<<

Review 569

Convergence of Multiple Mechanisms of Action

Authors S. K. Mani 1 * , P. G. Mermelstein 2 * , M. J. Tetel 3 * , G. Anesetti 4 *

Affi liations 1 Department of Molecular & Cellular Biology and Neuroscience, Baylor College of Medicine, Houston, TX, USA 2 Department of Neuroscience, University of Minnesota, Minneapolis, MN, USA 3 Neuroscience Program, Wellesley College, Wellesley, MA, USA 4 Departamento de Hostologia y Embriologia, Facultad de Medicine, Universidad de la Republica, Montevideo, Uruguay

Key words Abstract receptors can also be activated in a “-inde- ● ▶ ▼ pendent” manner by other factors including neu- ● ▶ Steroid modulate a wide array of rotransmitters. Recent studies indicate that rapid, ▶ ● signaling physiological processes including development, nonclassical steroid eff ects involve extranuclear ● ▶ cross-talk metabolism, and in various species. steroid receptors located at the membrane, which ● ▶ ● ▶ brain It is generally believed that these biological eff ects interact with cytoplasmic kinase signaling mol- are predominantly mediated by their binding to ecules and G-. The current review deals specifi c intracellular receptors resulting in con- with various mechanisms that function together formational change, dimerization, and recruit- in an integrated manner to promote hormone- ment of coregulators for -dependent dependent actions on the central and sympathetic genomic actions (classical mechanism). In addi- nervous systems. tion, to their cognate ligands, intracellular steroid

Abbreviations expression and function. Interestingly, not ▼ all the “classical” receptors are intranuclear and CBP CREB binding can be associated at the membrane. As described CRE CREB response element in this review, extranuclear ERs and PRs at the DAR (DAR) membrane or in the cytoplasm can interact with ER G proteins and signaling kinases, and other G received 13 . 12 . 2011 ERE Estrogen response element protein coupled receptors, to mediate rapid accepted 24 . 02 . 2012 mGluR Metabotropic glutamate receptor eff ects of the hormones. The rapid actions involv- PGMRC1 Progesterone membrane receptor ing cytoplasmic kinase signaling and/or extra- Bibliography nuclear steroid receptors can result in both DOI http://dx.doi.org/ component 1 10.1055/s-0032-1306343 PR Progestin receptor transcription-independent and transcription- Published online: PRE Progestin response element dependent actions. In addition, to their cognate March 27, 2012 SRCs Steroid receptor coactivators ligands, intranuclear steroid receptors (PRs) can Horm Metab Res 2012; also be activated in a “ligand-independent” man- 44: 569–576 ner by neurotransmitters (dopamine; DA). In this © Georg Thieme Verlag KG Introduction review, the distinct classical and nonclassical cel- Stuttgart · New York lular and molecular mechanisms of steroid hor- ISSN 0018-5043 ▼ Steroid hormones, (E 2 ) and progester- mone action will be discussed with specifi c Correspondence one (P), regulate important physiological proc- reference to E2 and P eff ects in central and sym- S. K. Mani esses including development, diff erentiation, pathetic nervous systems ( ● ▶ Fig. 1 ) . Department of Molecular & metabolism, reproduction, learning, and mem- Cellular Biology ory in various species. The biological eff ects of E2 Baylor College of Medicine and P are primarily mediated by binding to their Mechanisms of Progesterone Action in One Baylor Plaza classical intranuclear receptors, estrogen (ERs) Brain Houston Texas 77030 and progestin receptors (PRs) that act as ligand- ▼ USA inducible transcription factors and interact with Ovarian steroid hormones, E 2 and P regulate cel- Tel.: +1/713/798 6647 steroid receptor coregulators to modulate target lular functions in the central nervous system Fax: +1/713/790 1275 resulting in alterations in physiology and repro- [email protected] * All the authors contributed equally to this work.

Mani SK et al. Mechanisms of Action … Horm Metab Res 2012; 44: 569–576 570 Review

Fig. 1 Convergence of progesterone- and estradiol-mediated signaling in the nervous system. Through membrane, cytosolic, and intra- cellular receptors, these hormones aff ect surface membrane signaling, second messenger systems, and gene expression. Transcriptional co-factors are also intimately involved in the regulation of classical, steroid hormone-mediated as well as activity-dependent gene expression.

ductive behavior. As in other steroid-sensitive tissues, the regu- female sexual receptive behavior has been demonstrated

latory action of E2 on behavior is believed to involve the [ 18 , 19 ] . In addition, DA-initiated second messenger signaling activation of ERs, altering the expression of a number of , cascade was demonstrated to involve the activation of protein including PR gene. Progestins, including P, exert their physiolog- kinase A (PKA) and neuronal phosphoprotein, dopamine and

ical eff ects primarily by binding to E2 -induced, intracellular PRs, cAMP regulated phosphoprotein-32 (DARPP-32) [ 20 ] . This sign- which function as transcriptional factors, regulating the expres- aling mechanism could potentially lead to the alterations in the sion of genes and genomic neural networks to initiate and/or phosphorylation dynamics and activation of PRs and/or its sustain physiological response [ 1 , 2 ] . PRs undergo signifi cant coregulators as discussed below. conformational change upon binding by P, leading to their While genomic eff ects characterized by a delayed onset have nuclear translocation, dimerization and DNA binding [ 3 ] . When traditionally been assumed to be the primary pathway for P bound to DNA, PRs interact with basal transcriptional machin- action in the brain, recent studies suggest the involvement of ery, assisted by molecules to initiate chromatin “nonclassical” mechanisms of progesterone action. These non- remodeling [ 4 – 6 ] . Phosphorylation of the coactivators also plays classical short-latency eff ects of progesterone widely aff ect cell a crucial role in the activation of steroid receptors [ 7 , 8 ] . The role functioning, through modulation of putative cell surface recep- of coactivators is discussed below. tors, ion channels and mechanisms coupled to cytoplasmic

Spatial, temporal, and functional correlations indicate that E2 - second messenger signaling cascades, independent of gene tran- induced PRs function as transcriptional mediators and regulate scription [ 21 – 23 ] . In addition to P, several of its ring-A reduced transcription of target genes to aff ect reproductive behavior [ 2 ] . metabolites have been shown to facilitate lordosis response in

The time course of activation and termination of reproductive ovariectomized, E2 -primed female rats via activation of MAPK behavior has also been shown to parallel E2 -induced increase pathway [ 17 ] . A number of laboratories have reported the and decline in PRs in the and the preoptic areas of involvement of at least 4 extranuclear kinase systems, PKA, PKC, the brain. A wide body of literature has identifi ed diff erent neu- CaMKII, and PKG in the rapid P eff ects in the VMH and POA of the roanatomical sites in the regulation of female sexual behavior by female rat [ 16 , 17 , 21 , 24 – 27 ] . Since the initiation of these non- steroid hormones [ 9 ] . Studies using PR antagonists, protein and classical eff ects occurs rapidly (in seconds or minutes) and is RNA synthesis inhibitors, antisense to oligonucleotides to PRm- triggered at the membrane surface, the classical model of RNA and PR mutant mice indicate a requirement for classical nuclear PR-mediation is inadequate to account for these eff ects. genomic mode of activation involving intracellular PRs in Recent evidence suggests the involvement of 2 types of novel P-mediated facilitation of reproductive behavior [ 10 – 15 ] . membrane proteins unrelated to classical PRs, progesterone Studies have also demonstrated that PRs can be activated by fac- membrane receptor component 1 (PGMRC1) and progesterone tors other than their cognate ligand, P (ligand-independent membrane receptors (mPRs), in P signaling in several reproduc- activation). Second messenger molecules, including 3′,5′-cyclic tive tissues and in the brain [ 28 , 29 ] . Inhibition of MAPK signaling adenosine monophosphate (cAMP), 3′,5′-cyclic guanosine mono- pathway results in reduction of P, dibutyryl-cAMP (db-cAMP)-,

phosphate (cGMP), nitric oxide (NO), and neurotransmitter E 2 (PGE 2)-, or GnRH-facilitated female reproductive (dopamine, DA) can substitute for P in the facilitation of repro- behavior in rats [ 17 ] . These studies suggest that E2 and P actions ductive behavior [ 16 , 17 ] . Using PR antagonists, antisense oligo- in the brain could involve rapid activation of multiple signal nucleotides and null mutants for PRs, the critical requirement of transduction pathways that converge with classical genomic classical PRs as transcriptional mediators in the cross talk pathways. between P and DA-initiated pathways in the facilitation of

Mani SK et al. Mechanisms of Steroid Hormone Action … Horm Metab Res 2012; 44: 569–576 Review 571

Nuclear Receptor Coactivators: The p160 Steroid expressed at high levels in the hypothalamus, cortex and hip- Receptor Coactivator Family pocampus of rodents [ 34 – 43 ] and birds [ 44 – 47 ] . Interestingly, in ▼ contrast with the other members of the SRC family, SRC-3 is In the classic genomic mechanism of action discussed above, expressed sparsely in the hypothalamus [ 48 ] . In order for SRC-1 coregulators enhance (coactivators) or repress and SRC-2 to function with steroid receptors in brain, both the (corepressors) the steroid receptor transcriptional activity. Over coactivator and receptor must be expressed in the same cells. In 300 coactivators have been identifi ed, many of which have been support, SRC-1 and SRC-2 are expressed in the majority of estra- shown to function in physiology, behavior and human disease diol-induced PR cells in regions involved in metabolism and [ 30 ] . This review will focus on the role of the steroid receptor behavior, including the ventromedial hypothalamus (VMH), coactivator family in metabolism and brain and behavior. medial preoptic area and arcuate nucleus in rodents [ 35 , 49 ] . A The steroid receptor coactivator (SRC) family of p160 proteins variety of studies reveal that infl uence the expression of consists of SRC-1 (NcoA-1), SRC-2 (GRIP1/TIF2/NCoA-2), and these coactivators in brain [ 35 , 43 , 50 – 52 ] . Moreover, protein- SRC-3 (AIB1/TRAM-1/ACTR/RAC3/pCIP). This SRC family of coac- protein interactions studies reveal that SRC-1 and SRC-2 from tivators physically interacts with steroid receptors, including rodent brain physically associate with ER and PR in a receptor receptors for (AR), (ER), progestins (PR), and subtype- and brain region-specifi c manner [ 53 , 54 ] . (GR), in a ligand-dependent manner [ 31 ] . The A variety of studies using antisense to SRC-1 or SRC-2 mRNA C-terminus of the SRCs contains 2 activation domains (AD-1 and indicate that these coactivators are important for hormone AD-2), while the N-terminus contains a third activation domain dependent brain development and behavior. SRC-1 is critical for (AD-3) and a bHLH-PAS motif (basic helix loop helix-Per Arnt normal development of hormone-dependent sexual diff erentia- Sims), which is the most conserved domain within this family of tion of the brain and adult sexual behavior [ 40 ] . In the adult proteins. The activation domains interact with secondary coac- brain, SRC-1 and SRC-2 function in the VMH to modulate ER- tivators known as co-coactivators. These co-coactivators act as mediated transactivation of the behaviorally-relevant PR gene bridging molecules between the receptor and the general tran- [ 48 , 55 ] . In addition, SRC-1 functions in the VMH to infl uence scription machinery and modify chromatin within the promoter distinct ER- and PR-dependent aspects of female sexual behavior and enhancer regions by histone acetylation and methylation [ 56 ] . Interestingly, reduction of SRC-1 expression in this brain [31 ] . region by antisense altered PR function and reduced PR-depend- ent proceptivity (behavior by the female to solicit interaction by the male), but not PR-dependent receptivity [ 56 ] . These fi ndings The p160 SRC Family in Metabolism suggest that reduction of SRC-1 by antisense disrupted the activ- ▼ ity of PR signaling pathway(s) that infl uence proceptivity, while All 3 members of the p160 family of coactivators are involved in alternate PR signaling pathways, that regulate PR-dependent metabolic . SRC-1 is critical in maintaining energy receptivity, remained intact and functional. These coactivators, balance by regulating both energy intake and expenditure [ 32 ] . and others, have also been found to be essential in hormone In support, SRC-1 knockout mice have decreased energy expend- action in brain and behavior of birds [ 57 , 58 ] . iture and a reduced thermogenic capacity and thus, are prone to The p160 family of nuclear receptor coactivators has a critical obesity. One proposed mechanism of action for SRC-1 in metab- role in modulating steroid receptor action at the cellular level. In olism is through its interactions with PPARγ coactivator-1α the future, it will be important to determine the role of these (PGC-1α), an important regulator of mitochondrial biogenesis coactivators in nonclassical and membrane steroid signaling in and oxidative phosphorylation [ 32 ] . In contrast to SRC-1, SRC-2 brain discussed above and below, respectively. A better under- knockout mice are leaner than wild type mice and have an standing of how these important coactivators function in brain increase in adaptive thermogenesis [ 33 ] . It has been suggested is crucial to understanding the complex regulation of behavior that the absence of SRC-2 results in enhanced interaction and physiology by steroids. between SRC-1 and PGC-1α, which increases thermogenic activ- ity. Thus, it has been proposed that the ratio of SRC-1 and SRC-2 plays a role in maintaining the balance of energy expenditure Membrane Estrogen Receptors Activate and adipogenesis through controlling PGC-1α activity [ 33 ] . Metabotropic Glutamate Receptor Signaling SRC-3 knockouts have lower body fat content compared with ▼ wild-type mice [ 32 ] . SRC-3 mediates the transcriptional activity While we continue to appreciate the diversity of signaling of PPARγ2, which is important for adipocyte diff erentiation [ 33 ] . regarding the actions of nuclear ERα, ERβ and their associated In support, adipocyte diff erentiation and adipogenesis are proteins, estrogen receptors are also traffi cked to the surface impaired in mouse embryonic fi broblasts from cells isolated membrane, whereby they regulate various intracellular signal- from SRC-3 knockout mice or from 3T3-L1 adipocyte cells with ing pathways. Membrane-initiated estrogen action, both inside decreased levels of SRC-3 [ 33 ] . Taken together, these studies sug- and outside of the nervous system has been described for quite gest that the 3 members of the p160 SRC family play an impor- some time [ 59 , 60 ] . Yet, the identity of these membrane associ- tant role in energy homeostasis. ated receptors has for years remained controversial [ 61 ] . Several studies have suggested unique estrogen receptors acting at the membrane to initiate intracellular signaling [ 62 – 64 ] , while oth- The p160 SRC Family in Brain and Behavior ers have reported that these G-protein coupled receptors, local- ▼ ized to , are not activated by E2 and have A variety of recent studies indicate that 2 of the p160 SRC family no eff ect on intracellular signaling [ 65 ] . That said, in most sys- members, SRC-1 and SRC-2, are important for hormone action in tems, membrane-initiated steroid hormone action appears due brain and the regulation of behavior [ 34 ] . SRC-1 and SRC-2 are to the activation of a subpopulation of ERα and ERβ that have

Mani SK et al. Mechanisms of Steroid Hormone Action … Horm Metab Res 2012; 44: 569–576 572 Review

been modifi ed to traffi c and act at the neuronal surface [ 66 , 67 ] . to CREB phosphorylation were similarly segregated by expres- Determining how ERα and ERβ act at the membrane to aff ect sion of either caveolin-1 or caveolin-3. Surprisingly, however, intracellular signaling and how these estrogen receptors are the mGluRs responsible for membrane estrogen receptor signal- traffi cked to the membrane is critical to understanding estrogen ing in striatum were diff erent. In substitution of mGluR1a, ERα action in brain. was functionally coupled to mGluR5 in striatal neurons, whereas Recent studies have focused on estradiol-mediated phosphor- ERα and ERβ were paired with mGluR3 as opposed to in hippo- ylation (i. e., activation) of the CREB [ 68 , 69 ] . campus where these ERs activated mGluR2. These results are Phosphorylation of CREB is an important convergence point in interesting as each of these 4 mGluRs is expressed in both hip- , and is involved in various forms of neuronal plas- pocampus and striatum. These data suggest ERs may be promis- ticity, including learning and memory, drug addiction and noci- cuous regarding their coupling to various -coupled ception [ 70 ] , processes also under the infl uence of estrogens receptors. [ 61 ] . Work across multiple laboratories has indicated that acti- T r a ffi cking of ERα and ERβ to the membrane appears dependent vation of surface estrogen receptors leads to CREB phosphoryla- upon palmitoylation. Palmitoylation is the reversible addition of tion via stimulation of the MAPK/ERK signaling pathway the fatty acid palmitate to a protein. Palmitoylation aff ects the [ 71 – 73 ] . Furthermore, estrogen receptor knockout mice do not subcellular distribution and function of the modifi ed protein, exhibit estrogen-dependent CREB phosphorylation, indicating including promoting protein localization to membranes. Palmi- the dependence upon ERα and ERβ [ 74 ] . toylation occurs most commonly on residues, and is To elucidate the signaling mechanisms by which estradiol aff ects mediated by a family of proteins called palmitoyl acyltrans- CREB phosphorylation, initial studies utilized primary cultures ferases (PATs). There are 23 unique PATs, each derived from a from rat hippocampus [ 75 ] . Estradiol stimulation of membrane separate gene (DHHC 1–9, 11–24). Previous work in cell lines ERα was found to initiate mGluR1 signaling. These actions of demonstrated ERα incorporates radiolabeled palmitate [ 82 ] . estradiol/ERα on mGluR1 were independent of glutamate and Substitution of the palmitoyl cysteine residue with alanine only occurred in cultures generated from female rats. ERα acti- blocks both radiolabeling of ERα as well as membrane-initiated vation of mGluR1 signaling led to CREB phosphorylation via Gq- ERα responsiveness [ 82 – 84 ] . In neurons, recent fi ndings indicate mediated stimulation of phospholipase C (PLC), protein kinase C that palmitoylation of ERα and ERβ is essential for membrane ER (PKC) and trisphosphate (IP3). Interestingly, this was not localization, association with caveolin proteins, and activation the only membrane-initiated signaling pathway sensitive to mGluR signaling [ 81 ] . estradiol. In these same neurons, estradiol-mediated activation Using a simplifi ed model system (i. e., cultured neurons) it has of ERα or ERβ would lead to activation of mGluR2. Activation of been possible to identify this potential mechanism of estrogen these Gi/o G protein-coupled receptors by estradiol would lead action in brain. Work from a number of labs has expanded the to an attenuation of L-type calcium channel-mediated CREB importance of these studies to the whole animal, to additional phosphorylation. This was consistent, as well as provided a brain regions, and correspondingly, to additional behaviors. The mechanism, regarding previous fi ndings of estradiol inhibition fi rst report demonstrating the importance of ER/mGluR signal- of L-type calcium channels [ 76 ] . ing related to membrane estrogen action on the regulation of rat While CREB phosphorylation is only one of many downstream lordosis [ 85 ] . Specifi cally, within the arcuate nucleus, ERα acti- targets of mGluR activation, it was somewhat surprising to vation of mGluR1 signaling is essential for full sexual receptivity. observe estradiol regulating opposing processes within the In a separate study, regulation of the estrous cycle by membrane same cell. That is, at least, if gonadal estrogens are the principal ERs is dependent on ERα activation of mGluR1 in hypothalamic activators of ER/mGluR signaling. However, recent work has pro- astrocytes [ 86 ] . Examining dorsal root ganglion neurons in a vided several clues that brain-derived estrogen (i. e., neuroestro- model of nociception, estrogen receptor activation of mGluR2/3 gen) may be the critical mediator of rapid estrogen signaling attenuates ATP-induced increases in intracellular calcium via [77 ] . Specifi cally, not only does estradiol directly aff ect the elec- inhibition of L-type calcium channels [ 87 ] . Additionally, estro- trochemical state of the cell, but also synthesis of estradiol from gen-induced masculinization of the medial preoptic area is aromatase localized to presynaptic boutons suggests estradiol dependent upon mGluR [ 88 ] . ER/mGluR signaling has also been may act as a neurotransmitter [ 78 – 81 ] . A universal characteris- demonstrated to aff ect spinogenesis [ 89 ] , and recent fi ndings tic of neurotransmission is that the ligand release point is proxi- indicate ER/mGluR coupling is responsible for estrogen-induced mal to the location of the cognate receptor. Thus, if ER/mGluR release of endocannabinoids in hippocampal neurons [ 90 ] and signaling acts as similar to a traditional neurotransmitter sys- the potentiation of dopamine release in the female striatum fol- tem, these signaling proteins must also be spatially restricted. In lowing the administration of a psychostimulant [ 91 ] . Over the fact, caveolin-1 expression was found to be essential for the next several years, it is likely that other membrane actions of functional coupling of ERα with mGluR1a, whereas, caveolin-3 estradiol will also be shown dependent upon activation of is necessary for ERα and ERβ activation of mGluR2 [ 68 ] . Hence, mGluRs. the spatial segregation of diff erent ER/mGluR signaling path- ways can allow for independent activation and inhibition of CREB phosphorylation. Estrogens and Sympathetic Innervation in Female Because membrane estrogen receptor signaling is widespread Reproductive System both inside and outside of the nervous system, it was deter- ▼ mined whether ERs were coupled to mGluR signaling in other In addition to the variety of mechanisms by which estrogens act brain regions. Similar to previous results in hippocampal cul- in brain to regulate female reproductive behavior discussed tures, bidirectional eff ects of estradiol on CREB phosphoryla- above, estradiol infl uences the function of sympathetic neurons tion, dependent on ER/mGluR signaling, were found in striatal associated with reproductive organs in a variety of ways. The neurons [ 68 ] . Moreover, opposing ER/mGluR processes related sympathetic system includes preganglionic neurons located in

Mani SK et al. Mechanisms of Steroid Hormone Action … Horm Metab Res 2012; 44: 569–576 Review 573

the intermediolateral column at the thoracic and lumbar levels gens during the rodent estrous cycle and pregnancy induce of the spinal cord, which synapse with postganglionic neurons changes in sympathetic nerve density, including a progressive located in sympathetic ganglia. Axonal projections of these neu- loss of sympathetic activity at late gestation [ 110 ] . In the ovary, rons are dependent on several guidance and survival signals estradiol treatment induces an augmented synthesis of NGF provided by target organs [ 92 ] . These neurons display immuno- associated with an increase in content that can reactivity for one or both of the ER subtypes [ 93 , 94 ] . While induce long-lasting eff ects on the sympathetic innervation of almost all sympathetic neurons express ERβ, only about a third the . These changes alter ovarian morphology and func- of these neurons express ERα [ 95 ] . In some instances sympa- tion; induce the development and maintenance of cystic folli- thetic neurons in lumbar paravertebral and prevertebral ganglia cles, and cause anovulation and alterations in estrous cyclicity display cytoplasmic ER-immunoreactivity [ 94 , 96 ] . This cyto- [ 111 , 112 ] . Similar changes are found in ovaries from women plasmic distribution has been attributed to the presence of ERα with polycystic ovary syndrome (PCOS), a common cause of isoforms lacking nuclear localization signals and thus unable to infertility during reproductive age [ 113 ] . In animal models of respond to estrogens [ 97 ] . The membrane ER, GPR30, may also PCOS, the eff ects induced by estradiol exposure can be partially mediate signaling. Immunohistochemical and in situ hybridiza- reversed by surgical sympathectomy [ 111 , 114 ] . A potential con- tion studies reveal expression of GPR30 in some regions of brain, tribution of the sympathetic system to PCOS has been suggested spinal cord and peripheral tissues [ 98 ] . However, it is not known given that polycystic ovaries show an increase in the number of if GPR30 are expressed or function in postganglionic sympa- sympathetic fi bers [ 115 ] and ovarian wedge resection or laparo- thetic neurons. scopic laser cauterization are eff ective in increasing the ovula- Sympathetic fi bers that innervate the ovary originate mainly tory response in women with PCOS [ 116 ] . These fi ndings raise from the complex celiac-mesenteric ganglia. Ovarian innerva- the possibility that sympathetic input may play a role in the tion is associated with growing follicles, interstitial tissue and development and maintenance of polycystic ovaries in PCOS ovarian vasculature [ 99 ] . Two of the major neurotransmitters women. contained in the ovarian nerves, norepinephrine (NE) and In summary, estradiol can regulate several aspects of sympa- vasoactive intestinal polypeptide (VIP), are considered modula- thetic neuronal function through a variety of mechanisms. Neu- tors of steroid production [ 100 ] . Several studies show the rele- ronal target tissues can be infl uenced by these estradiol-induced vance of neural sympathetic infl uence on follicular assembly, changes. While estrogens can have direct eff ects by acting acquisition of responsiveness to gonadotropins and regulation of through neuronal ERs, they can also act indirectly to elicit ovarian steroidogenesis [ 101 , 102 ] . changes in production of survival signals in target organs that Estradiol produced by growing ovarian follicles has signifi cant aff ect the function of subpopulations of neurons. Knowledge of paracrine and autocrine eff ects that can infl uence gonadal phys- the basic mechanisms of this crosstalk is essential in under- iology. Estrogen action is mediated by ERs in follicular structures standing the pathophysiological aspects involved in the devel- and the stroma of the ovary. ERβ expression in granulosa cells is opment and maintenance of complex hormone-dependent essential for cell diff erentiation and function [ 103 ] , while ERα disorders aff ecting human health. expressed in thecal and interstitial cells are crucial for steroido- genesis [ 104 ] . Some eff ects of estradiol on sympathetic innervation are medi- Conclusions ated by changes in the expression of neurotrophins, including ▼ nerve (NGF), a signaling molecule involved in sur- The research outlined in this review (e. g., mechanisms of PR vival and diff erentiation of sympathetic and sensorial neurons. activation, transcriptional coactivators, membrane/nuclear sig- In the rat ovary, neurotrophins are produced in cells of the fol- naling, neuronal feedback to the reproductive organs) refl ects licular wall and are responsible for the development of ovarian how far the fi eld of neuroendocrinology has progressed over the sympathetic innervation [ 105 ] . The actions of these neuro- last several years. Clearly, steroid hormone action in the nervous trophins are mediated by 2 receptors: 1) the 75 kDa low-affi nity system is not a simple on/off activational relay. Based upon the neurotrophin receptor (p75NGFR), which displays rapid associa- role these hormones play in a diversity of physiological tion and dissociation with most members of the neurotrophin responses, this is not surprising. Over the next several years, we family and 2) a high-affi nity tyrosine kinase (TrkA) receptor, can expect basic science to provide additional breakthroughs as which binds NGF more specifi cally but with slow kinetics [ 106 ] . these fi ndings are integrated into a comprehensive study of Estrogen-responsive elements have been identifi ed in many of endocrine and neuroendocrine responsiveness. This work will the genes that code for neurotrophins or their receptors [ 107 ] . ultimately impact the treatment of various hormonal and meta- NGF production induced by estradiol exposure may diff er bolic disorders. depending on the tissue being analyzed [ 108 ] . In addition, vari- ations in estrogen levels alter neurotrophin receptor expression in sympathetic neurons, thus modulating neurotrophin respon- Acknowledgements siveness. This neurotrophin response varies with the neuronal ▼ population analyzed, the doses administered and the physiolog- The studies contributed to this work were supported by Public ical status of animals (e. g., prepubertal, adult cyclic, or adult Health Service grants from National Institutes of Health HD pregnant rat) [ 94 , 109 ] . 062512 (SKM), National Institutes of Health R01 DK61935 (MJT), Mounting evidence indicates that changes in estrogen signaling National Institute of Health NS 041302 and core funding NS and adrenergic innervation can induce alterations in sympa- 062125 (PGM), and grants from National Science Foundation thetic tone. The most dramatic modifi cations are seen in the IBN 0080818 (MJT). myometrial layer of uterus, where variations in levels of estro-

Mani SK et al. Mechanisms of Steroid Hormone Action … Horm Metab Res 2012; 44: 569–576 574 Review

References 23 Schumacher M , Coirini H , Robert F , Guennoun R , El-Etr M. Genomic 1 Blaustein J D , Olster D H . Gonadal steroid hormone receptors and social and membrane actions of progesterone: implications for reproduc- behaviors. In: Balthazar J (ed.). Molecular and cellular basis of social tive physiology and behavior . Behav Brain Res 1999 ; 105 : 37 – 52 behavior in vertebrates. Berlin : Springer-Verlag , 1989 ; 31 – 104 24 Balasubramanian B , Portillo W , Reyna A , Chen J Z , Moore A N , Dash 2 Pfaff D W , Ogawa S , Kia K , Vasudevan N , Krebs C , Frohlich J , Kow L M . P K , Mani S K . Nonclassical mechanisms of progesterone action in the Genetic mechanisms in neural and hormonal controls over female brain: I. Protein kinase C activation in the hypothalamus of female reproductive behaviors . In: Pfaff D W , Arnold A P , Etgen A M , Fahrbach rats . 2008 ; 149 : 5509 – 5517 S E , Rubin R T (eds.). Hormones, Brain and Behavior . San Diego : Aca- 25 Balasubramanian B , Portillo W , Reyna A , Chen J Z , Moore A N , Dash demic Press , 2002 ; 441 – 510 P K , Mani S K . Nonclassical mechanisms of progesterone action in 3 Tsai M J , O’Malley B W . Molecular mechanisms of action of steroid/ the brain: II. Role of calmodulin-dependent protein kinase II in pro- receptor superfamily members . Annu Rev Biochem 1994 ; gesterone-mediated signaling in the hypothalamus of female rats. 63 : 451 – 486 Endocrinology 2008 ; 149 : 5518 – 5526 4 Horwitz K B , Tung L , Takimoto G S . Novel mechanisms of antiprogestin 26 Petitti N , Etgen A M . Progesterone depression of norepinephrine- action . Acta Oncol 1996 ; 35: 129 – 140 stimulated cAMP accumulation in hypothalamic slices . Brain Res 5 Katzenellenbogen J A , O’Malley B W , Katzenellenbogen B S . Tripartite Mol Brain Res 1989 ; 5 : 109 – 119 steroid pharmacology: interaction with multiple 27 Petitti N , Etgen A M . Alpha 1-adrenoceptor augmentation of beta- eff ector sites as a basis for the cell- and promoter-specifi c action of stimulated cAMP formation is enhanced by estrogen and reduced these hormones. Mol Endocrinol 1996 ; 10 : 119 – 131 by progesterone in rat hypothalamic slices . J Neurosci 1990 ; 10 : 6 McKenna N J , Lanz R , O’Malley B W . Nuclear receptor coregulators: cel- 2842 – 2849 lular and molecular biology . Endocr Rev 1999 ; 20 : 321 – 344 28 Falkenstein E , Meyer C , Eisen C , Scriba P C , Wehling M. Full-length cDNA 7 Rowan B G , Garrison N , Weigel N L , O’Malley B W . 8-Bromo-cyclic AMP sequence of a progesterone membrane-binding protein from porcine induces phosphorylation of two sites in SRC-1 that facilitate ligand- vascular smooth muscle cells . Biochem Biophys Res Commun 1996 ; independent activation of the chicken and are 229 : 86 – 89 critical for functional cooperation between SRC-1 and CREB binding 29 Zhu Y , Rice C D , Pang Y , Pace M , Thomas P . Cloning, expression, and protein. Mol Cell Biol 2000 ; 20 : 8720 – 8730 characterization of a membrane progestin receptor and evidence it 8 Rowan B G , Weigel N L , O’Malley B W . Phosphorylation of steroid is an intermediary in meiotic maturation of fi sh oocytes . Proc Natl receptor coactivator-1. Identifi cation of the phosphorylation sites Acad Sci USA 2003 ; 100 : 2231 – 2236 and phosphorylation through the mitogen-activated protein kinase 30 Lonard D M , Kumar R , O’Malley B W . Minireview: the SRC family of pathway . J Biol Chem 2000 ; 275 : 4475 – 4483 coactivators: an entree to understanding a subset of polygenic dis- 9 Blaustein J D , Erskine M S . Feminine sexual behavior: Cellular integra- eases? Mol Endocrinol 2010 ; 24 : 279 – 285 tion of hormonal and aff erent information in the rodent forebrain. 31 Johnson A B , O’Malley B W . Steroid receptor coactivators 1, 2 and 3: In: Pfaff D W , Arnold A P , Etgen A M , Fahrbach S E , Rubin R T (eds.). Critical regulators of nuclear receptor activity and steroid receptor Hormones, Brain and Behavior. New York: Academic Press, New York, modulator (SRM)-based cancer therapy . Mol Cell Endocrinol 2011 ; 2002 ; 139 – 214 348 : 430 – 439 10 Meisel R L , Pfa ff D W . RNA and protein synthesis inhibitors: eff ects on 32 Louet J F , O’Malley B W . Coregulators in adipogenesis: what could we sexual behavior in female rats . Brain Res Bull 1984 ; 12 : 187 – 193 learn from the SRC (p160) coactivator family? Cell Cycle 2007 ; 6 : 11 Meisel R L , Pfa ff D W . Specifi city and neural sites of action of anisomy- 2448 – 2452 cin in the reduction or facilitation of female sexual behavior in rats . 33 Chopra A R , Louet J F , Saha P , An J , DeMayo F , Xu J , York B , Karpen S , Horm Behav 1985 ; 19 : 237 – 251 Finegold M , Moore M , Chan L , Newgard C B , O’Malley B W . Absence of 12 Brown T J , Blaustein J D . Inhibition of sexual behavior in female guinea the SRC-2 coactivator results in a glycogenopathy resembling Von pigs by a progestin receptor antagonist . Brain Res 1984 ; 301 : 343 – 349 Gierke’s disease . Science 2008 ; 322 : 1395 – 1399 13 Mani S K , Allen J M , Clark J H , Blaustein J D , O’Malley B W . Convergent 34 Tetel M J , Auger A P , Charlier T D . Who’s in charge? Nuclear receptor pathways for steroid hormone- and neurotransmitter-induced rat coactivator and corepressor function in brain and behavior. Front sexual behavior . Science 1994 ; 265 : 1246 – 1249 Neuroendocrinol 2009 ; 30 : 328 – 342 14 Ogawa S , Olazabal U E , Parhar I S , Pfa ff D W . E ff ects of intrahypotha- 35 Tognoni C M , Chadwick J G Jr , Ackei fi C A , Tetel M J . Nuclear receptor lamic administration of antisense DNA for progesterone receptor coactivators are coexpressed with steroid receptors and regulated mRNA on reproductive behavior and progesterone receptor immu- by estradiol in mouse brain . Neuroendocrinology 2011 ; 94 : 49 – 57 noreactivity in female rat . J Neurosci 1994 ; 14 : 1766 – 1774 36 Misiti S , Schomburg L , Yen P M , Chin W W . Expression and hormo- 15 Pollio G , Xue P , Zanisi M , Nicolin A , Maggi A . Antisense oligonucleotide nal regulation of coactivator and corepressor genes. Endocrinology blocks progesterone-induced lordosis behavior in ovariectomized 1998 ; 139 : 2493 – 2500 rats . Brain Res Mol Brain Res 1993 ; 19 : 135 – 139 37 Shearman L P , Zylka M J , Reppert S M , Weaver D R . Expression of basic 16 Chu H P , Morales J C , Etgen A M . Cyclic GMP may potentiate lordosis helix-loop-helix/PAS genes in the mouse suprachiasmatic nucleus. behaviour by progesterone receptor activation . J Neuroendocrinol Neuroscience 1999 ; 89 : 387 – 397 1999; 11 : 107 – 113 38 Martinez de Arrieta C , Koibuchi N , Chin W W . Coactivator and core- 17 Gonzalez-Flores O , Gomora-Arrati P , Garcia-Juarez M , Gomez-Camarillo pressor gene expression in rat cerebellum during postnatal develop- M A , Lima-Hernandez F J , Beyer C , Etgen A M . Nitric oxide and ERK/ ment and the eff ect of altered thyroid status . Endocrinology 2000 ; MAPK mediation of estrous behavior induced by GnRH, PGE2 and 141 : 1693 – 1698 db-cAMP in rats . Physiol Behav 2009 ; 96 : 606 – 612 39 Meijer O C , Steenbergen P J , de Kloet E R . D i ff erential expression and 18 Mani S K , Allen J M , Lydon J P , Mulac-Jericevic B , Blaustein J D , DeMayo F J , regional distribution of steroid receptor coactivators SRC-1 and Conneely O , O’Malley B W . Dopamine requires the unoccupied proges- SRC-2 in brain and pituitary . Endocrinology 2000 ; 141 : 2192 – 2199 terone receptor to induce sexual behavior in mice. Mol Endocrinol 40 Auger A P , Tetel M J , McCarthy M M . Steroid receptor coactivator-1 1996 ; 10 : 1728 – 1737 mediates the development of sex specifi c brain morphology and 19 Mani S K , Blaustein J D , Allen J M , Law S W , O’Malley B W , Clark J H. Inhi- behavior . Proc Natl Acad Sci USA 2000 ; 97 : 7551 – 7555 bition of rat sexual behavior by antisense oligonucleotides to the 41 Ogawa H , Nishi M , Kawata M . Localization of nuclear coactivators progesterone receptor . Endocrinology 1994 ; 135 : 1409 – 1414 p300 and steroid receptor coactivator 1 in the rat hippocampus. 20 Mani S K , Fienberg A A , O’Callaghan J P , Snyder G L , Allen P B , Dash P K , Brain Res 2001 ; 890 : 197 – 202 Moore A N , Mitchell A J , Bibb J , Greengard P , O’Malley B W . Requirement 42 Nishihara E , Yoshida-Kimoya H , Chan C , Liao L , Davis R L , O’Malley for DARPP-32 in progesterone-facilitated sexual receptivity in female B W , Xu J . SRC-1 null mice exhibit moderate motor dysfunction and rats and mice . Science 2000 ; 287 : 1053 – 1056 delayed development of cerebellar Purkinje cells . J Neurosc 2003 ; 21 Gonzalez-Flores O , Gomora-Arrati P , Garcia-Juarez M , Gomez-Camarillo 23 : 213 – 222 M A , Lima-Hernandez F J , Beyer C , Etgen A M . Nitric oxide and ERK/ 43 McGinnis M Y , Lumia A R , Tetel M J , Molenda-Figuiera H A , Possidente MAPK mediation of estrous behavior induced by GnRH, PGE2 and B . E ff ects of anabolic androgenic steroids on the development and db-cAMP in rats . Physiol Behav 2009 ; 96 : 606 – 612 expression of running wheel activity and circadian rhythms in male 22 Boonyaratanakornkit V , Bi Y , Rudd M , Edwards D P . The role and mech- rats . Physiol Behav 2007 ; 92 : 1010 – 1018 anism of progesterone receptor activation of extranuclear signaling 44 Charlier T D , Lakaye B , Ball G F , Balthazart J . Steroid receptor coactiva- pathways in regulating gene transcription and cell cycle progression . tor SRC-1 exhibits high expression in steroid-sensitive brain areas Steroids 2008 ; 73 : 922 – 928 regulating reproductive behaviors in the quail brain . Neuroendo- crinology 2002 ; 76 : 297 – 315

Mani SK et al. Mechanisms of Steroid Hormone Action … Horm Metab Res 2012; 44: 569–576 Review 575

45 Charlier T D , Balthazart J , Ball G F . S e x d i ff erences in the distribution 66 Mermelstein P G . Membrane-localised oestrogen receptor alpha and of the steroid coactivator SRC-1 in the song control nuclei of male beta infl uence neuronal activity through activation of metabotropic and female canaries. Brain Res 2003 ; 959 : 263 – 274 glutamate receptors . J Neuroendocrinol 2009 ; 21 : 257 – 262 46 Niessen N A , Balthazart J , Ball G F , Charlier T D . Steroid receptor coac- 67 Levin E R . Minireview: Extranuclear Steroid Receptors: Roles in Modu- tivator 2 modulates steroid-dependent male sexual behavior and lation of Cell Functions . Mol Endocrinol 2011 ; 25 : 377 – 384 neuroplasticity in Japanese quail (Coturnix japonica). J Neurochem 68 Boulware M I , Kordasiewicz H , Mermelstein P G . Caveolin proteins are 2011 ; 119 : 579 – 593 essential for distinct eff ects of membrane estrogen receptors in neu- 47 Duncan K A , Jimenez P , Carruth L L . Distribution and sexually dimor- rons . J Neurosci 2007 ; 27 : 9941 – 9950 phic expression of steroid receptor coactivator-1 (SRC-1) in the zebra 69 Grove-Strawser D , Boulware M I , Mermelstein P G . Membrane estrogen fi nch brain . Gen Comp Endocrinol 2011 ; 170 : 408 – 414 receptors activate the metabotropic glutamate receptors mGluR5 and 48 Apostolakis E M , Ramamurphy M , Zhou D , Onate S , O’Malley B . A c u t e mGluR3 to bidirectionally regulate CREB phosphorylation in female disruption of select steroid receptor coactivators prevents reproduc- rat striatal neurons . Neuroscience 2010 ; 170 : 1045 – 1055 tive behavior in rats and unmasks genetic adaptation in knockout 70 Lonze B E , Ginty D D . Function and regulation of CREB family tran- mice . Mol Endocrinol 2002 ; 16 : 1511 – 1523 scription factors in the nervous system. Neuron 2002 ; 35 : 605 – 623 49 Tetel M J , Siegal N K , Murphy S D . Cells in behaviourally relevant brain 71 Gu Q , Moss R L . 17 beta-Estradiol potentiates kainate-induced currents regions coexpress nuclear receptor coactivators and ovarian steroid via activation of the cAMP cascade . J Neurosci 1996 ; 16 : 3620 – 3629 receptors . J Neuroendocrinol 2007 ; 19 : 262 – 271 72 Wade C B , Dorsa D M . Estrogen activation of cyclic adenosine 5′-mono- 50 Camacho-Arroyo I , Neri-Gomez T , Gonzalez-Arenas A , Guerra-Araiza C . phosphate response element-mediated transcription requires the Changes in the content of steroid receptor coactivator-1 and silenc- extracellularly regulated kinase/mitogen-activated protein kinase ing mediator for retinoid and thyroid hormone receptors in the rat pathway . Endocrinology 2003 ; 144 : 832 – 838 brain during the estrous cycle . J Steroid Biochem Mol Biol 2005 ; 73 Lee S J , Campomanes C R , Sikat P T , Green fi eld A T , Allen P B , McEwen B S . 94 : 267 – 272 Estrogen induces phosphorylation of cyclic AMP response element 51 Maerkel K , Durrer S , Henseler M , Schlumpf M , Lichtensteiger W . S e x u - binding (pCREB) in primary hippocampal cells in a time-dependent ally dimorphic gene regulation in brain as a target for endocrine manner . Neuroscience 2003 ; 124 : 549 – 560 disrupters: developmental exposure of rats to 4-methylbenzylidene 74 Abraham I M , Todman M G , Korach K S , Herbison A E . Critical in vivo roles camphor . Toxicol Appl Pharmacol 2007 ; 218 : 152 – 165 for classical estrogen receptors in rapid estrogen actions on intracel- 52 Mitev Y A , Wolf S S , Almeida O F , Patchev V K . Developmental expression lular signaling in mouse brain . Endocrinology 2004 ; 145 : 3055 – 3061 profi les and distinct regional estrogen responsiveness suggest a novel 75 Boulware M I , Weick J P , Becklund B R , Kuo S P , Groth R D , Mermelstein P G . role for the steroid receptor coactivator SRC-l as a discriminative Estradiol activates group I and II metabotropic glutamate receptor amplifi er of estrogen signaling in the rat brain . FASEB J 2003 ; 17 : signaling, leading to opposing infl uences on cAMP response element- 518 – 519 binding protein . J Neurosci 2005 ; 25 : 5066 – 5078 53 Yore M A , Im D , Webb L K , Zhao Y , Chadwick J G Jr , Molenda-Figueira 76 Mermelstein P G , Becker J B , Surmeier D J . Estradiol reduces calcium cur- H A , Haidacher S J , Denner L , Tetel M . Steroid receptor coactivator-2 rents in rat neostriatal neurons via a membrane receptor . J Neurosci expression in brain and physical associations with steriod receptors. 1996 ; 16 : 595 – 604 Neuroscience 2010 ; 169 : 1017 – 1028 77 Balthazart J , Ball G F . Is brain estradiol a hormone or a neurotransmit- 54 Molenda-Figueira H A , Murphy S D , Shea K L , Siegal N K , Zhao Y , Chad- ter? Trends Neurosci 2006 ; 29 : 241 – 249 wick J G Jr , Denner L A , Tetel J . Steroid receptor coactivator-1 from brain 78 Schlinger B A , Callard G V. Localization of aromatase in synaptosomal physically interacts diff erentially with steroid receptor subtypes . and microsomal subfractions of quail (Coturnix coturnix japonica) Endocrinology 2008 ; 149 : 5272 – 5279 brain . Neuroendocrinology 1989 ; 49 : 434 – 441 55 Molenda H A , Gri ffi n A L , Auger A P , McCarthy M M , Tetel M J . N u c l e a r 79 Hojo Y , Hattori T A , Enami T , Furukawa A , Suzuki K , Ishii H T , Mukai H , receptor coactivators modulate hormone-dependent gene expres- Morrison J H , Janssen W G , Kominami S , Harada N , Kimoto T , Kawato sion in brain and female reproductive behavior in rats . Endocrinol- S . Adult male rat hippocampus synthesizes estradiol from pregne- ogy 2002 ; 143 : 436 – 444 nolone by cytochromes P45017alpha and P450 aromatase localized 56 Molenda-Figueira H A , Williams C A , Gri ffi n A L , Rutledge E M , Blaustein in neurons . Proc Natl Acad Sci USA 2004 ; 101 : 865 – 870 J D , Tetel M J . Nuclear receptor coactivators function in estrogen recep- 80 Srivastava D P , Woolfrey K M , Liu F , Brandon N J , Penzes P . Estrogen tor- and progestin receptor-dependent aspects of sexual behavior in receptor b activity modulates synaptic signaling and structure. J Neu- female rats . Horm Behav 2006 ; 50 : 383 – 392 rosci 2010 ; 30 : 13454 – 13460 57 Charlier T D . Importance of steroid receptor coactivators in the modu- 81 Srivastava D P , Waters E M , Mermelstein P G , Kramar E A , Shors T J , Liu lation of steroid action on brain and behavior . Psychoneuroendo- F . Rapid Estrogen Signaling in the Brain: Implications for the Fine- crinology 2009 ; 34 : S20 – S29 Tuning of Neuronal Circuitry . J Neurosci 2011 ; 31 : 16056 – 16063 58 Duncan K A , Carruth L L . The song remains the same: coactivators and 82 Pedram A , Razandi M , Sainson R C , Kim J K , Hughes C C , Levin E R . A c o n - sex diff erences in the songbird brain . Front Neuroendocrinol 2011 ; served mechanism for steroid receptor translocation to the plasma 32 : 84 – 94 membrane . J Biol Chem 2007 ; 282 : 22278 – 22288 59 Szego C M , Davis J S . Adenosine 3′,5′-monophosphate in rat uterus: acute 83 Acconcia F , Ascenzi P , Bocedi A , Spisni E , Tomasi V , Trentalance A , Visca elevation by estrogen . Proc Natl Acad Sci USA 1967 ; 58 : 1711 – 1718 P , Marino M . Palmitoylation-dependent 60 Kelly M J , Moss R L , Dudley C A . D i ff erential sensitivity of preoptic- membrane localization: regulation by 17beta-estradiol . Mol Biol septal neurons to microelectrophoresed estrogen during the estrous Cell 2005 ; 16 : 231 – 237 cycle . Brain Res 1976 ; 114 : 152 – 157 84 Marino M , Ascenzi P , Acconcia F . S-palmitoylation modulates estro- 61 Micevych P E , Mermelstein P G . Membrane estrogen receptors acting gen receptor alpha localization and functions. Steroids 2006 ; 71 : through metabotropic glutamate receptors: an emerging mechanism 298 – 303 of estrogen action in brain . Mol Neurobiol 2008 ; 38 : 66 – 77 85 Dewing P , Boulware M I , Sinchak K , Christensen A , Mermelstein P G , 62 Filardo E J , Quinn J A , Bland KI , Frackelton A R Jr . Estrogen-induced acti- Micevych P . Membrane estrogen receptor-alpha interactions with vation of Erk-1 and Erk-2 requires the G protein-coupled receptor metabotropic glutamate receptor 1a modulate female sexual recep- homolog, GPR30, and occurs via trans-activation of the epidermal tivity in rats . J Neurosci 2007 ; 27 : 9294 – 9300 growth factor receptor through release of HB-EGF. Mol Endocrinol 86 Kuo J , Hamid N , Bondar G , Prossnitz E R , Micevych P . Membrane 2000 ; 14 : 1649 – 1660 estrogen receptors stimulate intracellular calcium release and pro- 63 Singh M , Setalo G Jr , Guan X , Frail D E , Toran-Allerand C D . Estrogen- gesterone synthesis in hypothalamic astrocytes . J Neurosci 2010 ; induced activation of the mitogen-activated protein kinase cascade 30 : 12950 – 12957 in the cerebral cortex of estrogen receptor-alpha knock-out mice . 87 Chaban V , Li J , McDonald J S , Rapkin A , Micevych P . Estradiol attenuates J Neurosci 2000 ; 20 : 1694 – 1700 the adenosine triphosphate-induced increase of intracellular calcium 64 Qiu J , Bosch M A , Tobias S C , Grandy D K , Scanlan T S , Ronnekleiv O K , Kelly through group II metabotropic glutamate receptors in rat dorsal root M J. Rapid signaling of estrogen in hypothalamic neurons involves ganglion neurons . J Neurosci Res 2011 ; 89 : 1707 – 1710 a novel G-protein-coupled estrogen receptor that activates protein 88 Wright C L , McCarthy M M. Prostaglandin E2-induced masculinization kinase C . J Neurosci 2003 ; 23 : 9529 – 9540 of brain and behavior requires protein kinase A, AMPA/kainate, and 65 Otto C , Rohde-Schulz B , Schwarz G , Fuchs I , Klewer M , Brittain D , Langer metabotropic glutamate receptor signaling . J Neurosci 2009 ; 29 : G , Bader B , Prelle K , Nubbemeyer R , Fritzemeier K H. G-protein-coupled 13274 – 13282 receptor 30 localizes to the endoplasmic reticulum and is not acti- vated by estradiol . Endocrinol 2008 ; 149 : 4846 – 4856

Mani SK et al. Mechanisms of Steroid Hormone Action … Horm Metab Res 2012; 44: 569–576 576 Review

89 Christensen A , Dewing P , Micevych P . Membrane-initiated estradiol 103 Couse J F , Yates M M , Deroo B J , Korach K S . Estrogen receptor-beta is signaling induces spinogenesis required for female sexual receptivity . critical to granulosa cell diff erentiation and the ovulatory response J Neurosci 2011 ; 31 : 17583 – 17589 to gonadotropins . Endocrinology 2005 ; 146 : 3247 – 3262 90 Woolley C S . Rapid synaptic actions of estrogens . Society of Neuro- 104 Woodruff T K , Mayo K E . To beta or not to beta: estrogen receptors and science Annual Meeting 308, 2011 ovarian function . Endocrinology 2005 ; 146 : 3244 – 3246 91 Becker J B . 2011 ; personal communication 105 Lara H E , Hill D F , Katz K H , Ojeda S R. The Gene Encoding Nerve Growth 92 Glebova N O , Ginty D D. Growth and survival signals controlling sym- Factor Is Expressed in the Immature Rat Ovary: Eff ect of Denerva- pathetic nervous system development . Annu Rev Neurosci 2005 ; tion and Hormonal Treatment . Endocrinology 1990 ; 126 : 357 – 363 28 : 191 – 222 106 Reichardt L F . Neurotrophin-regulated signalling pathways . Philo- 93 Vanderhorst V G , Gustafsson J A , Ulfhake B . Estrogen receptor-alpha and sophical Transactions of the Royal Society B: Biol Sci 2006 ; 361 : -beta immunoreactive neurons in the brainstem and spinal cord of 1545 – 1564 male and female mice: relationships to monoaminergic, cholinergic, 107 Sohrabji F , Miranda R C , Toran-Allerand C D . Identifi cation of a puta- and spinal projection systems . J Comp Neurol 2005 ; 488 : 152 – 179 tive estrogen response element in the gene encoding brain-derived 94 Anesetti G , Lombide P , Chavez-Genaro R . Prepubertal estrogen expo- neurotrophic factor . Proc Natl Acad Sci USA 1995 ; 92 : 11110 – 11114 sure modifi es neurotrophin receptor expression in celiac neurons 108 Bjorling D E , Beckman M , Clayton M K , Wang Z Y . Modulation of nerve and alters ovarian innervation . Auton Neurosci 2009 ; 145 : 35 – 43 growth factor in peripheral organs by estrogen and progesterone . 95 Papka R E , Storey-Workley M , Shughrue P J , Merchenthaler I , Collins J J , Neuroscience 2002 ; 110 : 155 – 167 Usip S , Saunders P T , Shupnik M . Estrogen receptor-alpha and beta- 109 Richeri A , Bianchimano P , Marmol N M , Viettro L , Cowen T , Brauer M M . immunoreactivity and mRNA in neurons of sensory and autonomic Plasticity in rat uterine sympathetic nerves: the role of TrkA and p75 ganglia and spinal cord . Cell Tissue Res 2001 ; 304 : 193 – 214 nerve growth factor receptors . J Anat 2005 ; 207 : 125 – 134 96 Vega Orozco A S , Daneri C , Anesetti G , Cabrera R , Sosa Z , Rastrilla A M . 110 Chavez-Genaro R , Lombide P , Anesetti G . A quantitative study of rat Involvement of the oestrogenic receptors in superior mesenteric uterine sympathetic innervation during pregnancy and post partum. ganglion on the ovarian steroidogenesis in rat . Reproduction 2012 ; Reprod Fertil Dev 2006 ; 18 : 525 – 531 143 : 183 – 193 111 Lara H E , Dissen G A , Leyton V , Paredes A , Fuenzalida H , Fiedler J L , Ojeda 97 Varayoud J , Ramos J G , Monje L , Bosquiazzo V , Muñoz-de-Toro M , Luque S R . An Increased Intraovarian Synthesis of Nerve Growth Factor and E H . The estrogen receptor α Σ3 mRNA splicing variant is diff erentially Its Low Affi nity Receptor Is a Principal Component of Steroid-Induced regulated by estrogen and progesterone in the rat uterus. J Endocri- Polycystic Ovary in the Rat . Endocrinology 2000 ; 141 : 1059 – 1072 nol 2005 ; 186 : 51 – 60 112 Sotomayor-Zarate R , Dorfman M , Paredes A , Lara H E . Neonatal expo- 98 Hazell GG J , Yao S T , Roper J A , Prossnitz E R , O’Carroll A-M , Lolait S J . sure to estradiol valerate programs ovarian sympathetic innervation Localisation of GPR30, a novel G protein-coupled oestrogen receptor, and follicular development in the adult rat . Biol Reprod 2008 ; 78 : suggests multiple functions in rodent brain and peripheral tissues. 673 – 680 J Endocrinol 2009 ; 202 : 223 – 236 113 Franks S , Stark J , Hardy K . Follicle dynamics and anovulation in poly- 99 Lawrence I E Jr , Burden H W . The origin of the extrinsic adrenergic cystic ovary syndrome. Human Reprod Update 2008 ; 14 : 367 – 378 innervation to the rat ovary . Anat Rec 1980 ; 196 : 51 – 59 114 Lara H E , Dorfman M , Venegas M , Luza S M , Luna S L , Mayerhofer A , 100 Ahmed C E , Dees W L , Ojeda S R . The immature rat ovary is innervated Guimaraes M A , ESAA Rosa , Ramirez V D . Changes in sympathetic nerve by vasoactive intestinal (VIP)-containing fi bers and responds activity of the mammalian ovary during a normal estrous cycle and to VIP with steroid secretion . Endocrinology 1986 ; 118 : 1682 – 1689 in polycystic ovary syndrome: Studies on norepinephrine release. 101 Lara H E , McDonald J K , Ahmed C E , Ojeda S R . Guanethidine-mediated Microsc Res Tech 2002 ; 59 : 495 – 502 destruction of ovarian sympathetic nerves disrupts ovarian develop- 115 Heider U , Pedal I , Spanel-Borowski K . Increase in nerve fi bers and loss ment and function in rats . Endocrinology 1990 ; 127 : 2199 – 2209 of mast cells in polycystic and postmenopausal ovaries . Fertil Steril 102 Mayerhofer A , Dissen G A , Costa M E , Ojeda S R . A role for neurotrans- 2001 ; 75 : 1141 – 1147 mitters in early follicular development: induction of functional fol- 116 Donesky B W , Adashi E Y . Surgically induced ovulation in the polycystic licle-stimulating hormone receptors in newly formed follicles of the ovary syndrome: wedge resection revisited in the age of laparoscopy. rat ovary . Endocrinology 1997 ; 138 : 3320 – 3329 Fertil Steril 1995 ; 63 : 439 – 463

Mani SK et al. Mechanisms of Steroid Hormone Action … Horm Metab Res 2012; 44: 569–576