafsson, J.A., Korach, K.S., Pfaff, D.W., 1999. Phys. 265, R1399–R1403.
Survival of reproductive behaviors in estrogen receptor beta gene-deficient Waring, D.W., Turgeon, J.L., 1992. A pathway for luteinizing hormone releasing-hormone
(betaERKO) male and female mice. Proc. Natl. Acad. Sci. U. S. A. 96, 12887–12892. self-potentiation: cross-talk with the progesterone receptor. Endocrinology 130,
Pardridge, W.M., 2012. Drug transport across the blood-brain barrier. J. Cereb. Blood 3275–3282.
Flow Metab. 32, 1959–1972. Waters, E.M., Thompson, L., Patel, P., Gonzales, A.D., Ye, H.Z., Filardo, E.J., Clegg, D.J.,
Paxinos, G., Watson, C., 2006. The Rat Brain in Stereotaxic Coordinates, 6th ed. Academic 2015. G-protein-coupled estrogen receptor 1 is anatomically positioned to modulate
Press, Sydney, Australia. synaptic. J. Neurosci. 35, 2384–2397.
Pfaff, D.W., 1973. Luteinizing hormone-releasing factor potentiates lordosis behavior in Weiner, R.I., Charles, A., 2001. Regulation of gonadotropin-releasing hormone release by
hypophysectomized ovariectomized female rats. Science 182, 1148–1149. cyclic AMP signalling pathways. Growth Hormon. IGF Res. 11, S9–S15.
Pleim, E.T., Cailliau, P.J., Weinstein, M.A., Etgen, A.M., Barfield, R.J., 1990. Facilitation Wierman, M.E., Kiseljak-Vassiliades, K., Tobet, S., 2011. Gonadotropin-releasing hor-
of receptive behavior in estrogen-primed female rats by the anti-progestin, RU 486. mone (GnRH) neuron migration: initiation, maintenance and cessation as critical
Horm. Behav. 24, 301–310. steps to ensure normal reproductive function. Front. Neuroendocrinol. 32, 43–52.
Qiu, J., Ronnekleiv, O.K., Kelly, M.J., 2008. Modulation of hypothalamic neuronal ac- Zemlan, F.P., Adler, N.T., 1977. Hormonal control of female sexual behavior in the rat.
tivity through a novel G-protein-coupled estrogen membrane receptor. Steroids 73, Horm. Behav. 9, 345–357.

87

You might also like