Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2016 May 15.
Published in final edited form as: Behav Brain Res. 2014 Aug 15;285:140–157. doi: 10.1016/j.bbr.2014.08.001

Regulation of object recognition and object placement by ovarian sex steroid hormones

Jennifer J Tuscher 1, Ashley M Fortress 1, Jaekyoon Kim 1, Karyn M Frick 1,*
PMCID: PMC4329280  NIHMSID: NIHMS623180  PMID: 25131507

Abstract

The ovarian hormones 17β-estradiol (E2) and progesterone (P4) are potent modulators of hippocampal memory formation. Both hormones have been demonstrated to enhance hippocampal memory by regulating the cellular and molecular mechanisms thought to underlie memory formation. Behavioral neuroendocrinologists have increasingly used the object recognition and object placement (object location) tasks to investigate the role of E2 and P4 in regulating hippocampal memory formation in rodents. These one-trial learning tasks are ideal for studying acute effects of hormone treatments on different phases of memory because they can be administered during acquisition (pre-training), consolidation (post-training), or retrieval (pre-testing). This review synthesizes the rodent literature testing the effects of E2 and P4 on object recognition (OR) and object placement (OP), and the molecular mechanisms in the hippocampus supporting memory formation in these tasks. Some general trends emerge from the data. Among gonadally intact females, object memory tends to be best when E2 and P4 levels are elevated during the estrous cycle, pregnancy, and in middle age. In ovariectomized females, E2 given before or immediately after testing generally enhances OR and OP in young and middle-aged rats and mice, although effects are mixed in aged rodents. Effects of E2 treatment on OR 7and OP memory consolidation can be mediated by both classical estrogen receptors (ERα and ERβ), and depend on glutamate receptors (NMDA, mGluR1) and activation of numerous cell signaling cascades (e.g., ERK, PI3K/Akt, mTOR) and epigenetic processes (e.g., histone H3 acetylation, DNA methylation). Acute P4 treatment given immediately after training also enhances OR and OP in young and middle-aged ovariectomized females by activating similar cell signaling pathways as E2 (e.g., ERK, mTOR). The few studies that have administered both hormones in combination suggest that treatment can enhance OR and OP, but that effects are highly dependent on factors such as dose and timing of administration. In addition to providing more detail on these general conclusions, this review will discuss directions for future avenues of research into the hormonal regulation of object memory.

Keywords: Estradiol, progesterone, hippocampus, object location, rat, mouse

1. Introduction

The object recognition (OR) task was first introduced in 1988 to provide a method of testing episodic memory in rodents that was similar to methods used in clinical neuropsychology [1]. The task capitalizes on rodent’s inherent predilection for novelty. In the most common version of the task, rodents explore two identical objects during a single training session. During the single test session, subjects are then allowed to explore an object identical to the training objects and a novel object. More time spent exploring the novel object indicates memory for the familiar object. In the years since its introduction, OR has become prevalent in rodent learning and memory studies, where it is used alone or as part of a test battery to investigate the effects of lesion, genetic, or pharmacological manipulations. The task has also evolved to assess spatial memory in rodents via a modified version referred to as object placement or object location (referred to herein as object placement) [2]. As such, OR is used to assess memory for the identity of objects (i.e., “what”) and object placement (OP) is used to assess memory for the location of objects (i.e., “where”). To this end, OR in rodents is generally considered a non-spatial memory task involving the hippocampus, perirhinal, entorhinal, and parahippocampal cortices [35], whereas OP in rodents is considered a spatial memory task that relies primarily on the hippocampus [6]. However, the brain regions involved in OR in rodents and other species have been the subject of intense debate, particularly the role of the hippocampus in mediating OR. Although this issue is not the primary focus of this review, rodent data from our laboratory and others do support a role for the hippocampus in OR, as will be discussed below. Therefore, this review is written from the perspective that the hippocampus is essential for memory formation in both OR and OP. Because the amount of data collected on hormonal regulation of object memory in rodents far outnumbers the amount of data collected in other species, this review will limit discussion to studies employing rats or mice as subjects.

OR and OP are particularly well suited for investigating the molecular processes underlying the formation of hippocampal-dependent memories in rodents. First, they take advantage of a rodent’s natural tendency to explore novel stimuli, while avoiding other potentially confounding variables. For example, no rule learning is required, nor are any rewarding or punishing stimuli involved that may influence motivational, rather than mnemonic, aspects of task performance [1; 7]. Therefore, memory can be measured in the absence of confounds due to the stress of nutrient restriction (as commonly used in the radial arm maze and T-maze), shock (as used in fear conditioning), or submersion in water (as used in the Morris water maze). Second, OR and OP are true one-trial learning tasks. This quality makes them ideal for studying the effects of acute drug or hormone treatments, which may be given pre- or post-training to investigate effects on different phases of learning and memory such as encoding, consolidation, and retrieval.

This unique combination of one-trial learning in a relatively stress-free environment has appealed in recent years to behavioral neuroendocrinologists seeking to identify the molecular mechanisms through which sex steroid hormones, such as 17β-estradiol (E2) and progesterone (P4), influence memory across the rodent lifespan. The low stress associated with OR and OP testing is advantageous for behavioral endocrinologists because corticosteroids released in response to more stressful tasks may interact with ovarian hormones and could confound the interpretation of results [8; 9]. Tasks like OR and OP that do not provoke a strong stress response allow for effects of ovarian hormones on memory to be more clearly identified in rodents. Furthermore, behavioral endocrinologists have found that E2 and P4 can very rapidly impact hippocampal function [1013], and therefore, the one-trial nature of OR and OP makes these tasks particularly useful for identifying the molecular mechanisms underlying hormonal regulation of memory consolidation. As such, both OR and OP have been widely used in the past decade to study the effects of sex steroid hormones on hippocampal learning and memory in rats and mice.

However, as will be seen below, investigators have taken very different approaches to studying hormonal regulation of object memory in rodents. Both rats and mice of various strains have been used, with studies using mice generally outnumbering those using rats. Although species differences could influence the effects of hormones on OR, OR does not appear to differ between male rats and mice [14], and effects of E2 and P4 on OR and OP are remarkably consistent among rat and mouse studies. However, an interesting new report showing that the effects of estrogen receptor agonists on OR differ in two mouse strains suggests that strain or species may influence the receptor mechanisms through which hormones regulate object memory [15]. It is also important to note that although several studies have used gonadally intact females, the vast majority of studies to date administered exogenous hormones to ovariectomized females because removal of the ovaries eliminates the ovarian hormone fluctuations generated by the estrous cycle. In most studies, treatment was started at the time of ovariectomy or within a week afterwards, but recent data show that long periods of ovarian hormone deprivation after ovariectomy eliminate the memory-enhancing effects of E2 on OR [16]. As such, timing of treatment to ovariectomy is an important variable for investigators to consider. Among exogenous hormone studies, most administered only E2. Therefore, much less is known about the effects of P4, alone or in combination with E2. Finally, a key difference among many studies is the timing of treatment relative to testing, as an increasing number of studies have administered E2 and P4 immediately after training to examine rapid effects of these hormones on object memory consolidation. In general, E2 or P4 given prior to, or immediately after, training enhance OR and OP, so the neural mechanisms underlying these effects are likely to be similar. As with any drug treatment, however, the effects of these hormones on object memory depend on many factors such as dose, route of administration, and age of subjects. Nevertheless, E2 and P4 generally enhance memory in the OR and OP tasks, as will be illustrated in the sections below.

The primary goal of this review is to survey literature examining the regulation of OR and OP in rats and mice by ovarian sex steroid hormones. To set the stage for this discussion, we will first address the role of the hippocampus in OR, provide an overview of ovarian hormone regulation of hippocampal function, and detail the protocols most commonly used in hormone studies. We will then describe the effects of natural estrous cycling and exogenous hormone administration on OR and OP. Finally, we will end by reviewing the molecular mechanisms known to mediate the memory-enhancing effects of E2 and P4 on OR and OP, and discuss how the reduction of ovarian hormone levels during aging affects memory in these tasks. Our intent is to highlight the importance of ovarian sex steroid hormones in regulating object memories in rodents and encourage investigators to consider the role that these hormones may play in their own experimental designs.

2. Role of the hippocampus in object recognition

Since their introduction in the 1980s and 1990’s, one-trial OR and OP tasks have gained traction as standard tests for rodent memory in behavioral neuroscience and behavioral neuroendocrinology alike. Sensitive to hormones, aging, and drug treatments, OR and OP are most commonly associated with measures of episodic hippocampal memory. In addition to its role in other well known tasks such as the Morris water maze [17], contextual fear conditioning [18], and radial arm maze [18], the hippocampus also plays a pivotal role in OR and OP [12; 1921]. Several researchers have conducted systems-level investigations to determine which brain regions are involved in mediating the non-spatial and spatial memory components of these tasks. For example, the hippocampus, cingulate cortex, and fornix have been implicated in modulating OP memory [2]. Although there is little debate regarding the role of the hippocampus in mediating spatial memory [6; 22; 23], the role of the hippocampus in OR remains controversial [4; 2329] and may depend on the extent to which spatial information is available. Despite compelling evidence implicating the entorhinal and perirhinal cortices in mediating OR memory [5; 24; 30], considerable data also suggest an important role of the hippocampus in recognition memory in rodents, non-human primates, and humans [1921; 31; 32]. The bulk of this evidence comes from inactivation studies demonstrating OR memory impairment when the hippocampus is lesioned or inactivated [3; 6; 19; 21; 33]. Importantly, impairments are not observed when lesions are made to the fornix or cortical regions adjacent to the hippocampus, suggesting a specific role for the hippocampus in OR memory [19]. Although some studies suggest that a substantial lesion or pharmacological inactivation of greater than 50% of the total hippocampal volume is required [6], a recent study demonstrated significant OR impairment in mice when only about 1% of the dorsal hippocampus was inactivated [21]. This striking finding suggests that even the smallest disruption to cellular circuitry in the dorsal hippocampus can significantly disrupt OR memory.

Object recognition memory is also disrupted when specific receptors or cell-signaling pathways are inhibited in the hippocampus. For example, pharmacological inactivation of the dorsal hippocampus by GABAA agonists, NMDA antagonists, or inhibitors of ERK/MAPK cell signaling, histone acetylation, and protein synthesis significantly impair OR memory in rats and mice [10; 20; 21; 34; 35]. Other studies report molecular and physiological alterations in the hippocampus resulting from OR training, including changes in synaptic physiology, neurotransmitter release, and activation of immediate early genes [21; 36; 37]. For example, long-term potentiation (LTP)-like changes occur in hippocampal CA3-CA1 during the consolidation period following OR training [36]. Moreover, mice do not demonstrate intact memory for the familiar object if these LTP-like changes are disrupted prior to presentation of the objects [36], suggesting that the hippocampus plays an essential role in OR memory consolidation. The presentation of a novel object during the testing phase facilitates the firing of CA1 neurons in the dorsal hippocampus and increases excitatory glutamate release [21]. Because these events occur only in the presence of a novel object, and not upon reexposure to two familiar objects [21], this finding suggests that the dorsal hippocampus is activated by novelty and plays a role in novelty detection in the OR task. Further support for a role of the hippocampus in OR is provided by a study demonstrating significantly more c-fos expression in the dentate gyrus of rats tested in OR than in the dentate of home cage control rats [37]. This immediate early gene expression suggests engagement of the hippocampus during OR training and/or testing. Collectively, these data support the involvement of the hippocampus in OR, and highlight the carefully orchestrated molecular processes required in this region for the successful formation and storage of recognition memories in the rodent brain.

3. Effects of ovarian hormones in the hippocampus: A general overview

Although the contributions of the hippocampus to OR may be the subject of continued debate, an increasing body of research supports a role for the hippocampus in mediating OR memory consolidation. As a result, OR has become a useful tool for studying the effects of various neuromodulators, including sex steroid hormones, on hippocampal memory. To provide context for a discussion of this work, this section will describe the ways in which ovarian sex steroid hormones regulate hippocampal function. For more detail on this subject than can be provided here, we recommend several recent reviews [3842].

3.1. Early development

Sex steroid hormones regulate brain function throughout the lifespan. These hormones play an integral organizational role in the sexual differentiation of the brain during the pre-natal and early post-natal periods in both males and females [4350]. Although early studies focused largely on brain regions related to sexual behavior, such as the hypothalamus and preoptic area, more recent findings demonstrate that sex steroid hormones also promote sexual differentiation of the hippocampus, basal forebrain, and cerebral cortex [51; 52]. Such differentiation can have long-term consequences for memory function later in life, as suggested by findings in rats indicating that neonatal hormone exposure contributes to a male advantage in hippocampal-dependent spatial memory in adulthood [53]. Indeed, this sex difference in adulthood can be reversed by gonadectomy in males and E2 treatment in females before postnatal day 10 [53], suggesting that early hormonal exposure shapes the hippocampus into a “male” or “female” pattern. However, it should be noted that the magnitude of sex differences in the hippocampus is considerably smaller than those associated with reproductive brain regions and behaviors [53; 54].

3.2. Hippocampal morphology

In addition to their effects on brain organization, sex steroid hormones exert activational effects on the brain throughout adulthood. In female mammals, estrogens and progestins are made primarily in the ovaries, although these hormones are also synthesized in other organs including the brain. In fact, levels of E2 are higher in the hippocampus than in serum in both male and female rats [55; 56]. Serum levels of estrogens and progesterone fluctuate substantially in response to the release of hormones from the hypothalamus and anterior pituitary. The rodent hormone cycle, called the estrous cycle, lasts 4–5 days and is divided into four phases (proestrus, estrus, metestrus, and diestrus) that each last about 24 hours [57]. The cycle is characterized by peaks of E2 and P4 just prior to ovulation during the proestrus phase. Levels of both hormones drop steeply after ovulation and are, therefore, quite low during the subsequent estrus phase of the cycle. The earliest work to demonstrate that the adult hippocampus is sensitive to ovarian hormones examined how dendritic spine density in the CA1 region of the hippocampus was affected by hormonal fluctuations during the estrous cycle or exogenous administration of E2 and P4. This work, published in the early 1990s by Bruce McEwen and colleagues, showed that dendritic spine density was 30% higher during proestrus than estrus [58]. Furthermore, bilateral removal of the ovaries significantly decreased spine synapse density relative to intact females, an effect that could be reversed within hours by acute E2 treatment alone or E2 plus P4 [59]. In addition to dramatic changes in spine number, spine synapses were also increased during proestrus [60] and by exogenous E2 treatment [61]. Interestingly, P4 had a biphasic effect on spine synapses, initially increasing CA1 spine density during the first 2–6 hours after injection, but then sharply decreasing spine density afterwards [61]. In the years since, these findings have been replicated and expanded upon by numerous labs [6269]. Collectively, these landmark findings provided the first evidence demonstrating that ovarian hormones could modify the CA1 synaptic morphology thought to support the formation of lasting memories.

3.3. Neural mechanisms through which estradiol affects hippocampal function

Numerous subsequent studies have expanded upon these seminal discoveries, demonstrating E2’s role as a potent regulator of cellular events in the hippocampus critical for synaptic plasticity and memory. Systemic, intracranial, or in vitro applications of E2 increase hippocampal expression of synaptic proteins such as synaptophysin, spinophilin, syntaxin, and postsynaptic density-95 [7072], increase intrinsic excitability [42], facilitate hippocampal LTP [73], and promote neurogenesis in the dentate gyrus [74]. Some of these effects may be due to the binding of E2 to its canonical intracellular receptors, ERα and ERβ. ERα and ERβ are found in dendritic spines, dendrites, axon terminals, and the nuclei of hippocampal pyramidal neurons [33; 75; 76]. Hippocampal ERα is also present within cholinergic axons and terminals [77], and in the cytoplasm and nucleus of GABAergic interneurons, where it facilitates an E2-induced decrease in GABAergic tone that promotes pyramidal neuron spinogenesis [78]. The classical mechanism of estrogen action involves the formation of an E2-ER complex in the cytoplasm, the translocation of the complex into the nucleus, and the binding of the complex to an estrogen response element (ERE) on the DNA to initiate gene transcription. However, E2 can also influence neuronal function by rapidly activating cell signaling cascades like extracellular signal-regulated kinase (ERK) and phosphatidylinositol 3-kinase (PI3K) [10; 12; 7981], induce post-translational epigenetic modifications such as histone acetylation and DNA methylation [35; 79], and initiate mammalian target of rapamycin (mTOR)-mediated protein synthesis [34]. The classical ERE-mediated mechanism is too slow to account for these effects, which can occur within five minutes of dorsal hippocampal E2 infusion [10; 34]. It is now well accepted that E2 can rapidly influence hippocampal function via interactions between ERs and neurotransmitter receptors (e.g., mGluRs, NMDA receptors) and/or by binding to novel ERs in the plasma membrane (e.g., GPER/GPR30, Gq-mER) [12; 80; 82]. Studies of ER localization in the hippocampus support the notion that canonical ERs are positioned within spines and axon terminals for rapid local modulation of hippocampal function [83]. For example, our own laboratory has demonstrated that rapid activation of hippocampal cell signaling, epigenetic processes, and mTOR-mediated protein synthesis is necessary for OR memory consolidation [10; 12; 34; 35] (see Section 7 below). Collectively, these studies demonstrate that E2 can influence hippocampal function in numerous ways to regulate hippocampal memory formation.

3.4. Neural mechanisms through which progesterone affects hippocampal function

Much less is known about the neural mechanisms through which P4 regulates hippocampal function and memory formation. However, like E2, P4 can also enhance hippocampal LTP [84], neurogenesis [85; 86], ERK signaling, and mTOR-dependent local protein synthesis [87]. Nevertheless, understanding how P4 influences memory is considerably more challenging than E2 because P4 serves as an obligatory precursor for the synthesis of other steroids including estrogens, androgens, and glucocorticoids. Thus, P4 may influence hippocampal function via conversion to other steroid hormones that subsequently bind to their cognate receptors. Furthermore, P4 is metabolized into neuroactive steroids like 3α,5α-tetrahydroprogesterone (3α,5α-THP or allopregnanolone) that can bind to the steroid binding site on GABAA receptors and regulate synaptic excitability [88]. In addition, P4 shares with E2 the complexity that it can bind to two types of receptors: canonical intracellular progesterone receptors (PRs), thought to regulate slow transcription-mediated (i.e., classical) events, and plasma membrane-bound receptors (mPRs), thought to mediate rapid cell signaling-initiated (i.e., non-classical) effects. As such, there are at least three ways in which P4 may affect hippocampal memory: 1) binding to intracellular PRs, which then translocate to the nucleus and initiate gene transcription at a P4 response element (PRE), 2) binding to mPRs and rapidly activating cell signaling cascades like ERK and PI3K, and 3) metabolism to E2, glucocorticoids, or 3α,5α-THP, which can then bind to estrogen receptors (ERs), GABAA receptors, and glucocorticoid receptors (GRs). Within the hippocampus, intracellular PRs are located within dendritic spines, dendrites, cell bodies, and axons of principal neurons in CA1, CA3, and the dentate gyrus, as well as in GABAergic interneurons and glia [83; 89]. Therefore, like ERs, PRs are positioned to act locally to regulate hippocampal function. Indeed, our laboratory has found that dorsal hippocampal infusion of P in ovariectomized mice activates ERK and mTOR cell signaling in the dorsal hippocampus within five minutes [87]. Interestingly, P4 has a biphasic effect on p42 ERK activation, at first increasing phospho-p42 ERK levels five minutes after infusion and then decreasing levels 15 minutes after infusion, before returning to baseline 30 minutes later [87]. Similarly, systemic P4 in ovariectomized rats at first increases, and then decreases, CA1 dendritic spine density before levels return to baseline [61]. Such biphasic effects of P4 on hippocampal function further complicate attempts to understand how P4 regulates hippocampal memory.

3.5. General effects of estradiol and progesterone on hippocampal memory

In the two decades since the initial demonstration that ovarian hormones regulate CA1 dendritic spine density, hundreds of studies have examined the effects of E2 and/or P4 on memory in rodents, non-human primates, and humans. As such, a thorough evaluation of this literature is beyond the scope of this article, and the reader is encouraged to consult any of the numerous reviews on this subject (e.g., [39; 9098]). However, some broad generalizations may be drawn, particularly among rodent studies. In rodents, E2 is generally thought to facilitate hippocampal memory, although its effects depend on many factors, including age, dose and duration of treatment, duration of hormone loss prior to treatment, method of inducing ovarian dysfunction, timing of treatment relative to testing, type of memory tested, and task difficulty [95; 99; 100]. The vast majority of studies have examined the effects of exogenously administered E2 on spatial memory in ovariectomized female rodents. Consistent with the beneficial effects of exogenous E2 on hippocampal plasticity, spinogenesis, and neurogenesis, exogenous E2 administered to young ovariectomized rats and mice generally enhances spatial memory in the Morris water maze, radial arm maze, and T-maze [9; 99116]. E2 also facilitates memory formation in non-spatial tasks, including social recognition [62], inhibitory avoidance [117119], and trace eyeblink conditioning [120]. In accord with these other tasks that assess hippocampal function, E2 generally enhances memory consolidation in the OR and OP tasks; these data will be detailed in the sections below.

The effects of P4 on hippocampal memory differ depending on its administration relative to training and testing. Chronic systemic administration of P4 prior to training impairs footshock avoidance learning and spatial working memory in young ovariectomized mice and rats [121; 122], perhaps due to the anxiolytic and analgesic effects of progesterone metabolites at GABAA receptors [123125]. On the other hand, acute administration of P4 prior to training (systemic) or immediately after training (intrahippocampal) has no effect on spatial memory in the Morris water maze [126; 127] or radial arm maze [128] in young ovariectomized rats. These data suggest that P4 treatment prior to training impairs or has no effect on memory in these behavioral tasks. In contrast, acute systemic or intrahippocampal administration of P4 immediately after training enhances Y-maze inhibitory avoidance and OR in young ovariectomized female rodents [127; 129; 130], and OR in ovariectomized middle-aged and aged mice [131], suggesting that P can facilitate memory consolidation if present only during the memory consolidation period immediately following learning. It should also be noted that P4 is often administered in conjunction with E2 because E2 levels surge prior to P4 levels during the natural estrous cycle. In some cases, systemic P4 administered immediately after training blocks the memory-enhancing effects of E2 (e.g., on spatial memory in the Morris water maze in aged female mice; [132]) and in other studies, the combination of E2 and P4 enhances memory (e.g., on OR in young ovariectomized mice; [133]). In general, combined E2 and P4 treatment enhances object memory, but effects are dose dependent. Data on the effects of P4, alone or in combination with E2, on OR and OP will be discussed more comprehensively in the sections below.

4. OR and OP protocols typically used in behavioral endocrinology

Since their initial introduction for use in rats, OR and OP have been extended for use in mice and modified in variety of ways to suit the needs of different investigators and their experimental questions [1; 134136]. Modifications include varying the number and size of the objects, size and shape of the testing arena, light levels in the testing arena, duration of habituation, and delay between training and testing. Although the experimental protocols for OR and OP differ somewhat among laboratories, most protocols include three stages: habituation, training (also referred to as a sample phase), and testing (also referred to as a discrimination phase) (Fig. 1). Habituation serves to familiarize the subject to the testing arena and environment. On average, habituation ranges from 5–10 min per day for 1–4 days prior to training. During the training or sample phase, two identical objects are presented for exploration, and the time spent exploring each object is recorded. The amount of time allowed for subjects to investigate the objects during training varies by protocol, with most investigators typically using a predetermined limit of 3–20 minutes. The interval between training and testing can be manipulated to assess the memory-impairing or memory-enhancing effects of experimental manipulations. For example, some experimenters use as little as five minutes, whereas others use up to seven days [63; 137139]. During testing, one of the familiar objects from training is replaced by a novel object (Fig. 1A), and time spent investigating each object is again recorded (other dependent variables include number of visits to the objects and total exploration time). Because rodents tend to explore novelty, they should spend more time exploring the new object if they remember the familiar object. Therefore, recognition memory is considered intact when a subject spends significantly more time exploring the new object than the familiar object or than chance. Training and testing for OP is identical to OR, except that one of the identical objects from training is moved to a new location in the arena during testing (Fig. 1B). Intact spatial memory is indicated if a subject spends significantly more time with the moved object during the testing phase than either the unmoved object or than chance. When used together, these one-trial tasks provide low stress methods to assess both spatial and non-spatial memory in rodents.

Figure 1.

Figure 1

Overview of the object memory testing protocols discussed in this review. Mice are first habituated to an empty arena prior to beginning behavioral training (Habituation). (A) In novel object recognition (OR), mice are then allowed to explore two identical novel objects placed in the arena (Training). Finally, retention is tested by presenting mice with one novel and one familiar object (Testing). Mice who remember the familiar object spend more time exploring the novel object relative to the familiar object or to chance. (B) Object placement (OP) uses the same apparatus and general procedure, but during testing, one training object moves to a new location in the arena, rather than being replaced with a new object. (C) Object pairs used in our laboratory’s OR and OP protocols.

OR and OP data can be analyzed using several different dependent measures. Discrimination ratios are often calculated when investigators do not set a minimum criterion for active exploration time, and instead set a maximum trial time regardless of the amount of active object exploration. Discrimination ratios can be calculated by dividing the time spent exploring the novel or moved object minus the time spent exploring the familiar or unmoved object, by the sum of the total exploration time for both objects (Tnovel-Tfamiliar/Tnovel+Tfamiliar). This ratio is sometimes multiplied by 100 to obtain a percentage. Regardless, it is essential to divide the difference in exploration time between the two objects by the total exploration time because exploration time will vary among subjects. Chance performance is indicated by ratios near 0, whereas ratios of approximately 0.5 or above indicate a preference for the novel object. Another commonly used discrimination ratio is calculated using the equation (Tnovel/(Tnovel+Tfamiliar)), which can also be multiplied by 100 to obtain a percentage. Values of greater than 0.5 indicate that the subject can successfully discriminate between the two objects. For both discrimination ratio equations, the resulting data are limited in range (e.g., between 0 and 100) and so cannot be truly normally distributed. Therefore, ratio data should be transformed (i.e., using arcsin or ln transformation) for use with parametric statistics [64].

Although the protocols that provide data suitable for discrimination ratio calculations do provide an indication of preference, they do not account for individual differences in the total time that subjects spend investigating the objects. For example, one mouse may spend only 20 sec investigating objects during a five-minute training period, whereas another may spend 120 sec. A subject that spends 120 sec exploring the objects is likely to form a more lasting memory of the objects than a subject that spends only 20 sec exploring the objects. Therefore, it can be difficult to estimate the extent to which exploration time during training affects exploration time during testing. To eliminate this confound, some investigators require a set amount of time spent investigating the objects during training only [21] or during both training and testing [140]. Using this method, time spent with the objects is compared to chance performance [10; 12; 34; 79]. For example, our laboratory requires that subjects explore the two objects for a combination of 30 seconds during training and testing, and gives subjects up to 20 minutes to reach this criterion. In this protocol, exploration time is compared using a one-sample t-test to the chance value of 15 seconds, which reflects an identical amount of time spent with both objects. We use this protocol because controlling for exploration time during training is important in our experimental designs. However, trial time is often fixed in studies using pre-training treatments to ensure that the time from treatment to testing and amount of drug in circulation during training and testing is the same for all subjects. Ultimately, whether interpreting the results of an experiment or designing an experiment of one’s own, it is important to consider the way in which the experiment was designed in order to understand how hormones, or any other experimental manipulation, affect memory measured by OR and OP.

5. Performance of gonadally intact female rodents in object memory tasks

5.1. The estrous cycle

Studying the influence of endogenous E2 and P4 levels on hippocampal memory in gonadally intact females is inherently challenging in light of the naturally fluctuating hormones levels across the 4–5 day estrous cycle and the multi-day protocols typically used to test learning and memory. As such, very few studies have examined the effects of E2 and P4 on hippocampal memory in gonadally intact females. However, some investigators have capitalized on the one-trial nature of OR and OP to test the effects of cyclic hormone fluctuations on hippocampal memory (Table 1). In these protocols, training and testing occurred on the same day to ensure that both phases were conducted within the same estrous phase. When tested one hour after training, female rats in any phase of the cycle displayed intact OR memory, whereas only those in estrus (low E2 and P4 levels) exhibited intact OP memory [141]. Other investigators report greater differences among cycle phases when OR and OP testing were conducted four hours after training. In these studies, rats or mice in proestrus (elevated E2 and P4 levels) displayed enhanced OR memory relative to their counterparts in estrus or diestrus (reduced E2 and P4 levels) [129; 142; 143]. Similarly, rats in proestrus exhibited better spatial memory in OP than rats in diestrus [143; 144]; however, rats in estrus did as well [144]. This curious finding, combined with the OP data from Sutcliffe et al., 2007, suggests that perhaps the P4 generated by the corpus luteum during estrus is more beneficial for spatial memory than for object recognition. Based on the data from these few studies, it is tempting to speculate that memory in OR and OP is facilitated during the estrous cycle when E2 and P4 are elevated, but definitive conclusions about cyclic fluctuations in object memory will require further study.

Table 1.

Effects of endogenous ovarian sex steroid hormones on novel object recognition and object placement

Species Age Novel object recognition Object placement Ref # Reference
Estrous cycling
Rat Young Proestrus outperformed diestrus and estrus; estrus outperformed diestrus --- [129] Walf et al., 2006
Rat Young Memory intact in all phases of the estrous cycle Memory intact during estrus, but not other stages [141] Sutcliffe et al., 2007
Rat Young --- Proestrus and estrus outperformed Diestrus [144] Frye et al., 2007
Rat Young Proestrus outperformed diestrus Proestrus outperformed diestrus [143] Paris & Frye, 2008
Mouse Young Memory intact in wild-type mice in proestrus, but not wild-type or ERβ knockout mice in diestrus --- [142] Walf et al., 2009
Pregnancy
Rat Young --- Rats in 3rd trimester, post-partum, or lactating outperformed rats in 1st trimester [144] Frye et al., 2007
Rat Young Pregnant outperformed non-pregnant Pregnant outperformed nonpregnant [143] Paris & Frye, 2008
Estropause
Rat Middle-aged Rats with intact reproductive function outperformed rats with declining reproductive function --- [210] Paris et al., 2011

5.2. Pregnancy

E2 and P4 are also naturally elevated during pregnancy, which influences hippocampal morphology and memory [93; 145]. For example, CA1 dendritic spine density is significantly higher in pregnant and lactating female rats than in virgin females at any stage of the estrous cycle [146]. Furthermore, reproductively experienced middle-aged female rats exhibit more cell proliferation in the dentate gyrus in response to exogenous E2 than virgin females of the same age, suggesting that reproductive experience may potentiate the hippocampal response to E2 later in life [147]. Behaviorally, pregnant females with high endogenous E2 and P4 levels exhibited enhanced memory in the OR and OP tasks relative to non-pregnant females [143], although other studies find that memory in different hippocampal-dependent tasks is impaired during pregnancy [93] (Table 1). Interestingly, number of pregnancies also plays a role in object memory, as multiparous rats (females with >2 pregnancies) demonstrate enhanced OP memory relative to primiparous (first-time pregnancy) rats [144]. Moreover, OP was enhanced in post-partum and lactating rats relative to rats in their first trimester [144], suggesting that the hormonal milieu in late pregnancy and the early post-partum period may facilitate spatial memory. Although these data are consistent with findings for other hippocampal-dependent spatial tasks such as the radial arm maze [148], other studies contradict these findings [93; 145].

5.3. Caveats

Inconsistencies among studies examining gonadally intact female rodents may be due to numerous factors, including the fact that so few labs are engaged in this research. With such a small literature, differences in methods, species, and strains are likely to be magnified in importance. However, these discrepancies also likely reflect the inherent difficulty of trying to examine the behavioral effects of hormones in subjects whose hormone state is in nearly constant flux across a pregnancy or a natural estrous cycle. For this reason, most investigators opt to eliminate natural hormonal variability by ovariectomizing their female subjects, thereby allowing investigators to focus their attention on the effects of exogenous hormone treatment in the absence of hormone fluctuations. As such, the literature on the effects of E2 and P4 on hippocampal memory in ovariectomized females is considerably more extensive than that on gonadally intact females. Although ovariectomy is easier than managing the natural cycle and allows for more control of hormone levels, it is unclear if these two models are truly comparable. For example, it is unknown if an exogenous E2 injection would have a similar effect on object memory in an ovariectomized female and a gonadally intact female in estrus. Serum E2 levels would be low in both females prior to injection, but one has ovarian tissue that can be stimulated by the exogenous injection and the other does not. It is unknown whether the presence of functioning ovaries makes a difference in the mnemonic responsiveness to exogenous hormones, but this is a key question because most women who take hormones possess an intact reproductive system. Unfortunately, this issue has not been addressed for object memory or any other type memory. Pharmacological depletion of ovarian follicles by 4-Vinylcyclohexene-diepoxide (VCD) results in a gradual loss of ovarian function, so this treatment could be a useful alternative to traditional ovariectomy surgery. In middle-aged rats, VCD-induced follicular depletion was less detrimental for spatial working memory in a water-based radial arm maze task than ovariectomy surgery [99]. However, middle-aged VCD-treated rats given conjugated equine estrogens had significantly worse spatial working memory in the water radial arm maze than middle-aged ovariectomized rats [100], suggesting that VCD-treatment and ovariectomy have fundamentally different effects on the response to ovariectomy have fundamentally different effects on the response to exogenous E2 in aging females. Yet very few studies have used VCD, so it is premature to make any conclusions about the effects of this treatment on memory function. Nevertheless, it is important to note that this potential alternative to ovariectomy could help in understanding how ovarian function and ovarian loss may influence the mnemonic response to exogenous hormone treatment.

The majority of ovariectomy studies have examined effects of ovarian hormones on spatial tasks, such as the Morris water maze, radial arm maze, and T-maze. However, a substantial number of studies from the past decade have examined effects of E2 and P4 on OR and OP memory in adult female rats and mice of various ages. The remaining sections of this review will discuss these findings in detail.

6. Hormone replacement in young ovariectomized females

6.1. Effects of ovariectomy on OR and OP

Given the effects of circulating ovarian hormones on OR and OP in gonadally intact rodents, one might ask whether ovariectomy itself has detrimental effects on memory in these tasks. Few studies have addressed this issue directly, but the data indicate that ovariectomy impairs memory in both tasks. One study of ovariectomized rats and gonadally intact sham-operated rats tested memory in the OR and OP tasks weekly for seven weeks after surgery, which was conducted at approximately 2.5 months of age [149]. For both tasks, ovariectomy produced a significant and persistent deficit; for OR, the deficit became evident two weeks after surgery, and for OP, the deficit became evident four weeks after surgery [149]. For both tasks, the deficit persisted throughout the seven weeks of testing [149]. These deficits were associated with a reduction in pyramidal neuron dendritic spine density in hippocampal CA1 and the medial prefrontal cortex [149], suggesting a role for morphological alterations in the observed memory deficits. These findings are supported by other recent studies demonstrating that ovariectomy for extended periods impairs memory and reduces the response to exogenous E2. In one study, two month-old female mice ovariectomized one week prior to OR training outperformed mice that had been ovariectomized without E2 or P4 replacement for six or 12 weeks [150]. This impairment was reversed by five weeks of chronic systemic E2 treatment, suggesting that the young female mouse brain remains responsive to E2 for at least three months after ovariectomy. However, longer periods of ovarian hormone deprivation diminish responsiveness to E2, as illustrated by data from rats ovariectomized at two months of age for a duration of 9, 15, or 19 months prior to E2 treatment. Acute pre-training injections of E2 enhanced OR memory in rats ovariectomized for 9 or 15 months [16], which is consistent with other studies by these investigators demonstrating that E2 treatment 9 or 15 months after ovariectomy also increased LTP, CA1 dendritic spine density, GluN2B-mediated neurotransmission, and NMDA/AMPA ratio [16; 151153]. In contrast, E2 did not enhance memory or increase hippocampal function in rats ovariectomized for 19 months prior to E2 treatment [151153]. One potential explanation of this finding is that the rats were too old to respond to E2, as the rats were about 21 months old at the time of testing. However, E2 could enhance memory in 21 month-old rats ovariectomized only one month prior to treatment [16], suggesting a greater role of length of hormone deprivation than age on the response to E2 delivered prior to training. Overall, these studies demonstrate a detrimental effect of ovariectomy on memory in the OR and OP tasks, with greater impairments observed after longer lengths of ovarian hormone deprivation.

6.2. Effects of E2 on OR and OP in young ovariectomized females

The aforementioned detrimental effects of ovarian hormone deprivation are reversible with systemic or intracranial administration of exogenous E2 (Table 2). Systemic injections of E2 in young ovariectomized rats and mice in the range of 15 μg/kg to 0.2 mg/kg dose-dependently enhance OR [62; 64; 150; 154157], and OP [62; 64; 155] when given prior to training. These enhancements were observed after chronic or acute E2 treatments. However, such pre-training treatments can affect non-mnemonic aspects of task performance (e.g., motivation, anxiety, motor behavior) that can confound interpretation of behavioral outcomes and should be accounted for when interpreting results. Further, pre-training treatments do not permit a distinction between memory acquisition and consolidation [158]. Therefore, several laboratories, including our own, have administered E2 immediately post-training, which allows effects on memory consolidation to be isolated in the absence of non-mnemonic performance confounds. In these studies, systemic E2 (15 μg/kg to 0.2 mg/kg) administered immediately after training enhanced both OR [15; 80; 133; 155; 159164] and OP [155; 163; 164], indicating that E2 specifically enhances novel object recognition and spatial memory consolidation. Interestingly, immediate post-training systemic injections of other estrogens, including 17α-estradiol and the synthetic diethylstilbestrol, also enhanced OR and OP memory [155], suggesting that multiple forms of estrogens can facilitate object recognition and spatial memory. Moreover, E2’s ability to facilitate memory consolidation is restricted to an approximate one hour window after training, as rats and mice treated with E2 1–3 hours post-training do not remember the familiar or unmoved objects during OR or OP testing [10; 129; 144]. These data are significant, as they demonstrate that E2 can enhance OR and OP within the brief time window during which memory consolidation occurs in these tasks.

Table 2.

Effects of exogenous estradiol on object memory in young adult female rodents

Species Type of
hormone
ROA Timing of
administrationa
Task(s) Effect on NOR and/or OP Ref # Reference
Estradiol
Rat 17α-E2, E2, DES Systemic Pre-training NOR, OP Injection of 17α E2, E2, or DES (15 μg/kg) 30 min before training enhanced NOR and OP [155] Luine et al., 2003
Rat Estradiol Benzoate Systemic Pre-training NOR, OP Two injections of EB (50 μg/kg) two days before training enhanced NOR and OP [154] Jacome et al., 2010
Rat E2 Systemic Pre-training NOR Two injections of 10 μg E2 24 or 48 h before training enhanced NOR [156] Vedder et al., 2013
Mouse E2 Systemic Pre-training NOR, OP Injection of E2 (1.5–3 μg/kg) 15 min before training enhanced NOR and OP [62] Phan et al. 2012
Mouse E2 Systemic Pre-training NOR, OP Six or 10 weeks of E2 silastic (50 μg/25 μl) enhanced NOR and OP [157] Ismail & Blaustein, 2013
Mouse E2 Systemic Pre-training NOR Five weeks of E2 silastic (0.18 mg/4 µl) enhanced NOR [150] Fonseca et al., 2013
Mouse Estradiol Benzoate Systemic Pre-training OP EB (1 μg/day) for 5 days before training enhanced OP [69] Li et al., 2004
Rat E2, DES Systemic Post-training NOR, OP E2 (30 μg/kg) enhanced OP, DES (15 μg/kg) enhanced NOR, but not OP [155] Luine et al., 2003
Rat 17α-E2, E2 Systemic Post-training NOR, OP Doses of 5–20 μg/kg E2 enhanced NOR or OP; doses of 1–5 μg/kg 17α-E2 enhanced NOR or OP [164] Inagaki et al., 2010
Rat E2 Systemic Post-training NOR E2 (0.9 mg/kg) enhanced NOR, but not if delayed 1 hr after training [129] Walf et al., 2006
Rat E2 Systemic Post-training OP E2 (0.9 mg/kg) enhanced OP, but not if delayed 1.5 hr after training [144] Frye et al., 2007
Mouse Water-soluble E2 Systemic Post-training NOR E2 (0.2 mg/kg) enhanced NOR [159] Gresack & Frick, 2004
Mouse Water-soluble E2 Systemic Post-training NOR E2 (0.2 and 0.4 mg/kg) enhanced NOR [160] Gresack & Frick, 2006
Mouse Water-soluble E2 Systemic Post-training NOR E2 (0.2 mg/kg) enhanced NOR [161] Gresack et al., 2007
Mouse Water-soluble E2 Systemic Post-training NOR E2 (0.2 mg/kg) enhanced NOR [162] Gresack et al., 2007
Mouse Water-soluble E2 Systemic Post-training NOR E2 (0.2 mg/kg) enhanced NOR [80] Lewis et al., 2008
Mouse E2 Systemic Post-training NOR, OP E2 (0.1 mg/kg) enhanced NOR, OP in wild type mice, but not ERβ knockouts [163] Walf et al., 2008
Mouse Water-soluble E2 Systemic Post-training NOR E2 (0.2 mg/kg) enhanced NOR [133] Harburger et al, 2009
Mouse Water-soluble E2 Systemic Post-training NOR E2 (0.2 mg/kg) enhanced NOR in wild type and ERα knockout mice, but not in ERβ knockouts [178] Frick et al., 2010
Mouse Water-soluble E2 Systemic Post-training NOR E2 (0.2 mg/kg) enhanced NOR [15] Pereira et al., 2014
Mouse Water-soluble E2 Intrahippocampal Post-training NOR 5 μg E2 infused bilaterally into the dorsal hippocampus enhanced NOR, but not if delayed 3 hr after training [10] Fernandez et al, 2008
Mouse Water-soluble E2 Intrahippocampal Post-training NOR 5 μg E2 infused bilaterally into the dorsal hippocampus enhanced NOR [79] Zhao et al., 2010
Mouse Water-soluble E2 Intrahippocampal Post-training NOR 5 μg E2 infused bilaterally into the dorsal hippocampus enhanced NOR [35] Zhao et al., 2012
Mouse Water-soluble E2 Intrahippocampal Post-training NOR 5 μg E2 infused bilaterally into the dorsal hippocampus enhanced NOR [34] Fortress et al., 2013
Mouse Water-soluble E2 Intrahippocampal Post-training NOR, OP 5 μg E2 infused bilaterally into the dorsal hippocampus enhanced NOR and OP [12] Boulware et al., 2013
a

Post-training treatments were given immediately after training unless indicated otherwise in the "Effect on NOR and/or OP" column

Note: ROA=route of administration; NOR=novel object recognition; OP=object placement; EB=estradiol benzoate

It is important to note that systemic treatments have the primary disadvantage of affecting tissues throughout the body, so the role of the hippocampus in these memory enhancements is unclear. Therefore, our laboratory has conducted a series of studies in which we bilaterally infused 5 μg E2 directly into the dorsal hippocampus to pinpoint the role of this structure in hormonal regulation of OR and OP memory consolidation. We have found that bilateral dorsal hippocampal infusion of E2 immediately post-training enhances both OR and OP memory consolidation in young ovariectomized mice [10; 12; 34; 35; 79], thereby demonstrating that the dorsal hippocampus plays a major role in mediating E2’s effects on object recognition and spatial memory consolidation. A study from another laboratory recently confirmed this finding in two different strains of mice [15], including the C57BL/6 strain used for our studies.

Collectively, the literature on E2 and object memory in rodents shows that systemic or intrahippocampal administration of E2 consistently enhances memory in the OR and OP tasks when given prior to or after training. These effects generalize across various OR and OP protocols conducted in different labs in different rodent species (rats and mice). Importantly, E2 must be administered during acquisition or within a one-hour window immediately after training to enhance memory consolidation. Finally, E2 infusion restricted to the dorsal hippocampus enhances OR and OP memory consolidation, indicating a crucial role of this structure in the hormonal modulation of object memory in females.

6.3 Effects of P4 on OR and OP in young ovariectomized females

Although the majority of studies have focused on E2 replacement, some investigators have also examined the effects of P4, alone or in combination with E2, on OR and OP (Table 3). As mentioned above, P4 administered systemically prior to training impairs or has no effect on various forms of hippocampal memory in tasks such as the radial arm maze, footshock avoidance, and Morris water maze [121; 122; 126; 128]. These effects typically depend on dose and method of administration (e.g., cyclic or continuous). However, P4 administered systemically (4–20 mg/kg) immediately after training dose-dependently enhances OR in young ovariectomized female rodents [127; 129; 144; 165167]. These effects are supported by other work from our laboratory in which 0.01, 0.1, or 1 μg P4 infused directly into the dorsal hippocampus enhanced OR in a manner dependent on rapid hippocampal cell signaling [87; 168].

Table 3.

Effects of exogenous progestins on object memory in young adult female rodents

Species Type of
hormone
ROA Timing of
administrationa
Task(s) Effect on NOR and/or OP Ref #
Progesterone
Rat P4, 3α,5α-THP Systemic Post-training NOR P4 (4 mg/kg) enhanced NOR, but not if delayed 1 hr after training; 3α,5α-DHT (4 mg/kg) enhanced NOR [129]
Rat P4, 3α,5α-THP Systemic Post-training OP P4 (4 mg/kg) enhanced NOR, but not if delayed 1.5 hr after training; 3α,5α-DHT (4 mg/kg) enhanced OP [144]
Rat P4 Systemic Post-training NOR P4 (4 mg/kg) enhanced NOR [166]
Mouse P4 Systemic Post-training NOR P4 (10 mg/kg) enhanced NOR, but not if delayed 1.5 hr after training [167]
Mouse P4, 3α,5α-THP, MPA Systemic Post-training NOR, OP P4 and 3α,5α-DHT (4 mg/kg) post-training enhanced NOR and OP, but MPA (10 mg/kg) did not [165]
Mouse Water-soluble P4 Systemic Post-training NOR P4 (10 or 20 mg/kg) enhanced NOR [127]
Mouse Water-soluble P4 Intrahippocampal Post-training NOR P4 (0.01–1 μg) enhanced NOR, but not if delayed 2 hr after training [168]
Mouse Water-soluble P4 Intrahippocampal Post-training NOR P4 (0.1 μg) enhanced NOR [87]
Estradiol plus Progesterone
Rat E2, P4 Systemic Post-training NOR E2 (0.9 mg/kg) + P4 (4 mg/kg) enhanced NOR, but not if delayed 1 hr after training [129]
Rat E2, P4 Systemic Post-training OP E2 (0.9 mg/kg) + P4 (4 mg/kg) enhanced OP, but not if delayed 1.5 hr after training [144]
Mouse Water-soluble E2, P4 Systemic Post-training NOR E2 (0.2 mg/kg) + P4 (10 or 20 mg/kg) enhanced NOR; E2 (0.2 mg/kg) + P4 (5 mg/kg) did not enhance NOR [133]
a

Post-training treatments were given immediately after training unless indicated otherwise in the "Effect on NOR and/or OP" column

Note: ROA=route of administration; NOR=novel object recognition; OP=object placement; 3α,5α-THP=3α,5α-tetrahydroprogesterone; MPA=medroxyprogesterone acetate

In contrast, fewer studies have examined the effects of P4 on spatial memory tested in OP, and these data are conflicting. For example, in one study, 4 mg/kg P4 given immediately post-training enhanced OP in young ovariectomized mice [165], however, another post-training study found that 10 mg/kg did not enhance OP in young ovariectomized rats [167]. These differences could be due to numerous factors, including dose of P4 and/or species. However, the fact that ovariectomized female rats treated with the P4 metabolite 3α,5α-THP immediately after training also exhibited enhanced OR and OP memory relative to vehicle-treated rats [129; 144], may suggest a dose-dependent effect of P4 on OP. Clearly, more research is needed on this subject. Finally, P4 must be within two hours after object training in order to facilitate object memory consolidation [129; 144; 168], supporting the role of P4 in mediating memory formation during the memory consolidation window.

Several studies have co-administered E2 and P4 to mimic peak hormone levels observed during the proestrus phase of the estrous cycle. Ovariectomized rats treated with systemic E2 (0.9 mg/kg) and P4 (4 mg/kg) immediately after object training exhibited enhanced OR and OP memory relative to vehicle-treated rats [129; 144]. In ovariectomized mice, we found that systemic E2 (0.2 mg/kg) and P4 (10 or 20 mg/kg) given immediately after training enhanced OR memory consolidation [133]. However, a 5 mg/kg dose of P4 blocked the memory-enhancing effects of E2 in this study, highlighting the dose-dependency of P4’s effects on memory observed in our previous work [127], even when paired with E2. In general, these data suggest that high levels of E2 + P4 are beneficial for object memory consolidation in young female rodents, but more work is needed to elucidate optimal dose ranges and timing of injection, as well as the specificity of these effects to the hippocampus.

7. Molecular mechanisms in the dorsal hippocampus underlying the beneficial effects of E2 and P4 on object memory

Numerous molecular mechanisms within the hippocampus are likely involved in the hormonal regulation of object memories. As discussed in Section 3 above, E2 and P4 may act via classical or non-classical mechanisms to alter hippocampal spinogenesis, neurogenesis, excitability, synaptic plasticity, cell signaling, epigenetic processes, and gene expression. Many of the rapid effects of these hormones on object memory have recently been attributed to non-classical actions of E2 and P4 (see below). However, some effects may also involve classical actions of these hormones, which could lead to changes in hippocampal function that have been observed to last for months [115]. Effects of E2 and P4 outside of the hippocampus must be also considered. Abundant evidence suggests that E2 enhances cholinergic function in basal forebrain neurons that project to the hippocampus [97]. Basal forebrain cholinergic changes in response to E2 can occur within minutes of application (e.g., potassium-evoked acetylcholine release) or after many days or weeks of treatment (e.g., choline acetyltransferase mRNA or activity, high affinity choline uptake, cholinergic neuron number, muscarinic receptor binding) [169173], so both classical and non-classical mechanisms could mediate these effects. Although the importance of these basal forebrain cholinergic neurons to hippocampal memory has been subject to debate [174], potentiation of basal forebrain cholinergic function seems to play a role in the beneficial effects of E2 on hippocampal spatial memory in the radial arm and Morris water mazes [102; 106; 107; 175]. However, the role of basal forebrain cholinergic neurons to OR and OP memory is unclear, as the only study to examine this subject found that the effects of E2 on OR in mice were not related to changes in basal forebrain muscarinic receptors [176].

At present, it is unclear how much classical ER or PR mechanisms contribute to the effects of E2 and P4 on object memory because these mechanisms have not been specifically blocked in memory studies to date; receptor knockouts or antagonists also block non-classical effects of ERs and PRs. However, non-classical effects of E2 and P4 on cell signaling pathways or epigenetic processes can be pinpointed using inhibitors of specific rapid processes such as enzyme phosphorylation or histone acetylation (it is important to note that doses used in these inhibitor experiments do not affect memory on their own so that interactions between hormone and inhibitor can be observed). As such, there is a much better understanding of the non-classical mechanisms underlying rapid consolidation of OR and OP memories. These data will be the focus of the discussion below.

7.1. Estrogen receptors

The beneficial effects of estrogens on hippocampal memory are likely mediated through classical ERs (ERα and ERβ), as well as non-classical membrane receptors (e.g., GPER, ER-X, Gq-mER) [41; 82; 177]. Although some evidence does implicate non-classical ERs in mediating OR and OP memory [178; 179], the involvement of specific non-classical ERs remains unclear. Much more is known about the roles of classical ERs in object memory, which have been tested in young ovariectomized rodents using specific ER agonists and ER knockouts. The majority of behavioral studies using ER agonists implicate ERβ in mediating the memory-enhancing effects of E2 on OR and/or OP [12; 15; 64; 129; 154; 163; 178], although no effect of the ERβ agonist diarylpropionitrile (DPN) has been reported for OP in rats [144], or for OR and OP in Swiss mice [15] (Table 4). Interestingly, several studies found beneficial effects of ERβ agonists, but not ERα agonists, on OR and/or OP. For example, one report found that systemic pre-training delivery of the ERβ agonists Compound 19 (C19) or DPN enhanced both OR and OP, whereas the ERα agonist propyl pyrazole triol [180] did not [154; 178]. Moreover, unlike female wild type or ERα knockout mice, ERβ knockout mice do not exhibit enhanced OR or OP memory following systemic post-training treatment with E2 [163; 178], further supporting a role for ERβ, rather than ERα, in mediating object recognition and spatial memories. Consistent with this notion, E2-treated ERβ knockout mice are impaired in other hippocampal-dependent tasks, including the Morris water maze [181] and Y-maze [182]. However, other data suggest a possible role for ERα, particularly in the dorsal hippocampus [12; 15; 64; 144]. For example, systemic PPT and DPN dose-dependently enhanced OR and OP when given pre-training [64] or posttraining [129]. Recently, low does of PPT and DPN infused into the dorsal hippocampus immediately post-training were shown to enhance OR and OP in C57BL/6 mice [12; 15]. These behavioral effects are supported at the cellular level by studies demonstrating that systemic administration of estradiol benzoate, PPT, or DPN can upregulate hippocampal expression of synaptophysin and spinophilin [183], synaptic proteins that help support and maintain synaptic connections important for memory formation and storage. Collectively, this work suggests that either ERα or ERβ may mediate the effects of E2 on object memory, although the contradictory evidence for ERα necessitates more research to better pinpoint the involvement of ERα in object memory consolidation.

Table 4.

Effects of exogenous estrogen receptor agonists on object memory in young adult female rodents

Species Type of
hormone
ROA Timing of
administrationa
Task(s) Effect on NOR and/or OP Ref #
Rat PPT, DPN, C-19 Systemic Pre-training NOR, OP Two injections of DPN (3 mg/kg) or C-19 (5 mg/kg), but not PPT (3 or 5 mg/kg), two days before training enhanced NOR and OP [154]
Mouse PPT, DPN Systemic Pre-training NOR, OP PPT (75 µg) 15 min before training enhanced NOR and OP, effects of DPN depended on dose and task difficulty [64]
Rat PPT, DPN Systemic Post-training NOR PPT and DPN (0.9 mg/kg) enhanced NOR [129]
Rat PPT, DPN Systemic Post-training OP PPT (0.9 mg/kg), but not DPN (0.9 mg/kg), enhanced OP [144]
Mouse DPN Systemic Post-training NOR, OP DPN (0.1 mg/kg) enhanced NOR and OP in wild type mice, but not ERβ knockouts [163]
Mouse PPT, DPN Systemic Post-training NOR DPN (0.5 mg/kg), but not PPT (0.5 mg/kg), enhanced NOR [178]
Mouse PPT, DPN Intrahippocampal Post-training NOR, OP PPT (0.1 pg) and DPN (10 pg) enhanced NOR and OP [12]
Mouse PPT, DPN Intrahippocampal Post-training NOR PPT (0.1 pg) and DPN (10 pg) enhanced NOR in C57BL/6 mice; PPT, but not DPN, enhanced NOR in Swiss mice [15]
a

Post-training treatments were given immediately after training unless indicated otherwise in the "Effect on NOR and/or OP" column

Note: ROA=route of administration; NOR=novel object recognition; OP=object placement; PPT=propyl pyrazole triol; DPN=diarylpropionitrile C-19=compound 19

7.2. Cell signaling pathways

As discussed above, many effects of sex steroid hormones are thought to occur via rapid non-classical mechanisms that activate cell signaling in the brain. These data have been reviewed in detail elsewhere [39; 95; 177; 184] and, therefore, will be only briefly summarized here (see Fig. 2 for a schematic illustration). One way in which ERα and ERβ can initiate rapid cell signaling is by interacting with non-steroidal membrane receptors like neurotransmitter receptors. Our laboratory recently used bilateral dorsal hippocampal infusions of PPT and DPN given immediately post-training to demonstrate that ERα and ERβ enhance OR and OP in female mice in a manner dependent on concomitant activation of metabotropic glutamate receptor 1a (mGluR1a) [12]. We had previously shown that rapid activation of the ERK cell-signaling pathway in the dorsal hippocampus is necessary for E2 to enhance OR in young ovariectomized mice [10]. The importance of ERK phosphorylation in the effects of E2 on OR are underscored by other findings from our laboratory showing that dorsal hippocampal activation of PI3K/Akt signaling upstream from ERK and mammalian target of rapamycin (mTOR) signaling downstream from ERK are also necessary for E2 to enhance OR in young ovariectomized mice [34; 80]. The E2-induced activation of mTOR by ERK is notable given mTOR’s important role in initiating local protein synthesis in hippocampal dendrites [185]. Our recent work extended these findings to show that inhibition of mGluR1a activation prevented E2, PPT, and DPN from activating dorsal hippocampal ERK signaling and from enhancing OR and OP memory consolidation [12]. These data suggest that E2 activates cell-signaling cascades by binding to ERα or ERβ, which then interact with mGluR1a to initiate cell signaling. ERs may interact with additional glutamate receptors, such as NMDA receptors, as suggested by other data from our laboratory showing that post-training dorsal hippocampal administration of the NMDA receptor antagonist APV prevents systemic E2 (0.2 mg/kg) from enhancing OR memory in young ovariectomized mice [80]. Moreover, inhibiting cell signaling downstream from NMDA receptors with the cAMP inhibitor Rp-cAMPs also prevented systemic E2 from enhancing OR, suggesting that ERα or ERβ may also interact with NMDA receptors to the initiate rapid cell signaling necessary for hippocampal object memory consolidation.

Figure 2.

Figure 2

Schematic illustration of the molecular mechanisms in the hippocampus thus far identified to be required for E2 to enhance object memory consolidation. In the classical response, E2 binds ERα and ERβ, which then translocate into the nucleus, bind to the estrogen response element (ERE) on DNA, and interact with co-regulatory proteins (including histone acetyltransferases, HAT) to influence transcription. E2 also affects cell signaling in several non-classical ways. It can bind to ERs that interact with metabotropic glutamate receptors (mGluRs) at the membrane and activate extracellular regulated kinase (ERK) signaling. It can also interact with NMDA receptors and membrane-bound ERs (mER) to activate the protein kinase A (PKA), phosphoinositol-3-kinase (PI3K), and mammalian target of rapamycin (mTOR) signaling pathways. mTOR signaling regulates the protein synthesis necessary for memory formation. Activation of ERK increases H3 acetylation. Both H3 acetylation and DNA methylation are necessary for E2 to enhance memory consolidation. Adapted with permission from Frick, 2013 and Fortress and Frick, in press [177; 184].

7.3. Epigenetics

ERK activation can regulate cellular processes in numerous ways. As mentioned above, ERK activates mTOR, which phosphorylates core components of the protein synthesis machinery to initiate protein translation [186]. Perhaps better known is the ability of ERK to regulate gene transcription by phosphorylating the transcription factor cAMP response-element binding protein (CREB; [187]), which interacts with histone acetyltransferase enzymes to promote transcription [188]. Indeed, ERK-dependent regulation of epigenetic processes such as histone acetylation is essential for long-term hippocampal memory formation [35; 189]. DNA is tightly coiled around 4 core histone proteins (H2A, H2B, H3, H4), each of which has a tail that can be post-translationally modified to produce transcriptionally permissive or repressive states [190]. Histone acetyltransferases [191] add acetyl groups to histone tails, whereas histone deacetylases (HDACs) remove them [191]. OR memory consolidation is enhanced by HDAC inhibitors [79; 192] and impaired by HAT inhibitors [35]. Contextual fear conditioning or ERK activation increases acetylation of H3 in the hippocampus, and inhibiting ERK activation in the dorsal hippocampus blocks H3 acetylation [189]. Our laboratory recently demonstrated that E2 increases H3 acetylation in an ERK-dependent manner, and that the ability of post-training dorsal hippocampal infusion of E2 to enhance OR memory consolidation is dependent on histone acetylation [35; 79]. E2 also influences OR memory by suppressing negative regulators of memory, such as HDAC2 and HDAC3. These HDACs exert detrimental effects on hippocampal memory by removing acetyl groups from histone tails, thereby preventing the relaxed chromatin state that is essential for gene transcription [193195]. HDAC2 levels are significantly reduced in the dorsal hippocampus after E2 treatment in young ovariectomized mice [35; 79], and both HDAC2 and HDAC3 levels are reduced after E2 treatment in middle-aged females [196]. As such, E2 promotes a suite of histone alterations that facilitate acetylation and the transcriptionally permissive state that allows for gene transcription and object memory enhancement.

E2’s effects on OR memory consolidation are also regulated, in part, by DNA methylation, an epigenetic process that generally silences gene transcription by adding methyl groups to certain cytosine residues on the DNA. Three DNA methyltransferases (DNMTs) catalyze the methylation reaction: DNMT1 is a maintenance enzyme that moves existing methyl groups during replication, whereas DNMT3A and DNMT3B are de novo methyltransferases that add new methyl marks to the DNA [197]. Interestingly, blocking hippocampal DNMT activation with the DNMT inhibitor 5-AZA immediately, but not 3 hours, after training enhances OR memory consolidation in young ovariectomized mice [79]. These data suggest that preventing the methylation of genes necessary for memory formation (e.g., reelin) is instrumental for OR memory. With respect to E2, we found that dorsal hippocampal infusion of E2 increases DNMT3B mRNA and protein in the dorsal hippocampus, and the ability of post-training dorsal hippocampal infusion of E2 to enhance OR memory consolidation is blocked by 5-AZA [79]. These data suggest that E2 may enhance OR memory by methylating memory suppressor genes like those for Hdac2 or Hdac3. Collectively, these data suggest that E2 may mediate it’s beneficial effects on memory by increasing acetylation of memory supporting genes and increasing methylation of memory suppressing genes, processes that may ultimately act in concert to facilitate the transcription and translation of genes important for neural plasticity and the consolidation of object memories.

7.4. Progesterone

The molecular mechanisms through which P4 affects hippocampal memory consolidation are not nearly as well characterized as those of E2. However, some data suggest that P4 regulates OR memory via similar cell signaling mechanisms as E2. For example, P4 promotes rapid activation of the ERK [87; 198], PI3K [198], and PKA [199] cell signaling pathways. Our laboratory has shown that the ability of a post-training dorsal hippocampal infusion of P4 to enhance OR memory consolidation in young ovariectomized mice is dependent on rapid dorsal hippocampal activation of ERK and mTOR [87]. These findings suggest that P4 may regulate memory via similar cell signaling mechanisms as E2. However, potential progesterone receptor mechanisms mediating these effects are unclear. Our unpublished work suggests that dorsal hippocampal activation of either classical PRs or mPRs enhances OR memory consolidation in young ovariectomized mice [200], although possibly through different cell-signaling mechanisms. That is, mPRs appear to activate ERK/mTOR signaling, whereas classical PRs appear to activate canonical Wnt signaling [200]. Finally, it is possible that P4’s effects on memory and cell signaling are entirely independent of PRs, and rather stem from the actions of P4 metabolites such as allopregnanolone, androgens, and estrogens on GABAA, androgen, and estrogen receptors, respectively. As such, much more work must be conducted to gain a better understanding of the molecular mechanisms underlying P4’s effects on object memory consolidation.

8. Regulation of object memory in aging females

In women, menopause occurs in the early 50’s and is characterized by the cessation of reproductive function due to a sharp decline in circulating estrogens and progestins. In rodents, regular estrous cycling also declines in middle age due to age-related reductions in E2 and P4 levels, and the transition from normal to abnormal cycling has been associated with the onset of hippocampal memory decline [201; 202]. Ovarian hormones play a significant role in hippocampal neuroprotection [203206], so the loss of these hormones during aging renders the hippocampus particularly susceptible to neurodegeneration and dysfunction [207209]. Therefore, it should come as no surprise that memory in hippocampal tasks suffers in females as a consequence of aging. Most relevant to this discussion, one recent study capitalized on variability in the timing of reproductive senescence in rats to examine the relationship between reproductive cycling and OR memory. This study found that rats in early middle-age (12 months old) with compromised reproductive status (declining fertility, fecundity, and cycle regularity) and lower circulating levels of ovarian hormones, exhibited worse memory in the OR task than age-matched female rats with maintained reproductive status [210]. Furthermore, as will be discussed below, the responsiveness of the brain to ovarian hormone administration appears to diminish with age.

Ovariectomized rodents in middle age (approximately 14–19 months of age) appear to be as responsive to exogenous E2 as young females (Table 5). For example, systemic E2 or conjugated equine estrogens given pre- or post-training generally enhance OR and OP in middle-aged rats and mice [16; 160; 162; 211213] [but see 146]. Furthermore, dorsal hippocampal infusion of E2 given immediately post-training enhances OR memory in a manner dependent on ERK and PI3K activation, as in young females [34; 81]. Additional data from our group suggests that E2 regulates histone acetylation and Bdnf gene expression in a manner similar to young females [177]. On the other hand, systemic or intrahippocampal E2 administered immediately post-training fails to enhance OR or activate dorsal hippocampal cell signaling in aged (20+ months of age) female mice [81; 161; 162; 214], suggesting a loss of responsiveness to E2 in old age. E2 treatment has also proved unsuccessful in improving the performance of aged females in other types of hippocampal-dependent tasks, such as the radial arm maze [161; 214], T-maze active avoidance [215], and spatial water maze [161; 216]. This is not to say that the aged female brain cannot respond to E2 under the right conditions, as there are many instances in which E2 given chronically prior to training can improve memory in aged females (e.g., [16; 95; 217]). But whether E2 enhances memory in aged females depends on numerous methodological issues as previously discussed [95]. Collectively, however, most studies indicate that the aged female brain exhibits a reduced level of responsiveness to ovarian hormones (Table 5), perhaps due to a reduction in the number and sensitivity of ERα and ERβ in the hippocampus that may compromise E2’s ability to signal through these receptors [218220]. The behavioral findings support the existence of a critical period during early menopause in which estrogen replacement can effectively improve memory [221224]. In the clinical literature, this “critical period hypothesis” suggests that initiating estrogen therapy during early menopause or middle age is essential to prevent or reduce cognitive decline [94; 221; 222]. The data from post-training treatments in OR support the existence of this critical period in rodents, and so this task could be instrumental in determining the neurobiological origins of the critical period, as well as to developing treatments to extend the critical period in aged women.

Table 5.

Effects of exogenous estradiol and progesterone on object memory in middle-aged & aged female rodents

Species Age at testing Type of
hormone
ROA Timing of
administration
a
Task(s) Effect on NOR and/or OP Ref # Reference
Estradiol
Mouse Middle-aged (16–17 months) E2 Systemic Pre-training NOR Chronic E2 in drinking water (1000, 1500, 2500 nM) for 5 weeks before training enhanced NOR [211] Fernandez & Frick, 2004
Rat Middle-aged (11 and 17 months) E2 Systemic Pre-training NOR In mice OVXed at 7–9 weeks of age, E2 enhanced NOR when given via silastics for 9 or 15 months and via acute pre-training injection [16] Vedder et al., 2014
Rat Middle-aged (13 months) E2 Systemic Post-training NOR Conjugated equine estrogens (0.625 mg/kg) enhanced NOR [213] Walf & Frye, 2008
Mouse Middle-aged (17 months) Water-soluble E2 Systemic Post-training NOR E2 (0.2 mg/kg) enhanced NOR [161] Gresack et al., 2007
Mouse Middle-aged (17 months) Water-soluble E2 Intrahippocampal Post-training NOR E2 (0.2 mg/kg) enhanced NOR [81] Fan et al., 2010
Mouse Aged (21 months) Water-soluble E2 Systemic Pre-training NOR E2 (0.2 mg/kg) injected every day or twice/week from 18 months through 21 months of age did not enhance NOR [214] Gresack & Frick, 2006
Rat Aged (20 months) E2 Systemic Pre-training OP Chronic E2 silastics implanted at 14 months enhanced OP tested at 20 months [212] Walf et al., 2009
Rat Aged (21 months) E2 Systemic Pre-training NOR In mice OVXed at 7–9 weeks of age, E2 did not enhance NOR when given via silastics for 19 months and via acute pre-training injection; however, acute E2 enhanced NOR in rats OVXed at 20 months and tested at 21 months [16] Vedder et al., 2014
Mouse Aged (22 months) Water-soluble E2 Systemic Post-training NOR E2 (0.2 mg/kg) did not enhance NOR [162] Gresack et al., 2007
Mouse Aged (22 months) Water-soluble E2 Systemic Post-training NOR E2 (0.2 mg/kg) did not enhance NOR [161] Gresack et al., 2007
Progesterone
Mouse Young middle-age (9 months) P4 Systemic Pre-training NOR, OP Chronic P4 silastics implanted at 6 months enhanced NOR and OP tested at 9 months [236] Frye & Walf, 2008
Mouse Middle-aged (16 months) Water-soluble P4 Systemic Post-training NOR P4 (10 or 20 mg/kg) enhanced NOR tested 24 hr, but not 48 hr, after training [131] Lewis, Orr, and Frick, 2008
Mouse Middle-aged/Aged (18–22 months) P4 Systemic Post-training NOR P4 (10 mg/kg) enhanced NOR in females and males [226] Frye & Walf, 2008
Mouse Aged (20–24 months) P4 Systemic Post-training OP P4 (10 mg/kg) enhanced OP [225] Frye & Walf, 2010
Mouse Aged (21 months) Water-soluble P4 Systemic Post-training NOR P4 (10 mg/kg) enhanced NOR tested 24 hr after training; P4 (5 or 10 mg/kg) enhanced NOR tested 48 hr after training [131] Lewis, Orr, and Frick, 2008
a

Post-training treatments were given immediately after training unless indicated otherwise in the "Effect on NOR and/or OP" column

Note: ROA=route of administration; NOR=novel object recognition; OP=object placement; OVXed=ovariectomized

Few studies have examined the mnemonic effects of P4 in middle-aged or aged rodents, but those that have generally report beneficial effects. In middle-aged ovariectomized mice, chronic pre-training or acute post-training treatment with P enhanced OR [131; 167]. In addition, the P4 metabolite 3α,5α-THP also significantly enhances OR memory in middle-aged female rats [210]. Similarly, in aged gonadally-intact or ovariectomized mice, post-training injection of P4 significantly improved both OR and OP memory relative to vehicle-treated mice [131; 167; 225]. Interestingly, systemic P4 treatment immediately after training facilitated OR memory consolidation in both middle-aged and aged female mice 24 hours later, but only in aged mice 48 hours later [131]. These data suggest that aged females may be more sensitive than middle-aged females to the memory-enhancing effects of P4. Consistent with this notion, P4 also improves the memory of aged female mice in other hippocampal-dependent memory tasks including T-maze, water maze, and contextual fear conditioning [226]. Such evidence that P4, but not E2, can reverse hippocampal memory deficits in aged rodents suggests that P4 may regulate memory through mechanisms that differ or are less sensitive to aging than those used by E2. Such differential sensitivity to E2 and P4 is worth exploring with respect to the development of future treatments to reduce memory decline in postmenopausal women.

9. Conclusions and future directions

E2 and P4 are important regulators of object recognition and spatial memory in rodents, as illustrated by their effects on memory in the OR and OP tasks. Our laboratory and others have made great strides in recent years using these tasks as tools to understand the extent to which E2 and P4 regulate hippocampal memory formation. We have begun to identify the essential receptors, cell signaling pathways, and epigenetic processes necessary for E2 and P4 to enhance OR and OP, but we have considerably more work to do to identify the complex molecular mechanisms underlying hormonal regulation of object memory. Vital issues to address in future years include determining the extent to which rapid changes in cell signaling and epigenetics translate into structural alterations that maintain long-term memories, and defining the essential genes and gene products that support the synaptic plasticity underlying hormonal regulation of memory consolidation. As discussed above, many other key questions have yet to be resolved. For example, does ovarian function play a significant role in the response to hormone treatment? How do hormones regulate object memory during the estrous cycle, pregnancy, and reproductive senescence? Does progesterone regulate object memory itself via progesterone receptors or, rather, via its conversion to neurosteroid and/or sex steroid metabolites? How might the effects of clinically utilized hormone preparations on object memory differ from the E2 and P4 used in rodent studies? The answers to these questions would fundamentally advance our understanding of the key neural mechanisms through which E2 and P4 regulate memory formation, and provide sorely needed insight into the etiology and symptomatology of mental illnesses for which women are at increased risk, such as depression, anxiety disorders, schizophrenia, and dementia [227230].

Along these lines, dysfunction in multiple cognitive brain regions is characteristic of several neuropsychiatric and neurodegenerative diseases that predominantly affect women (e.g., depression, anxiety disorders). Yet studies of hormonal regulation of object memory in rodents have focused largely on the hippocampus. Therefore, another important future direction is to identify the brain regions involved in hormonal modulation of OR and OP. The dorsal hippocampus is a key modulator for object-based memory tasks, as illustrated by the many examples presented above in which direct dorsal hippocampal infusions of hormones or other drugs altered memory consolidation in OR and OP. However, far too few studies have examined hormonal regulation in other brain regions that mediate OR and OP memory. One such study recently reported that E2 infused into the perirhinal cortex/entorhinal cortex region enhanced OR memory in young ovariectomized rats [231]. Surely, other medial temporal lobe regions, as well as other cortical regions (e.g., prefrontal cortex) also play key roles. As such, we would encourage more studies examining hormonal regulation of other brain regions to gain a more complete understanding of how hormones regulate memory formation in the OR and OP tasks.

Another important area in which object memory tasks may prove useful is in understanding the etiology of sex differences in cognitive function. Women are significantly more likely than men to develop age-related memory decline, Alzheimer’s disease, depression, anxiety, and mood disorders [224; 227; 228; 232; 233]. Further, for many of these disorders, women with lower endogenous levels of E2 have exacerbated symptoms compared to women with higher levels of E2 and men [218220]. Yet why women are at increased risk for developing certain mental illnesses is unclear. Much of this uncertainty stems from the fact that many of the cellular and molecular mechanisms through which sex steroid hormones like E2 and P4 affect brain function remain unknown. As such, understanding the neural mechanisms through which E2 and P regulate memory function may shed light on sex differences in risk of mental illness. Few studies have examined sex differences in OR and OP, and these are somewhat contradictory. For example, our laboratory reported that male mice outperform female mice in two different OR tasks and an OP task [140]. However, others report a female advantage in OR under conditions of high E2 and P levels [234] or high object similarity [235]. As such, more research on sex differences in these tasks would help establish the parameters under which sex differences may be observed, and could provide insights into how sex differences contribute to increased susceptibility for developing certain mental illnesses.

In conclusion, this review has illustrated the importance of ovarian sex steroid hormones in regulating object memory. As should be clear from the discussion above, many questions remain to be answered to more fully understand how hormones regulate the various aspects of object memory. Significant progress in resolving these issues could allow researchers to identify putative drug targets that might lead to novel therapeutics for maintaining mental health in women. As such, it will be imperative to continue this work in future years, and we encourage researchers to consider the potential contributions of these hormones to their own experiments.

Research Highlights.

  • 17β-Estradiol (E2) and progesterone (P4) potently regulate hippocampal memory.

  • Object recognition (OR) and object placement (OP) are enhanced by E2 and P4.

  • E2 and P4 rapidly activate the molecular processes underlying object memory.

  • Both endogenous and exogenous E2 and P4 affect object memory across the lifespan.

  • This review discusses regulation of object memory by E2 and P4 in female rodents.

Acknowledgements

The University of Wisconsin-Milwaukee supported this writing of this review. The empirical work from our laboratory described in this review was supported by grants from the National Institute on Aging (AG022525), National Institute of Mental Health (MH065460), Alzheimer’s Association (IIRG-03�6051), American Foundation for Aging Research/Pfizer (Grant in Hormones and Aging), and University of Wisconsin-Milwaukee (Research Growth Initiative Award) to K.M.F., an Ellison Medical Foundation/AFAR Postdoctoral Fellowship to A.M.F., the University of Wisconsin-Milwaukee, and Yale University.

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.Ennaceur A, Delacour J. A new one-trial test for neurobiological studies of memory in rats. 1: Behavioral data. Behav Brain Res. 1988;31:47–59. doi: 10.1016/0166-4328(88)90157-x. [DOI] [PubMed] [Google Scholar]
  • 2.Ennaceur A, Neave N, Aggleton JP. Spontaneous object recognition and object location memory in rats: The effects of lesions in the cingulate cortices, the medial prefrontal cortex, the cingulum bundle and the fornix. Exp Brain Research. 1997;113:509–519. doi: 10.1007/pl00005603. [DOI] [PubMed] [Google Scholar]
  • 3.Hammond RS, Tull LE, Stackman RW. On the delay-dependent involvement of the hippocampus in object recognition memory. Neurobiol Learn Mem. 2004;82:26–34. doi: 10.1016/j.nlm.2004.03.005. [DOI] [PubMed] [Google Scholar]
  • 4.Squire LR, Wixted JT, Clark RE. Recognition memory and the medial temporal lobe: a new perspective. Nat Rev Neurosci. 2007;8:872–883. doi: 10.1038/nrn2154. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Wilson DI, Langston RF, Schlesiger MI, Wagner M, Watanabe S, Ainge JA. Lateral entorhinal cortex is critical for novel object-context recognition. Hippocampus. 2013;23:352–366. doi: 10.1002/hipo.22095. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Broadbent NJ, Squire LR, Clark RE. Spatial memory, recognition memory, and the hippocampus. Proc Natl Acad Sci U S A. 2004;101:14515–14520. doi: 10.1073/pnas.0406344101. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.McGaugh JL. Involvement of hormonal and neuromodulatory systems in the regulaton of memory storage. Annu Rev Neurosci. 1989;12:255–287. doi: 10.1146/annurev.ne.12.030189.001351. [DOI] [PubMed] [Google Scholar]
  • 8.McLaughlin KJ, Wilson JO, Harman J, Wright RL, Wieczorek L, Gomez J, et al. Chronic 17beta-estradiol or cholesterol prevents stress-induced hippocampal CA3 dendritic retraction in ovariectomized female rats: possible correspondence between CA1 spine properties and spatial acquisition. Hippocampus. 2010;20:768–786. doi: 10.1002/hipo.20678. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Bowman RE, Ferguson D, Luine VN. Effects of chronic restraint stress and estradiol on open field activity, spatial memory, and monoaminergic neurotransmitters in ovariectomized rats. Neuroscience. 2002;113:401–410. doi: 10.1016/s0306-4522(02)00156-2. [DOI] [PubMed] [Google Scholar]
  • 10.Fernandez SM, Lewis MC, Pechenino AS, Harburger LL, Orr PT, Gresack JE, et al. Estradiol-induced enhancement of object memory consolidation involves hippocampal ERK activation and membrane-bound estrogen receptors. J Neurosci. 2008;28:8660–8667. doi: 10.1523/JNEUROSCI.1968-08.2008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.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. 2005;25:5066–5078. doi: 10.1523/JNEUROSCI.1427-05.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Boulware MI, Heisler JD, Frick KM. The memory-enhancing effects of hippocampal estrogen receptor activation involve metabotropic glutamate receptor signaling. J Neurosci. 2013;33:15184–15194. doi: 10.1523/JNEUROSCI.1716-13.2013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Zhao L, Brinton RD. Estrogen receptor α and β differentially regulate intracellular Ca2+ dynamics leading to ERK phosphorylation and estrogen neuroprotection in hippocampal neurons. Brain Res. 2007;1172:48–59. doi: 10.1016/j.brainres.2007.06.092. [DOI] [PubMed] [Google Scholar]
  • 14.Stranahan AM. Similarities and differences in spatial learning and object recognition between young male C57Bl/6J mice and Sprague-Dawley rats. Behav Neurosci. 2011;125:791–795. doi: 10.1037/a0025133. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Pereira LM, Bastos CP, de Souza JM, Ribeiro FM, Pereira GS. Estradiol enhances object recognition memory in Swiss female mice by activating hippocampal estrogen receptor alpha. Neurobiol Learn Mem. 2014;114C:1–9. doi: 10.1016/j.nlm.2014.04.001. [DOI] [PubMed] [Google Scholar]
  • 16.Vedder LC, Bredemann TM, McMahon LL. Estradiol replacement extends the window of opportunity for hippocampal function. Neurobiol Aging. 2014;35:2183–2192. doi: 10.1016/j.neurobiolaging.2014.04.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Morris RG, Garrud P, Rawlins JN, O'Keefe J. Place navigation impaired in rats with hippocampal lesions. Nature. 1982;297:681–683. doi: 10.1038/297681a0. [DOI] [PubMed] [Google Scholar]
  • 18.Refolo LM, Salton SRJ, Anderson JP, Mehta P, Robakis NK. Nerve and epidermal growth factors induce the release of the Alzheimer amyloid precursor from PC 12 cells. Biochem Biophys Res Commun. 1989;164:664–670. doi: 10.1016/0006-291x(89)91511-8. [DOI] [PubMed] [Google Scholar]
  • 19.Clark RE, Zola SM, Squire LR. Impaired recognition memory in rats after damage to the hippocampus. J Neurosci. 2000;20:8853–8860. doi: 10.1523/JNEUROSCI.20-23-08853.2000. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Baker KB, Kim JJ. Effects of stress and hippocampal NMDA receptor antagonism on recognition memory in rats. Learn Mem. 2002;9:58–65. doi: 10.1101/lm.46102. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Cohen SJ, Munchow AH, Rios LM, Zhang G, Asgeirsdottir HN, Stackman RW., Jr The rodent hippocampus is essential for nonspatial object memory. Curr Biol. 2013;23:1685–1690. doi: 10.1016/j.cub.2013.07.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Duva CA, Floresco SB, Wunderlich GR, Lao TL, Pinel JP, Phillips AG. Disruption of spatial but not object-recognition memory by neurotoxic lesions of the dorsal hippocampus in rats. Behav Neurosci. 1997;111:1184–1196. doi: 10.1037//0735-7044.111.6.1184. [DOI] [PubMed] [Google Scholar]
  • 23.Forwood SE, Winters BD, Bussey TJ. Hippocampal lesions that abolish spatial maze performance spare object recognition memory at delays of up to 48 hours. Hippocampus. 2005;15:347–355. doi: 10.1002/hipo.20059. [DOI] [PubMed] [Google Scholar]
  • 24.Winters BD, Forwood SE, Cowell RA, Saksida LM, Bussey TJ. Double dissociation between the effects of peri-postrhinal cortex and hippocampal lesions on tests of object recognition and spatial memory: heterogeneity of function within the temporal lobe. J Neurosci. 2004;24:5901–5908. doi: 10.1523/JNEUROSCI.1346-04.2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Ainge JA, Heron-Maxwell C, Theofilas P, Wright P, de Hoz L, Wood ER. The role of the hippocampus in object recognition in rats: examination of the influence of task parameters and lesion size. Behav Brain Res. 2006;167:183–195. doi: 10.1016/j.bbr.2005.09.005. [DOI] [PubMed] [Google Scholar]
  • 26.Broadbent NJ, Gaskin S, Squire LR, Clark RE. Object recognition memory and the rodent hippocampus. Learn Mem. 2010;17:5–11. doi: 10.1101/lm.1650110. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Mumby DG. Perspectives on object-recognition memory following hippocampal damage: lessons from studies in rats. Behav Brain Res. 2001;127:159–181. doi: 10.1016/s0166-4328(01)00367-9. [DOI] [PubMed] [Google Scholar]
  • 28.Mumby DG, Tremblay A, Lecluse V, Lehmann H. Hippocampal damage and anterograde object-recognition in rats after long retention intervals. Hippocampus. 2005;15:1050–1056. doi: 10.1002/hipo.20122. [DOI] [PubMed] [Google Scholar]
  • 29.Oliveira AM, Hawk JD, Abel T, Havekes R. Post-training reversible inactivation of the hippocampus enhances novel object recognition memory. Learn Mem. 2010;17:155–160. doi: 10.1101/lm.1625310. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Barker GR, Bird F, Alexander V, Warburton EC. Recognition memory for objects, place, and temporal order: a disconnection analysis of the role of the medial prefrontal cortex and perirhinal cortex. J Neurosci. 2007;27:2948–2957. doi: 10.1523/JNEUROSCI.5289-06.2007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Zola SM, Squire LR, Teng E, Stefanacci L, Buffalo EA, Clark RE. Impaired recognition memory in monkeys after damage limited to the hippocampal region. J Neurosci. 2000;20:451–463. doi: 10.1523/JNEUROSCI.20-01-00451.2000. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.McKee RD, Squire LR. On the development of declarative memory. J Exp Psychol Learn Mem Cogn. 1993;19:397–404. doi: 10.1037//0278-7393.19.2.397. [DOI] [PubMed] [Google Scholar]
  • 33.Milner TA, Ayoola K, Drake CT, Herrick SP, Tabori NE, McEwen BS, et al. Ultrastructural localization of estrogen receptor beta immunoreactivity in the rat hippocampal formation. J Comp Neurol. 2005;491:81–95. doi: 10.1002/cne.20724. [DOI] [PubMed] [Google Scholar]
  • 34.Fortress AM, Fan L, Orr PT, Zhao Z, Frick KM. Estradiol-induced object recognition memory consolidation is dependent on activation of mTOR signaling in dorsal hippocampus. Learn Mem. 2013;20:147–155. doi: 10.1101/lm.026732.112. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Zhao Z, Fan L, Fortress AM, Boulware MI, Frick KM. Hippocampal histone acetylation regulates object recognition and the estradiol-induced enhancement of object recognition. J Neurosci. 2012;32:2344–2351. doi: 10.1523/JNEUROSCI.5819-11.2012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Clarke JR, Cammarota M, Gruart A, Izquierdo I, Delgado-Garcia JM. Plastic modifications induced by object recognition memory processing. Proc Natl Acad Sci U S A. 2010;107:2652–2657. doi: 10.1073/pnas.0915059107. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Barbosa FF, Santos JR, Meurer YS, Macedo PT, Ferreira LM, Pontes IM, et al. Differential cortical c-Fos and zif-268 expression after object and spatial memory processing in a standard or episodic-like object recognition task. Front Behav Neurosci. 2013;7:112. doi: 10.3389/fnbeh.2013.00112. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Dumitriu D, Rapp PR, McEwen BS, Morrison JH. Estrogen and the aging brain: An elixir for the weary cortical network. Ann N Y Acad Sci. 2010;1204:104–112. doi: 10.1111/j.1749-6632.2010.05529.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Frick KM. Building a better hormone therapy? How understanding the rapid effects of sex steroid hormones could lead to new therapeutics for age-related memory decline. Behav Neurosci. 2012;126:29–53. doi: 10.1037/a0026660. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Luine V, Frankfurt M. Interactions between estradiol, BDNF and dendritic spines in promoting memory. Neuroscience. 2013;239:34–45. doi: 10.1016/j.neuroscience.2012.10.019. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Micevych P, Christensen A. Membrane-initiated estradiol actions mediate structural plasticity and reproduction. Front Neuroendocrinol. 2012;33:331–341. doi: 10.1016/j.yfrne.2012.07.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Woolley CS. Acute effects of estrogen on neuronal physiology. Ann Rev Pharm Toxicol. 2007;47:657–680. doi: 10.1146/annurev.pharmtox.47.120505.105219. [DOI] [PubMed] [Google Scholar]
  • 43.Williams C, Meck WH. The organizational effects of gonadal steroids on sexually dimorphic spatial ability. Psychoneuroendocrinology. 1991;16:155–176. doi: 10.1016/0306-4530(91)90076-6. [DOI] [PubMed] [Google Scholar]
  • 44.McCarthy MM. Estradiol and the developing brain. Physiol Rev. 2008;88:91–124. doi: 10.1152/physrev.00010.2007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.McCarthy MM, Auger AP, Bale TL, De Vries GJ, Dunn GA, Forger NG, et al. The epigenetics of sex differences in the brain. J Neurosci. 2009;29:12815–12823. doi: 10.1523/JNEUROSCI.3331-09.2009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.McCarthy MM, Nugent BM. Epigenetic contributions to hormonally-mediated sexual differentiation of the brain. J Neuroendocrinol. 2013;25:1133–1140. doi: 10.1111/jne.12072. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Kurian JR, Olesen KM, Auger AP. Sex differences in epigenetic regulation of the estrogen receptor-alpha promoter within the developing preoptic area. Endocrinology. 2010;151:2297–2305. doi: 10.1210/en.2009-0649. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Murray EK, Hien A, de Vries GJ, Forger NG. Epigenetic control of sexual differentiation of the bed nucleus of the stria terminalis. Endocrinology. 2009;150:4241–4247. doi: 10.1210/en.2009-0458. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Matsuda KI, Mori H, Nugent BM, Pfaff DW, McCarthy MM, Kawata M. Histone deacetylation during brain development is essential for permanent masculinization of sexual behavior. Endocrinology. 2011;152:2760–2767. doi: 10.1210/en.2011-0193. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Chung WC, Swaab DF, De Vries GJ. Apoptosis during sexual differentiation of the bed nucleus of the stria terminalis in the rat brain. J Neurobiol. 2000;43:234–243. [PubMed] [Google Scholar]
  • 51.Tsai HC, Zhang F, Adamantidis A, Stuber GD, Bonci A, de Lecea L, et al. Phasic firing in dopaminergic neurons is sufficient for behavioral conditioning. Science. 2009;324:1080–1084. doi: 10.1126/science.1168878. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.McEwen BS, Alves SE. Estrogen actions in the central nervous system. Endocr Rev. 1999;20:279–307. doi: 10.1210/edrv.20.3.0365. [DOI] [PubMed] [Google Scholar]
  • 53.Williams C, Barnett AM, Meck WH. Organizational effects of early gonadal secretions on sexual differentiation in spatial memory. Behav Neurosci. 1990;104:84–97. doi: 10.1037//0735-7044.104.1.84. [DOI] [PubMed] [Google Scholar]
  • 54.McCarthy MM, Konkle AT. When is a sex difference not a sex difference? Front Neuroendocrinol. 2005;26:85–102. doi: 10.1016/j.yfrne.2005.06.001. [DOI] [PubMed] [Google Scholar]
  • 55.Hojo Y, Hattori TA, Enami T, Furukawa A, Suzuki K, Ishii HT, et al. Adult male rat hippocampus synthesizes estradiol from pregnenolone by cytochromes P45017alpha and P450 aromatase localized in neurons. Proc Natl Acad Sci U S A. 2004;101:865–870. doi: 10.1073/pnas.2630225100. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.MacLusky NJ, Clark AS, Naftolin F, Goldman-Rakic PS. Estrogen formation in the mammalian brain: possible role of aromatase in sexual differentiation of the hippocampus and neocortex. Steroids. 1987;50:459–474. doi: 10.1016/0039-128x(87)90032-8. [DOI] [PubMed] [Google Scholar]
  • 57.Long JA, Evans HM. The oestrous cycle in the rat and its associated phenomena. Berkeley, CA: University of California Press; 1922. [Google Scholar]
  • 58.Woolley CS, Gould E, Frankfurt M, McEwen BS. Naturally occurring fluctuation in dendritic spine density on adult hippocampal pyramidal neurons. J Neurosci. 1990;10:4035–4039. doi: 10.1523/JNEUROSCI.10-12-04035.1990. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.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]
  • 60.Woolley CS, McEwen BS. Estradiol mediates fluctuation in hippocampal synapse density during the estrous cycle in the adult rat. J Neurosci. 1992;12:2549–2554. doi: 10.1523/JNEUROSCI.12-07-02549.1992. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.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]
  • 62.Phan A, Gabor CS, Favaro KJ, Kaschack S, Armstrong JN, MacLusky NJ, et al. Low doses of 17beta-estradiol rapidly improve learning and increase hippocampal dendritic spines. Neuropsychopharmacology. 2012;37:2299–2309. doi: 10.1038/npp.2012.82. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Luine VN, Frankfurt M. Estrogens facilitate memory processing through membrane mediated mechanisms and alterations in spine density. Front Neuroendocrinol. 2012;33:388–402. doi: 10.1016/j.yfrne.2012.07.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Phan A, Lancaster KE, Armstrong JN, MacLusky NJ, Choleris E. Rapid effects of estrogen receptor alpha and beta selective agonists on learning and dendritic spines in female mice. Endocrinology. 2011;152:1492–1502. doi: 10.1210/en.2010-1273. [DOI] [PubMed] [Google Scholar]
  • 65.Pozzo-Miller LD, Inoue T, Murphy DD. Estradiol increases spine density and NMDA-dependent Ca2+ transients in spines of CA1 pyramidal neurons from hippocampal slices. J Neurophysiol. 1999;81:1404–1411. doi: 10.1152/jn.1999.81.3.1404. [DOI] [PubMed] [Google Scholar]
  • 66.Prange-Kiel J, Fester L, Zhou L, Jarry H, Rune GM. Estrus cyclicity of spinogenesis: underlying mechanisms. J Neural Transm. 2009;116:1417–1425. doi: 10.1007/s00702-009-0294-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Segal M, Murphy D. Estradiol induces formation of dendritic spines in hippocampal neurons: functional correlates. Horm Behav. 2001;40:156–159. doi: 10.1006/hbeh.2001.1688. [DOI] [PubMed] [Google Scholar]
  • 68.MacLusky NJ, Luine VN, Hajszan T, Leranth C. The 17alpha and 17beta isomers of estradiol both induce rapid spine synapse formation in the CA1 hippocampal subfield of ovariectomized female rats. Endocrinology. 2005;146:287–293. doi: 10.1210/en.2004-0730. [DOI] [PubMed] [Google Scholar]
  • 69.Li C, Brake WG, Romeo RD, Dunlop JC, Gordon M, Buzescu R, et al. Estrogen alters hippocampal dendritic spine shape and enhances synaptic protein immunoreactivity and spatial memory in female mice. Proc Natl Acad Sci U S A. 2004;101:2185–2190. doi: 10.1073/pnas.0307313101. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Brake WG, Alves SE, Dunlop JC, Lee SJ, Bulloch K, Allen PB, et al. Novel target sites for estrogen action in the dorsal hippocampus: An examination of synaptic proteins. Endocrinology. 2001;142:1284–1289. doi: 10.1210/endo.142.3.8036. [DOI] [PubMed] [Google Scholar]
  • 71.Akama KT, McEwen BS. Estrogen stimulates postsynaptic density-95 rapid protein synthesis via the Akt/protein kinase B pathway. J Neurosci. 2003;23:2333–2339. doi: 10.1523/JNEUROSCI.23-06-02333.2003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Sato K, Akaishi T, Matsuki N, Ohno Y, Nakazawa K. beta-Estradiol induces synptogenesis in the hippocampus by enhancing brain-derived neurotrophic factor release from dentate gyrus granule cells. Brain Res. 2007;1150:108–120. doi: 10.1016/j.brainres.2007.02.093. [DOI] [PubMed] [Google Scholar]
  • 73.Foy MR, Xu J, Xie X, Brinton RD, Thompson RF, Berger TW. 17β-estradiol enhances NMDA receptor-mediated EPSPs and long-term potentiation. J Neurophysiol. 1999;81:925–929. doi: 10.1152/jn.1999.81.2.925. [DOI] [PubMed] [Google Scholar]
  • 74.McClure RE, Barha CK, Galea LA. 17beta-Estradiol, but not estrone, increases the survival and activation of new neurons in the hippocampus in response to spatial memory in adult female rats. Horm Behav. 2013;63:144–157. doi: 10.1016/j.yhbeh.2012.09.011. [DOI] [PubMed] [Google Scholar]
  • 75.Milner TA, McEwen BS, Hayashi S, Li CJ, Reagan LP, Alves SE. Ultrastructural evidence that hippocampal alpha estrogen receptors are located at extranuclear sites. J Comp Neurol. 2001;429:355–371. [PubMed] [Google Scholar]
  • 76.Waters EM, Yildirim M, Janssen WG, Lou WY, McEwen BS, Morrison JH, et al. Estrogen and aging affect the synaptic distribution of estrogen receptor beta-immunoreactivity in the CA1 region of female rat hippocampus. Brain Res. 2011;1379:86–97. doi: 10.1016/j.brainres.2010.09.069. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Towart LA, Alves SE, Znamensky V, Hayashi S, McEwen BS, Milner TA. Subcellular relationships between cholinergic terminals and estrogen receptor-alpha in the dorsal hippocampus. J Comp Neurol. 2003;463:390–401. doi: 10.1002/cne.10753. [DOI] [PubMed] [Google Scholar]
  • 78.Murphy DD, Cole NB, Greenberger V, Segal M. Estradiol increases dendritic spine density by reducing GABA neurotransmission in hippocampal neurons. J Neurosci. 1998;18:2550–2559. doi: 10.1523/JNEUROSCI.18-07-02550.1998. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Zhao Z, Fan L, Frick KM. Epigenetic alterations regulate the estradiol-induced enhancement of memory consolidation. Proc Natl Acad Sci U S A. 2010;107:5605–5610. doi: 10.1073/pnas.0910578107. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Lewis MC, Kerr KM, Orr PT, Frick KM. Estradiol-induced enhancement of object memory consolidation involves NMDA receptors and protein kinase A in the dorsal hippocampus of female C57BL/6 mice. Behav Neurosci. 2008;122:716–721. doi: 10.1037/0735-7044.122.3.716. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Fan L, Zhao Z, Orr PT, Chambers CH, Lewis MC, Frick KM. Estradiol-induced object memory consolidation in middle-aged female mice requires dorsal hippocampal extracellular signal-regulated kinase and phosphatidylinositol 3-kinase activation. J Neurosci. 2010;30:4390–4400. doi: 10.1523/JNEUROSCI.4333-09.2010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Srivastava DP, Evans PD. G-protein oestrogen receptor 1: trials and tribulations of a membrane oestrogen receptor. J Neuroendocrinol. 2013;25:1219–1230. doi: 10.1111/jne.12071. [DOI] [PubMed] [Google Scholar]
  • 83.Mitterling KL, Spencer JL, Dziedzic N, Shenoy S, McCarthy K, Waters EM, et al. Cellular and subcellular localization of estrogen and progestin receptor immunoreactivities in the mouse hippocampus. J Comp Neurol. 2010;518:2729–2743. doi: 10.1002/cne.22361. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Foy MR, Akopian G, Thompson RF. Progesterone regulation of synaptic transmission and plasticity in rodent hippocampus. Learn Mem. 2008;15:820–822. doi: 10.1101/lm.1124708. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Liu M, Dziennis S, Hurn PD, Alkayed NJ. Mechanisms of gender-linked ischemic brain injury. Restor Neurol Neurosci. 2009;27:163–179. doi: 10.3233/RNN-2009-0467. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Zhao Y, Wang J, Liu C, Jiang C, Zhao C, Zhu Z. Progesterone influences postischemic synaptogenesis in the CA1 region of the hippocampus in rats. Synapse. 2011;65:880–891. doi: 10.1002/syn.20915. [DOI] [PubMed] [Google Scholar]
  • 87.Orr PT, Rubin AJ, Fan L, Kent BA, Frick KM. The progesterone-induced enhancement of object recognition memory consolidation involves activation of the extracellular signal-regulated kinase (ERK) and mammalian target of rapamycin (mTOR) pathways in the dorsal hippocampus. Horm Behav. 2012;61:487–495. doi: 10.1016/j.yhbeh.2012.01.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Belelli D, Lambert JJ. Neurosteroids: endogenous regulators of the GABA(A) receptor. Nat Rev Neurosci. 2005;6:565–575. doi: 10.1038/nrn1703. [DOI] [PubMed] [Google Scholar]
  • 89.Waters EM, Torres-Reveron A, McEwen BS, Milner TA. Ultrastructural localization of extranuclear progestin receptors in the rat hippocampal formation. J Comp Neurol. 2008;511:34–46. doi: 10.1002/cne.21826. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Maki PM. Minireview: effects of different HT formulations on cognition. Endocrinology. 2012;153:3564–3570. doi: 10.1210/en.2012-1175. [DOI] [PubMed] [Google Scholar]
  • 91.Foster TC. Role of estrogen receptor alpha and beta expression and signaling on cognitive function during aging. Hippocampus. 2012;22:656–669. doi: 10.1002/hipo.20935. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.Hammond R, Gibbs RB. GPR30 is positioned to mediate estrogen effects on basal forebrain cholinergic neurons and cognitive performance. Brain Res. 2011;1379:53–60. doi: 10.1016/j.brainres.2010.11.098. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.Galea LA, Uban KA, Epp JR, Brummelte S, Barha CK, Wilson WL, et al. Endocrine regulation of cognition and neuroplasticity: our pursuit to unveil the complex interaction between hormones, the brain, and behaviour. Can J Exp Psychol. 2008;62:247–260. doi: 10.1037/a0014501. [DOI] [PubMed] [Google Scholar]
  • 94.Sherwin BB, Henry JF. Brain aging modulates the neuroprotective effects of estrogen on selective aspects of cognition in women: a critical review. Front Neuroendocrinol. 2008;29:88–113. doi: 10.1016/j.yfrne.2007.08.002. [DOI] [PubMed] [Google Scholar]
  • 95.Frick KM. Estrogens and age-related memory decline in rodents: what have we learned and where do we go from here? Horm Behav. 2009;55:2–23. doi: 10.1016/j.yhbeh.2008.08.015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96.Daniel JM. Effects of oestrogen on cognition: What have we learned from basic research? J Neuroendocrinol. 2006;18:787–795. doi: 10.1111/j.1365-2826.2006.01471.x. [DOI] [PubMed] [Google Scholar]
  • 97.Gibbs RB. Estrogen therapy and cognition: A review of the cholinergic hypothesis. Endocr Rev. 2010;31:224–253. doi: 10.1210/er.2009-0036. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98.Choleris E, Clipperton-Allen AE, Phan A, Valsecchi P, Kavaliers M. Estrogenic involvement in social learning, social recognition and pathogen avoidance. Front Neuroendocrinol. 2012;33:140–159. doi: 10.1016/j.yfrne.2012.02.001. [DOI] [PubMed] [Google Scholar]
  • 99.Acosta JI, Mayer L, Talboom JS, Tsang CW, Smith CJ, Enders CK, et al. Transitional versus surgical menopause in a rodent model: etiology of ovarian hormone loss impacts memory and the acetylcholine system. Endocrinology. 2009;150:4248–4259. doi: 10.1210/en.2008-1802. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100.Acosta JI, Mayer LP, Braden BB, Nonnenmacher S, Mennenga SE, Bimonte-Nelson HA. The cognitive effects of conjugated equine estrogens depend on whether menopause etiology is transitional or surgical. Endocrinology. 2010;151:3795–3804. doi: 10.1210/en.2010-0055. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101.Bimonte HA, Denenberg VH. Estradiol facilitates performance as working memory load increases. Psychoneuroendocrinology. 1999;24:161–173. doi: 10.1016/s0306-4530(98)00068-7. [DOI] [PubMed] [Google Scholar]
  • 102.Daniel JM, Dohanich GP. Acetylcholine mediates the estrogen-induced increase in NMDA receptor binding in CA1 of the hippocampus and the associated improvement in working memory. J Neurosci. 2001;21:6949–6956. doi: 10.1523/JNEUROSCI.21-17-06949.2001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103.Sandstrom NJ, Williams CL. Memory retention is modulated by acute estradiol and progesterone replacement. Behav Neurosci. 2001;115:384–393. [PubMed] [Google Scholar]
  • 104.Bohacek J, Daniel JM. Increased daily handling of ovariectomized rats enhances performance on a radial-maze task and obscures effects of estradiol replacement. Horm Behav. 2007;52:237–243. doi: 10.1016/j.yhbeh.2007.04.010. [DOI] [PubMed] [Google Scholar]
  • 105.Daniel JM, Fader AJ, Spencer AL, Dohanich GP. Estrogen enhances performance of female rats during acquisition of a radial arm maze. Horm Behav. 1997;32:217–225. doi: 10.1006/hbeh.1997.1433. [DOI] [PubMed] [Google Scholar]
  • 106.Fader AJ, Hendricson AW, Dohanich GP. Estrogen improves performance of reinforced T-maze alternation and prevents the amnestic effects of scopolamine administered systemically or intrahippocampally. Neurobiol Learn Mem. 1998;69:225–240. doi: 10.1006/nlme.1998.3820. [DOI] [PubMed] [Google Scholar]
  • 107.Fader AJ, Johnson PEM, Dohanich GP. Estrogen improves working but not reference memory and prevents amnestic effects of scopolamine on a radial-arm maze. Pharm Biochem Behav. 1999;62:711–717. doi: 10.1016/s0091-3057(98)00219-6. [DOI] [PubMed] [Google Scholar]
  • 108.Gibbs RB. Estrogen replacement enhances acquisition of a spatial memory task and reduces deficits associated with hippocampal muscarinic receptor inhibition. Horm Behav. 1999;36:222–233. doi: 10.1006/hbeh.1999.1541. [DOI] [PubMed] [Google Scholar]
  • 109.Garza-Meilandt A, Cantu RE, Claiborne BJ. Estradiol's effects on learning and neuronal morphology vary with route of administration. Behav Neurosci. 2006;120:905–916. doi: 10.1037/0735-7044.120.4.905. [DOI] [PubMed] [Google Scholar]
  • 110.Holmes MM, Wide JK, Galea LAM. Low levels of estradiol facilitate, whereas high levels of estradiol impair, working memory performance on the radial arm maze. Behav Neurosci. 2002;116:928–934. doi: 10.1037//0735-7044.116.5.928. [DOI] [PubMed] [Google Scholar]
  • 111.Luine VN, Richards ST, Wu VY, Beck KD. Estradiol enhances learning and memory in a spatial memory task and effects levels of monoaminergic neurotransmitters. Horm Behav. 1998;34:149–162. doi: 10.1006/hbeh.1998.1473. [DOI] [PubMed] [Google Scholar]
  • 112.O'Neal MF, Means LW, Poole MC, Hamm RJ. Estrogen affects performance of ovariectomized rats in a two-choice water-escape working memory task. Psychoneuroendocrinology. 1996;21:51–65. doi: 10.1016/0306-4530(95)00032-1. [DOI] [PubMed] [Google Scholar]
  • 113.Sandstrom NJ, Williams CL. Spatial memory retention is enhanced by acute and continuous estradiol replacement. Horm Behav. 2004;45:128–135. doi: 10.1016/j.yhbeh.2003.09.010. [DOI] [PubMed] [Google Scholar]
  • 114.Wide JK, Hanratty K, Ting J, Galea LA. High level estradiol impairs and low level estradiol facilitates non-spatial working memory. Behav Brain Res. 2004;155:45–53. doi: 10.1016/j.bbr.2004.04.001. [DOI] [PubMed] [Google Scholar]
  • 115.Rodgers SP, Bohacek J, Daniel JM. Transient estradiol exposure during middle age in ovariectomized rats exerts lasting effects on cognitive function and the hippocampus. Endocrinology. 2010;151:1194–1203. doi: 10.1210/en.2009-1245. [DOI] [PubMed] [Google Scholar]
  • 116.Bohacek J, Bearl AM, Daniel JM. Long-term ovarian hormone deprivation alters the ability of subsequent oestradiol replacement to regulate choline acetyltransferase protein levels in the hippocampus and prefrontal cortex of middle-aged rats. J Neuroendocrinol. 2008;20:1023–1027. doi: 10.1111/j.1365-2826.2008.01752.x. [DOI] [PubMed] [Google Scholar]
  • 117.Frye CA, Rhodes ME. Enhancing effects of estrogen on inhibitory avoidance performance may be in part independent of intracellular estrogen receptors in the hippocampus. Brain Res. 2002;956:285–293. doi: 10.1016/s0006-8993(02)03559-x. [DOI] [PubMed] [Google Scholar]
  • 118.Singh M, Meyer EM, Millard WJ, Simpkins JW. Ovarian steroid deprivation results in a reversible learning impairment and compromised cholinergic function in female Sprague-Dawley rats. Brain Res. 1994;644:305–312. doi: 10.1016/0006-8993(94)91694-2. [DOI] [PubMed] [Google Scholar]
  • 119.Foster TC, Sharrow KM, Kumar A, Masse J. Interaction of age and chronic estradiol replacement on memory and markers of brain aging. Neurobiol Aging. 2003;24:839–852. doi: 10.1016/s0197-4580(03)00014-9. [DOI] [PubMed] [Google Scholar]
  • 120.Leuner B, Mendolia-Loffredo S, Shors TJ. High levels of estrogen enhance associative memory formation in ovariectomized females. Psychoneuroendocrinology. 2004;29:883–890. doi: 10.1016/j.psyneuen.2003.08.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 121.Bimonte-Nelson HA, Singleton RS, Williams BJ, Granholm AC. Ovarian hormones and cognition in the aged female rat: II. progesterone supplementation reverses the cognitive enhancing effects of ovariectomy. Behav Neurosci. 2004;118:707–714. doi: 10.1037/0735-7044.118.4.707. [DOI] [PubMed] [Google Scholar]
  • 122.Farr SA, Flood JF, Scherrer JF, Kaiser FE, Taylor GT, Morley JE. Effect of ovarian steroids on footshock avoidance learning and retention in female mice. Physiol Behav. 1995;58:715–723. doi: 10.1016/0031-9384(95)00124-2. [DOI] [PubMed] [Google Scholar]
  • 123.Bitran D, Hilvers RJ, Kellogg CK. Anxiolytic effects of 3 alpha-hydroxy-5 alpha[beta]-pregnan-20-one: endogenous metabolites of progesterone that are active at the GABAA receptor. Brain Res. 1991;561:157–161. doi: 10.1016/0006-8993(91)90761-j. [DOI] [PubMed] [Google Scholar]
  • 124.Bitran D, Hilvers RJ, Kellogg CK. Ovarian endocrine status modulates the anxiolytic potency of diazepam and the efficacy of gamma-aminobutyric acid-benzodiazepine receptor-mediated chloride ion transport. Behav Neurosci. 1991;105:653–662. doi: 10.1037//0735-7044.105.5.653. [DOI] [PubMed] [Google Scholar]
  • 125.Frye CA, Duncan JE. Progesterone metabolites, effective at the GABAA receptor complex, attenuate pain sensitivity in rats. Brain Res. 1994;643:194–203. doi: 10.1016/0006-8993(94)90025-6. [DOI] [PubMed] [Google Scholar]
  • 126.Chesler EJ, Juraska JM. Acute administration of estrogen and progesterone impairs the acquisition of the spatial Morris water maze in ovariectomized rats. Horm Behav. 2000;38:234–242. doi: 10.1006/hbeh.2000.1626. [DOI] [PubMed] [Google Scholar]
  • 127.Harburger LL, Pechenino AS, Saadi A, Frick KM. Post-training progesterone dose-dependently enhances object, but not spatial, memory consolidation. Behav Brain Res. 2008;194:174–180. doi: 10.1016/j.bbr.2008.07.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 128.Sato T, Tanaka K, Ohnishi Y, Teramoto T, Irifune M, Nishikawa T. Effects of estradiol and progesterone on radial maze performance in middle-aged female rats fed a low-calcium diet. Behav Brain Res. 2004;150:33–42. doi: 10.1016/S0166-4328(03)00249-3. [DOI] [PubMed] [Google Scholar]
  • 129.Walf AA, Rhodes ME, Frye CA. Ovarian steroids enhance object recognition in naturally cycling and ovariectomized, hormone-primed rats. Neurobiol Learn Mem. 2006;86:35–46. doi: 10.1016/j.nlm.2006.01.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 130.Frye CA, Lacey EH. Progestins influence performance on cognitive tasks independent of changes in affective behavior. Psychobiology. 2000;28:550–563. [Google Scholar]
  • 131.Lewis MC, Orr PT, Frick KM. Differential effects of acute progesterone administration on spatial and object memory in middle-aged and aged female C57BL/6 mice. Horm Behav. 2008;54:455–462. doi: 10.1016/j.yhbeh.2008.05.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 132.Harburger LL, Bennett JC, Frick KM. Effects of estrogen and progesterone on spatial memory consolidation in aged females. Neurobiol Aging. 2007;28:602–610. doi: 10.1016/j.neurobiolaging.2006.02.019. [DOI] [PubMed] [Google Scholar]
  • 133.Harburger LL, Saadi A, Frick KM. Dose-dependent effects of post-training estradiol plus progesterone treatment on object memory consolidation and hippocampal extracellular signal-regulated kinase activation in young ovariectomized mice. Neuroscience. 2009;160:6–12. doi: 10.1016/j.neuroscience.2009.02.024. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 134.Dodart JC, Mathis C, Ungerer A. Scopolamine-induced deficits in a two-trial object recognition task in mice. Neuroreport. 1997;8:1173–1178. doi: 10.1097/00001756-199703240-00023. [DOI] [PubMed] [Google Scholar]
  • 135.Messier C. Object recognition in mice: improvement of memory by glucose. Neurobiol Learn Mem. 1997;67:172–175. doi: 10.1006/nlme.1996.3755. [DOI] [PubMed] [Google Scholar]
  • 136.Steckler T, Weis C, Sauvage M, Mederer A, Holsboer F. Disrupted allocentric but preserved egocentric spatial learning in transgenic mice with impaired glucocorticoid receptor function. Behav Brain Res. 1999;100:77–89. doi: 10.1016/s0166-4328(98)00115-6. [DOI] [PubMed] [Google Scholar]
  • 137.Wiescholleck V, Manahan-Vaughan D. Persistent deficits in hippocampal synaptic plasticity accompany losses of hippocampus-dependent memory in a rodent model of psychosis. Front Integr Neurosci. 2013;7:12. doi: 10.3389/fnint.2013.00012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 138.Mathiasen JR, DiCamillo A. Novel object recognition in the rat: a facile assay for cognitive function. Chapter 5:Unit 5 59. Curr Protoc Pharmacol. 2010 doi: 10.1002/0471141755.ph0559s49. [DOI] [PubMed] [Google Scholar]
  • 139.Heyward FD, Walton RG, Carle MS, Coleman MA, Garvey WT, Sweatt JD. Adult mice maintained on a high-fat diet exhibit object location memory deficits and reduced hippocampal SIRT1 gene expression. Neurobiol Learn Mem. 2012;98:25–32. doi: 10.1016/j.nlm.2012.04.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 140.Frick KM, Gresack JE. Sex differences in the behavioral response to spatial and object novelty in adult C57BL/6 mice. Behav Neurosci. 2003;117:1283–1291. doi: 10.1037/0735-7044.117.6.1283. [DOI] [PubMed] [Google Scholar]
  • 141.Sutcliffe JS, Marshall KM, Neill JC. Influence of gender on working and spatial memory in the novel object recognition task in the rat. Behav Brain Res. 2007;177:117–125. doi: 10.1016/j.bbr.2006.10.029. [DOI] [PubMed] [Google Scholar]
  • 142.Walf AA, Koonce C, Manley K, Frye CA. Proestrous compared to diestrous wildtype, but not estrogen receptor beta knockout, mice have better performance in the spontaneous alternation and object recognition tasks and reduced anxiety-like behavior in the elevated plus and mirror maze. Behav Brain Res. 2009;196:254–260. doi: 10.1016/j.bbr.2008.09.016. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 143.Paris JJ, Frye CA. Estrous cycle, pregnancy, and parity enhance performance of rats in object recognition or object placement tasks. Reproduction. 2008;136:105–115. doi: 10.1530/REP-07-0512. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 144.Frye CA, Duffy CK, Walf AA. Estrogens and progestins enhance spatial learning of intact and ovariectomized rats in the object placement task. Neurobiol Learn Mem. 2007;88:208–216. doi: 10.1016/j.nlm.2007.04.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 145.Pawluski JL, Brummelte S, Barha CK, Crozier TM, Galea LA. Effects of steroid hormones on neurogenesis in the hippocampus of the adult female rodent during the estrous cycle, pregnancy, lactation and aging. Front Neuroendocrinol. 2009;30:343–357. doi: 10.1016/j.yfrne.2009.03.007. [DOI] [PubMed] [Google Scholar]
  • 146.Kinsley CH, Trainer R, Stafisso-Sandoz G, Quadros P, Marcus LK, Hearon C, et al. Motherhood and the hormones of pregnancy modify concentrations of hippocampal neuronal dendritic spines. Horm Behav. 2006;49:131–142. doi: 10.1016/j.yhbeh.2005.05.017. [DOI] [PubMed] [Google Scholar]
  • 147.Barha CK, Galea LA. Motherhood alters the cellular response to estrogens in the hippocampus later in life. Neurobiol Aging. 2011;32:2091–2095. doi: 10.1016/j.neurobiolaging.2009.12.004. [DOI] [PubMed] [Google Scholar]
  • 148.Pawluski JL, Vanderbyl BL, Ragan K, Galea LA. First reproductive experience persistently affects spatial reference and working memory in the mother and these effects are not due to pregnancy or 'mothering' alone. Behav Brain Res. 2006;175:157–165. doi: 10.1016/j.bbr.2006.08.017. [DOI] [PubMed] [Google Scholar]
  • 149.Wallace M, Luine V, Arellanos A, Frankfurt M. Ovariectomized rats show decreased recognition memory and spine density in the hippocampus and prefrontal cortex. Brain Res. 2006;1126:176–182. doi: 10.1016/j.brainres.2006.07.064. [DOI] [PubMed] [Google Scholar]
  • 150.Fonseca CS, Gusmao ID, Raslan AC, Monteiro BM, Massensini AR, Moraes MF, et al. Object recognition memory and temporal lobe activation after delayed estrogen replacement therapy. Neurobiol Learn Mem. 2013;101:19–25. doi: 10.1016/j.nlm.2012.12.016. [DOI] [PubMed] [Google Scholar]
  • 151.Smith CC, McMahon LL. Estrogen-induced increase in the magnitude of long-term potentiation occurs only when the ratio of NMDA transmission to AMPA transmission is increased. J Neurosci. 2005;25:7780–7791. doi: 10.1523/JNEUROSCI.0762-05.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 152.Smith CC, McMahon LL. Estradiol-induced increase in the magnitude of long-term potentiation is prevented by blocking NR2B-containing receptors. J Neurosci. 2006;26:8517–8522. doi: 10.1523/JNEUROSCI.5279-05.2006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 153.Smith CC, Vedder LC, Nelson AR, Bredemann TM, McMahon LL. Duration of estrogen deprivation, not chronological age, prevents estrogen's ability to enhance hippocampal synaptic physiology. Proc Natl Acad Sci U S A. 2010;107:19543–19548. doi: 10.1073/pnas.1009307107. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 154.Jacome LF, Gautreaux C, Inagaki T, Mohan G, Alves S, Lubbers LS, et al. Estradiol and ERβ agonists enhance recognition memory, and DPN, an ERβ agonist, alters brain monoamines. Neurobiol Learn Mem. 2010;94:488–498. doi: 10.1016/j.nlm.2010.08.016. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 155.Luine VN, Jacome LF, MacLusky NJ. Rapid enhancement of visual and place memory by estrogens in rats. Endocrinology. 2003;144:2836–2844. doi: 10.1210/en.2003-0004. [DOI] [PubMed] [Google Scholar]
  • 156.Vedder LC, Smith CC, Flannigan AE, McMahon LL. Estradiol-induced increase in novel object recognition requires hippocampal NR2B-containing NMDA receptors. Hippocampus. 2013;23:108–115. doi: 10.1002/hipo.22068. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 157.Ismail N, Blaustein JD. Pubertal immune challenge blocks the ability of estradiol to enhance performance on cognitive tasks in adult female mice. Psychoneuroendocrinology. 2013;38:1170–1177. doi: 10.1016/j.psyneuen.2012.11.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 158.McGaugh JL. Dissociating learning and performance: Drug and hormone enhancement of memory storage. Brain Res Bull. 1989;23:339–345. doi: 10.1016/0361-9230(89)90220-7. [DOI] [PubMed] [Google Scholar]
  • 159.Gresack JE, Frick KM. Environmental enrichment reduces the mnemonic and neural benefits of estrogen. Neuroscience. 2004;128:459–471. doi: 10.1016/j.neuroscience.2004.06.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 160.Gresack JE, Frick KM. Post-training estrogen enhances spatial and object memory consolidation in female mice. Pharm Biochem Behav. 2006;84:112–119. doi: 10.1016/j.pbb.2006.04.013. [DOI] [PubMed] [Google Scholar]
  • 161.Gresack JE, Kerr KM, Frick KM. Life-long environmental enrichment differentially affects the mnemonic response to estrogen in young, middle-aged, and aged female mice. Neurobiol Learn Mem. 2007;88:393–408. doi: 10.1016/j.nlm.2007.07.015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 162.Gresack JE, Kerr KM, Frick KM. Short-term environmental enrichment decreases the mnemonic response to estrogen in young, but not aged, female mice. Brain Res. 2007;1160:91–101. doi: 10.1016/j.brainres.2007.05.033. [DOI] [PubMed] [Google Scholar]
  • 163.Walf AA, Koonce CJ, Frye CA. Estradiol or diarylpropionitrile administration to wild type, but not estrogen receptor beta knockout, mice enhances performance in the object recognition and object placement tasks. Neurobiol Learn Mem. 2008;89:513–521. doi: 10.1016/j.nlm.2008.01.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 164.Inagaki T, Gautreaux C, Luine V. Acute estrogen treatment facilitates recognition memory consolidation and alters monoamine levels in memory-related brain areas. Horm Behav. 2010;58:415–426. doi: 10.1016/j.yhbeh.2010.05.013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 165.Frye CA, Koonce CJ, Walf AA. Progesterone, compared to medroxyprogesterone acetate, to C57BL/6, but not 5alpha-reductase mutant, mice enhances object recognition and placement memory and is associated with higher BDNF levels in the hippocampus and cortex. Neurosci Lett. 2013;551:53–57. doi: 10.1016/j.neulet.2013.07.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 166.Frye CA, Llaneza DC, Walf AA. Progesterone can enhance consolidation and/or performance in spatial, object and working memory tasks in Long-Evans rats. Anim Behav. 2009;78:279–286. doi: 10.1016/j.anbehav.2009.04.017. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 167.Frye CA, Walf AA. Progesterone to ovariectomized mice enhances cognitive performance in the spontaneous alternation, object recognition, but not placement, water maze, and contextual and cued conditioned fear tasks. Neurobiol Learn Mem. 2008;90:171–177. doi: 10.1016/j.nlm.2008.03.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 168.Orr PT, Lewis MC, Frick KM. Dorsal hippocampal progesterone infusions enhance object recognition in young female mice. Pharm Biochem Behav. 2009;93:177–182. doi: 10.1016/j.pbb.2009.05.012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 169.Gibbs RB. Effects of gonadal hormone replacement on measures of basal forebrain cholinergic function. Neuroscience. 2000;101:931–938. doi: 10.1016/s0306-4522(00)00433-4. [DOI] [PubMed] [Google Scholar]
  • 170.Gibbs RB. Effects of estrogen on basal forebrain cholinergic neurons vary as a function of dose and duration of treatment. Brain Res. 1997;757:10–16. doi: 10.1016/s0006-8993(96)01432-1. [DOI] [PubMed] [Google Scholar]
  • 171.Gibbs RB. Fluctuations in relative levels of choline acetyltransferase mRNA in different regions of the rat basal forebrain across the estrous cycle: Effects of estrogen and progesterone. J Neurosci. 1996;16:1049–1055. doi: 10.1523/JNEUROSCI.16-03-01049.1996. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 172.Dohanich GP, Witcher JA, Weaver DR, Clemens LG. Alteration of muscarinic binding in specific brain areas following estrogen treatment. Brain Res. 1982;241:347–350. doi: 10.1016/0006-8993(82)91075-7. [DOI] [PubMed] [Google Scholar]
  • 173.Gibbs RB, Hashash A, Johnson DA. Effects of estrogen on potassium-stimulated acetylcholine release in the hippocampus and overlying cortex of adult rats. Brain Res. 1997;749:143–146. doi: 10.1016/s0006-8993(96)01375-3. [DOI] [PubMed] [Google Scholar]
  • 174.Baxter MG, Bucci DJ. Selective immunotoxic lesions of basal forebrain cholinergic neurons: twenty years of research and new directions. Behav Neurosci. 2013;127:611–618. doi: 10.1037/a0033781. [DOI] [PubMed] [Google Scholar]
  • 175.Packard MG, Teather LA. Posttraining estradiol injections enhance memory in ovariectomized rats: Cholinergic blockade and synergism. Neurobiol Learn Mem. 1997;68:172–188. doi: 10.1006/nlme.1997.3785. [DOI] [PubMed] [Google Scholar]
  • 176.Vaucher E, Reymond I, Najaffe R, Kar S, Quirion R, Miller MM, et al. Estrogen effects on object memory and cholinergic receptors in young and old female mice. Neurobiol Aging. 2002;23:87–95. doi: 10.1016/s0197-4580(01)00250-0. [DOI] [PubMed] [Google Scholar]
  • 177.Fortress AM, Frick KM. Epigenetic regulation of estrogen-dependent memory. Front Neuroendocrinol. 2014 doi: 10.1016/j.yfrne.2014.05.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 178.Frick KM, Fernandez SM, Harburger LL. A new approach to understanding the molecular mechanisms through which estrogens affect cognition. Biochim Biophys Acta Gen Subj. 2010;1800:1045–1055. doi: 10.1016/j.bbagen.2009.11.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 179.Kim J, Boulware M, Frick KM. Role of g-protein-coupled estrogen receptor (gper/gpr30) in hippocampal memory and cell signaling in female mice. Soc Neurosci Abstr. 2013 Abstract #376.05. [Google Scholar]
  • 180.Hamilton RT, Rettberg JR, Mao Z, To J, Zhao L, Appt SE, et al. Hippocampal responsiveness to 17β-estradiol and equol after long-term ovariectomy: Implication for a therapeutic window of opportunity. Brain Res. 2011;1379:11–22. doi: 10.1016/j.brainres.2011.01.029. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 181.Rissman EF, Heck AL, Leonard JE, Shupnik MA, Gustafsson JA. Disruption of estrogen receptor beta gene impairs spatial learning in female mice. Proc Natl Acad Sci U S A. 2002;99:3996–4001. doi: 10.1073/pnas.012032699. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 182.Liu F, Day M, Muniz LC, Bitran D, Arias R, Revilla-Sanchez R, et al. Activation of estrogen receptor-beta regulates hippocampal synaptic plasticity and improves memory. Nat Neurosci. 2008;11:334–343. doi: 10.1038/nn2057. [DOI] [PubMed] [Google Scholar]
  • 183.Spencer JL, Waters EM, Romeo RD, Wood GE, Milner TA, McEwen BS. Uncovering the mechanisms of estrogen effects on hippocampal function. Front Neuroendocrinol. 2008;29:219–237. doi: 10.1016/j.yfrne.2007.08.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 184.Frick KM. Epigenetics, oestradiol and hippocampal memory consolidation. J Neuroendocrinol. 2013;25:1151–1162. doi: 10.1111/jne.12106. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 185.Cammalleri M, Lutjens R, Berton F, King AR, Simpson C, Francesconi W, et al. Time-restricted role for dendritic activation of the mTOR-p70S6K pathway in the induction of late-phase long-term potentiation in the CA1. Proc Natl Acad Sci U S A. 2003;100:14368–14373. doi: 10.1073/pnas.2336098100. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 186.Hoeffer CA, Klann E. mTOR signaling: at the crossroads of plasticity, memory and disease. Trends Neurosci. 2010;33:67–75. doi: 10.1016/j.tins.2009.11.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 187.Roberson ED, English JD, Adams JP, Selcher JC, Kondratick C, Sweatt JD. The mitogen-activated protein kinase cascade couples PKA and PKC to cAMP response element binding protein phosphorylation in area CA1 of hippocampus. J Neurosci. 1999;19:4337–4348. doi: 10.1523/JNEUROSCI.19-11-04337.1999. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 188.Selvi BR, Cassel JC, Kundu TK, Boutillier AL. Tuning acetylation levels with HAT activators: therapeutic strategy in neurodegenerative diseases. Biochim Biophys Acta. 2010;1799:840–853. doi: 10.1016/j.bbagrm.2010.08.012. [DOI] [PubMed] [Google Scholar]
  • 189.Levenson JM, O'Riordan KJ, Brown KD, Trinh MA, Molfese DL, Sweatt JD. Regulation of histone acetylation during memory formation in the hippocampus. J Biol Chem. 2004;279:40545–40559. doi: 10.1074/jbc.M402229200. [DOI] [PubMed] [Google Scholar]
  • 190.Luger K, Mäder AW, Richmond RK, Sargent DF, Richmond TJ. Crystal structure of the nucleosome core particle at 2.8 A resolution. Nature. 1997;389:251–260. doi: 10.1038/38444. [DOI] [PubMed] [Google Scholar]
  • 191.Serruya MD, Hatsopoulos NG, Paninski L, Fellows MR, Donoghue JP. Instant neural control of a movement signal. Nature. 2002;416:141–142. doi: 10.1038/416141a. [DOI] [PubMed] [Google Scholar]
  • 192.Stefanko DP, Barrett RM, Ly AR, Reolon GK, Wood MA. Modulation of long-term memory for object recognition via HDAC inhibition. Proc Natl Acad Sci U S A. 2009;106:9447–9452. doi: 10.1073/pnas.0903964106. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 193.Guan JS, Haggarty SJ, Giacometti E, Dannenberg JH, Joseph N, Gao J, et al. HDAC2 negatively regulates memory formation and synaptic plasticity. Nature. 2009;459:55–60. doi: 10.1038/nature07925. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 194.McQuown SC, Barrett RM, Matheos DP, Post RJ, Rogge GA, Alenghat T, et al. HDAC3 is a critical negative regulator of long-term memory formation. J Neurosci. 2011;31:764–747. doi: 10.1523/JNEUROSCI.5052-10.2011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 195.Haettig J, Stefanko DP, Multani ML, Figueroa DX, McQuown SC, Wood MA. HDAC inhibition modulates hippocampus-dependent long-term memory for object location in a CBP-dependent manner. Learn Mem. 2011;18:71–79. doi: 10.1101/lm.1986911. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 196.Fortress AM, Kim J, Poole RL, Gould TJ, Frick KM. Estradiol-induced epigenetic changes and memory consolidation in middle-aged mice. Learn Mem. 2014 doi: 10.1101/lm.034033.113. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 197.Denis H, Ndlovu MN, Fuks F. Regulation of mammalian DNA methyltransferases: a route to new mechanisms. EMBO Rep. 2011;12:647–656. doi: 10.1038/embor.2011.110. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 198.Singh M. Ovarian hormones elicit phosphorylation of Akt and extracellular-signal regulated kinase in explants of the cerebral cortex. Endocrine. 2001;14:407–415. doi: 10.1385/ENDO:14:3:407. [DOI] [PubMed] [Google Scholar]
  • 199.Petralia SM, Frye CA. In the ventral tegmental area, cyclic AMP mediates the actions of progesterone at dopamine type 1 receptors for lordosis of rats and hamsters. J Neuroendocrinol. 2006;18:902–914. doi: 10.1111/j.1365-2826.2006.01488.x. [DOI] [PubMed] [Google Scholar]
  • 200.Fortress AM, Heisler JD, Boulware MI, Frick KM. Intracellular and membrane progesterone receptors facilitate object recognition memory consolidation, but potentially through different molecular mechanisms. Soc Neurosci Abstr. 2012 Abstract #92.17. [Google Scholar]
  • 201.Frick KM, Burlingame LA, Arters JA, Berger-Sweeney J. Reference memory, anxiety, and estrous cyclicity in C57BL/6NIA mice are affected by age and sex. Neuroscience. 2000;95:293–307. doi: 10.1016/s0306-4522(99)00418-2. [DOI] [PubMed] [Google Scholar]
  • 202.Markowska AL. Sex dimorphisms in the rate of age-related decline in spatial memory: Relevance to alterations in the estrous cycle. J Neurosci. 1999;19:8122–8133. doi: 10.1523/JNEUROSCI.19-18-08122.1999. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 203.Jayaraman A, Pike CJ. Differential effects of synthetic progestagens on neuron survival and estrogen neuroprotection in cultured neurons. Mol Cell Endocrinol. 2014;384:52–60. doi: 10.1016/j.mce.2014.01.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 204.Simpkins JW, Singh M. More than a decade of estrogen neuroprotection. Alzheimers Dement. 2008;4:S131–S136. doi: 10.1016/j.jalz.2007.10.009. [DOI] [PubMed] [Google Scholar]
  • 205.Baudry M, Bi X, Aguirre C. Progesterone-estrogen interactions in synaptic plasticity and neuroprotection. Neuroscience. 2013;239:280–294. doi: 10.1016/j.neuroscience.2012.10.051. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 206.Singh M, Su C. Progesterone-induced neuroprotection: factors that may predict therapeutic efficacy. Brain Res. 2013;1514:98–106. doi: 10.1016/j.brainres.2013.01.027. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 207.Decker MW, Gallagher M. Scopolamine-disruption of radial arm maze performance: Modification by noradrenergic depletion. Brain Res. 1987;417:59–69. doi: 10.1016/0006-8993(87)90179-x. [DOI] [PubMed] [Google Scholar]
  • 208.Erickson CA, Barnes CA. The neurobiology of memory changes in normal aging. Exp Gerontol. 2003;38:61–69. doi: 10.1016/s0531-5565(02)00160-2. [DOI] [PubMed] [Google Scholar]
  • 209.Rosenzweig ES, Barnes CA. Impact of aging on hippocampal function: Plasticity, network dynamics, and cognition. Prog Neurobiol. 2003;69:143–179. doi: 10.1016/s0301-0082(02)00126-0. [DOI] [PubMed] [Google Scholar]
  • 210.Paris JJ, Walf AA, Frye CA. II. Cognitive performance of middle-aged female rats is influenced by capacity to metabolize progesterone in the prefrontal cortex and hippocampus. Brain Res. 2011 doi: 10.1016/j.brainres.2010.10.099. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 211.Fernandez SM, Frick KM. Chronic oral estrogen affects memory and neurochemistry in middle-aged female mice. Behav Neurosci. 2004;118:1340–1351. doi: 10.1037/0735-7044.118.6.1340. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 212.Walf AA, Paris JJ, Frye CA. Chronic estradiol replacement to aged female rats reduces anxiety-like and depression-like behavior and enhances cognitive performance. Psychoneuroendocrinology. 2009;34:909–916. doi: 10.1016/j.psyneuen.2009.01.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 213.Walf AA, Frye CA. Conjugated equine estrogen enhances rats' cognitive, anxiety, and social behavior. Neuroreport. 2008;19:789–792. doi: 10.1097/WNR.0b013e3282fe209c. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 214.Gresack JE, Frick KM. Effects of continuous and intermittent estrogen treatments on memory in aging female mice. Brain Res. 2006;1115:135–147. doi: 10.1016/j.brainres.2006.07.067. [DOI] [PubMed] [Google Scholar]
  • 215.Savonenko AV, Markowska AL. The cognitive effects of ovariectomy and estrogen replacement are modulated by aging. Neuroscience. 2003;119:821–830. doi: 10.1016/s0306-4522(03)00213-6. [DOI] [PubMed] [Google Scholar]
  • 216.Talboom JS, Williams BJ, Baxley ER, West SG, Bimonte-Nelson HA. Higher levels of estradiol replacement correlate with better spatial memory in surgically menopausal young and middle-aged rats. Neurobiol Learn Mem. 2008;90:155–163. doi: 10.1016/j.nlm.2008.04.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 217.Frick KM, Fernandez SM, Bulinski SC. Estrogen replacement improves spatial reference memory and increases hippocampal synaptophysin in aged female mice. Neuroscience. 2002;115:547–558. doi: 10.1016/s0306-4522(02)00377-9. [DOI] [PubMed] [Google Scholar]
  • 218.Yamaguchi-Shima N, Yuri K. Age-related changes in the expression of ER-β mRNA in the female rat brain. Brain Res. 2007;1155:34–41. doi: 10.1016/j.brainres.2007.04.016. [DOI] [PubMed] [Google Scholar]
  • 219.Bohacek J, Daniel JM. The ability of oestradiol administration to regulate protein levels of oestrogen receptor alpha in the hippocampus and prefrontal cortex of middle-aged rats is altered following long-term ovarian hormone deprivation. Journal of Neuroendocrinol. 2009;21:640–647. doi: 10.1111/j.1365-2826.2009.01882.x. [DOI] [PubMed] [Google Scholar]
  • 220.Zhang QG, Han D, Wang RM, Dong Y, Yang F, Vadlamudi RK, et al. C terminus of Hsc70-interacting protein (CHIP)-mediated degradation of hippocampal estrogen receptor-alpha and the critical period hypothesis of estrogen neuroprotection. Proc Natl Acad Sci U S A. 2011;108:E617–E624. doi: 10.1073/pnas.1104391108. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 221.Daniel JM. Estrogens, estrogen receptors, and female cognitive aging: the impact of timing. Horm Behav. 2013;63:231–237. doi: 10.1016/j.yhbeh.2012.05.003. [DOI] [PubMed] [Google Scholar]
  • 222.Maki PM. Hormone therapy and cognitive function: is there a critical period for benefit? Neuroscience. 2006;138:1027–1030. doi: 10.1016/j.neuroscience.2006.01.001. [DOI] [PubMed] [Google Scholar]
  • 223.Sherwin BB. Estrogen and cognitive aging in women. Neuroscience. 2006;138:1021–1026. doi: 10.1016/j.neuroscience.2005.07.051. [DOI] [PubMed] [Google Scholar]
  • 224.Zandi PP, Carlson MC, Plassman BL, Welsh-Bohmer KA, Mayer LS, Steffens DC, et al. Hormone replacement therapy and incidence of Alzheimer disease in older women. J Am Med Assoc. 2002;288:2123–2129. doi: 10.1001/jama.288.17.2123. [DOI] [PubMed] [Google Scholar]
  • 225.Frye CA, Walf AA. Progesterone enhances learning and memory of aged wildtype and progestin receptor knockout mice. Neurosci Lett. 2010;472:38–42. doi: 10.1016/j.neulet.2010.01.051. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 226.Frye CA, Walf AA. Progesterone enhances performance of aged mice in cortical or hippocampal tasks. Neurosci Lett. 2008;437:116–120. doi: 10.1016/j.neulet.2008.04.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 227.Kessler RC, Chiu WT, Demler O, Merikangas KR, Walters EE. Prevalence, severity, and comorbidity of 12-month DSM-IV disorders in the National Comorbidity Survey Replication. Arch Gen Psychiatry. 2005;62:617–627. doi: 10.1001/archpsyc.62.6.617. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 228.Meinhard N, Kessing LV, Vinberg M. The role of estrogen in bipolar disorder, a review. Nord J Psychiatry. 2014;68:81–87. doi: 10.3109/08039488.2013.775341. [DOI] [PubMed] [Google Scholar]
  • 229.Milad MR, Zeidan MA, Contero A, Pitman RK, Klibanski A, Rauch SL, et al. The influence of gonadal hormones on conditioned fear extinction in healthy humans. Neuroscience. 2010;168:652–658. doi: 10.1016/j.neuroscience.2010.04.030. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 230.Graham BM, Milad MR. Blockade of estrogen by hormonal contraceptives impairs fear extinction in female rats and women. Biol Psychiatry. 2013;73:371–378. doi: 10.1016/j.biopsych.2012.09.018. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 231.Gervais NJ, Jacob S, Brake WG, Mumby DG. Systemic and intra-rhinal-cortical 17-beta estradiol administration modulate object-recognition memory in ovariectomized female rats. Horm Behav. 2013;64:642–652. doi: 10.1016/j.yhbeh.2013.08.010. [DOI] [PubMed] [Google Scholar]
  • 232.Naftolin F, Ryan KJ, Davies IJ, Reddy VV, Flores F, Petro Z, et al. The formation of estrogens by central neuroendocrine tissues. Recent Prog Horm Res. 1975;31:295–319. doi: 10.1016/b978-0-12-571131-9.50012-8. [DOI] [PubMed] [Google Scholar]
  • 233.Yaffe K, Barnes D, Lindquist K, Cauley J, Simonsick EM, Penninx B, et al. Endogenous sex hormone levels and risk of cognitive decline in an older biracial cohort. Neurobiol Aging. 2007;28:171–178. doi: 10.1016/j.neurobiolaging.2006.10.004. [DOI] [PubMed] [Google Scholar]
  • 234.Cost KT, Williams-Yee ZN, Fustok JN, Dohanich GP. Sex differences in object-in-place memory of adult rats. Behav Neurosci. 2012;126:457–464. doi: 10.1037/a0028363. [DOI] [PubMed] [Google Scholar]
  • 235.Bettis T, Jacobs LF. Sex differences in object recognition are modulated by object similarity. Behav Brain Res. 2012;233:288–292. doi: 10.1016/j.bbr.2012.04.028. [DOI] [PubMed] [Google Scholar]
  • 236.Frye CA, Walf AA. Effects of progesterone administration and APP-swe+ PSEN1Deltae9 mutation for cognitive performance of mid-aged mice. Neurobiol Learn Mem. 2008;89:17–26. doi: 10.1016/j.nlm.2007.09.008. [DOI] [PubMed] [Google Scholar]

RESOURCES