Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2019 May 1.
Published in final edited form as: Steroids. 2017 Nov 28;133:53–59. doi: 10.1016/j.steroids.2017.11.013

Membrane Estrogen Receptor Signaling Impacts the Reward Circuitry of the Female Brain to Influence Motivated Behaviors

Katherine R Tonn Eisinger 1, Erin B Larson 1, Marissa I Boulware 2, Mark J Thomas 1, Paul G Mermelstein 1
PMCID: PMC5864533  NIHMSID: NIHMS926027  PMID: 29195840

Abstract

Within the adult female, estrogen signaling is well-described as an integral component of the physiologically significant hypothalamic-pituitary-gonadal axis. In rodents, the timing of ovulation is intrinsically entwined with the display of sexual receptivity. For decades, the importance of estradiol activating intracellular estrogen receptors within the hypothalamus and midbrain/spinal cord lordosis circuits has been appreciated. These signaling pathways primarily account for the ability of the female to reproduce. Yet, often overlooked is that the desire to reproduce is also tightly regulated by estrogen receptor signaling. This lack of emphasis can be attributed to an absence of nuclear estrogen receptors in brain regions associated with reward, such as the nucleus accumbens, which are associated with motivated behaviors. This review outlines how membrane-localized estrogen receptors affect metabotropic glutamate receptor signaling within the rodent nucleus accumbens. In addition, we discuss how, as estrogens drive increased motivation for reproduction, they also produce the untoward side effect of heightening female vulnerability to drug addiction.

Keywords: Estrogen Receptors, Drug Addiction, Sex, mGluR, Brain

Estrogen regulation of sexual behavior

The actions of estradiol on brain function and behavior have been studied for many years [1]. Within the adult female, estrogen signaling plays an essential role in reproduction. Primarily synthesized in the ovaries, estradiol synthesis and its subsequent signaling is a principal component regulating the hypothalamus-pituitary-gonadal (HPG) axis [25]. Even before the identification of estrogen receptor alpha (ERα) [6,7], estradiol was demonstrated to accumulate in brain regions involved with reproduction [8,9]. Cloning of the receptor further promulgated the hypothesis that the principal responsibility of estrogen signaling within the brain is to regulate female reproduction, as its distribution is primarily found in the rodent ventromedial hypothalamus, medial preoptic area and central gray region of the midbrain, all essential for the display of lordosis [10].

Coordinating the ability to display lordosis with ovulation is essential for rodent reproduction. But the ability to procreate is a process dissociable from the desire to have sex. Often underappreciated is that estradiol not only synchronizes the ability to display lordosis with ovulation, but, in parallel, also drives the desire to copulate (Figure 1). Simply, sexual behavior is a motivated behavior. Like most motivated behaviors, it requires the integration of sensorimotor circuitry and information about reinforcement in order to properly evaluate a situation, determine its salience, and execute the appropriate behavior [1113]. Failure to recognize that estrogens coordinate the desire to copulate with both ovulation and the ability to display lordosis is most likely due to the limited nuclear estrogen receptor expression in reward-related regions of the brain [8,14,15]. Even when the necessity of estrogen to impact sexual drive has been considered, often it has been theorized that estradiol modifies the motivational system indirectly, through the estrogen receptor-expressing hypothalamic regions [16]. And notably, while in humans the desire and ability to engage in sex has been divorced from ovulation, women report heightened sexual drive following the rise in circulating estrogens around the time of ovulation [17,18].

Figure 1.

Figure 1

In females, estrogen receptor signaling coordinates multiple pathways to enable reproduction. These pathways time ovulation with both the ability and desire to copulate. By altering the brain reward system, estrogen receptor signaling also impacts other motivated behaviors, such as vulnerability to drugs of abuse.

Estradiol regulates the female motivational system

Over the last several years it has become increasingly apparent that the female motivational system is both exquisitely and directly sensitive to estradiol, and that this system plays an important role in sex behavior [19]. Similar to what has been reported in males [20,21], sexual behavior drives dopamine release in the nucleus accumbens [22,23] and this dopamine signal is reinforcing [24]. Accordingly, females who engage in paced mating behavior develop a conditioned place preference [25] and will work for access to a mate [26]. Inhibiting signaling in this brain region blocks the reinforcement properties of sex [27], and lesions of the nucleus accumbens impacts sexual reward in female rats [2831].

Estradiol regulates both the activity and the structure of the nucleus accumbens in order to accommodate these changes when the animal is in a position to copulate. The nucleus accumbens is responsible for much of the complex processing needed for motivated behaviors. Current models posit that glutamatergic afferents from regions including the hippocampus, prefrontal cortex, and amygdala provide signals needed for prediction, dopaminergic inputs from the ventral tegmental area provide reinforcement information, and GABAergic inputs engage in action selection and subsequent motor output [3234]. With the motivational circuitry system largely devoid of nuclear estrogen receptor signaling, the mechanism by which estradiol supports plasticity in the nucleus accumbens is just now being fully elucidated.

In addition to estradiol binding intracellular estrogen receptors that act as ligand-gated transcription factors, estrogens can also have immediate effects on cellular function [35]. For decades, the mechanism(s) behind these rapid effects of estrogens remained controversial. Since at least the 1980s, some have theorized that ERα (and upon its discovery, estrogen receptor beta, ERβ [36]) also acts at the cellular membrane. In some of the first studies to support this hypothesis, estrogen receptors were found to translocate to the membrane of Xenopus oocytes [37,38]. Since then, membrane-localized estrogen receptors have been reported in various cell types and across multiple brain regions [3941]. Some of the most compelling evidence for classical estrogen receptor involvement in rapid estradiol signaling came from experiments using estrogen receptor knockout mice in which ERα and/or ERβ (dependent on brain region) were found responsible for the rapid activation of the MAPK/ERK signaling pathway [42].

Estradiol signals through ER/mGluR complexes at the membrane

Mechanistically, membrane-localized ERα and ERβ were found to activate G-protein signaling [43], though current evidence suggests this is not through direct interactions with G-proteins. (This should not be confused with the estrogen-sensitive GPCR, GPR30/GPER [44].) Evidence from our lab and others shows that membrane-associated ERα and ERβ functionally couple to various group I and II metabotropic glutamate receptors (mGluRs) in order to activate second messenger signaling [40,4548]. Through this mechanism, estradiol can activate mGluR signaling in the absence of glutamate. The particular estrogen receptor-mGluR (ER/mGluR) pairing is cell-type specific, but more than one pairing can occur within the same cell [45,49]. Interestingly, while ER/mGluR signaling is initiated at the membrane, it too can regulate gene expression and long-term changes to neuronal function [50]. Because ER/mGluR signaling is hypothesized to be directly responsible for altering the female motivational circuit, details regarding its mechanism of action are described below. This is not to imply that ER/mGluR signaling is limited to just this neuronal system. In fact, classical lordosis behavior driven by estrogen modulation of hypothalamic function also has an essential ER/mGluR component [51].

In order for ERα and ERβ to be trafficked to the surface membrane, palmitoylation of the receptor must occur. Palmitoylation is a reversible lipid modification that controls membrane tethering of otherwise cytosolic proteins [5254]. Palmitoyl acyltransferases (PATs) are responsible for adding the 16-carbon fatty acid palmitate to target proteins, which increases the hydrophobicity of the protein to facilitate association with lipid membranes and lipophilic proteins. Palmitoylation of a single site on ERα and ERβ is required for membrane-initiated estrogen signaling [5557]. Furthermore, two PATs, DHHC7 and DHHC21, are essential for estrogen receptor trafficking and membrane localization [58]. Knockdown of either of these PATs eliminates ERα and ERβ mGluR signaling in neurons [57]. The reversible nature of the palmitoyl group addition sets up the possibility for palmitoylation-depalmitoylation estrogen receptor cycling to dynamically modulate the membrane signaling responses [59]. Whether this also plays a role with β-arrestin-dependent cycling of ERα/mGluR1a in and out of neuronal membranes [60] remains to be seen.

At the plasma membrane, estrogen receptors must interact with caveolin proteins in order to signal to mGluRs. Caveolins are small integral membrane proteins that spatially organize signaling proteins, including mGluRs, into functional microdomains. In peripheral tissue, caveolins form large oligomers – caveolae – that create invaginations in the plasma membrane. Caveolae are not observed in the brain, but caveolin proteins are still expressed. Modifications to caveolin expression and/or activity interfere with ER/mGluR signaling [47,49,61]. There are three isoforms of caveolin, and all of these are expressed in the brain [49]. Interestingly, different caveolin isoforms produce different ER/mGluR interactions: CAV1 is necessary for ERα pairing with group I mGluRs, whereas CAV3 is necessary for ERα and ERβ coupling to group II mGluRs [49]. Hence, CAV1 and CAV3 organize unique signaling microdomains that link membrane-associated ERs to specific mGluR partners in order to produce divergent effects. Furthermore, caveolin proteins appear to play a dual role in the context of ER/mGluR signaling in that not only do they confer signaling specificity through protein-protein interactions, but, as with palmitoylation, they also likely facilitate trafficking of ERs to the plasma membrane [61,62]. This dynamic interplay of various signaling partners can also be seen following the mutation of the ERα palmitoylation site, interfering with the ability of the estrogen receptor to associate with CAV1 [55]. Figure 2 provides such an example.

Figure 2.

Figure 2

Mutation of the ERα palmitoylation site prevents CAV1 association and membrane localization. (A) HEK293 cells transfected with ERα or palmitoylation-null ERα (ERα Mut) along with CAV1. Western blot revealed that elimination of the ERα palmitoylation site reduced CAV1-ERα co-IP without affecting overall expression of either ERα or CAV1. Untransfected HEK293 cells were run in lane two (i.e. negative control). (B) Membrane (MF) and cytosolic (CF) fractions of HEK293 cells transfected with ERα or ERα Mut indicate the necessity of ERα palmitoylation for membrane localization. Flotillin (Fltn) was used as membrane fraction control.

This functional coupling of ERs with mGluRs provides an avenue through which estradiol can exert far-reaching effects. Importantly, this relationship between estrogen receptors and mGluRs appears unique to females across many brain regions, and appears to exist even in the absence of ER activation. Co-immunoprecipitation studies have indicated a physical association between estrogen receptors and mGluRs [51]. The transactivation hypothesis is further supported by the fact in cultured neurons generated from female rat pups, the estrogen receptor antagonist, ICI 182,780, will attenuate DHPG-induced group I mGluR activation, which leads to increased CREB phosphorylation (Figure 3). Furthermore, disrupting ERα interacting with group I mGluRs via use of a dominant-negative CAV1 construct eliminates the effect. Additional potential off-target effects of ICI 182,780 were ruled out using male-derived cultures, which lack ER/mGluR coupling [45]. In this preparation, ICI 182,780 had no effect on DHPG-induced CREB phosphorylation.

Figure 3.

Figure 3

Transactivation of mGluRs by estrogen receptors. (A) Model of ERα physical association with group I mGluRs via CAV1. (B) Administration of the estrogen receptor antagonist ICI 182,780 (ICI) attenuates DHPG-mediated group I mGluR-dependent CREB phosphorylation (pCREB) in cultured neurons from female rat pups. Transfecting a separate population of neurons with a dominant-negative form of CAV1 tagged with EGFP (EGFP-dnCAV1), eliminates the effect of ICI. (C) ICI has no effect on DHPG-mediated group I mGluR-dependent pCREB in cultures from male rat pups. Group data is the average fluorescent intensity within the nucleus of pyramidal hippocampal neurons across two coverslips. The findings were replicated twice using different cultures.

Estradiol alters the dendritic structure of nucleus accumbens neurons

One of the most striking effects of ER/mGluR signaling is the change of functional circuitry of the nucleus accumbens. This can be seen through structural changes in medium spiny neurons (MSNs), the predominant neuronal subtype in this brain region. MSNs are the output neurons of the nucleus accumbens, and integrate multiple inputs [63,64], processing these signals in order to influence motor and cognitive behaviors through projections to other brain regions [65,66]. Repeated exposure to psychostimulants enhances both glutamatergic and dopaminergic input to the nucleus accumbens [32,67]. MSNs are aptly named for the high density of spines lining their dendrites. These spines receive glutamatergic inputs on the spine heads, and dopamine inputs on the spine necks. The high density of spines indicate the vast number of inputs MSNs receive, and changes in spine density are an essential component of synaptic plasticity [6873].

Estradiol injections in adult ovariectomized rats decrease MSN dendritic spine density in the core region of the nucleus accumbens, indicating a decrease in putative glutamatergic input [74,75]. Pre-treatment with an mGluR5 antagonist, MPEP, eliminates the estradiol-induced change, indicating that this estradiol effect on structure is mediated by ERα/mGluR5 [75]. In contrast, estradiol increases the spine densities on nucleus accumbens shell MSNs though an ERα/mGluR1a mechanism [75]. Estradiol increasing dendritic spines via mGluR1a has been demonstrated/hypothesized in various other brain regions, including the hippocampus and hypothalamus [74,7679]. Through direct mGluR receptor activation, mGluR5 signaling will decrease MSN spine densities throughout the nucleus accumbens, whereas mGluR1a activation results in increased spine densities [80]. Additionally, follow-up experiments determined that ER/mGluR changes in MSN spine densities required mGluR-mediated endocannabinoid release and activation of CB1 receptors [81]. Current experiments are examining which of the specific glutamatergic afferents into the nucleus accumbens are strengthened/weakened following ER/mGluR signaling, ultimately producing increased sexual drive.

Influence of estradiol on sexual motivation generalizes to other motivated behaviors

It is important to note that the neurobiological circuitry driving different motivated behaviors is not distinct from behavior to behavior. Hence, estradiol promotion of lordosis through increased sexual drive impacts other motivated behaviors. And while increased motivation to copulate during ovulation is biologically advantageous, enhanced responsiveness to other potentially rewarding stimuli is not necessarily so. This is most readily observed in models of drug addiction, as drugs of abuse exert their effects through co-opting the brain’s natural reward system. Interactions between hormones and responses to drugs of abuse are revealed in part by the sex differences observed between men and women in addiction. For example, 16–17% of drug users progress to an addicted state [82], but women appear more likely to reach this addicted state than men [83,84]. Additionally, women begin psychostimulant use at younger ages, have increased acute responses, more rapidly escalate use, and progress to addiction faster [8587]. This is especially evident with psychostimulants, but is also seen with opioids, nicotine, alcohol, and marijuana [88,89]. Women also report that psychostimulants produce greater subjective effects when estradiol levels are elevated, due either to natural hormonal cycles or to exogenous administration [86,9092]. These effects have been reproduced in rodent studies, where ovariectomy eliminated sex differences in models of addiction, and estradiol replacement restored those differences [89,9395].

One of the consequences of neuronal plasticity in the motivational system is that sex behavior and exposure to drugs of abuse cross-sensitize. This also solidifies the notion that the motivational pathways for reproduction and drugs of abuse are interrelated [96]. For example, prior sexual experience produces increased dopamine release in the nucleus accumbens during subsequent mating [97] and increases drug-induced locomotor responsiveness [98]. The reverse has also been demonstrated; that is, previous exposure to drugs of abuse enhances motivation to engage in sex behavior [99].

ER/mGluR signaling influences responses to drugs of abuse

Modulation of neurotransmission systems in the nucleus accumbens by drugs of abuse produces long-lasting changes in MSN excitability and structure; these changes are seen as the neurobiological basis for drug addiction [100102]. Thus, it follows that estradiol-induced plasticity within the female nucleus accumbens could impact the effects of drugs of abuse. Much of our current work in females has focused on the role ER/mGluR signaling plays in regulating various aspects of drug addiction. This work builds on a growing body of evidence linking group I mGluRs (especially mGluR5) to responses to drugs of abuse [103]. We find that estradiol replacement facilitates cocaine-induced locomotor sensitization in ovariectomized female rats in an mGluR5-dependent manner [104]. This ERα/mGluR5 mediated effect also relies on endocannabinoid signaling and CB1 receptor activation [81].

Estradiol not only affects locomotor responses to cocaine; interactions between ER/mGluR-induced plasticity and the immense reinforcing properties of drugs of abuse also lead to increased vulnerability to addiction and relapse. In order to explore the effects of estradiol within a model more closely related to addiction, we examined the effects of estradiol and their dependence on mGluR activation using an extended access self-administration paradigm. Estradiol increased cocaine intake in female rats undergoing self-administration; this effect was abolished by the mGluR5 antagonist, MPEP [105].

To illustrate the effects of estradiol-induced plasticity on relapse potential, we performed a conditioned place preference (CPP) study using three groups of ovariectomized female mice (Figure 4). After showing no initial place preference, all animals underwent a four-day training period in which they received a daily injection of either cocaine or saline, and were then restricted to one side of the chamber or the other. Following the training period, all mice displayed a strong preference for the cocaine-associated side of the chamber. Animals then underwent five days of extinction training in which they no longer received cocaine. On the sixth day, the preference for the cocaine-associated side of the chamber was eliminated. Finally, animals were subjected to a reinstatement trial in which they were given a low dose of cocaine or saline prior to being placed back in the CPP chamber. This concentration of cocaine was previously found not to induce reinstatement of CPP in male mice. Saline-treated animals that were administered estradiol 24 hours prior to the reinstatement trial exhibited no reinstatement. Oil-treated animals that were administered the sub-threshold dose of cocaine also did not reinstate. However, estradiol-treated animals given this same dose of cocaine exhibited a robust reinstatement of CPP, suggesting that estrogen potentiation of cocaine action [106108], increases the likelihood for drug-relapse behavior. Notably, while estradiol has been found to directly increase striatal dopamine release [109], we have yet to find conditions where estradiol, in the absence of other trigger events, will induce relapse behavior on its own.

Figure 4.

Figure 4

Estradiol facilitates reinstatement of cocaine-mediated conditioned place preference, a model of drug relapse behavior. Three groups of ovariectomized mice (n=5–6/group) underwent cocaine-mediated conditioned place preference (CPP) and extinction, followed by a test for reinstatement of CPP. Before conditioning (baseline) there was no preference for either side of the chamber. Following two pairings of cocaine (7.5 mg/kg, s.c.) with a particular side of the chamber, all animals displayed CPP (cocaine preference). Following six days of access to both sides of the chamber with no drug pairing, all three groups of animals extinguished CPP (extinction). The following day, only animals that received estradiol (2.5 μg, s.c.) 24 hours before, and a subthreshold dose of cocaine (5 mg/kg) immediately prior to testing, reinstated CPP (reinstatement). Animals receiving either estradiol or cocaine alone did not exhibit reinstatement.

Summary and conclusions

Within adult females, estrogen receptor signaling plays an essential role in reproduction. For most species, females will only display sexual receptivity during ovulation. This has led to the false presumption that these animals have sex in order to reproduce. Further, when compared to women who will engage in intercourse outside periods of fertility, it was presumed that humans were one of the few species to engage in sex for pleasure. We now believe the converse is closer to the truth. That is, for most species, estradiol not only ties ovulation with the ability to engage in sexual intercourse, but also synchronizes ovulation with the motivation to engage in sex. In contrast, humans, who comprehend the consequences of intercourse, are one of the few species that (at times) engage in sex for reproduction. This misunderstanding regarding the role of motivation in sex behavior can be at least partially attributed to the lack of nuclear estrogen receptors in the reward circuitry of the brain, leading to a lack of evidence for estradiol impacting the motivational system. We now know that in females, both ERα and ERβ functionally couple to various group I and II mGluRs across multiple brain regions, including the nucleus accumbens. One of the functional consequences of ER/mGluR signaling in the nucleus accumbens is alteration of neurotransmission, at least partially due to changes in dendritic connectivity. Details regarding how the estradiol-induced changes in dendritic structure ultimately impact sex drive are still forthcoming, but we do know that another functional consequence of ER/mGluR signaling in this brain region is that estradiol produces periods of heightened vulnerability to drug abuse.

Acknowledgments

This work was supported by NIH Grants DA035008 (PGM) and DA041808 (MJT and PGM). KRTE was supported by training grant DA007234 (PGM).

Footnotes

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

References

  • 1.Beach FA. Hormones and behavior; a survey of interrelationships between endocrine secretions and patterns of overt response. Paul B. Hoeber; Oxford, England: 1948. [Google Scholar]
  • 2.Couse JF, Yates MM, Walker VR, Korach KS. Characterization of the Hypothalamic-Pituitary-Gonadal Axis in Estrogen Receptor (ER) Null Mice Reveals Hypergonadism and Endocrine Sex Reversal in Females Lacking ERα But Not ERβ. Mol Endocrinol. 2003;17:1039–1053. doi: 10.1210/me.2002-0398. [DOI] [PubMed] [Google Scholar]
  • 3.Gharib SD, Wierman ME, Shupnik MA, Chin WW. Molecular Biology of the Pituitary Gonadotropins. Endocr Rev. 1990;11:177–199. doi: 10.1210/edrv-11-1-177. [DOI] [PubMed] [Google Scholar]
  • 4.Herbison AE. Multimodal Influence of Estrogen upon Gonadotropin-Releasing Hormone Neurons. Endocr Rev. 1998;19:302–330. doi: 10.1210/edrv.19.3.0332. [DOI] [PubMed] [Google Scholar]
  • 5.Petersen SL, Ottem EN, Carpenter CD. Direct and Indirect Regulation of Gonadotropin-Releasing Hormone Neurons by Estradiol. Biol Reprod. 2003;69:1771–1778. doi: 10.1095/biolreprod.103.019745. [DOI] [PubMed] [Google Scholar]
  • 6.Greene GL, Gilna P, Waterfield M, Baker A, Hort Y, Shine J. Sequence and expression of human estrogen receptor complementary DNA. Science. 1986;231:1150–1154. doi: 10.1126/science.3753802. [DOI] [PubMed] [Google Scholar]
  • 7.Green S, Walter P, Kumar V, Krust A, Bornert JM, Argos P, Chambon P. Human oestrogen receptor cDNA: sequence, expression and homology to v-erb-A. Nature. 1986;320:134–139. doi: 10.1038/320134a0. [DOI] [PubMed] [Google Scholar]
  • 8.Pfaff D, Keiner M. Atlas of estradiol-concentrating cells in the central nervous system of the female rat. J Comp Neurol. 1973;151:121–157. doi: 10.1002/cne.901510204. [DOI] [PubMed] [Google Scholar]
  • 9.Stumpf WE, Sar M. Steroid hormone target sites in the brain: The differential distribution of estrogen, progestin, androgen and glucocorticosteroid. J Steroid Biochem. 1976;7:1163–1170. doi: 10.1016/0022-4731(76)90050-9. [DOI] [PubMed] [Google Scholar]
  • 10.Pfaff DW, Schwartz-Giblin S, McCarthy MM, Kow LM. Cellular and molecular mechanisms of female reproductive behaviors. In: Knobil E, Neill JD, editors. Physiol Reprod. 2nd. Raven Press, Ltd; New York, New York: 1994. [Google Scholar]
  • 11.Everitt BJ. Sexual motivation: A neural and behavioural analysis of the mechanisms underlying appetitive and copulatory responses of male rats. Neurosci Biobehav Rev. 1990;14:217–232. doi: 10.1016/S0149-7634(05)80222-2. [DOI] [PubMed] [Google Scholar]
  • 12.Berridge KC. Motivation concepts in behavioral neuroscience. Physiol Behav. 2004;81:179–209. doi: 10.1016/j.physbeh.2004.02.004. [DOI] [PubMed] [Google Scholar]
  • 13.Yoest KE, Cummings JA, Becker JB. Estradiol, Dopamine and Motivation. Cent Nerv Syst Agents Med Chem - Cent Nerv Syst Agents. 2014;14:83–89. doi: 10.2174/1871524914666141226103135. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Simerly RB, Swanson LW, Chang C, Muramatsu M. Distribution of androgen and estrogen receptor mRNA-containing cells in the rat brain: An in situ hybridization study. J Comp Neurol. 1990;294:76–95. doi: 10.1002/cne.902940107. [DOI] [PubMed] [Google Scholar]
  • 15.Shughrue PJ, Lane MV, Merchenthaler I. Comparative distribution of estrogen receptor-α and -β mRNA in the rat central nervous system. J Comp Neurol. 1997;388:507–525. doi: 10.1002/(sici)1096-9861(19971201)388:4<507::aid-cne1>3.0.co;2-6. doi:10.1002/(SICI)1096-9861(19971201)388:4<507::AID-CNE1>3.0.CO;2-6. [DOI] [PubMed] [Google Scholar]
  • 16.Pfaff DW. Drive: Neurobiological and Molecular Mechanisms of Sexual Motivation. MIT Press; 1999. [Google Scholar]
  • 17.Adams DB, Gold AR, Burt AD. Rise in Female-Initiated Sexual Activity at Ovulation and Its Suppression by Oral Contraceptives. N Engl J Med. 1978;299:1145–1150. doi: 10.1056/NEJM197811232992101. [DOI] [PubMed] [Google Scholar]
  • 18.Wilcox AJ, Day Baird D, Dunson DB, McConnaughey DR, Kesner JS, Weinberg CR. On the frequency of intercourse around ovulation: evidence for biological influences. Hum Reprod. 2004;19:1539–1543. doi: 10.1093/humrep/deh305. [DOI] [PubMed] [Google Scholar]
  • 19.Micevych PE, Meisel RL. Integrating Neural Circuits Controlling Female Sexual Behavior. Front Syst Neurosci. 2017;11 doi: 10.3389/fnsys.2017.00042. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Pfaus JG, Damsma G, Nomikos GG, Wenkstern DG, Blaha CD, Phillips AG, Fibiger HC. Sexual behavior enhances central dopamine transmission in the male rat. Brain Res. 1990;530:345–348. doi: 10.1016/0006-8993(90)91309-5. [DOI] [PubMed] [Google Scholar]
  • 21.Wenkstern D, Pfaus JG, Fibiger HC. Dopamine transmission increases in the nucleus accumbens of male rats during their first exposure to sexually receptive female rats. Brain Res. 1993;618:41–46. doi: 10.1016/0006-8993(93)90426-N. [DOI] [PubMed] [Google Scholar]
  • 22.Meisel RL, Camp DM, Robinson TE. A microdialysis study of ventral striatal dopamine during sexual behavior in female Syrian hamsters. Behav Brain Res. 1993;55:151–157. doi: 10.1016/0166-4328(93)90111-3. [DOI] [PubMed] [Google Scholar]
  • 23.Mermelstein PG, Becker JB. Increased extracellular dopamine in the nucleus accumbens and striatum of the female rat during paced copulatory behavior. Behav Neurosci. 1995;109:354–365. doi: 10.1037//0735-7044.109.2.354. [DOI] [PubMed] [Google Scholar]
  • 24.Meisel RL, Joppa MA. Conditioned place preference in female hamsters following aggressive or sexual encounters. Physiol Behav. 1994;56:1115–1118. doi: 10.1016/0031-9384(94)90352-2. [DOI] [PubMed] [Google Scholar]
  • 25.Jenkins WJ, Becker JB. Female rats develop conditioned place preferences for sex at their preferred interval. Horm Behav. 2003;43:503–507. doi: 10.1016/S0018-506X(03)00031-X. [DOI] [PubMed] [Google Scholar]
  • 26.Matthews TJ, Grigore M, Tang L, Doat M, Kow LM, Pfaff DW. Sexual Reinforcement in the Female Rat. J Exp Anal Behav. 1997;68:399–410. doi: 10.1901/jeab.1997.68-399. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Meisel RL, Joppa MA, Rowe RK. Dopamine receptor antagonists attenuate conditioned place preference following sexual behavior in female Syrian hamsters. Eur J Pharmacol. 1996;309:21–24. doi: 10.1016/0014-2999(96)00389-5. [DOI] [PubMed] [Google Scholar]
  • 28.Dohanich GP, McEwen BS. Cholinergic limbic projections and behavioral role of basal forebrain nuclei in the rat. Brain Res Bull. 1986;16:477–482. doi: 10.1016/0361-9230(86)90176-0. [DOI] [PubMed] [Google Scholar]
  • 29.Rivas FJ, Mir D. Effects of nucleus accumbens lesion on female rat sexual receptivity and proceptivity in a partner preference paradigm. Behav Brain Res. 1990;41:239–249. doi: 10.1016/0166-4328(90)90111-Q. [DOI] [PubMed] [Google Scholar]
  • 30.Rivas FJ, Mir D. Accumbens lesion in female rats increases mount rejection without modifying lordosis. Rev Esp Fisiol. 1991;47:1–6. [PubMed] [Google Scholar]
  • 31.Guarraci FA, Megroz AB, Clark AS. Effects of ibotenic acid lesions of the nucleus accumbens on paced mating behavior in the female rat. Behav Neurosci. 2002;116:568–576. doi: 10.1037//0735-7044.116.4.568. [DOI] [PubMed] [Google Scholar]
  • 32.Nestler EJ. Molecular basis of long-term plasticity underlying addiction. Nat Rev Neurosci. 2001;2:119–128. doi: 10.1038/35053570. [DOI] [PubMed] [Google Scholar]
  • 33.Sesack SR, Grace AA. Cortico-Basal Ganglia Reward Network: Microcircuitry. Neuropsychopharmacology. 2010;35:27–47. doi: 10.1038/npp.2009.93. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Lüscher C, Malenka RC. Drug-Evoked Synaptic Plasticity in Addiction: From Molecular Changes to Circuit Remodeling. Neuron. 2011;69:650–663. doi: 10.1016/j.neuron.2011.01.017. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Szego CM, Davis JS. Adenosine 3′,5′-monophosphate in rat uterus: acute elevation by estrogen. Proc Natl Acad Sci U S A. 1967;58:1711–1718. doi: 10.1073/pnas.58.4.1711. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.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. 1996;93:5925–5930. doi: 10.1073/pnas.93.12.5925. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Sadler SE, Maller JL. Identification of a steroid receptor on the surface of Xenopus oocytes by photoaffinity labeling. J Biol Chem. 1982;257:355–361. [PubMed] [Google Scholar]
  • 38.Sadler SE, Bower MA, Maller JL. Studies of a plasma membrane steroid receptor in Xenopus oocytes using the synthetic progestin RU 486. J Steroid Biochem. 1985;22:419–426. doi: 10.1016/0022-4731(85)90448-0. [DOI] [PubMed] [Google Scholar]
  • 39.Razandi M, Pedram A, Greene GL, Levin ER. Cell membrane and nuclear estrogen receptors (ERs) originate from a single transcript: studies of ERalpha and ERbeta expressed in Chinese hamster ovary cells. Mol Endocrinol Baltim Md. 1999;13:307–319. doi: 10.1210/mend.13.2.0239. [DOI] [PubMed] [Google Scholar]
  • 40.Micevych PE, Mermelstein PG. Membrane Estrogen Receptors Acting Through Metabotropic Glutamate Receptors: An Emerging Mechanism of Estrogen Action in Brain. Mol Neurobiol. 2008;38:66–77. doi: 10.1007/s12035-008-8034-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Pedram A, Razandi M, Kim JK, O’Mahony F, Lee EY, Luderer U, Levin ER. Developmental Phenotype of a Membrane Only Estrogen Receptor α (MOER) Mouse. J Biol Chem. 2009;284:3488–3495. doi: 10.1074/jbc.M806249200. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Ábrahám IM, Todman MG, Korach KS, Herbison AE. Critical in Vivo Roles for Classical Estrogen Receptors in Rapid Estrogen Actions on Intracellular Signaling in Mouse Brain. Endocrinology. 2004;145:3055–3061. doi: 10.1210/en.2003-1676. [DOI] [PubMed] [Google Scholar]
  • 43.Mermelstein PG, Becker JB, Surmeier DJ, Mermelstein PG, Becker JB, Surmeier DJ. Estrogen reduces calcium currents in rat neostriatal neurons via a membrane receptor. J Neurosci. 16:595–604. doi: 10.1523/JNEUROSCI.16-02-00595.1996. [DOI] [PMC free article] [PubMed] [Google Scholar]; ResearchGate. 1996;16:595–604. [Google Scholar]
  • 44.Filardo EJ, Quinn JA, Frackelton AR, Bland KI. Estrogen Action Via the G Protein-Coupled Receptor, GPR30: Stimulation of Adenylyl Cyclase and cAMP-Mediated Attenuation of the Epidermal Growth Factor Receptor-to-MAPK Signaling Axis. Mol Endocrinol. 2002;16:70–84. doi: 10.1210/mend.16.1.0758. [DOI] [PubMed] [Google Scholar]
  • 45.Boulware MI, Weick JP, Becklund BR, Kuo SP, Groth RD, Mermelstein PG. Estradiol activates group I and II metabotropic glutamate receptor signaling, leading to opposing influences on cAMP response element-binding protein. J Neurosci Off J Soc Neurosci. 2005;25:5066–5078. doi: 10.1523/JNEUROSCI.1427-05.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Kuo J, Hariri OR, Bondar G, Ogi J, Micevych P. Membrane Estrogen Receptor-α Interacts with Metabotropic Glutamate Receptor Type 1a to Mobilize Intracellular Calcium in Hypothalamic Astrocytes. Endocrinology. 2009;150:1369–1376. doi: 10.1210/en.2008-0994. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Grove-Strawser D, Boulware MI, Mermelstein PG. Membrane estrogen receptors activate the metabotropic glutamate receptors mGluR5 and mGluR3 to bidirectionally regulate CREB phosphorylation in female rat striatal neurons. Neuroscience. 2010;170:1045–1055. doi: 10.1016/j.neuroscience.2010.08.012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Huang GZ, Woolley CS. Estradiol Acutely Suppresses Inhibition in the Hippocampus through a Sex-Specific Endocannabinoid and mGluR-Dependent Mechanism. Neuron. 2012;74:801–808. doi: 10.1016/j.neuron.2012.03.035. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Boulware MI, Kordasiewicz H, Mermelstein PG. Caveolin Proteins Are Essential for Distinct Effects of Membrane Estrogen Receptors in Neurons. J Neurosci. 2007;27:9941–9950. doi: 10.1523/JNEUROSCI.1647-07.2007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Meitzen J, Mermelstein PG. Estrogen receptors stimulate brain region specific metabotropic glutamate receptors to rapidly initiate signal transduction pathways. J Chem Neuroanat. 2011;42:236–241. doi: 10.1016/j.jchemneu.2011.02.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Dewing P, Boulware MI, Sinchak K, Christensen A, Mermelstein PG, Micevych P. Membrane Estrogen Receptor-α Interactions with Metabotropic Glutamate Receptor 1a Modulate Female Sexual Receptivity in Rats. J Neurosci. 2007;27:9294–9300. doi: 10.1523/JNEUROSCI.0592-07.2007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Wedegaertner PB, Chu DH, Wilson PT, Levis MJ, Bourne HR. Palmitoylation is required for signaling functions and membrane attachment of Gq alpha and Gs alpha. J Biol Chem. 1993;268:25001–25008. [PubMed] [Google Scholar]
  • 53.Topinka JR, Bredt DS. N-Terminal Palmitoylation of PSD-95 Regulates Association with Cell Membranes and Interaction with K+ Channel Kv1.4. Neuron. 1998;20:125–134. doi: 10.1016/S0896-6273(00)80440-7. [DOI] [PubMed] [Google Scholar]
  • 54.Linder ME, Deschenes RJ. Palmitoylation: policing protein stability and traffic. Nat Rev Mol Cell Biol. 2007;8:74–84. doi: 10.1038/nrm2084. [DOI] [PubMed] [Google Scholar]
  • 55.Acconcia F, Ascenzi P, Fabozzi G, Visca P, Marino M. S-palmitoylation modulates human estrogen receptor-α functions. Biochem Biophys Res Commun. 2004;316:878–883. doi: 10.1016/j.bbrc.2004.02.129. [DOI] [PubMed] [Google Scholar]
  • 56.Pedram A, Razandi M, Sainson RCA, Kim JK, Hughes CC, Levin ER. A Conserved Mechanism for Steroid Receptor Translocation to the Plasma Membrane. J Biol Chem. 2007;282:22278–22288. doi: 10.1074/jbc.M611877200. [DOI] [PubMed] [Google Scholar]
  • 57.Meitzen J, Luoma JI, Boulware MI, Hedges VL, Peterson BM, Tuomela K, Britson KA, Mermelstein PG. Palmitoylation of Estrogen Receptors Is Essential for Neuronal Membrane Signaling. Endocrinology. 2013;154:4293–4304. doi: 10.1210/en.2013-1172. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Pedram A, Razandi M, Deschenes RJ, Levin ER. DHHC-7 and -21 are palmitoylacyltransferases for sex steroid receptors. Mol Biol Cell. 2012;23:188–199. doi: 10.1091/mbc.E11-07-0638. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Tabatadze N, Smejkalova T, Woolley CS. Distribution and Posttranslational Modification of Synaptic ERα in the Adult Female Rat Hippocampus. Endocrinology. 2013;154:819–830. doi: 10.1210/en.2012-1870. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Wong AM, Abrams MC, Micevych PE. β-Arrestin Regulates Estradiol Membrane-Initiated Signaling in Hypothalamic Neurons. PLoS ONE. 2015;10 doi: 10.1371/journal.pone.0120530. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Stern CM, Mermelstein PG. Caveolin regulation of neuronal intracellular signaling. Cell Mol Life Sci. 2010;67:3785–3795. doi: 10.1007/s00018-010-0447-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Christensen A, Micevych P. CAV1 siRNA Reduces Membrane Estrogen Receptor-α Levels and Attenuates Sexual Receptivity. Endocrinology. 2012;153:3872–3877. doi: 10.1210/en.2012-1312. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Groenewegen HJ, Wright CI, Beijer AVj, Voorn P. Convergence and Segregation of Ventral Striatal Inputs and Outputs. Ann N Y Acad Sci. 1999;877:49–63. doi: 10.1111/j.1749-6632.1999.tb09260.x. [DOI] [PubMed] [Google Scholar]
  • 64.Kelley AE. Memory and Addiction: Shared Neural Circuitry and Molecular Mechanisms. Neuron. 2004;44:161–179. doi: 10.1016/j.neuron.2004.09.016. [DOI] [PubMed] [Google Scholar]
  • 65.Smith RJ, Lobo MK, Spencer S, Kalivas PW. Cocaine-induced adaptations in D1 and D2 accumbens projection neurons (a dichotomy not necessarily synonymous with direct and indirect pathways) Curr Opin Neurobiol. 2013;23:546–552. doi: 10.1016/j.conb.2013.01.026. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Yager LM, Garcia AF, Wunsch AM, Ferguson SM. The ins and outs of the striatum: Role in drug addiction. Neuroscience. 2015;301:529–541. doi: 10.1016/j.neuroscience.2015.06.033. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.D’Souza MS. Glutamatergic transmission in drug reward: implications for drug addiction. Front Neurosci. 2015;9 doi: 10.3389/fnins.2015.00404. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.O’Donnell P, Grace AA. Synaptic interactions among excitatory afferents to nucleus accumbens neurons: hippocampal gating of prefrontal cortical input. J Neurosci. 1995;15:3622–3639. doi: 10.1523/JNEUROSCI.15-05-03622.1995. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Mulder AB, Hodenpijl MG, da Silva FHL. Electrophysiology of the Hippocampal and Amygdaloid Projections to the Nucleus Accumbens of the Rat: Convergence, Segregation, and Interaction of Inputs. J Neurosci. 1998;18:5095–5102. doi: 10.1523/JNEUROSCI.18-13-05095.1998. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Sesack SR, Carr DB, Omelchenko N, Pinto A. Anatomical Substrates for Glutamate-Dopamine Interactions. Ann N Y Acad Sci. 2003;1003:36–52. doi: 10.1196/annals.1300.066. [DOI] [PubMed] [Google Scholar]
  • 71.Papp E, Borhegyi Z, Tomioka R, Rockland KS, Mody I, Freund TF. Glutamatergic input from specific sources influences the nucleus accumbens-ventral pallidum information flow. Brain Struct Funct. 2011;217:37–48. doi: 10.1007/s00429-011-0331-z. [DOI] [PubMed] [Google Scholar]
  • 72.Stuber GD, Sparta DR, Stamatakis AM, van Leeuwen WA, Hardjoprajitno JE, Cho S, Tye KM, Kempadoo KA, Zhang F, Deisseroth K, Bonci A. Excitatory transmission from the amygdala to nucleus accumbens facilitates reward seeking. Nature. 2011;475:377–380. doi: 10.1038/nature10194. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Britt JP, Benaliouad F, McDevitt RA, Stuber GD, Wise RA, Bonci A. Synaptic and Behavioral Profile of Multiple Glutamatergic Inputs to the Nucleus Accumbens. Neuron. 2012;76:790–803. doi: 10.1016/j.neuron.2012.09.040. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Staffend NA, Loftus CM, Meisel RL. Estradiol reduces dendritic spine density in the ventral striatum of female Syrian hamsters. Brain Struct Funct. 2010;215:187–194. doi: 10.1007/s00429-010-0284-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Peterson BM, Mermelstein PG, Meisel RL. Estradiol mediates dendritic spine plasticity in the nucleus accumbens core through activation of mGluR5. Brain Struct Funct. 2014:1–8. doi: 10.1007/s00429-014-0794-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Gould E, Woolley CS, Frankfurt M, McEwen BS. Gonadal steroids regulate dendritic spine density in hippocampal pyramidal cells in adulthood. J Neurosci. 1990;10:1286–1291. doi: 10.1523/JNEUROSCI.10-04-01286.1990. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Woolley CS, McEwen BS. Estradiol mediates fluctuation in hippocampal synapse density during the estrous cycle in the adult rat [published erratum appears in J Neurosci 1992 Oct;12(10):following table of contents] J Neurosci. 1992;12:2549–2554. doi: 10.1523/JNEUROSCI.12-07-02549.1992. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Woolley CS, McEwen BS. Roles of estradiol and progesterone in regulation of hippocampal dendritic spine density during the estrous cycle in the rat. J Comp Neurol. 1993;336:293–306. doi: 10.1002/cne.903360210. [DOI] [PubMed] [Google Scholar]
  • 79.Christensen A, Dewing P, Micevych P. Membrane-Initiated Estradiol Signaling Induces Spinogenesis Required for Female Sexual Receptivity. J Neurosci. 2011;31:17583–17589. doi: 10.1523/JNEUROSCI.3030-11.2011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Gross KS, Brandner DD, Martinez LA, Olive MF, Meisel RL, Mermelstein PG. Opposite Effects of mGluR1a and mGluR5 Activation on Nucleus Accumbens Medium Spiny Neuron Dendritic Spine Density. PLOS ONE. 2016;11:e0162755. doi: 10.1371/journal.pone.0162755. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Peterson BM, Martinez LA, Meisel RL, Mermelstein PG. Estradiol impacts the endocannabinoid system in female rats to influence behavioral and structural responses to cocaine. Neuropharmacology. 2016;110(Part A):118–124. doi: 10.1016/j.neuropharm.2016.06.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.SAMHSA. Behavioral Health Trends in the United States: Results from the 2014 National Survey on Drug Use and Health. 2014 [Google Scholar]
  • 83.Wagner FA, Anthony JC. Male–female differences in the risk of progression from first use to dependence upon cannabis, cocaine, and alcohol. Drug Alcohol Depend. 2007;86:191–198. doi: 10.1016/j.drugalcdep.2006.06.003. [DOI] [PubMed] [Google Scholar]
  • 84.Becker JB, Perry AN, Westenbroek C. Sex differences in the neural mechanisms mediating addiction: a new synthesis and hypothesis. Biol Sex Differ. 2012;3:14. doi: 10.1186/2042-6410-3-14. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Etten MLV, Neumark YD, Anthony JC. Male-female differences in the earliest stages of drug involvement. Addiction. 1999;94:1413–1419. doi: 10.1046/j.1360-0443.1999.949141312.x. [DOI] [PubMed] [Google Scholar]
  • 86.Justice AJH, de Wit H. Acute effects of d-amphetamine during the follicular and luteal phases of the menstrual cycle in women. Psychopharmacology (Berl) 1999;145:67–75. doi: 10.1007/s002130051033. [DOI] [PubMed] [Google Scholar]
  • 87.Van Etten ML, Anthony JC. Male-Female Differences in Transitions from First Drug Opportunity to First Use: Searching for Subgroup Variation by Age, Race, Region, and Urban Status. J Womens Health Gend Based Med. 2001;10:797–804. doi: 10.1089/15246090152636550. [DOI] [PubMed] [Google Scholar]
  • 88.Carroll ME, Lynch WJ, Roth ME, Morgan AD, Cosgrove KP. Sex and estrogen influence drug abuse. Trends Pharmacol Sci. 2004;25:273–279. doi: 10.1016/j.tips.2004.03.011. [DOI] [PubMed] [Google Scholar]
  • 89.Becker JB, Hu M. Sex differences in drug abuse. Front Neuroendocrinol. 2008;29:36–47. doi: 10.1016/j.yfrne.2007.07.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Evans SM, Haney M, Foltin RW. The effects of smoked cocaine during the follicular and luteal phases of the menstrual cycle in women. Psychopharmacology (Berl) 2002;159:397–406. doi: 10.1007/s00213-001-0944-7. [DOI] [PubMed] [Google Scholar]
  • 91.Terner JM, de Wit H. Menstrual cycle phase and responses to drugs of abuse in humans. Drug Alcohol Depend. 2006;84:1–13. doi: 10.1016/j.drugalcdep.2005.12.007. [DOI] [PubMed] [Google Scholar]
  • 92.Maria MMMS, Flanagan J, Brady K. Ovarian Hormones and Drug Abuse. Curr Psychiatry Rep. 2014;16:511. doi: 10.1007/s11920-014-0511-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.Sircar R, Kim D. Female Gonadal Hormones Differentially Modulate Cocaine-Induced Behavioral Sensitization in Fischer, Lewis, and Sprague-Dawley Rats. J Pharmacol Exp Ther. 1999;289:54–65. [PubMed] [Google Scholar]
  • 94.Anker JJ, Carroll ME. Biol Basis Sex Differ Psychopharmacol. Springer; Berlin, Heidelberg: 2010. Females Are More Vulnerable to Drug Abuse than Males: Evidence from Preclinical Studies and the Role of Ovarian Hormones; pp. 73–96. https://link.springer.com/chapter/10.1007/7854_2010_93 (accessed September 26, 2017) [DOI] [PubMed] [Google Scholar]
  • 95.Segarra AC, Agosto-Rivera JL, Febo M, Lugo-Escobar N, Menéndez-Delmestre R, Puig-Ramos A, Torres-Diaz YM. Estradiol: A key biological substrate mediating the response to cocaine in female rats. Horm Behav. 2010;58:33–43. doi: 10.1016/j.yhbeh.2009.12.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96.Hedges VL, Staffend NA, Meisel RL. Neural mechanisms of reproduction in females as a predisposing factor for drug addiction, Front. Neuroendocrinol. 2010;31:217–231. doi: 10.1016/j.yfrne.2010.02.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97.Kohlert JG, Meisel RL. Sexual experience sensitizes mating-related nucleus accumbens dopamine responses of female Syrian hamsters. Behav Brain Res. 1999;99:45–52. doi: 10.1016/S0166-4328(98)00068-0. [DOI] [PubMed] [Google Scholar]
  • 98.Bradley KC, Meisel RL. Sexual behavior induction of c-Fos in the nucleus accumbens and amphetamine-stimulated locomotor activity are sensitized by previous sexual experience in female Syrian hamsters. J Neurosci Off J Soc Neurosci. 2001;21:2123–2130. doi: 10.1523/JNEUROSCI.21-06-02123.2001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99.Afonso VM, Mueller D, Stewart J, Pfaus JG. Amphetamine pretreatment facilitates appetitive sexual behaviors in the female rat. Psychopharmacology (Berl) 2009;205:35–43. doi: 10.1007/s00213-009-1511-x. [DOI] [PubMed] [Google Scholar]
  • 100.Kourrich S, Calu DJ, Bonci A. Intrinsic plasticity: an emerging player in addiction. Nat Rev Neurosci. 2015;16:173–184. doi: 10.1038/nrn3877. [DOI] [PubMed] [Google Scholar]
  • 101.Russo SJ, Dietz DM, Dumitriu D, Morrison JH, Malenka RC, Nestler EJ. The addicted synapse: mechanisms of synaptic and structural plasticity in nucleus accumbens. Trends Neurosci. 2010;33:267–276. doi: 10.1016/j.tins.2010.02.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102.Golden SA, Russo SJ. Mechanisms of Psychostimulant-Induced Structural Plasticity. Cold Spring Harb Perspect Med. 2012;2:a011957. doi: 10.1101/cshperspect.a011957. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103.Pomierny-Chamioło L, Rup K, Pomierny B, Niedzielska E, Kalivas PW, Filip M. Metabotropic glutamatergic receptors and their ligands in drug addiction. Pharmacol Ther. 2014;142:281–305. doi: 10.1016/j.pharmthera.2013.12.012. [DOI] [PubMed] [Google Scholar]
  • 104.Martinez LA, Peterson BM, Meisel RL, Mermelstein PG. Estradiol facilitation of cocaine-induced locomotor sensitization in female rats requires activation of mGluR5. Behav Brain Res. 2014;271:39–42. doi: 10.1016/j.bbr.2014.05.052. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105.Martinez LA, Gross KS, Himmler BT, Emmitt NL, Peterson BM, Zlebnik NE, Olive MF, Carroll ME, Meisel RL, Mermelstein PG. Estradiol Facilitation of Cocaine Self-Administration in Female Rats Requires Activation of mGluR5. eNeuro. 2016;3 doi: 10.1523/ENEURO.0140-16.2016. ENEURO.0140-16.2016. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106.Becker JB. Direct effect of 17β-estradiol on striatum: Sex differences in dopamine release. Synapse. 1990;5:157–164. doi: 10.1002/syn.890050211. [DOI] [PubMed] [Google Scholar]
  • 107.Castner SA, Xiao L, Becker JB. Sex differences in striatal dopamine: in vivo microdialysis and behavioral studies. Brain Res. 1993;610:127–134. doi: 10.1016/0006-8993(93)91225-H. [DOI] [PubMed] [Google Scholar]
  • 108.Walker QD, Johnson ML, Van Swearingen AED, Arrant AE, Caster JM, Kuhn CM. Individual differences in psychostimulant responses of female rats are associated with ovarian hormones and dopamine neuroanatomy. Neuropharmacology. 2012;62:2267–2277. doi: 10.1016/j.neuropharm.2012.01.029. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109.Cummings JA, Jagannathan L, Jackson LR, Becker JB. Sex differences in the effects of estradiol in the nucleus accumbens and striatum on the response to cocaine: Neurochemistry and behavior. Drug Alcohol Depend. 2014;135:22–28. doi: 10.1016/j.drugalcdep.2013.09.009. [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES