Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2015 Jun 18.
Published in final edited form as: Biochim Biophys Acta. 2010 Jan 25;1800(10):1084–1089. doi: 10.1016/j.bbagen.2010.01.008

Contributions of estrogen receptor-α and estrogen receptor-β to the regulation of behavior

Marc J Tetel a, Donald W Pfaff b,*
PMCID: PMC4472314  NIHMSID: NIHMS516092  PMID: 20097268

Abstract

Studies of the mechanisms by which estrogens influence brain function and behavior have advanced from the explication of individual hormone receptors, neural circuitry and individual gene expression. Now, we can report patterns of estrogen receptor subtype contributions to patterns of behavior. Moreover, new work demonstrates important contributions of nuclear receptor coactivator expression in the central nervous system. In this paper, our current state of knowledge is reviewed.

Keywords: Estrogen receptor, Hypothalamus, Preoptic area, Lordosis, Aggression, Progestin receptor, Sexual differentiation, Steroid receptor coactivator-1 (SRC-1)


Gene products coding for nuclear hormone receptors have afforded neuroscientists unusual opportunities for the analysis of brain mechanisms that regulate behavior. Brain researchers benefit from the relatively simple chemistry of steroid hormones and from the large numbers of reagents effective in the manipulation of hormone-dependent systems of cells. Success during the recent scientific generations of work in this field has allowed us to understand how sex steroid actions in the brain coordinate behavioral regulation with other factors in the body and in the environment. The upshot is that we understand mechanisms with physical dimensions ranging from Angstrom units (ligand binding domains of estrogen receptors) to light years (the seasonality of reproduction in many animals).

Of particular interest are gene duplication products among steroid hormone receptors, both of which the ancestral hormone can activate both as the normal ligand, but which are genetically and chemically distinguishable. Estrogens elicit many of their effects on behavior and physiology by binding to its intracellular receptors, estrogen receptor- α (ERα) and ERβ, which are transcribed from different genes [1,2] and are members of the steroid/nuclear receptor superfamily of transcriptional activators [3,4]. While estrogens can function in the brain by binding to receptors on the cell membrane and rapidly activate cytoplasmic signaling pathways [58], this review will focus on the effects of estrogens on behavior and physiology mediated by ERα and ERβ via classic, genomic mechanisms of action.

Using mice created by homologous recombination to achieve null mutations for ERα (ERαKO) or ERβ (ERβKO), we and others have conducted a large number of behavioral and histochemical assays in genetic females and genetic males. Several of the assays simply depend on the classical ER, without a discernible contribution from ERβ. During other assays, expression of ERβ appears to act as a brake on ERα function. In still other assays, ERβ and ERα can substitute for each other.

The contributions of ERα and ERβ to behaviors and physiological events are complicated and in no way uniform. Below we have summarized results gathered to date, and labeled each according to the relative contributions of the ER subtypes. The extreme scenarios are that ERα and ERβ act in an obligatory synergistic fashion {α+β} or that they exert equal and opposite function {α vs. β}. The data suggest that different patterns of neural function require different patterns of contributions by ERα and ERβ, such that: ERα and ERβ can substitute for each other {α or β}; the contributions of ERα and ERβ are not related {α/β contributions nr}; ERβ acts as a brake on ERα function and reduces the effects of ERα {α−β=α′}. While we and others have used many behavioral or histochemical assays, the references below concentrate on data related directly to the brain's regulation of reproduction. Then, our main inference is stated at the end of this paper. For an in depth review of ERα and ERβ expression in brain regions known to regulate behaviors associated with reproduction, including sexual behavior, aggression and anxiety, please see Bodo et al. [9].

1. Behavior

Perhaps the most striking result was the first reported: that putting an ERαKO female into the cage of a sexually experienced male had two surprising results: (i) that she behaves exactly as a male would, with respect to immediate aggressive behavior, and (ii) that she is treated like an intruder male by the resident male [10]. In a broader framework, comparisons among results of the different labs cited below attest to the reliability of neuroendocrine and behavioral results in this field.

1.1. Female sexual behavior {α−β=α′}

Gene expression from ERα raises levels of generalized arousal in female mice [11] eventuating in significantly higher locomotor activity [12], important for the initiation of courtship behaviors. In turn, studies using ERαKO mice and RNA interference indicate that the ERα gene product is essential for the normal performance of lordosis behavior [1316]. In contrast, while ERβKO females are subfertile [17], their lordosis behavior was equal to that of wt females and somewhat elevated in that they exhibited strong sexual receptivity the day after estrus [14,16]. These results indicate that female sexual behavior is dependent on the classical ERα, and not ERβ which appears to reduce the effects of ERα on sexual receptivity.

1.2. Male sexual behavior {α/β contributions nr}

While adult ERβKO males exhibit normal male sex behavior in regard to mounts, intromissions and ejaculations [16,18], this ER subtype appears to be important for development of male sex behavior. Peripubertal ERβKO males display their first ejaculations at a later age than their wt littermates [19]. In adults, reduced success in progress toward ejaculation is seen in ERαKO males [20]; that is, latencies through the series of intromissions are increased so that the temporal interval from first intromission to eventual ejaculation is increased — ejaculation is delayed. Finally, knocking out both ERα and ERβ completely abolishes male sexual behaviors, including mounting. [21]. Thus it appears that both ER subtypes contribute to the normal display of male sexual behavior. We infer from these data that even under circumstances in which ERβ by itself is not necessary for male sex behavior, it still can add synergistically to ERα, such that the double knockout completely abolishes males sexual behaviors.

1.3. Aggression in male ERKO mice {α−β=α′}

Early work has shown that ERαKO males are less aggressive to a male intruder than wt males [20,22]. Another study found that while wt males treated with testosterone (T) only attack T-treated male intruders, ERαKO males display equal aggression towards T-treated male, and estradiol (E)-treated female, intruders [23]. This inability of ERαKO males to distinguish intruders, possibly due to impaired processing of chemosensory information, suggests that ERα is important in normal social preferences. Furthermore, gonadally intact ERβKO males exhibit higher levels of aggression compared to wt males [20]. In gonadectomized males, estradiol benzoate (EB) treatment induces higher levels of aggression in ERβKO animals than wt [24]. These data suggest that the effects of estrogens on male aggression are stimulated by ERα and suppressed by ERβ.

1.4. Anxiety in females {α−β=α′}

A variety of studies indicate that ovarian hormones influence anxiety in female rodents [2527]. To test the function of the two ER subtypes in anxiety, ovariectomized rats were given DPN (diarylpropionitrile, an ERβ selective agonist), PPT (Tris(4-hydroxyphenyl)-4-propyl-1H-pyrazole, an ERα selective agonist), E or vehicle and tested for anxiety-related behaviors [28]. DPN treatment decreased anxiety-like behaviors in elevated plus maze and open field tests. Anxiogenic behaviors, including the time spent grooming, were decreased by DPN and increased by PPT [28]. It should be pointed out that the selectivity of DPN and PPT for ERβ and ERα, respectively, are not absolute [2931]. However, in support of this anxiolytic effect of ERβ, ERβKO females display higher levels of anxiety than wt littermates by spending less time exploring the distal portion of the open arm of a maze [32,33]. This effect in ovariectomized ERβKO mice was observed whether mice were treated with chronic E or vehicle [32]. Taken together, these findings indicate that ERβ is important during development and/or adulthood in mediating the anxiolytic effects of estrogens on anxiety.

1.5. Stress in male rats {α/β contributions nr}

The paraventricular nucleus (PVN) of the hypothalamus is critical in the modulation of stress and expresses high levels of androgen receptors (AR) [34] and ERβ [3537], but very little ERα [35]. To examine the role of ERβ and AR in the regulation of the hypothalamic–pituitary–adrenal response to stress, gonadectomized male rats were implanted with pellets containing E, dihydrotestosterone (DHT, which binds AR and is not aromatized to estradiol), 5alpha-androstane-3beta,17beta-diol (3β-diol, which binds ERβ with a higher affinity than ERα; [29], DPN or PPT into the PVN [38]. Following restraint stress, animals treated with DHT, 3β-diol or DPN had reduced levels of stress-induced release of corticosterone and ACTH, while E or PPT treatment increased the release of these stress hormones. These data suggest that ERβ functions to decrease HPA activity, while stimulation of ERα enhances it. The ER antagonist, tamoxifen, partially blocked the effects of 3β-diol and DHT, while an AR antagonist did not. The authors suggest that these effects are mediated through ERβ, since 3β-diol binds ERβ with a higher affinity than ERα [29]. In addition, administration into the PVN of DHT, 3β-diol or DPN reduced the restraint-induced increase in c-fos mRNA in this brain region [38]. These findings suggest that ERβ inhibits the HPA axis in males and that DHT may suppress the stress response, in part, via conversion to 3β-diol and its subsequent binding to ERβ.

2. Sexual differentiation of the brain

The gonadal steroids elicit profound organizational effects on the developing brain that result in adult sexually dimorphic brain physiology and behavior. During the perinatal period, the testes secrete testosterone that masculinizes (increased adult male sexual behavior) and defeminizes (decreased adult female sexual behavior) adult reproductive behavior. In the brain, testosterone can be converted to DHT or estradiol, which binds to AR and ER, respectively. Thus, testosterone can ultimately have both androgenic and estrogenic effects on the developing and adult brain.

2.1. Sexual differentiation of dopamine neurons in the anteroventral periventricular area {α or β}

In the hypothalamus of rodents, the anteroventral periventricular area (AVPV) has a sexually dimorphic subpopulation of dopaminergic neurons that is larger in females than males and is thought to be involved in the normal display of the preovulatory surge [39]. Testicular feminized male (tfm) mice, which have disrupted AR, and wt males had equal numbers of tyrosine-hydroxylase (TH)-immunoreactive neurons (indicative of dopamine neurons) in the AVPV, suggesting that AR do not regulate the expression of dopamine neurons in this brain region [40]. However, ERαKO male mice had more TH-immunoreactive neurons than wt males, indicating that disruption of the ERα gene feminized the number of dopaminergic neurons in the AVPV [40]. In an extension of this work, both ERαKO and ERβKO male mice had levels of TH-immunoreactive neurons that were higher than wt males and equal to wt female mice [41]. In addition, treatment of wt female mice during postnatal days 1–3 with either estradiol, DPN or PPT reduced the number of TH-immunoreactive neurons in the AVPV [41]. Taken together, these studies suggest that both ERα and ERβ, and not AR, are important in the sexual differentiation of the dopaminergic system in the AVPV of male and female mice.

3. Estrogen-responsive genes

3.1. Estradiol-induction of PR: females {α|β contributions nr}, males {α+β}, and developing female brain {α−β=α′}

A classic example of an estrogen-responsive gene is the estradiol-induction of PR in a variety of tissues, including brain. While PR are present in low levels in the brains of ovariectomized rodents, E-priming dramatically increases the expression of PR in the medial preoptic area (MPOA), ventromedial hypothalamus (VMH), arcuate and midbrain central gray [4252]. Interestingly, the developing rat brain is extremely sensitive to estradiol, given that doses as low as 100 ng of E are sufficient to induce PR [53]. In adult female guinea pigs virtually all of the E-induced PR cells express ERα [49,54], while about 30% of the E-induced PR cells in rat brain express ERβ [37].

In females, it appears that ERα is the major contributor to E-induction of PR in the brain. In support, E-induction of PR in the VMH and POA of ERβKO females was equal to wt animals [14,55], suggesting an important role for ERα in these regions. However, while E-induction of PR is greatly diminished in ERαKO female mice, it is not abolished in all areas, including the POA and VMH [14,56,57]. This residual induction of PR by estradiol may be due to ERβ (in the absence of ERα) or ERα splice variants that have been detected in ERαKO mice [56]. In contrast to females, while E treatment did not induce PR expression in the VMH and POA of wt males, it did increase PR levels in both brain regions of ERβKO males [55]. These findings suggest an important role for ERβ in the sexually dimorphic responses to estrogens in reproductively-relevant brain regions. In addition, these data suggest that ERβ can modulate the responsiveness of ERα in some cells.

Emilie Rissman's lab has used heterozygous breeders to generate mice with varying numbers of functional and disrupted ER genes to determine the contribution of each ER subtype to E-induction of PR in the MPOA [58]. Male and female mice with either 0, 1 or 2 functional copies of ERα and/or ERβ were gonadectomized and received implants of estradiol or blank capsules for 5 days. In females, maximal induction of PR by E in the MPOA required at least one functional copy of the ERα gene. In contrast to females, one copy of each ERα and ERβ genes are required for the full induction of PR in the male MPOA. These data support the concept that the role of ERβ in PR induction is sexually dimorphic and that there is functional dependence of the two ERs in PR induction in the male brain.

These findings in the adult brain suggest that ERβ can alter the activity of ERα. This concept is supported by a study in developing rat brain using ER subtype selective agonists [59]. In the developing female ventromedial nucleus of the hypothalamus (VMN), selective activation of ERα with PPT induces PR to higher levels than estradiol alone, which activates both ER subtypes. In contrast, in the MPN, which expresses high levels of ERα and low amounts of ERβ [60], PPT and estradiol induced PR to the same extent. In addition, selective activation of ERβ by DPN did not induce PR in the MPN. Moreover, activation of ERβ diminishes the ERα-mediated increase of PR in the VMN, but not the MPN. These findings suggest that activation of ERβ by estradiol in the VMN inhibits ERα transcriptional activity, and thus suppresses ERα-mediated transactivation of the PR gene in this brain region [59]. This suppressive effect of ERβ may provide a mechanism to protect the VMN from defeminization by estradiol during important periods of development, thus ensuring proper feminization of the VMN and subsequent adult female sexual behavior. This idea is supported by work in mice showing that activation of ERα or ERβ during development alters female sexual behavior in adulthood [61].

3.2. Estrogen and androgen regulation of AVP {α|β contributions nr}

Arginine vasopressin (AVP), a neurotransmitter involved in aggression, is regulated by androgens and estrogens. Testosterone, and to a lesser extent estradiol, up-regulates AVP expression in brain regions involved in aggression [62,63]. To investigate the roles of ERα and AR in gonadal hormone regulation of AVP and aggression, mice lacking a functional AR (tfm) were compared with ERαKO mice crossed with tfm (resulting in a double AR/ERαKO), ERαKO and wt mice [64]. Gonadectomized mice were treated with estradiol and analyzed by resident-intruder aggression tests. Wt and tfm males displayed aggressive behaviors, while ERαKO males and AR/ERαKO males did not, suggesting that ERα functions in males to regulate aggression as discussed above. AVP levels in the lateral septum revealed that wt and tfm males had the highest levels, followed by ERαKO males, and lowest levels in wt females and AR/ERαKO males. AR/ERαKO males expressed the lowest levels of AVP in the medial amygdala, with all other genotypes expressing equal amounts in this region [64]. In contrast to the lateral septum, estradiol reduces AVP expression in the male PVN [65]. This down-regulation of AVP by estradiol is abolished in ERβKO males, indicating that ERβ is critical for normal AVP expression in this brain region [65]. Interestingly, another study using a neuronal cell line found that DHT, and its metabolite androstane-3β,17β-diol, bind to ERβ to stimulate AVP promoter activity [66]. It will be essential for future studies to more fully investigate the function of ERβ in AVP expression in the brain. Taken together, these findings indicate that both ERα and AR are required for maximal expression of AVP in the lateral septum and medial amygdala, while ERβ is critical in AVP expression in the PVN.

4. ERα and ERβ interactions and mechanisms

In order to better understand the functional differences of ERα and ERβ, one must understand the mechanisms of action of these two subtypes. The ER subtypes have high homology in the DNA binding domain (97%), but much lower homology in the AF-1 and ligand binding domain (60%) [67]. These structural differences allow for variation in the binding affinity and specificity for different ligands of the two ER subtypes [29,68]. In addition, these differences in ligand binding, and the subsequent distinct changes in receptor conformation, enable the ER subtypes to vary in their ability to interact with different estrogen response elements [69]. Thus, ERα and ERβ can have profoundly different effects in the transduction of estrogen signaling.

4.1. ERα and ERβ interactions with nuclear receptor coactivators

In addition to binding to different ligands, ERα and ERβ may elicit distinct transcriptional effects through differential interactions with nuclear receptor coactivators. Nuclear receptor coactivators enhance steroid receptor transcriptional activity in vitro [70,71]. Moreover, these coactivators have been found to be critical in estrogen action in brain and behavior [72,73]. For example, steroid receptor coactivator- 1 (SRC-1), a member of the p160 family of coactivators, is critical for maximal ER-mediated transactivation of the PR gene in the VMN [74,75] and the display of estrogen-dependent behaviors [76]. Recent studies using pull-down assays have explored the physical interactions between the ER subtypes and SRC-1 from female rat brains [77]. SRC-1 from hypothalamus or hippocampus interacted with ERα and ERβ when bound to estradiol, which was confirmed by mass spectrometry. SRC-1 may function with ERα in the hypothalamus to mediate expression of female sexual behavior [1316], and with both ER subtypes in the hippocampus to differentially modulate estrogen's effects on cognition [9,78] and stress [9,79].

The different functions of the ER subtypes in the brain may be explained in part by the lower transcriptional activity of ERβ observed in particular cell lines [80]. These differences in transcriptional abilities between ERα and ERβ may be attributed to differential recruitment of coactivators, or differences in the ability of the same coactivator to facilitate transcription of the ER subtypes [81]. Interestingly, while SRC-1 from the hippocampus interacted equally with the ER subtypes, SRC-1 from hypothalamus interacted more with ERα than with ERβ. These findings suggest that ERα is a more efficient transcriptional activator of SRC-1 dependent signaling pathways in the hypothalamus than ERβ [77]. In support, previous work indicates that SRC-1 function in the hypothalamus is important for normal expression of ER-mediated female sexual receptivity [76], which as discussed above is ERα-dependent [1316,82]. These differential interactions of SRC-1 from hypothalamus or hippocampus with the ER subtypes suggest that these brain regions have distinct expression patterns of coregulators involved in these important protein–protein interactions. In addition, it is possible that SRC-1 undergoes differential phosphorylation in these two brain regions, leading to distinct patterns of interaction with the ER subtypes. These findings suggest the importance of using biologically-relevant tissue, in contrast to the use of cell lines alone, in investigating receptor–coactivator interactions. It may be that other cofactors and proteins that are present in tissue, such as the brain, are important for appropriate coactivator interactions with receptor. Understanding how nuclear receptor coactivators function with various steroid receptors, and their subtypes, is critical to understanding how hormones act in different brain regions to profoundly influence physiology and behavior.

It has been suggested that the lack of ERβ results in ERβKO mice that are hypersensitive to E [55]. In support, E-induces elevated levels of PR in the brains of ERβKO males [55] and the uteri of ERβKO females [83]. In addition, sexual behavior and learning studies suggest that ERβKO females are hypersensitive to E [16,78]. ERβ repression of ERα activity may involve the sequestration of shared nuclear receptor coactivators, that is relieved due to the absence of ERβ in ERβKO mice.

4.2. DNA as an allosteric ligand

Another level of ER signaling regulation may be exerted by DNA on the ER subtypes. In a recent elegant series of studies, Meijsing et al. [84] demonstrate that DNA can act as an allosteric ligand of glucocorticoid receptors (GR). Different GR binding sequences, which are imperfect palindromic, hexameric half sites separated by 3-base pair spacers, that differ by as little as one base pair, can elicit distinct conformations in GR and regulate its transcriptional activity. As suggested by the authors, this idea of DNA sequences acting as allosteric ligands may modulate signaling of other nuclear receptors, including ER [84]. In support, the AF-2 domain of ER binds to different coregulatory peptides when ER is bound to distinct DNA sequences [69,85]. Thus, in addition to the type of ligand that is available to ER, specific DNA sequences that bind the ER subtypes may modulate the conformation of ERα and/or ERβ in the cell and influence the coregulators that it recruits, thus altering the transcriptional activity of the receptors.

5. Conclusions

Taken together, the complex dependencies of results in this field of neuroendocrinology serve to defeat simplistic thinking about gene/behavior relationships. Even with these clear genetic manipulations and straightforward behavioral assays, unexpected results have emerged. For example, with respect to the impact of an ERβKO on aggressive behaviors by male mice, it became clear that the strength of the phenotype depends upon the age at which the male is tested, the KO effect being strongest just after puberty [86]. Even more surprising, the relation between a given gene and a given behavior (in this case, aggressive behavior) can be exactly the opposite between males and females (summarized in [87]). In addition, given the importance of coactivators in steroid receptor signaling, it is important to consider the possible effects on the signaling of one receptor subtype when the other subtype is knocked out. For example, consider a population of cells that coexpress ERα and ERβ [37]. In ERαKO mice, it is possible that in this population of cells, ERβ signaling may be enhanced due to decreased competition for common coactivators now that ERα has been eliminated. While we are not aware of any published data supporting this idea, it would be important to consider and explore these potential cell-specific effects in future studies.

We note that the prize-winning discovery of George Beadle and Edward Tatum, based on their work in 1941 with the fungus Neurospora, depended on their enunciation of the “one gene – one enzyme” concept. As distinct from their conclusion, and as inferred in Pfaff et al., 2002 [88], we believe that the strategic advantages of studying steroid hormone action in the brain have allowed us to state, as modern neuroscientists, that patterns of gene expression govern patterns of behavioral functions.

To ultimately understand the differential role of these ER subtypes on complex behaviors, it will be critical to identify and explore the different molecules that function in ER transcription in the brain. Elucidating the myriad of modifications of chromatin elicited by these coactivators and other regulatory molecules in brain will be essential. Moreover, understanding how these proteins assemble at the promoter, in a brain region and cell-type specific fashion, will be an exciting challenge for neuroendocrinologists to tackle in the near future.

Acknowledgements

Studies contributed by the authors' laboratories were supported by grants from National Science Foundation IBN 0080818 and National Institutes of Health R01 DK61935 (MJT) and NIH MH-38273 and HD-05751 (DWP).

References

  • 1.Jensen EV, Suzuki T, Kawasima T, Stumpf WE, Jungblut PW, de Sombre ER. A two-step mechanism for the interaction of estradiol with rat uterus. Proc. Natl. Acad. Sci. U. S. A. 1968;59:632–638. doi: 10.1073/pnas.59.2.632. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Kuiper GGJM, Enmark E, Pelto-Huikko M, Nilsson S, Gustafsson J. Cloning of a novel estrogen receptor expressed in rat prostate and ovary. Proc. Natl. Acad. Sci. U. S. A. 1996;93:5925–5930. doi: 10.1073/pnas.93.12.5925. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Mangelsdorf DJ, Thummel C, Beato M, Herrlich P, Schütz G, Umesono K, Blumberg B, Kastner P, Mark M, Chambon P, Evans RM. The nuclear receptor superfamily: the second decade. Cell. 1995;83:835–839. doi: 10.1016/0092-8674(95)90199-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Tsai MJ, O'Malley BW. Molecular mechanisms of action of steroid/thyroid receptor superfamily members. Ann. Rev. Biochem. 1994;63:451–486. doi: 10.1146/annurev.bi.63.070194.002315. [DOI] [PubMed] [Google Scholar]
  • 5.Vasudevan N, Pfaff DW. Non-genomic actions of estrogens and their interaction with genomic actions in the brain. Front. Neuroendocrinol. 2008;29:238–257. doi: 10.1016/j.yfrne.2007.08.003. [DOI] [PubMed] [Google Scholar]
  • 6.Micevych P, Kuo J, Christensen A. Physiology of membrane oestrogen receptor signalling in reproduction. J. Neuroendocrinol. 2009;21:249–256. doi: 10.1111/j.1365-2826.2009.01833.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Mermelstein PG. Membrane-localised oestrogen receptor alpha and beta influence neuronal activity through activation of metabotropic glutamate receptors. J. Neuroendocrinol. 2009;21:257–262. doi: 10.1111/j.1365-2826.2009.01838.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Kelly MJ, Ronnekleiv OK. Membrane-initiated estrogen signaling in hypothalamic neurons. Mol. Cell. Endocrinol. 2008;290:14–23. doi: 10.1016/j.mce.2008.04.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Bodo C, Rissman EF. New roles for estrogen receptor beta in behavior and neuroendocrinology. Front. Neuroendocrinol. 2006;27:217–232. doi: 10.1016/j.yfrne.2006.02.004. [DOI] [PubMed] [Google Scholar]
  • 10.Ogawa S, Taylor JA, Lubahn DB, Korach KS, Pfaff DW. Reversal of sex roles in genetic female mice by disruption of estrogen receptor gene. Neuroendocrinology. 1996;64:467–470. doi: 10.1159/000127154. [DOI] [PubMed] [Google Scholar]
  • 11.Garey J, Goodwillie A, Frohlich J, Morgan M, Gustafsson JA, Smithies O, Korach KS, Ogawa S, Pfaff DW. Genetic contributions to generalized arousal of brain and behavior. Proc. Natl. Acad. Sci. U. S. A. 2003;100:11019–11022. doi: 10.1073/pnas.1633773100. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Ogawa S, Chan J, Gustafsson JA, Korach KS, Pfaff DW. Estrogen increases locomotor activity in mice through estrogen receptor alpha: specificity for the type of activity. Endocrinology. 2003;144:230–239. doi: 10.1210/en.2002-220519. [DOI] [PubMed] [Google Scholar]
  • 13.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]
  • 14.Kudwa AE, Rissman EF. Double oestrogen receptor alpha and beta knockout mice reveal differences in neural oestrogen-mediated progestin receptor induction and female sexual behaviour. J. Neuroendocrinol. 2003;15:978–983. doi: 10.1046/j.1365-2826.2003.01089.x. [DOI] [PubMed] [Google Scholar]
  • 15.Musatov S, Chen W, Pfaff DW, Kaplitt MG, Ogawa S. RNAi-mediated silencing of estrogen receptor alpha in the ventromedial nucleus of hypothalamus abolishes female sexual behaviors. Proc. Natl. Acad. Sci. U. S. A. 2006;103:10456–10460. doi: 10.1073/pnas.0603045103. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Ogawa S, Chan J, Chester AE, Gustafsson JA, Korach KS, Pfaff DW. Survival of reproductive behaviors in estrogen receptor beta gene- deficient (betaERKO) male and female mice. Proc. Natl. Acad. Sci. U. S. A. 1999;96:12887–12892. doi: 10.1073/pnas.96.22.12887. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Krege JH, Hodgin JB, Couse JF, Enmark E, Warner M, Mahler JF, Sar M, Korach KS, Gustafsson J, Smithies O. Generation and reproductive phenotypes of mice lacking estrogen receptor-beta. Proc. Natl. Acad. Sci. U.S.A. 1998;95:15677–15682. doi: 10.1073/pnas.95.26.15677. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Kudwa AE, Bodo C, Gustafsson JA, Rissman EF. A previously uncharacterized role for estrogen receptor beta: defeminization of male brain and behavior. Proc. Natl. Acad. Sci. U. S. A. 2005;102:4608–4612. doi: 10.1073/pnas.0500752102. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Temple JL, Scordalakes EM, Bodo C, Gustafsson JA, Rissman EF. Lack of functional estrogen receptor beta gene disrupts pubertal male sexual behavior. Horm. Behav. 2003;44:427–434. doi: 10.1016/j.yhbeh.2003.09.002. [DOI] [PubMed] [Google Scholar]
  • 20.Ogawa S, Lubahn DB, Korach KS, Pfaff DW. Behavioral effects of estrogen receptor gene disruption in male mice. Proc. Natl. Acad. Sci. U. S. A. 1997;94:1476–1481. doi: 10.1073/pnas.94.4.1476. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Ogawa S, Chester AE, Hewitt SC, Walker VR, Gustafsson JA, Smithies O, Korach KS, Pfaff DW. Abolition of male sexual behaviors in mice lacking estrogen receptors alpha and beta (alpha beta ERKO) Proc. Natl. Acad. Sci. U. S. A. 2000;97:14737–14741. doi: 10.1073/pnas.250473597. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Ogawa S, Washburn TF, Taylor J, Lubahn DB, Korach KS, Pfaff DW. Modifications of testosterone-dependent behaviors by estrogen receptor-alpha gene disruption in male mice. Endocrinology. 1998;139:5058–5069. doi: 10.1210/endo.139.12.6358. [DOI] [PubMed] [Google Scholar]
  • 23.Scordalakes EM, Rissman EF. Aggression in male mice lacking functional estrogen receptor alpha. Behav. Neurosci. 2003;117:38–45. [PubMed] [Google Scholar]
  • 24.Nomura M, Andersson S, Korach KS, Gustafsson JA, Pfaff DW, Ogawa S. Estrogen receptor-beta gene disruption potentiates estrogen-inducible aggression but not sexual behaviour in male mice. Eur. J. Neurosci. 2006;23:1860–1868. doi: 10.1111/j.1460-9568.2006.04703.x. [DOI] [PubMed] [Google Scholar]
  • 25.Morgan MA, Pfaff DW. Estrogen's effects on activity, anxiety, and fear in two mouse strains. Behav. Brain Res. 2002;132:85–93. doi: 10.1016/s0166-4328(01)00398-9. [DOI] [PubMed] [Google Scholar]
  • 26.Frick KM, Burlingame LA, Arters JA, Berger-Sweeney J. Reference memory, anxiety and estrous cyclicity in C57BL/6NIA mice are affected by age and sex. Neuroscience. 2000;95:293–307. doi: 10.1016/s0306-4522(99)00418-2. [DOI] [PubMed] [Google Scholar]
  • 27.Galeeva AY, Pivina SG, Tuohimaa P, Ordyan NE. Involvement of nuclear progesterone receptors in the formation of anxiety in female mice. Neurosci. Behav. Physiol. 2007;37:843–848. doi: 10.1007/s11055-007-0090-8. [DOI] [PubMed] [Google Scholar]
  • 28.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]
  • 29.Kuiper GG, Lemmen JG, Carlsson B, Corton JC, Safe SH, van der Saag PT, van DB, Gustafsson JA. Interaction of estrogenic chemicals and phytoestrogens with estrogen receptor beta. Endocrinology. 1998;139:4252–4263. doi: 10.1210/endo.139.10.6216. [DOI] [PubMed] [Google Scholar]
  • 30.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]
  • 31.Harrington WR, Sheng S, Barnett DH, Petz LN, Katzenellenbogen JA, Katzenellenbogen BS. Activities of estrogen receptor alpha- and beta-selective ligands at diverse estrogen responsive gene sites mediating transactivation or transrepression. Mol. Cell. Endocrinol. 2003;206:13–22. doi: 10.1016/s0303-7207(03)00255-7. [DOI] [PubMed] [Google Scholar]
  • 32.Imwalle DB, Gustafsson JA, Rissman EF. Lack of functional estrogen receptor beta influences anxiety behavior and serotonin content in female mice. Physiol. Behav. 2005;84:157–163. doi: 10.1016/j.physbeh.2004.11.002. [DOI] [PubMed] [Google Scholar]
  • 33.Walf AA, Koonce CJ, Frye CA. Estradiol or diarylpropionitrile decrease anxiety-like behavior of wildtype, but not estrogen receptor beta knockout, mice. Behav. Neurosci. 2008;122:974–981. doi: 10.1037/a0012749. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Zhou L, Blaustein JD, De Vries GJ. Distribution of androgen receptor immunoreactivity in vasopressin- and oxytocin-immunoreactive neurons in the male rat brain. Endocrinology. 1994;134:2622–2627. doi: 10.1210/endo.134.6.8194487. [DOI] [PubMed] [Google Scholar]
  • 35.Shughrue PJ, Lane MV, Merchenthaler I. Comparative distribution of estrogen receptor-alpha and -beta mRNA in the rat central nervous system. J. Neurol. 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]
  • 36.Alves SE, Lopez V, McEwen BS, Weiland NG. Differential colocalization of estrogen receptor beta (ERbeta) with oxytocin and vasopressin in the paraventricular and supraoptic nuclei of the female rat brain: an immunocytochemical study. Proc. Natl. Acad. Sci. U. S. A. 1998;95:3281–3286. doi: 10.1073/pnas.95.6.3281. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Greco B, Allegretto EA, Tetel MJ, Blaustein JD. Coexpression of ER beta with ER alpha and progestin receptor proteins in the female rat forebrain: effects of estradiol treatment. Endocrinology. 2001;142:5172–5181. doi: 10.1210/endo.142.12.8560. [DOI] [PubMed] [Google Scholar]
  • 38.Lund TD, Hinds LR, Handa RJ. The androgen 5alpha-dihydrotestosterone and its metabolite 5alpha-androstan-3beta, 17beta-diol inhibit the hypothalamopituitary-adrenal response to stress by acting through estrogen receptor beta-expressing neurons in the hypothalamus. J. Neurosci. 2006;26:1448–1456. doi: 10.1523/JNEUROSCI.3777-05.2006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Simerly RB, Swanson LW, Gorski RA. The distribution of monoaminergic cells and fibers in a periventricular preoptic nucleus involved in the control of gonadotropin release: immunohistochemical evidence for a dopaminergic sexual dimorphism. Brain Res. 1985;330:55–64. doi: 10.1016/0006-8993(85)90007-1. [DOI] [PubMed] [Google Scholar]
  • 40.Simerly RB, Zee MC, Pendleton JW, Lubahn DB, Korach KS. Estrogen receptor-dependent sexual differentiation of dopaminergic neurons in the preoptic region of the mouse. Proc. Natl. Acad. Sci. U. S. A. 1997;94:14077–14082. doi: 10.1073/pnas.94.25.14077. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Bodo C, Kudwa AE, Rissman EF. Both estrogen receptor-alpha and -beta are required for sexual differentiation of the anteroventral periventricular area in mice. Endocrinology. 2006;147:415–420. doi: 10.1210/en.2005-0834. [DOI] [PubMed] [Google Scholar]
  • 42.MacLusky NJ, McEwen BS. Oestrogen modulates progestin receptor concentrations in some rat brain regions but not in others. Nature. 1978;274:276–278. doi: 10.1038/274276a0. [DOI] [PubMed] [Google Scholar]
  • 43.Warembourg M. Radioautographic study of the rat brain, uterus and vagina after [3 H]R-5020 injection. Mol. Cell Endocrinol. 1978;12:67–79. doi: 10.1016/0303-7207(78)90102-8. [DOI] [PubMed] [Google Scholar]
  • 44.MacLusky NJ, McEwen BS. Progestin receptors in rat brain: distribution and properties of cytoplasmic progestin-binding sites. Endocrinology. 1980;106:192–202. doi: 10.1210/endo-106-1-192. [DOI] [PubMed] [Google Scholar]
  • 45.Blaustein JD, Feder HH. Cytoplasmic progestin receptors in female guinea pig brain and their relationship to refractoriness in expression of female sexual behavior. Brain Res. 1979;177:489–498. doi: 10.1016/0006-8993(79)90466-9. [DOI] [PubMed] [Google Scholar]
  • 46.Lauber AH, Romano GJ, Pfaff DW. Sex difference in estradiol regulation of progestin receptor messenger RNA in rat mediobasal hypothalamus as demonstrated by In situ hybridization. Neuroendocrinology. 1991;53:608–613. doi: 10.1159/000125781. [DOI] [PubMed] [Google Scholar]
  • 47.Ogawa S, Olazabal UE, Parhar IS, Pfaff DW. Effects of intrahypothalamic administration of antisense DNA for progesterone receptor mRNA on reproductive behavior and progesterone receptor immunoreactivity in female rat. J. Neurosci. 1994;14:1766–1774. doi: 10.1523/JNEUROSCI.14-03-01766.1994. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Blaustein JD, King JC, Toft DO, Turcotte J. Immunocytochemical localization of estrogen-induced progestin receptors in guinea pig brain. Brain Res. 1988;474:1–15. doi: 10.1016/0006-8993(88)90664-6. [DOI] [PubMed] [Google Scholar]
  • 49.Warembourg M, Jolivet A, Milgrom E. Immunohistochemical evidence of the presence of estrogen and progesterone receptors in the same neurons of the guinea pig hypothalamus and preoptic area. Brain Res. 1989;480:1–15. doi: 10.1016/0006-8993(89)91561-8. [DOI] [PubMed] [Google Scholar]
  • 50.Beyer C, Damm N, Brito V, Kuppers E. Developmental expression of progesterone receptor isoforms in the mouse midbrain. Neuroreport. 2002;13:877–880. doi: 10.1097/00001756-200205070-00028. [DOI] [PubMed] [Google Scholar]
  • 51.Scott REM, Wu-Peng XS, Pfaff DW. Regulation and expression of progesterone receptor mRNA isoforms A and B in the male and female rat hypothalamus and pituitary following oesterogen treatment. J. Neuroendocrinol. 2002;14:175–183. doi: 10.1046/j.0007-1331.2001.00750.x. [DOI] [PubMed] [Google Scholar]
  • 52.Brinton RD, Thompson RF, Foy MR, Baudry M, Wang J, Finch CE, Morgan TE, Pike CJ, Mack WJ, Stanczyk FZ, Nilsen J. Progesterone receptors: form and function in brain. Front. Neuroendocrinol. 2008;29:313–339. doi: 10.1016/j.yfrne.2008.02.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Chung WC, Pak TR, Weiser MJ, Hinds LR, Andersen ME, Handa RJ. Progestin receptor expression in the developing rat brain depends upon activation of estrogen receptor alpha and not estrogen receptor beta. Brain Res. 2006;1082:50–60. doi: 10.1016/j.brainres.2006.01.109. [DOI] [PubMed] [Google Scholar]
  • 54.Blaustein JD, Turcotte JC. Estradiol-induced progestin receptor immunoreactivity is found only in estrogen receptor-immunoreactive cells in guinea pig brain. Neuroendocrinology. 1989;49:454–461. doi: 10.1159/000125152. [DOI] [PubMed] [Google Scholar]
  • 55.Temple JL, Fugger HN, Li X, Shetty SJ, Gustafsson J, Rissman EF. Estrogen receptor beta regulates sexually dimorphic neural responses to estradiol. Endocrinology. 2001;142:510–513. doi: 10.1210/endo.142.1.8054. [DOI] [PubMed] [Google Scholar]
  • 56.Moffatt CA, Rissman EF, Shupnik MA, Blaustein JD. Induction of progestin receptors by estradiol in the forebrain of estrogen receptor-alpha gene-disrupted mice. J. Neurosci. 1998;18:9556–9563. doi: 10.1523/JNEUROSCI.18-22-09556.1998. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Shughrue PJ, Lubahn DB, Negro-Vilar A, Korach KS, Merchenthaler I. Responses in the brain of estrogen receptor alpha-disrupted mice. Proc. Natl. Acad. Sci. U. S. A. 1997;94:11008–11012. doi: 10.1073/pnas.94.20.11008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Kudwa AE, Gustafsson JA, Rissman EF. Estrogen receptor B modulates estradiol induction of progestin receptor immunoreactivity in male, but not in female, mouse medial preoptic area. Endocrinology. 2004;145:4500–4506. doi: 10.1210/en.2003-1708. [DOI] [PubMed] [Google Scholar]
  • 59.Gonzales KL, Tetel MJ, Wagner CK. Estrogen receptor (ER) beta modulates ERalpha responses to estrogens in the developing rat ventromedial nucleus of the hypothalamus. Endocrinology. 2008;149:4615–4621. doi: 10.1210/en.2008-0511. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.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]
  • 61.Kudwa AE, Michopoulos V, Gatewood JD, Rissman EF. Roles of estrogen receptors alpha and beta in differentiation of mouse sexual behavior. Neuroscience. 2006;138:921–928. doi: 10.1016/j.neuroscience.2005.10.018. [DOI] [PubMed] [Google Scholar]
  • 62.De Vries GJ, Crenshaw BJ, Al-Shamma HA. Gonadal steroid modulation of vasopressin pathways. Ann. N. Y. Acad. Sci. 1992;652:387–396. doi: 10.1111/j.1749-6632.1992.tb34369.x. [DOI] [PubMed] [Google Scholar]
  • 63.De Vries GJ, Wang Z, Bullock NA, Numan S. Sex differences in the effects of testosterone and its metabolites on vasopressin messenger RNA levels in the bed nucleus of the stria terminalis of rats. J. Neurosci. 1994;14:1789–1794. doi: 10.1523/JNEUROSCI.14-03-01789.1994. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Scordalakes EM, Rissman EF. Aggression and arginine vasopressin immunoreactivity regulation by androgen receptor and estrogen receptor alpha. Genes Brain Behav. 2004;3:20–26. doi: 10.1111/j.1601-183x.2004.00036.x. [DOI] [PubMed] [Google Scholar]
  • 65.Nomura M, McKenna E, Korach KS, Pfaff DW, Ogawa S. Estrogen receptor-beta regulates transcript levels for oxytocin and arginine vasopressin in the hypothalamic paraventricular nucleus of male mice. Brain Res. Mol. Brain Res. 2002;109:84–94. doi: 10.1016/s0169-328x(02)00525-9. [DOI] [PubMed] [Google Scholar]
  • 66.Pak TR, Chung WC, Hinds LR, Handa RJ. Estrogen receptor-{beta} mediates dihydrotestosterone-induced stimulation of the arginine vasopressin promoter in neuronal cells. Endocrinology. 2007;148:3371–3382. doi: 10.1210/en.2007-0086. [DOI] [PubMed] [Google Scholar]
  • 67.Kuiper GG, Enmark E, Pelto-Huikko M, Nilsson S, Gustafsson JA. Cloning of a novel receptor expressed in rat prostate and ovary. Proc. Natl. Acad. Sci. U. S. A. 1996;93:5925–5930. doi: 10.1073/pnas.93.12.5925. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Kuiper GGJM, Carlsson B, Grandien K, Enmark E, Häggblad J, Nilsson S, Gustafsson J. Comparison of the ligand binding specificity and transcript tissue distribution of estrogen receptors alpha and beta. Endocrinology. 1997;138:863–870. doi: 10.1210/endo.138.3.4979. [DOI] [PubMed] [Google Scholar]
  • 69.Hall JM, McDonnell DP, Korach KS. Allosteric regulation of estrogen receptor structure, function, and coactivator recruitment by different estrogen response elements. Mol. Endocrinol. 2002;16:469–486. doi: 10.1210/mend.16.3.0814. [DOI] [PubMed] [Google Scholar]
  • 70.Rosenfeld MG, Lunyak VV, Glass CK. Sensors and signals: a coactivator/corepressor/epigenetic code for integrating signal-dependent programs of transcriptional response. Genes Dev. 2006;20:1405–1428. doi: 10.1101/gad.1424806. [DOI] [PubMed] [Google Scholar]
  • 71.O'Malley BW. Molecular biology. Little molecules with big goals. Science. 2006;313:1749–1750. doi: 10.1126/science.1132509. [DOI] [PubMed] [Google Scholar]
  • 72.Tetel MJ. Nuclear receptor coactivators: essential players in steroid hormone action in brain and behavior. J. Neuroendocrinol. 2009;21:229–237. doi: 10.1111/j.1365-2826.2009.01827.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Tetel MJ, Auger AP, Charlier TD. Who's in charge? Nuclear receptor coactivator and corepressor function in brain and behavior. Front. Neuroendocrinol. 2009;30:328–342. doi: 10.1016/j.yfrne.2009.04.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Molenda HA, Griffin AL, Auger AP, McCarthy MM, Tetel MJ. Nuclear receptor coactivators modulate hormone-dependent gene expression in brain and female reproductive behavior in rats. Endocrinology. 2002;143:436–444. doi: 10.1210/endo.143.2.8659. [DOI] [PubMed] [Google Scholar]
  • 75.Apostolakis EM, Ramamurphy M, Zhou D, Onate S, O'Malley B. Acute disruption of select steroid receptor coactivators prevents reproductive behavior in rats and unmasks genetic adaptation in knockout mice. Mol. Endocrinol. 2002;16:1511–1523. doi: 10.1210/mend.16.7.0877. [DOI] [PubMed] [Google Scholar]
  • 76.Molenda-Figueira HA, Williams CA, Griffin AL, Rutledge EM, Blaustein JD, Tetel MJ. Nuclear receptor coactivators function in estrogen receptor- and progestin receptor-dependent aspects of sexual behavior in female rats. Horm. Behav. 2006;50:383–392. doi: 10.1016/j.yhbeh.2006.04.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Molenda-Figueira HA, Murphy SD, Shea KL, Siegal NK, Zhao Y, Chadwick JG, Denner LA, Tetel MJ. Steroid receptor coactivator-1 from brain physically interacts differentially with steroid receptor subtypes. Endocrinology. 2008;149:5272–5279. doi: 10.1210/en.2008-0048. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Fugger HN, Foster TC, Gustafsson J, Rissman EF. Novel effects of estradiol and estrogen receptor alpha and beta on cognitive function. Brain Res. 2000;883:258–264. doi: 10.1016/s0006-8993(00)02993-0. [DOI] [PubMed] [Google Scholar]
  • 79.Isgor C, Cecchi M, Kabbaj M, Akil H, Watson SJ. Estrogen receptor beta in the paraventricular nucleus of hypothalamus regulates the neuroendocrine response to stress and is regulated by corticosterone. Neuroscience. 2003;121:837–845. doi: 10.1016/s0306-4522(03)00561-x. [DOI] [PubMed] [Google Scholar]
  • 80.Delaunay F, Pettersson K, Tujague M, Gustafsson JA. Functional differences between the amino-terminal domains of estrogen receptors alpha and beta. Mol. Pharmacol. 2000;58:584–590. doi: 10.1124/mol.58.3.584. [DOI] [PubMed] [Google Scholar]
  • 81.Wong C, Komm B, Cheskis BJ. Structure–function evaluation of ER alpha and beta interplay with SRC family coactivators. ER selective ligands. Biochemistry. 2001;40:6756–6765. doi: 10.1021/bi010379h. [DOI] [PubMed] [Google Scholar]
  • 82.Rissman EF, Early AH, Taylor JA, Korach KS, Lubahn DB. Estrogen receptors are essential for female sexual receptivity. Endocrinology. 1997;138:507–510. doi: 10.1210/endo.138.1.4985. [DOI] [PubMed] [Google Scholar]
  • 83.Weihua Z, Saji S, Makinen S, Cheng G, Jensen EV, Warner M, Gustafsson JA. Estrogen receptor (ER) beta, a modulator of ERalpha in the uterus. Proc. Natl. Acad. Sci. U. S. A. 2000;97:5936–5941. doi: 10.1073/pnas.97.11.5936. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Meijsing SH, Pufall MA, So AY, Bates DL, Chen L, Yamamoto KR. DNA binding site sequence directs glucocorticoid receptor structure and activity. Science. 2009;324:407–410. doi: 10.1126/science.1164265. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Heery DM, Kalkhoven E, Hoare S, Parker MG. A signature motif in transcriptional co-activators mediates binding to nuclear receptors. Nature. 1997;387:733–736. doi: 10.1038/42750. [DOI] [PubMed] [Google Scholar]
  • 86.Nomura M, Durbak L, Chan J, Smithies O, Gustafsson JA, Korach KS, Pfaff D, Ogawa S. Genotype/age interactions on aggressive behavior in gonadally intact estrogen receptor-αknockout (αERKO) male mice. Horm. Behav. 2002;41:288–296. doi: 10.1006/hbeh.2002.1773. [DOI] [PubMed] [Google Scholar]
  • 87.Ogawa S, Choleris E, Pfaff D. Genetic influences on aggressive behaviors and arousability in animals. Ann. N. Y. Acad. Sci. 2004;1036:257–266. doi: 10.1196/annals.1330.016. [DOI] [PubMed] [Google Scholar]
  • 88.Pfaff D, Ogawa S, Kia K, Vasudevan N, Krebs C, Frohlich J, Kow L-M. Genetic mechanisms in neural and hormonal controls over female reproductive behaviors. In: Pfaff DW, Arnold AP, Etgen AM, Fahrbach SE, Rubin RT, editors. Hormones, Behavior and Brain. Vol. 3. San Diego: Academic Press; 2002. pp. 441–510. [Google Scholar]

RESOURCES