ArticlePDF AvailableLiterature Review

Abstract and Figures

Although hormones such as glucocorticoids have been broadly accepted in recent decades as general neuromodulators of memory processes, sex steroid hormones such as the potent oestrogen 17β-oestradiol have been less well recognized by the scientific community in this capacity. The predominance of females in studies of oestradiol and memory and the general (but erroneous) perception that oestrogens are ‘female’ hormones have probably prevented oestradiol from being more widely considered as a key memory modulator in both sexes. Indeed, although considerable evidence supports a crucial role for oestradiol in regulating learning and memory in females, a growing body of literature indicates a similar role in males. This Review discusses the mechanisms of oestradiol signalling and provides an overview of the effects of oestradiol on spatial, object recognition, social and fear memories. Although the primary focus is on data collected in females, effects of oestradiol on memory in males will be discussed, as will sex differences in the molecular mechanisms that regulate oestrogenic modulation of memory, which may have important implications for the development of future cognitive therapeutics. Sex steroid hormones such as the potent oestrogen 17β-oestradiol have only recently started to be acknowledged as important neuromodulators. Taxier, Gross and Frick review 17β-oestradiol signalling in the brain and its effects on different types of memory.
Content may be subject to copyright.
Oestrogens, androgens and progestogens are most
commonly associated with their roles in reproduction,
despite their involvement in a broad range of physio-
logical and neural functions. One particular realm in
which these sex steroid hormones exert wide-ranging
effects is that of learning and memory. In particular,
considerable effort has been devoted to understanding
how 17β-oestradiol (E2), the most potent and prevalent
circulating oestrogen, regulates the function of the hip-
pocampus, a bilateral medial temporal lobe structure
with extensive connections to numerous cortical and
subcortical brain regions. Although the hippocampus is
not the only brain region important for memory, damage
to or dysfunction of this structure leads to deficits in the
formation and retention of numerous types of memories,
including those used for spatial navigation, recognizing
objects and conspecifics, and recalling fear-associated
contexts. However, the roles of sex steroids in regulat-
ing the function of mnemonic brain regions such as the
hippocampus have rarely been considered by scientists
other than neuroendocrinologists.
In the early 1990s, a series of articles describing the
effects of oestrogens on neuronal morphology and
plasticity in the hippocampus were paradigm shifting
for the field of neuroendocrinology15. These publi-
cations demonstrated that dendritic spine density on
CA1 pyramidal neurons in the hippocampus fluctuates
across the reproductive (oestrous) cycle of female rats,
and is increased in bilaterally ovariectomized female rats
by exogenous E2 treatment3. Other contemporaneous
work showed that E2 potentiates hippocampal excit-
atory synaptic plasticity57 and protects hippocampal
neurons from drug-induced excitoxicity8. Collectively,
this body of work unlocked a new research frontier that
has broadened understanding of how E2 regulates hip-
pocampal function to modulate memory. Since their
publication, these findings have inspired much research
examining the actions of E2 and other sex steroid hor-
mones in non-reproductive, procognitive brain regions,
including the hippocampus and prefrontal cortex (PFC).
A subset of this newer research has unveiled very rapid
actions of E2 on molecular and cellular mechanisms in
the hippocampus, PFC, amygdala and other regions that
facilitate memory-consolidation processes.
Although the myriad effects of oestrogens such as
E2 on hippocampal function have been intensely studied
by neuroendocrinologists for nearly three decades, oes-
trogens have yet to be widely accepted in neuroscience
as general neuromodulators of cognitive processes. As
oestrogens exert numerous effects in multiple cognitive
brain areas in both sexes913, elucidating how these hor-
mones regulate memory could result in better mental
health outcomes in people of both sexes by identifying
potential therapeutic targets for neurodegenerative dis-
eases, mood disorders and other conditions in which
memory dysfunction features prominently. Effective
treatments for memory loss are sorely lacking, highlight-
ing an urgent need to characterize the neural mecha-
nisms underlying memory formation, including those
mediated by oestrogens. Moreover, such information
Retention
Storage of acquired and
consolidated information that
enables subsequent recall or
retrieval of the information.
Ovariectomized
Ovariectomy involves surgical
removal of the ovaries to
eliminate ovarian hormone
cycling. Subjects that have
undergone ovariectomy are
considered ovariectomized.
Consolidation
Process through which learned
information is encoded and
stored to form a memory that
can be recalled at a later time.
Oestradiol as a neuromodulator
of learning and memory
LisaR.Taxier , KellieS.Gross and KarynM.Frick ✉
Abstract | Although hormones such as glucocorticoids have been broadly accepted in recent
decades as general neuromodulators of memory processes, sex steroid hormones such as the
potent oestrogen 17β-oestradiol have been less well recognized by the scientific community in
this capacity. The predominance of females in studies of oestradiol and memory and the general
(but erroneous) perception that oestrogens are ‘female’ hormones have probably prevented
oestradiol from being more widely considered as a key memory modulator in both sexes. Indeed,
although considerable evidence supports a crucial role for oestradiol in regulating learning and
memory in females, a growing body of literature indicates a similar role in males. This Review
discusses the mechanisms of oestradiol signalling and provides an overview of the effects of
oestradiol on spatial, object recognition, social and fear memories. Although the primary focus
is on data collected in females, effects of oestradiol on memory in males will be discussed, as will
sex differences in the molecular mechanisms that regulate oestrogenic modulation of memory,
which may have important implications for the development of future cognitive therapeutics.
Department of Psychology,
University of Wisconsin-
Milwaukee, Milwaukee,
WI, USA.
e-mail: frickk@uwm.edu
https://doi.org/10.1038/
s41583-020-0362-7
REVIEWS
NATURE REVIEWS
|
NEUROSCIENCE
will greatly inform our general understanding of how
memories are formed. Thus, more broadly considering
oestrogens and related sex steroid hormones as impor-
tant neuromodulators of memory formation will provide
fundamental insights into the neurobiology of memory
and new avenues for drug development.
The goal of this Review is to highlight the impor-
tance of E2 as a critical modulator of synaptic plas-
ticity and memory circuitry. Although other sex
steroid hormones can regulate memory, the influence of
E2 on learning and me mory is the most extensive ly doc-
umented, and thus E2 is the focus of this Review. The
discussion will centre on the effects of E2 in rats and
mice because their size and short lifespans make them
convenient mammalian systems in which to explore
molecular mechanisms of E2 action in the brain, and
thus the preponderance of data are from these species.
However, non-human primate studies (reviewed in1113)
have also provided insights into oestrogenic regulation
of memory and neuronal morphology that corroborate
findings from rodent studies, highlighting the trans-
lational potential of this work. Here we first outline
molecular mechanisms underlying E2 signalling in the
rodent brain, including both classical and non-classical
signalling. Next, we describe several forms of learning
and memory modulated by E2 in rodent s, and the mech-
anisms thus far identified that underlie this mediation.
Although findings from female rodents are the primary
focus, data from males are also discussed, as are sex
differences in the neural mechanisms through which
E2 influences memory. Last, we conclude by discuss-
ing the broad health implications of considering E2 as
a neuromodulator, and speculate about the next steps
necessary to further advance knowledge of oestro-
genic regulation of memory. We hope readers will be
convinced of the importance of E2 as a general neuro-
modulator that, like stress hormones and growth fac-
tors, should be considered in mainstream models of the
neurobiological mechanisms underlying memory.
E2 signalling
Unlike neurotransmitters, which are stored in vesicles
after synthesis for later release, steroid hormones are
generated in response to a stimulus and released imme-
diately. Sex steroid biosynthesis begins with the catabo-
lism of cholesterol to progesterone, which is then broken
down into the androgens testosterone and androstenedi-
one, which are subsequently converted to the oestrogens
E2 and oestrone, respectively14,15. In this synthesis path-
way, E2 is generated from testosterone via the enzyme
aromatase. The primary sources of sex steroids in both
sexes are the gonads, which synthesize progestogens,
androgens and oestrogens in differing sex-dependent
amounts (with more androgens in males, and more
progestogens and oestrogens in females). Hormones
synthesized in the gonads can exert paracrine effects
on adjacent cells in the gonads, or endocrine effects on
distant tissues via the bloodstream16. However, other tis-
sues synthesize sex steroid hormones as well14, including
adipose tissue, the adrenal glands and numerous brain
areas, including the hippocampus1619 (BOX1). In the
brain, steroids including E2 exert rapid paracrine or
synaptocrine effects on neighbouring cells20,21.
In female mammals, circulating E2 is synthesized
by the ovaries, which release fluctuating levels into the
bloodstream as part of the reproductive cycle. During
the human menstrual cycle and rodent oestrous cycle,
oestrogen levels rise to promote follicle maturation in
advance of ovulation, peak to stimulate ovulation and
then return to the baseline as the degenerating folli-
cle secretes progesterone in preparation for implanta-
tion of a fertilized egg. In mice and rats, the oestrous
cycle consists of four approximately 12–24-h-long
stages: proestrus, oestrus, metoestrus and dioestrus22,23.
E2 levels surge during proestrus and remain high
through early oestrus, after which they plunge in late
oestrus and remain low throughout metoestrus, ris-
ing again during late dioestrus. These fluctuations
are reflected in the hippocampus, where E2 levels are
substantially higher during proestrus than during
other phases24, as is CA1 dendritic spine density2 and
neurogenesis25. Hippocampal E2 levels in ovariectomized
rats are similar to those of ovary-intact rats in dioestrus
and metoestrus24, reflecting hippocampal E2 synthesis in
the absence of the ovaries. As discussed in BOX1, denovo
hippocampal E2 synthesi s is activ ity depen dent and cr u-
cial for rapid synaptic plasticity and memory formation.
Oestrogen receptor (ER) expression is also influenced by
the oestrous cycle26.
Box 1 | A role for hippocampally synthesized oestrogens in memory
Although the gonads are a primary source of oestrogens in both sexes, oestrogens are
synthesized in numerous tissues, including the brain. Of most relevance to learning and
memory, the enzyme aromatase, which converts testosterone into 17β-oestradiol (E2),
is widely expressed in the brain and has been shown to produce E2 locall y in region s
such as the hypothalamus and hippocampus17,18,200,237,238. In the hippocampus,
neuron-derived denovo E2 supports multiple aspects of synaptic plasticity, including
synaptogenesis and long-term potentiation19,204,239,240. In hippocampal cultures from
female rats, pharmacologically blocking denovo E2 synthesi s with th e aromatas e inhibi tor
letrozole results in reduced spine density, decreased expression of synaptic proteins and
impaired long-term potentiation19,204,239.
Neural E2 synthesis seems to be regulated by neuronal activity. For example,
activation of NMDA receptors in cultured hippocampal neurons or in hippocampal
slices increased E2 synthesi s200,203, and exposure to a learning stimulus in ovariectomized
mice increased denovo E2 synthe sis in th e hippoca mpus, an e ffect blo cked by l etrozole226.
Invivo, systemic letrozole treatment decreased CA1 dendritic spine density and levels
of hippocampal synaptic proteins in both ovary-intact and ovariectomized females240,
demonstrating that neuron-derived E2 contributes to hippocampal synaptic plasticity
regardle ss of other so urces of the hor mone.
The central importance of denovo hippocampal E2 synthesis in both sexes is
illustrated by studies in which letrozole infusion into the dorsal hippocampus of
gonadectomized male or female mice impaired consolidation of spatial and object
recogni tion memory215,226. Similarly, hippocampal implants of the aromatase inhibitor
1,4,6-androstatriene-3,17-dione impaired spatial memory in male zebra finches241,
suggesting that the importance of hippocampal E2 synthesis is conserved across
species. Supporting this conclusion are recent data showing oral letrozole treatment
impaired spatial working memory and reduced hippocampal intrinsic excitability
in male and female marmosets228.
Aromatase outside the hippocampus also seems to be important for memory
consolidation, as infusion of letrozole into the perirhinal cortex of gonad-intact male
mice impaired short-term and long-term memory in an object placement memory task224.
In humans, letrozole is an approved treatment for oestrogen receptor (ER)-positive breast
cancer; however, women taking letrozole may experience a number of adverse side
effects that affect memory242. For example, women taking letrozole for ER-positive breast
cancer exhibited episodic memory deficits, supporting the idea that brain-synthesized
oestrogens are crucial in memory processes227.
www.nature.com/nrn
REVIEWS
Classical and non-classical signalling
Early research demonstrated that sex steroid recep-
tors act as transcription factors, dimerizing and
translocating to the nucleus on ligand activation to
bind hormone response elements and regulate gene
transcription27. E2 binds two ER subtypes, ERα and ERβ,
to accomplish this ‘classical’ mechanism of steroid hor-
mone action, resulting in slow transcriptional changes
that become evident within hours to days28. However,
data from the 1960s revealed that E2 could increase
cellular concentrations of cyclic AMP (cAMP) in uter-
ine tissue within seconds29, a time course too rapid for
classical genomic signalling. Similarly rapid effects on
the order of seconds to minutes were later found in the
brain, where E2 altered neuronal activity in the preoptic
area of the hypothalamus30. More recent studies have
characterized myriad ‘non-classical, membrane-initiated
mechanisms through which oestrogen rapidly triggers
cell signalling and epigenetic processes to produce
downstream alterations in gene expression, local protein
synthesis, actin polymerization, synaptic physiology and
dendritic spine morphology3135.
Non-classical oestrogen signalling is initiated at
the cell membrane and influences cellular processes
through the activation of downstream second messen-
ger pathways. The identities of the ERs responsible for
this signalling have been controversial, as the early char-
acterization of ERα and ERβ as purely nuclear recep-
tors seemed to exclude a role in membrane-initiated
signalling. However, technological advances have since
permitted observation of these receptors at the cell
membrane3640, and studies using overexpression or dele-
tion of ERα and ERβ demonstrated that these classical
receptors are indeed responsible for many of the defin-
ing effects of rapid E2 action, including increased cAMP
response-element binding protein (CREB) phospho-
rylation and activation of extracellular signal-regulated
kinase (ERK)–mitogen-activated protein kinase
(MAPK) signalling38,41,42. Evidence of the classical ERs
(that is, ERα and ERβ) in dendritic spines and axon ter-
minals of neurons4345 suggests that these receptors could
be directly involved in synaptic signalling.
How classical ERs initiate non-classical oestrogen
signalling is still not thoroughly understood. The field
continues to struggle with technical limitations, such as
insufficient antibody validation
46
, in studying the subcellu-
lar localization of ERs, and many questions remain about
the nature of the interaction between classical ERs and the
cell membrane. The ERα and ERβ structures do not con-
tain prototypical transmembrane domains
47,48
, indicating
that these receptors are probably not fully inserted into
the membrane. However, ERs undergo post-translational
lipidation
4951
and associate with caveolins
52,53
. Both of
these factors contribute to the ability of ERs to localize to
and associate with the cell membrane
50,5355
.
In addition, how ERα and ERβ, which do not have
inherent kinase activity, can interact with and activate
downstream second messengers is unclear. One expla-
nation is that ERs localized to the membrane co-opt
the signalling machinery of other membrane receptors.
At the membrane, ERα and ERβ can physically inter-
act with and activate metabotropic glutamate receptors
(mGluRs) independently of glutamate release, resulting
in rapid ERK and CREB phosphorylation56,57. Functional
coupling of ERs to mGluRs is observed throughout the
nervous system and seems to contribute to the effects
of E2 on memory consolidation, motivated behaviour
and sexual receptivity5759. ERs also interact with receptor
tyrosine kinases at the membrane, activating signalling
at insulin-like growth factor receptor 1 and the neuro-
trophin receptor TRKB through both rapid and gene
expression-dependent mechanisms6063. Activation of
these receptors subsequently engages neuroprotec-
tive and plasticity-related signalling cascades6266. The
integration of ERs into a signalling complex with other
receptors, G proteins and kinases at the cell membrane
is facilitated by multiple scaffold and adaptor proteins.
These include: caveolins, which facilitate ER interaction
with mGluRs as well as membrane association50,53,55;
p130CAS, which couples ERs to kinases such as SRC
and phosphoinositide 3-kinase (PI3K)67; and striatin,
which binds caveolins and G proteins at the membrane68.
Non-classical E2 signalling also occurs through
recently identified membrane-localized ERs. Although
multiple putative ERs exist, the most well-defined and
most studied is G protein-coupled ER (GPER; also
known as GPR30). Characterized in the early years of
the first decade of the twenty-first century, GPER is
a seven-transmembrane-domain G protein-coupled
receptor activated by oestrogens69,70. It most frequently
interacts with Gαs subunits, leading to increased levels
of cAMP and activation of the ERK, protein kinase A
(PKA) and PI3K signalling pathways6972. GPER is highly
expressed in the brain, including in forebrain regions
such as the cortex, hippocampus and hypothalamus73,74,
and pharmacological targeting of GPER has implicated
it in mediating oestrogen effects on cognitive7578, social79
and reproductive80,81 behaviours. Given the considerable
overlap of GPER and classical ER expression in regions
such as the hippocampus and PFC, as well as similar-
ities in the outcomes of their activation, an intriguing
question is whether these ER subtypes use parallel or
distinct mechanisms to modulate neuronal function and
behaviour. Current evidence suggests that the answer to
this question is complex, with classical ERs and GPER
often producing similar or complementary outcomes but
through distinct molecular mechanisms78,82.
Intracellular cascades
E2 activation of membrane-localized receptors triggers
numerous intracellular signalling events that ultimately
influence synaptic plasticity in cognitive brain regions.
These downstream cellular and molecular mechanisms
have been best elucidated in the hippocampus (FIG.1).
Activation of membrane-localized ERs in cultured hip-
pocampal neurons leads to rapid calcium influx83,84 and
activation of phospholipase C and adenylyl cyclase sig-
nalling via G protein activation56,85. In turn, these events
activate small GTPases such as RHOA35,82 and a host of
kinase pathways, including the ERK–MAPK56,84,8689,
PI3K–AKT62,8992, PKA9395, protein kinase C (PKC)56,96
and JUN amino-terminal kinase (JNK)78 pathways. The
activation of these second messenger cascades influences
processes that regulate synaptic structure and function,
Hormone response
elements
Short DNA sequence within
the promoter region of a
gene that binds a hormone
receptor complex to enable
gene transcription.
Caveolins
Integral membrane proteins
that form functional
microdomains of receptors
and their associated signalling
proteins at the plasma
membrane.
Sexual receptivity
A positive state of responsivity
towards the initiation of
sexual behaviour by another
individual. Often indicated
by a species-specific
mating posture.
NATURE REVIEWS
|
NEUROSCIENCE
REVIEWS
such as local protein synthesis, actin polymerization, ion
channel dynamics and gene expression.
E2 activation of ERK and AKT stimulates local protein
translation by activating mechanistic target of rapamycin
(mTOR) signalling and promoting the phosphorylation
of translational initiation factors such as 4E-binding
protein 1 (4E-BP1)32,82,97,98, and this new protein syn-
thesis is required for E2-induced spinogenesis in the
hippocampus99. Recruitment of actin-remodelling path-
ways also contributes to E2-induced structural plasticity:
E2 activation of RHOA elicits phosphorylation of LIM
domain kinase (LIMK) and its substrate, the actin-binding
protein cofilin, to promote actin polymerization35,100103.
LIMK–cofilin activation and enhanced actin polym-
erization contribute to E2 effects on spine structure
and synaptic transmission35,100,103. Moreover, although
membrane-initiated E2 signalling is often referred to as
‘non-genomic’ to differentiate it from the classical effects
of E2, the intracellular signalling cascades initiated by sur-
face ERs ultimately influence gene expression. E2 activa-
tion of ERK signalling rapidly induces phosphorylation of
the transcription factor CREB56,83,88,104 and produces epi-
genetic modifications such as histone acetylation31,105,106
both effects that can increase expression of genes with
neurotrophic effects, such as Bdnf106.
Synaptic function
E2-induced changes in intracellular signalling ulti-
mately alter synaptic function. In the hippocampus,
the general result of E2 exposure is a rapid increase in
excitatory neurotransmission. Application of E2 to hip-
pocampal slices increases intrinsic excitability of CA1
pyramidal neurons107,108, enhances baseline excitatory
neurotransmission5,6,35,109111 and enhances long-term
potentiation (LTP) at CA3–CA1 synapses35,109,111114.
The effects of E2 on synaptic function correlate with
increases in dendritic spine density5,110,113,114 and res ult pr i-
marily from regulation of ionotropic glutamate receptors.
In hippocampal neurons, E2 in fluences the phosphor yla-
tion and subcellular trafficking of both AMPA receptors
(AMPARs) and NMDA receptors (NMDARs)112,115120.
AMPAR-mediated excitatory postsynaptic potentials
can be positively modulated by E2 (REFS6,35); however,
many of the effects of E2 on synaptic plasticity rely on
NMDAR-dependent mechanisms5,109,110,120,121. In particu-
lar, NMDARs containing the NR2B subunit are required
for E2 enhancement of LTP120,121. E2 can also increase
hippocampal excitability by suppressing inhibitory
GABAergic signalling; however, this mechanism differs
by sex122,123.
In sum, considerable data support the view that the
rapid cell-signalling mechanisms resulting from acti-
vation of surface-localized ERs enhance the multiple
forms of synaptic plasticity thought to be cellular sub-
strates for learning and memory. As we discuss in the
next section, the activation of intracellular signalling and
the resulting effects on neuronal structure and function
are necessary for the beneficial effects of E2 on memory
in female rodents.
E2 modulation of memory in females
The numerous classical and non-classical effects of E2 on
brain function provide abundant opportunities for this
hormone to influence learning and memory. Copious
data supporting a modulatory influence of E2 on hip-
pocampal plasticity have led most rodent researchers
to focus on forms of learning and memory mediated
by the hippocampus, such as spatial and object-based
memories. However, oestrogens also regulate other
types of learning and memory, including social learn-
ing, social discrimination and fear memory mediated by
the amygdala, perirhinal cortex, PFC and other regions.
Thus, the following sections will focus not only on the
hippocampus but also on these brain regions as well.
Studies of ER-null mice
114,124126
or using viral vectors to
modulate ERα expression
127
have suggested key roles for
ERα and ERβ in various forms of memory. However, phar-
macological manipulations are the most common method
of interrogating the roles of E
2
and ERs in memory pro-
cesses (TABLE1). Although the compounds used afford
better temporal specificity than genetic manipulations,
they lack absolute specificity for one ER over the other.
However, most can be used at dos es that promote prefer-
ential binding to a single ER. Ongoing synthesis of more
potent and selective ER compounds will undoubtedly
help pinpoint discrete functions of specific ERs
128
.
Most studies examining the neuromodulatory role
of E2 have been conducted in bilaterally ovariectomized
Local protein synthesis
mTOR activation
p4E-BP1 Kinase activity
MEK–ERK
PI3K–AKT
PKA
PKC
JNK (GPER only)
Gene expression
pCREB
Histone acetylation
DNA methylation
Cytoskeletal regulation
pLIMK–pcofilin
Actin polymerization
Dendritic spine density
Ion channel
dynamics
Phosphorylation
Trafficking to
synapses
Local protein synthesis
mTOR activation
p4E-BP1
ER
E2
E2
AMPAR or
NMDAR
mGluR
RTK
GPER
Fig. 1 | Membrane-initiated oestrogen signalling and downstream intracellular
events. I ntracellu lar proce sses are in itiated b y 17β-oestradiol (E2) binding to G protein-
coupled oestrogen receptor (GPER), or functional interaction between the canonical
oestrogen receptors (ERα and ER β) and other receptors located at the membrane (such as
metabotropic glutamate receptors (mGluRs)). Several kinase cascades, key for the memory-
enhancing effects of E2, rapidly increase activity in response to membrane-initiated
signalling events. ERα and ERβ seem to trigger similar kinases, whereas GPER activates
distinct signalling pathways such as the JUN amino-terminal kinase (JNK) pathway. In turn,
kinase activity facilitates additional regulatory processes, including protein synthesis,
ion channel phosphorylation and trafficking, gene expression and cytoskeletal regulation.
AMPAR, AMPA receptor; ERK, extracellular signal-regulated kinase; MEK, mitogen-
activated protein kinase/extracellular signal-regulated kinase; mTOR, mechanistic
target of rapamycin; NMDAR, NMDA receptor; p4E-BP1, phosphorylated 4E-binding
protein 1; pCREB, phosphorylated cyclic AMP response element-binding protein;
PI3K, phosphoinositide 3-kinase; PKA, protein kinase A; PKC, protein kinase C; pLIMK,
phosphorylated LIM domain kinase; RTK, receptor tyrosine kinase.
www.nature.com/nrn
REVIEWS
female rodents, although some reports have examined
memory in naturally cycling females or in males (with or
without gonadectomy). Although the widespread use of
ovariectomized females has limitations that require cau-
tious considerations about extrapolating effects to the
natural cycle, this model has permitted more systematic
investigations of oestrogen actions than is possible in the
presence of daily ovarian hormone fluctuations. Much of
this work has used young rodents (2–3 months old), but
a rich literature also exists on the mnemonic effects of
E2 in middle-aged rodents (for example, 14–19 months
old) and aged rodents (older than 20 months)129,130
(BOX2). Besides animal age, there are several other
experimental variables that influence behavioural and
mechanistic outcomes. The timing of administration
relative to testing, the length of administration (for
example, long (that is, over several days) or short (that
is, a single dose)) and the dose can differ across studies,
making direct comparisons across studies challenging.
Moreover, when E2 is administered systemically, the site
of E2 action is often unknown. Indeed, given that E2 is
also synthesized in the brain (BOX1), differentiating the
effects of brain-derived E2 from systemically adminis-
tered E2, or from gonadal E2 in ovary-intact rodents,
poses a unique challenge. In addition, the age at ovariec-
tomy and the duration between ovariectomy and first
E2 treatment have also been demonstrated to affect oes-
trogenic regulation of memory129,130. The age at testing
and the duration of oestrogen deprivation are particu-
larly important, as ER expression in the rodent hip-
pocampus declines not only with age but also several
months after bilateral ovariectomy131,132. Combined,
these data highlight the importance of experimental
parameters when one is designing studies or interpreting
data. The following sections mirror the preponderance
of studies using young ovariectomized rodents (but see
BOX2 and reviews129,133135 for additional information
relevant to ageing female and male rodents). As a com-
prehensive synthesis of all types of memory modulated
by E2 is beyond the scope of this Review, the following
text focuses on select forms of memory and behavioural
tasks that have proved especially useful to pinpoint the
neural mechanisms underlying E2-facilitated memory.
Spatial and object memory
The tasks most commonly used to assess hippocampal-
dependent spatial and object memory include the Morris
water maze (MWM), the radial arm maze (RAM), and
object recognition (OR) and object placement (OP; also
known as object location) tasks (FIG.2). In the MWM and
RAM, animals use extramaze cues to traverse a round
or wheel-shaped maze to escape from water or locate a
food or water reward. Memory in these tasks is typically
measured across multiple days, enabling within-subject
and between-subject performance to be assessed over
time. Whereas both the MWM and the RAM involve
explicit motivational stimuli to compel performance, the
OR and OP tasks rely on the animal’s inherent proclivity
for novelty. That is, mice and rats will spend more time
investigating a novel object, or a familiar object in a novel
location, than a previously investigated object in a famil-
iar location. One advantage of these single-trial learning
tasks is that they permit learning assessment while mini-
mizing appetitive or aversive confounders. Moreover, the
rapidity with which object tasks are learned allows
the effects of short-term hormone administration to
be associated with specific neural events and discrete
phases of memory formation (including acquisition,
consolidation and retention).
Morris water maze. Studies using the MWM to test oes-
trogenic regulation of spatial reference memory present
conflicting data on the role of E
2
in this form of memory.
For example, long-term administration of systemic E
2
to
ovariectomized rats or high E
2
levels during the rat oes-
trous cycle can impair spatial learning and memory
136138
,
whereas high E
2
levels in cycling mice were associated with
enhanced spatial learning
139
, and ovariectomized rats sys-
temically administered E
2
72 a nd 48 h before testing exhib-
ited better memory for a hidden platform
140
. Moreover,
high plasma E
2
levels in ovary-intact female meadow
voles correlated with a longer latency to locate the escape
platform during acquisition trials, whereas lower plasma
E
2
lev els wer e asso ciated with b etter learni ng
141
. Given the
varied reported effects of E
2
level on spatial MWM learn-
ing in different rodent studies, the E
2
dose and species may
determine how E
2
affects spatial memory in this task.
Although few studies have examined the short-term
effects of intracranially administered E2 on MWM per-
formance, existing research suggests beneficial effects.
For example, E2 infusion into the dorsal hippocampus of
young ovariectomized rats immediately, but not 3 h, after
spatial MWM training facilitates retention 24 h later,
Gonadectomy
Surgical removal of the
gonads (ovaries or testes);
because ‘ovariectomy’ is the
preferred term for females,
‘gonadectomy’ is most
commonly used for males.
Acquisition
A process through which
information is learned through
physical or sensory interaction
with environmental stimuli.
Spatial reference memory
Memory for locations that
do not change over time
(for example, the layout of
buildings on a college campus).
Used for navigating through
an environment.
Ta bl e 1 | Compounds used to investigate the role of oestrogen receptors in memory
Target Action Compounds Effects on memory
ERαAgonist PPT Objec t recognit ion57,154,173
Spati al memory57,173,224
Socia l recognit ion173,175
Antagonist MPP dihydrochloride Spati al memory225
ERβAgonist DPN; WAY200070
(benzoxazole); ERB 041;
ISP358-2
Objec t recognit ion57
Spati al memory57,224
Socia l recognit ion175
Fear gen eralizati on198
Antagonist PHTPP Objec t recognit ion225
Spati al memory225
ERα and
ERβAntagonist ICI 182,780 (fulvestrant) Objec t recognit ion87
Spati al memory224
Fear gen eralizati on198
GPER Agonist G1 Objec t recognit ion78,103
Spati al memory76,78,103
Socia l recognit ion175
Antagonist G15 Objec t recognit ion78
Spati al memory78,224
Aromatase Inhibitor Letrozole; anastrozole; ATD Objec t recognit ion215,226
Spat ial me mory215,224,226228
ATD, 1,4,6-androstatriene-3,17-dione; DPN, diarylpropionnitrile; ERα, oestrogen
receptor-α; ERβ, oestrogen receptor-β; GPER, G protein-coupled oestrogen receptor;
MPP, 1,3-bis(4-hydroxyphenyl)-4-methyl-5-[4-(2-piperidinylethoxy) phenol]-1H-pyrazole
dihydrochloride; PHTPP, 4-[2-phenyl-5,7-bis(trifluoromethyl)pyrazolo [1,5-a]pyrimidin-3-yl]
phenol; PPT, propyl pyrazole triol.
NATURE REVIEWS
|
NEUROSCIENCE
REVIEWS
indicating that E2 specifically enhances spatial memory
consolidation142.
Radial arm maze. The RAM allows researchers to dis-
tinguish the effects of E2 on spatial reference memory
and spatial working memory (FIG.2). Broadly, long-term
systemic administration of E2 in ovariectomized rats
enhances spatial working memory in the RAM, an effect
that manifests itself most prominently after several days
of E2 admini stration143145. Consistent with this, long-term
systemic E2 treatment in rats rescued ovariectomy-
induced deficits in working memory but not reference
memory146. Similarly, intracerebroventricular E2 infu-
sion reduced working memory errors in ovariectomized
rats147. Therefore, E2 appears to facilitate spatial working
memory but not spatial reference memory in the RAM
among young ovariectomized rodents.
Many brain regions, including the PFC, are required
to coordinate the effects of E2 in the RAM, although the
effects of E2 in these brain regions differ by dose. For
example, spatial working memory in ovariectomized rats
was facilitated by infusion of a high, but not a low, dose
of E2 into the PFC, and infusion of a low, but not a high,
dose into the dorsal hippocampus148. Thus, the effects of
E2 on spatial working memory in the RAM may depend
on both the dose and the brain region148.
Single-trial learning. It is difficult to disaggregate the
rapid non-classical effects of E2 from its long -term classi-
cal effects in tasks such as the MWM and RAM, because
learning in these tasks requires multiple trials across sev-
eral days. Moreover, both tasks involve motivational com-
ponents (for example, escape stress and thirst or hunger)
to compel performance, which may alter how the brain
responds to E2. By contrast, single-trial learning tasks
such as OR and OP tasks afford more precise assessment
of the neural mechanisms through which E2 facilitates
memory formation. As illustrated below, the findings
from studies using these tasks to assess the effects of E2 on
spatial and recognition memory are considerably more
consistent than those from maze studies.
As in the MWM and RAM, memory in the OR and
OP tasks is negatively affected by ovariectomy149,150.
However, unlike in the MWM and RAM, exogenous
E2 consistently improves spatial and OR memory in
the object tasks. For example, systemic administration
of E2 immediately after training enhances memory
consolidation in the OR and OP tasks among young
ovariectomized rats and mice151153. However, systemic
administration of E2 does not enhance memory when
injected 2 h after training152, suggesting that the win-
dow in which E2 exerts procognitive effects on OR and
OP memory consolidation is time limited and probably
dependent on non-classical mechanisms.
E2 infuse d imme diately, but n ot 3 h, after training into
the dorsal hippocampus similarly enhances memory
consolidation in the OR and OP tasks, indicating that
the dorsal hippocampus has a crucial role in mediating the
rapid effects of E2 on memory consolidation57,87,154,155.
The perirhinal cortex also plays a part, as E2 infused
immediately after training into the perirhinal cortex
of ovariectomized rats enhanced their preference for
a novel object over a familiar object compared with
vehicle-treated controls156. However, perirhinal E2 infu-
sions also impaired object memory consolidation in a
delayed non-match-to-sample task, complicating the role
of this brain region in mediating the influence of E2 on
OR memory156,157. Overall, these data suggest that E2 gen-
erally enhances OR and OP memory consolidation in
young ovariectomized rodents when it is administered
either before training or immediately after training.
Molecular mechanisms. The memory-enhancing effects
of E2 in the hippocampus involve numerous cellular
and molecular events in female rodents. Several cell-
signalling cascades are rapidly activated in response to E2,
Box 2 | The effects of oestradiol in ageing rodents
Ageing female rodents are frequently used to examine the effects of oestrogen
deprivation and replacement during the transition to reproductive senescence (see
REFS133,134,243 for recent reviews). In humans, menopause onset coincides with follicular
depletion, whereas oestropause, the menopausal parallel in rodents, is driven by
alterations in the function of the hypothalamus–pituitary–gonad axis244,245. Given this
difference, researchers should be cautious when interpreting studies using rodent
models of reproductive ageing. Nevertheless, the ageing rodent model has proved useful
in understanding the effects of oestrogen deprivation, the timing of hormone therapy
administration and age-associated alterations in responsivity to 17β-oestradiol (E2).
Broadly, E2 deprivation as a consequence of ageing or artificial long-term ovarian
hormone depletion lowers sensitivity to the memory-enhancing effects of E2. In spatial
memory tasks such as the Morris water maze (MWM), systemic E2 administration typically
enhances memory in middle-aged (14–19-month-old) but not aged (20-month-old
or older) ovariectomized female rodents246249. However, E2 can still facilitate memory
in aged females under certain experimental conditions. Long-term, high doses of
E2 elicit ed better memory in the MWM am ong aged o variect omized fe male rats250 an d
aged female mice251. Similarly, 3 weeks of E2 administration enhanced object memory
in aged ovariectomized female mice252.
Age and duration of oestrogen deprivation also modulate the effects of systemic E2
treatment on spatial reference and working memory. For example, long-term systemic
E2 treatment enhanced spatial working memory in middle-aged ovariectomized rats
in a water-motivated radial arm maze (RAM) task253. However, daily systemic injections
of E2 given to ageing female mice ovariectomized at 17.5 months did not affect spatial
reference or working memory in a water-motivated RAM task, whereas intermittent
E2 treatment mimicking normal hormonal cycling impaired spatial reference and working
memory254. As with the MWM, long-term E2 treatment enhanced spatial reference memory
in aged ovariectomized rats, but only when rats were primed with additional short-term
injections of E2 (REF.255). These data suggest not only an age-related loss of responsivity
to the memory-enhancing effects of E2 but als o a detrim ental eff ect of E2 on memory in
aged rodents. Supporting the idea that the duration of hormone loss affects responsivity
to E2, systemic E2 implants administered to 12-month and 17-month-old mice immediately
after ovariectomy enhanced spatial working memory in a RAM256, whereas such
E2 treatment given to 17-month-old mice 5 months after ovariectomy no longer
enhanced spatial working memory. These data support the well-accepted notion that
the ageing brain is less responsive to E2 after long peri ods of oes trogen dep rivation .
As in the maze tasks, ageing female rodents or rodents subjected to delayed oestrogen
replacement after ovariectomy exhibit diminished sensitivity to the beneficial effects
of E2 in obje ct memory consolid ation. For example, rats treate d with sys temic E2 9 or
15 months after ovariectomy exhibited intact memory for a previously seen object91.
However, increasing the treatment delay to 19 months after ovariectomy eliminated the
memory-facilitating effects of E2 (REF.162), consistent with other findings that E2 typically
enhances object recognition and object placement memory in middle-aged, but not
aged, female rodents248,257. Combined, these data suggest that E2 is less efficacious
when given to aged rodents, especially when postovariectomy E2 treatm ent is del ayed.
Several potential mechanisms may underlie decreased sensitivity to E2 in older rodents.
For ex ample, E2-induced hippocampal transcription is reduced in aged female
rodents258, and several studies suggest that the expression of oestrogen receptor-α and
oestrogen receptor-β dimini shes with age131,259,260. The precise mechanisms underlying
decreased responsivity to E2 in adv anced age remain to b e determi ned.
Spatial working memory
Memory for locations that
change over time (for example,
the locations of your keys
or your car in your campus
car park).
Delayed non-match-to-
sample task
Test of memory for items that
differ from an initial stimulus
array, assessed at some delay
after the original stimulus
presentation.
www.nature.com/nrn
REVIEWS
including the ERK, mTOR and PI3K–AKT pathways;
activity in these pathways is required for E2 to enhance
memory consolidation in the OR and OP tasks among
ovariectomized mice57,87,91,97,158. Within the dorsal hip-
pocampus, these signalling cascades are initiated by E2
binding to hippocampal ERα and ERβ but not to
GPER57,78. Indeed, although GPER activation in the
dorsal hippocampus enhances OR and OP memory
consolidation in ovariectomized females, these effects
are independent of E2 signalling78.
a Morris water maze
c Object recognition and object placement
b Radial arm maze
Standard working memory Working or reference memory
Training Testing Training Context test
Delay
d Social discrimination e Fear conditioning
Training Testing
Delay
Training Testing Tone test Extinction
Delay
(repeated exposure
to context without
CS presentation)
Fig. 2 | Behavioural approaches to studying oestrogenic effects on
memory. a | The Morris water maze tests a rodent’s ability to navigate in
space and remember the location of spatial cues in the environment229,230.
Rodents are placed in a large round pool of water made opaque with
non-toxic paint or powdered milk, and must use extramaze spatial cues to
navigate to a platform hidden just below the surface of the water229,230.
Measures of performance include time to reach the platform, distance swum
and swim speed. During probe trials in which the hidden platform is
inaccessible to subjects for a portion of or throughout the trial, memory for
the platform location can be assessed by measuring the number of times that
animals cross the platform location and/or the time spent close to the
location of the platform. b | The radial arm maze also tests spatial navigation
abilities. Food-restricted or water-restricted subjects traverse a wheel-
shaped maze in which they must retrieve food or water rewards from the
ends of 8 or 12 long arms that radiate from a round central platform231,232.
Rewards are not replaced, so animals should visit each arm only once. In the
standard version, all arms are baited to measure working memory, which is
memory for information that changes from trial to trial. In a common
variation, half of the arms are baited to measure working memory and half
are never baited to measure reference memory, or memory for information
that does not change from trial to trial. c | Object recognition and object
placement tasks typically consist of a training trial, during which animals
explore objects in an open field for 5–20 minutes, followed by a delay
(minutes to days) and a single testing phase, during which a new object is
introduced (object recognition) or a training object is moved (object
placement)233,234. Because rodents are drawn to novelty, animals that
remember the identity and location of the training objects will spend more
time than chance with a novel or moved object during testing. d | Social
recognition can be tested using a habituation–dishabituation task or a
social discrimination task. In habituation–dishabituation, subjects are
repeatedly exposed to the same stimulus conspecific, leading to a decrease in
investigative behaviour (habituation) as the stimulus animal becomes known.
Animals are then exposed to a novel conspecific, and if social recognition
memory is intact, investigative behaviour returns (dishabituation). In social
discrimination, animals undergo a test trial where they are simultaneously
exposed to a familiar conspecific, seen previously in habituation trials, and a
novel conspecific. Increased investigative behaviour of the novel conspecific
is indicative of intact social recognition memory. e | Fear memory is most
commonly studied with use of classical conditioning paradigms that pair a
stimulus (a cue or context) with a foot shock, generating a learned association
that leads to fear responses (such as freezing) on subsequent presentations of
the conditioned stimulus (CS). Cued and contextual fear conditioning differ
in their underlying neurocircuitry, with cued fear being independent of
hippocampal function and contextual fear requiring hippocampal input235.
Extinction can be tested with repeated presentation of the context in the
absence of CS presentation236. Figure adapted from REF.236, Springer.
NATURE REVIEWS
|
NEUROSCIENCE
REVIEWS
The ability of E2 to activate dorsal hippocampal cell
signalling is closely tied to its ability to mediate epige-
netic processes associated with facilitated memory. ERK
activation is necessary for E2 to increase acetylation of
histone H3 within the dorsal hippocampus of ovariect-
omized mice31,106. Dorsal hippocampal E2 infusion also
decreases levels of histone deacetylases 2 and 3 in the
hippocampus of ovariectomized mice, thereby pro-
viding a mechanism for increased H3 acetylation in
the E2-treated hippocampus31,106. Importantly, dorsal
hippocampal infusion of a histone acetyltransferase
inhibitor prevents E2 from enhancing OR memory con-
solidation in ovariectomized mice, demonstrating a cru-
cial role for histone acetylation in the mnemonic effects
of E2 (REFS31,105).
In ovariectomized females, systemic E2 (REF.159)
and intrahippocampally administered E2 increase the
dendritic spine density of CA1 pyramidal neurons,
an effect dependent on ERK and mTOR signalling99.
Consistent with these data, elevated E2 levels robustly
influence hippocampal synaptic plasticity by facilitating
LTP121,160,161, which correlates with enhanced memory in
hippocampus-dependent tasks120,162.
Although much remains to be learned, researchers
are well on their way towards characterizing the neuro-
biological mechanisms through which E2 modulates
spatial and object memory consolidation.
Social cognition
The term ‘social cognition’ encompasses various pro-
cesses, but perhaps the most well studied in animals
is social recognition. Memory for conspecifics and past
social interactions allows the formation of meaningful
relationships and selection of appropriate behavioural
responses in future social interactions, which are cru-
cial abilities in social species163. Social discrimination
is heavily influenced by neuroendocrine mechanisms;
however, much of this research has focused on the
neuropeptides oxytocin and vasopressin, so only more
recently have sex steroid hormones become appreciated
as modulators of social memory164.
Ovary-intact female mice trained on a habituation–
dishabituation task (FIG.2) during proestrus show
better discrimination of a novel conspecific 24 h later rel-
ative to females trained in dioestrus, suggesting that ele-
vated E2 levels during training may improve long-term
social recognition165. Other studies specifically testing
the effects of exogenous E2 on social recognition in
ovariectomized rodents have found that systemically
administered E2 given before training improves social
memory in a habituation–dishabituation task166168.
Many of these studies use long-term E2 replacement
before training and then test memory at extended time-
points, suggesting that classical actions of E2 are involved.
Indeed, E2 action at ERα increases transcription of the
oxytocin receptor in the medial amygdala169, a region
where oxytocin signalling is crucial for social recogni-
tion memory170. However, rapid membrane-initiated
E2 signalling also contributes substantially to these
effects. In a modified social discrimination task where
acquisition and recall occur within 40 min of drug
administration, systemic administration of E2 enhanced
social recognition memory in ovariectomized rats33.
The relatively short period between training and testing
makes it unlikely that classical E2 mechanisms have a role
in this effect, thereby implicating membrane-initiated
signalling as the key mediator. Furthermore, rapid
E2-induced CA1 plasticity correlates with this memory
enhancement; within 40 min of systemic or intracranial
administration, E2 increased CA1 dendritic spine density
in ovariectomized mice, which was surprisingly asso-
ciated with reduced AMPAR-mediated signalling33,119.
This change in excitatory signalling may reflect the
generation of new silent synapses.
The effects of E2 on social recognition memory are
mediated by both classical receptors and GPER. ERα
plays a particularly important role, with several studies
reporting that ERα-null or ERα-deficient female rodents
show impaired social recognition memory165,169,171,172.
Systemic injection of the ERα-selective agonist propyl
pyrazole triol (PPT) improved memory among ovariecto-
mized mice in a social discrimination task within 40 min,
suggesting that rapid action of membrane-localized ERα
is important for this effect173. The GPER agonist G1
induces similarly rapid improvements in social recog-
nition memory when systemically injected in ovariecto-
mized mice77. However, whether ERα and GPER initiate
similar cellular and molecular mechanisms to facilitate
social recognition memory is unclear. The facilita-
tive effects of ERα and GPER on social recognition do
not seem to extend fully to ERβ. Social recognition
memory in ERβ-null female mice is either unaffected
or only modestly impaired165,169,171, and treatment with
the ERβ-selective agonist diarylpropionitrile (DPN) in
ovariectomized mice either impairs or does not improve
social recognition memory173.
Similarly to spatial and OR memory, the hippocam-
pus is a critical locus of E2 action for social recognition
memory. Infusions of E2, PPT or G1 directly into the
dorsal hippocampus of ovariectomized mice induce a
rapid enhancement of social recognition memory that is
correlated with increased CA1 spine density and mod-
ulation of excitatory neurotransmission119,174. However,
E2 also modulates the function of other brain regions
crucial for social recognition memory. For example,
knockdown of ERα in the medial amygdala of ovariecto-
mized female rats abolishes social recognition memory172,
and infusion of ERα and GPER agonists into the medial
amygdala of ovariectomized mice rapidly enhances social
recognition175. An integrated understanding of how
E2 works ac ross multiple brain regi ons, re ceptor subtyp es
and signalling mechanisms to influence social cogni-
tion and recognition behaviour remains an area ripe for
future research.
Fear memory
The study of fear conditioning and its underlying cir-
cuitry and mechanisms is a cornerstone of the learning
and memory field. Nevertheless, despite its prominence,
researchers have until relatively recently largely ignored
how sex steroid hormones such as E2 affect fear learning.
Existing work reveals an interesting, and at times con-
tradictory, role for E2 in modulating both the acquisition
and the extinction of fear memory.
Silent synapses
Immature synapses containing
few AMPA receptors, which
could allow greater synaptic
potentiation and learning
facilitation on interaction
with a training stimulus.
Extinction
Process whereby a learned
association between two
stimuli (for example, shock
occurs in context A) becomes
unlearned through repetitive
exposure to one stimulus
(context A) without the
other (shock).
www.nature.com/nrn
REVIEWS
Fear acquisition. Studies examining how E2 modulates
fear acquisition report both enhancing and impairing
effects. For example, long-term systemic E2 administra-
tion enhanced cued and context-dependent fear condi-
tioning in ovariectomized mice176,177 and fear-potentiated
startle in ovariectomized rats178, suggesting that
E2 enhances acquisition of fear learning. However, other
findings reveal impaired contextual fear conditioning
(CFC) in proestrus rats relative to males or rats in
oestrus179. Similarly, ovary-intact or ovariectomized rats
given E2 for 2 days before CFC froze less than did males
or untreated ovariectomized rats180, suggesting E2 may
impair fear learning.
These seemingly contradictory findings probably
result from differences in experimental approaches, with
the dose and timing of E2 administration as important
variables. High-dose systemic E2 administration days or
weeks before CFC enhanced acquisition in ovariecto-
mized mice, with lower doses of E2 having no effect 181,182.
By contrast, when E2 was injected into ovariectomized
rats 30 min before CFC training, high doses impaired
acquisition, whereas a low dose enhanced learning183.
These results suggest important differences in how
E2 dose and treatment duration interact to impact fear
learning, but further research is needed to more clearly
define these relationships.
Fear extinction. Research on the role of E2 in modulat-
ing fear extinction paints a clearer picture than does the
fear acquisition literature, with evidence from passive
avoidance tasks184, conditioned taste aversion185 and both
cued and conditioned fear conditioning186188 supporting
the conclusion that E2 improves extinction learning. In
ovary-intact female rats, fear extinction training during
proestrus produces more rapid learning across trials186
and reduced freezing during extinction recall testing187
compared with extinction during metoestrus. E2 seems
to be both necessary and sufficient for these effects, as
systemic E2 treatment given to metoestrus or ovariect-
omized rats enhances extinction learning186,187, whereas
systemic blockade of ERs in proestrus rats impairs
extinction recall187. Similarly, reducing circulating
E2 levels via hormon al cont racept ives impai rs extinc tion
recall in cycling female rats189.
As with fear acquisition, the influence of E2 on
extinction is dose dependent. In cycling female rats,
E2 exhi bits an invert ed-U d ose–resp onse effe ct on exti nc-
tion learning, such that rats with very low (untreated
metoestrus) or very high (proestrus or metoestrus given
a high dose of E2) levels of E2 exhibited poor extinction
recall, and rats with moderate E2 levels (untreated proes-
trus or metoestrus given a low dose of E2) extinguished
recall well190.
ERβ is an important mediator of the effects of E2 on
fear extinction. Systemic injection of the ERβ-selective
agonist DPN before extinction training improves
extinction learning in ovariectomized or metoestrus
rats, whereas the ERα-selective agonist PPT has no
effect186,188. Infusion of DPN into the dorsal hippocam-
pus of ovariectomized rats enhanced contextual fear
extinction learning, suggesting that this brain region is
an important locus for ERβ activity in this paradigm186.
However, E2 has more pervasive actions in the neuro-
circuitry of fear extinction beyond the hippocampus.
The amygdala, a central region in fear extinction, shows
widespread expression of ER subtypes16 and structural
and synaptic plasticity following E2 treatment191,192. The
infralimbic cortex sends excitatory projections that regu-
late amygdala subregions during extinction193, and is also
sensitive to oestrogens194. E2 see ms to modulate th e acti v-
ity and connectivity of these regions during extinction
recall; studies using FOS as a marker of neuronal activ-
ity report that metoestrus rats treated with E2 displayed
less amygdalar activation188,195 and greater infralimbic
cortex activation188 following extinction recall than did
untreated controls. Both the oestrous-cycle phase and
exogenous E2 treatment influence the activity of infral-
imbic cortex–amygdala projections in female rats195,196,
suggesting that E2 increases infralimbic cortex-driven
activation of an inhibitory circuitry in the amygdala that
reduces fear responses.
Fear generalization. In addition to fear acquisition
and extinction, E2 also influences fear generalization.
Ovariectomized rats systemically treated with E2 show
increased generalization of fear to neutral contexts rel-
ative to untreated controls197. Similarly to extinction,
this effect seems to depend primarily on hippocampal
ERβ signalling, as infusion of the ERβ-selective agonist
DPN, but not ERα-selective PPT, into the hippocampus
of ovariectomized rats increases fear generalization198.
That E2 can enhance both fear expression (via acqui-
sition and generalization) as well as extinction may at
first seem contradictory, but is not surprising given
that these processes share certain underlying molecular
mechanisms. Moreover, fear acquisition, generalization
and extinction all require new learning. Thus, the effects
of E2 on all aspects of fear learning highlight its actions as
a general promoter of learning and memory.
To ge t h er w it h fi n d in g s f r om t h e s p at i al , ob j e ct a nd
social memory tasks discussed earlier, the fear memory
data suggest that E2 is uniquely positioned as a general
neuromodulator that broadly facilitates new learning of
many sorts.
E2 modulation of memory in males
Although oestrogens are often (incorrectly) considered
‘female’ hormones, accumulating research findings
indicate a primary role for E2 in enhancing cognition
in male rodents. Indeed, E2 levels in the male rat hip-
pocampus are higher than in cycling females199,200.
Emerging findings, reviewed below, imply that E2 may
influence memory via distinct molecular mechanisms
in males and females. Why might these differences
matter if the beneficial effects on memory are simi-
lar in both sexes? From a drug discovery standpoint,
these data suggest the potential need to develop sex-
specific treatments for memory dysfunction in humans.
Tre at me nt s tha t t ar get me ch an is ms t hr ou gh wh ic h
E2 modulates memory in one sex (for example, ERK in
females) but not in the other are bound to fail in half
the population. Additional research to determine which
oestrogenic mechanisms are sex specific and which are
common to both sexes will substantially advance the
Contextual fear conditioning
Model of fear learning in
which repeated exposure to
foot shocks in one context
eventually elicits fear
(freezing) of the context
in the absence of shock.
FOS
A transcription factor
encoded by an immediate
early gene that is activated
rapidly and transiently in
response to neuronal activity,
leading to expression of
memory-related genes.
Generalization
Process whereby a stimulus–
response association learned
in one context (for example,
a stimulus induces fear)
becomes transferred to
another, similar context.
NATURE REVIEWS
|
NEUROSCIENCE
REVIEWS
development of effective memory therapeutics for both
men and women.
Sex differences in hippocampal E2 signall ing
In the hippocampus, E2 acts as a neuromodulator in both
males and females, but an emerging body of literature has
found notable sex differences in how E2 influences hip-
pocampal neurotransmission and the molecular under-
pinnings of these effects. In both sexes, E2 potentiates
excitatory neurotransmission in the hippocampus6,35,111,
and although some molecules and pathways, such as
SRC, ERK–MAPK, calcium/calmodulin-dependent pro-
tein kinase II and TRKB signalling, contribute to these
effects in both sexes35,201,202, others are sex specific. For
example, potentiation of excitatory postsynaptic currents
and LTP in the hippocampus depend on PKA in female
rats but not in males202. The sexes also differ in the ER
subtypes that mediate neural excitation, as hippocampal
glutamatergic neurotransmission is regulated presynap-
tically by ERβ in females and ERα in males, and postsyn-
aptically by GPER in females and ERβ in males111. Other
work has similarly shown differential contributions of
ERα versus ERβ in facilitating LTP between males and
females35,201. Differing roles of ER subtypes in modulat-
ing excitatory signalling may arise from sex differences
in subcellular ER localization, which in turn could
lead to sex-specific recruitment of signalling kinases
by E2 (REFS167,201).
Unlike excitator y signalling , inhibitor y h ippocam-
pal neurotransmission seems to be modulated by E2
only in females and through a sex-specific mechanism.
In the ovariectomized female rat hippocampus, ERα
activates postsynaptic mGluR1 signalling to stimulate
endocannabinoid release, which inhibits presynaptic
GABAergic terminals122. The functional coupling of
ERs with mGluR subtypes was shown to occur only in
females56, and further investigation found that although
ER–mGluR complexes can exist in both sexes, E2 acti vates
mGluR-dependent signalling only in females123.
A similar sex difference has been found with locally
synthesized E2 in the hippocampus. Although male and
female rodents both express aromatase and synthesize E2
in the hippocampus17,200,203, systemic aromatase inhibi-
tion reduces hippocampal synapse number and severely
impairs LTP induction in female mice, but has no effect
on synapse number and only modestly reduces LTP
in male mice204. Similarly, aromatase-null female mice
were shown to have reduced hippocampal spine density
compared with wild-type female mice, whereas spine
density was unaffected in aromatase-null male mice205.
However, in a forebrain-specific aromatase-null mouse,
both males and females exhibited reduced hippocampal
spine density206. Understanding the cellular and molecu-
lar consequences of local oestrogen synthesis in the male
hippocampus will require further investigation207.
Oestrogenic modulation of cognition in males
Despite differences in the mechanisms of E2 signalling
in cognition-related regions of the brain, E2 exerts sim-
ilar mnemonic benefits in males and females. Systemic
injection or dorsal hippocampal infusion of E2 immedi-
ately after training enhances memory consolidation in
the OR and OP tasks in gonad-intact and gonadecto-
mized male rats and mice208,209. Notably, sex differences
in the mechanisms that drive the beneficial effects of E2
on these tasks have been found: in contrast to its cen-
tral importance to the memory-enhancing effects of
E2 in ovariectomized mice, dorsal hippocampal ERK
phosphorylation is not necessary for E2 to facilitate
spatial and object memory consolidation in male mice,
regardless of gonadal status209.
In the MWM, gonad-intact adult male rats and aged
male mice systemically treated with E2 immediately after
training exhibited enhanced spatial memory compared
with vehicle-treated males210,211. Gonad-intact adult
males receiving E2 long term also made fewer working
memory errors in the RAM than did vehicle-treated
controls212.
Local synthesis of oestrogens is also important for
memory processes in male rodents, as aromatase-null
male mice exhibit deficits in social recognition213.
Furthermore, short-term oral aromatase inhibition
increases errors in a working memory task in male
rats214, and short-term intrahippocampal aromatase
inhibition prevents object and spatial memory consoli-
dation in gonadectomized male mice215. Therefore, the
data thus far suggest that hippocampally synthesized E2
is important for memory formation in male rodents, as
has been observed in female rodents. Although much
more remains to be learned, these selected findings sup-
port a beneficial effect of E2 on memory formation in
male rodents.
Conclusions and future directions
This Review has illustrated some of the myriad ways
in which E2, acting in several brain regions via mul-
tiple ERs, can facilitate numerous forms of learning
and memory (FIG.3). Three decades of research has
demonstrated that E2 is a potent regulator of the neural
mechanisms that are critically important for memory
formation, including cell signalling, gene expression,
protein synthesis, extrinsic and intrinsic excitability,
dendritic spine morphology and neurogenesis. Thus,
researchers studying the neurobiology of learning and
memory should recognize oestrogens as essential neuro-
modulators akin to other well-accepted modulatory
factors such as glucocorticoids and growth factors.
Widespread acceptance of oestrogenic neuromod -
ulation may have been historically slow to take hold
because of the strong association between oestrogens
and female reproduction, which has led to the erroneous
perceptions that oestrogens are ‘female’ hormones and
that cyclic oestrogen fluctuations in females confound
experimental outcomes due to increased variability.
In fact, oestrogens are important modulators of mem-
ory and neural function in both sexes216,217. Depending
on the behaviour being assessed, ovary-intact females
do not necessarily exhibit more behavioural variability
than males218, which argues for greater consideration of
the modulatory influence of oestrogens in females and in
males. Future progress on this issue may stem from the
US National Institutes of Health’s 2016 policy requiring
vertebrate animal researchers to consider sex as a biolog-
ical variable219221, as more explicit comparisons between
www.nature.com/nrn
REVIEWS
the sexes could lead to new insights into hormonal regu-
lation of cognition. Although some investigators were
initially resistant to this policy, attitudes towards the
requirement are improving222. Greater appreciation of
oestrogens as neuromodulators that exert wide-ranging
effects in all individuals — not just females — is crucial
for the advancement of both basic and clinical science.
To further advance knowledge of oestrogenic
modulation in both sexes, future studies should use
newer technologies, including single-cell sequencing,
‘om i cs’ -l e ve l an al ys es an d ta rg et ed ge ne ti c ma ni pu la -
tions, to pinpoint crucial molecules and cellular pro-
cesses that underlie oestrogenic facilitation of learning
and memory. For example, a multiplexed CRISPR–Cas9
gene-editing approach could be used to simultaneously
target multiple oestrogen-responsive genes implicated
in neurodegenerative disease to understand their role in
memory function223. Future research must also evolve
from a focus on oestrogenic effects in individual brain
regions to addressing how oestrogens concurrently
influence multiple brain areas within memory cir-
cuits. Recent work using multiplexed chemogenetic
silencing has shown that the memory-enhancing effects
of E2 in the dorsal hippocampus requires concurrent
activity of the dorsal hippocampus and PFC155. Thus,
determining how brain regions interact synergisti-
cally to support oestrogen-mediated memory pro-
cesses is a crucial next step for the field. As part of this
approach, researchers should also consider cell-type
specificity, as memory-modulating effects of E2 on cell
types such as inhibitory neurons and glial cells have been
overshadowed by a predominant focus on excitatory
neurons.
By leveraging new technologies and asking circuit-
level and cell type-specific questions in both males
and females, scientists will discover fundamental new
insights into the ways in which E2-induced modulation
of brain function influences memory formation. Given
the dearth of effective treatments for memory dysfunc-
tion in various disorders, this information could provide
valuable new avenues for therapeutic development that
benefits both sexes.
Published online xx xx xxxx
Memory process ER subtype Site of oestrogenic action Behavioural output
Spatial and object
memory
ERα
ERβ
GPER
Social memory ERα
GPER
Spatial reference memory
Spatial reference memory
Spatial working memory
Object recognition and
object placement memory
Fear memory ERβ Fear aquisition
Fear aquisition
Fear extinction learning
and recall
Fear generalization
Social recognition memory
PRC
HPC
HPC
MeA
PFC
HPC
PFC
Amy
Fig. 3 | Summary of oestrogenic actions on memory processes. This schematic shows the receptors and brain regions
involved in the effects of 17β-oestradiol (E2) on memory processes and the behavioural output of the actions of E2 in
young female rodents. Spatial reference memory142, working memory148 and ob ject memo ry57,87,154,155,173 are facilitated
by E2 (but see the discussion in the main text), and are largely dependent on the hippocampus (HPC) and prefrontal cortex
(PFC). The perirhinal cortex (PRC) is also involved in E2-mediated object recognition memory156,157. Social recognition
memory is facilitated by oestrogen receptor-α (ERα) and G protein-coupled oestrogen receptor (GPER) signalling in the
HPC and medial amygdala (MeA)119,172174. Fear acquisition, fear extinction learning and recall, and fear generalization are
regulat ed by ERβ in th e PFC, amy gdala (Am y) and HPC186,188,195,196,198.
NATURE REVIEWS
|
NEUROSCIENCE
REVIEWS
1. Gould, E., Woolley, C. S., Frankfurt, M. & McEwen, B. S.
Gonadal steroids regulate dendritic spine density in
hippocampal pyramidal cells in adulthood. J. Neurosci.
10, 1286–1291 (1990).
2. Woolley, C. & McEwen, B. Estradiol mediates
fluctuation in hippocampal synapse density during
the estrous cycle in the adult rat. J. Neurosci. 12,
2549–2554 (1992).
This foundational study links dynamic changes
in hippocampal spine density to oestrous cyclicity,
such that elevated spine density is correlated with
higher levels of E2.
3. Woolley, C. S. & McEwen, B. S. Roles of estradiol and
progesterone in regulation of hippocampal dendritic
spine density during the estrous cycle in the rat.
J. Comp. Neurol. 336, 293–306 (1993).
4. Wool ley, C . S. & McE wen, B. S. E strad iol regul ates
hippocampal dendritic spine density via an N-methyl-
D-aspartate receptor-dependent mechanism.
J. Neurosci. 14, 7680–7687 (1994).
5. Woolley, C. S., Weiland, N. G., McEwen, B. S. &
Schwartzkroin, P. A. Estradiol increases the sensitivity
of hippocampal CA1 pyramidal cells to NMDA
receptor-mediated synaptic input: correlation with
dendritic spine density. J. Neurosci. 17, 1848–1859
(1997).
6. Wong, M. & Moss, R. Long-term and short-term
electrophysiological effects of estrogen on the synaptic
properties of hippocampal CA1 neurons. J. Neurosci.
12, 3217–3225 (1992).
7. Gu, Q. & Moss, R. L. 17β-Estradiol potentiates
kainate-induced currents via activation of the cAMP
cascade. J. Neurosci. 16, 3620–3629 (1996).
8. Azcoitia, I., Sierra, A. & Garcia-Segura, L. M. Estradiol
prevents kainic acid-induced neuronal loss in the rat
dentate gyrus. Neuroreport 9, 3075–3079 (1998).
9. Frick, K. M., Tuscher, J. J., Koss, W. A., Kim, J. &
Taxier, L. R. Estrogenic regulation of memory
consolidation: a look beyond the hippocampus,
ovaries, and females. Physiol. Behav. 187, 57–66
(2018).
10. Rossetti, M. F., Cambiasso, M. J., Holschbach, M. A. &
Cabrera, R. Oestrogens and progestagens: synthesis
and action in the brain. J. Neuroendocrinol. https://
doi.org/10.1111/jne.12402 (2016).
11. Hara, Y., Waters, E. M., McEwen, B. S. & Morrison, J. H.
Estrogen effects on cognitive and synaptic health
over the lifecourse. Physiol. Rev. 95, 785–807
(2015).
12. Morrison, J. H. & Baxter, M. G. The aging cortical
synapse: hallmarks and implications for cognitive
decline. Nat. Rev. Neurosci. 13, 240–250 (2012).
13. Dumitriu, D., Rapp, P. R., McEwen, B. S. &
Morrison, J. H. Estrogen and the aging brain: an elixir
for the weary cortical network. Ann. N. Y. Acad. Sci.
1204, 104–112 (2010).
14. Miller, W. L. & Auchus, R. J. The molecular
biology, biochemistry, and physiology of human
steroidogenesis and its disorders. Endocr. Rev. 32,
81–1 51 (2 011).
15. Compagnone, N. A. & Mellon, S. H. Neurosteroids:
biosynthesis and function of these novel
neuromodulators. Front. Neuroendocrinol. 21,
1–56 (2000).
16. Österlund, M., Kuiper, G. G. J. M., Gustafsson, J.-Å.
& Hurd, Y. L. Differential distribution and regulation
of estrogen receptor-α and -β mRNA within the female
rat brain. Mol. Brain Res. 54, 175–180 (1998).
17. Prange-Kiel, J., Wehrenberg, U., Jarry, H. & Rune, G. M.
Para/autocrine regulation of estrogen receptors
in hippocampal neurons. Hippocampus 13, 226–234
(2003).
18. Stanic´, D. etal. Characterization of aromatase
expression in the adult male and female mouse
brain. I. Coexistence with oestrogen receptors
α and β, and androgen receptors. PLoS ONE 9, e90451
(2014).
19. Kretz, O. etal. Hippocampal synapses depend on
hippocampal estrogen synthesis. J. Neurosci. 24,
5913–5921 (2004).
This study provides early evidence that locally
synthesized oestrogens are critical for maintenance
of hippocampal synapses.
20. Balthazart, J. & Ball, G. F. Is brain estradiol a
hormone or a neurotransmitter? Tre n d s N e u r o s c i . 29,
241–249 (2006).
21. Remage-Healey, L., Saldanha, C. J. & Schlinger, B. A.
Estradiol synthesis and action at the synapse: evidence
for “synaptocrine” signaling. Front. Endocrinol. 2, 28
(2011).
22. Allen, E. The oestrous cycle in the mouse. Am. J. Anat.
30, 297–371 (1922).
23. Long, J. A. & Evans, H. M. The Oestrous Cycle in the
Rat and Its Associated Phenomena. (University of
California Press, 1922).
24. Kato, A. etal. Female hippocampal estrogens
have a significant correlation with cyclic fluctuation
of hippocampal spines. Front. Neural. Circuits https://
doi.org/10.3389/fncir.2013.00149 (2013).
25. Pawluski, J. L., Brummelte, S., Barha, C. K.,
Crozier, T. M. & Galea, L. A. M. Effects of steroid
hormones on neurogenesis in the hippocampus
of the adult female rodent during the estrous cycle,
pregnancy, lactation and aging. Front. Neuroendocrinol.
30, 343–357 (2009).
26. Mendoza-Garcés, L. etal. Differential expression
of estrogen receptors in two hippocampal regions
during the estrous cycle of the rat. Anat. Rec. 294,
1913–1919 (2011).
27. Balthazart, J., Choleris, E. & Remage-Healey, L.
Steroid and the brain: 50 years of research, conceptual
shifts and the ascent of non-classical and membrane-
initiated actions. Horm. Behav. 99, 1–8 (2018).
28. Vasudevan, N. & Pfaff, D. W. Non-genomic actions of
estrogens and their interaction with genomic actions
in the brain. Front. Neuroendocrinol. 29, 238–257
(2008).
29. Szego, C. M. & Davis, J. S. Adenosine
3,5-monophosphate in rat uterus: acute elevation by
estrogen. Proc. Natl Acad. Sci. USA 58, 1711–1718
(1967).
30. Kelly, M. J., Moss, R. L. & Dudley, C. A.
Differential sensitivity of preoptic-septal neurons to
microelectrophoressed estrogen during the estrous
cycle. Brain Res. 114, 152–157 (1976).
31. Zhao, Z., Fan, L. & Frick, K. M. Epigenetic alterations
regulate estradiol-induced enhancement of memory
consolidation. Proc. Natl Acad. Sci. USA 107,
5605–5610 (2010).
32. Akama, K. T. & McEwen, B. S. Estrogen stimulates
postsynaptic density-95 rapid protein synthesis via
the Akt/protein kinase B pathway. J. Neurosci. 23,
2333–2339 (2003).
33. Phan, A. etal. Low doses of 17β-estradiol rapidly
improve learning and increase hippocampal dendritic
spines. Neuropsychopharmacology 37, 2299–2309
(2012).
34. Woolley, C. S. Acute effects of estrogen on neuronal
physiology. Annu. Rev. Pharmacol. Toxicol. 47,
657–680 (2007).
35. Kramár, E. A. etal. Cytoskeletal changes underlie
estrogen’s acute effects on synaptic transmission
and plasticity. J. Neurosci. 29, 12982–12993
(2009).
36. Pappas, T. C., Gametchu, B. & Watson, C. S.
Membrane estrogen receptors identified by multiple
antibody labeling and impeded-ligand binding.
FAS EB J . 9, 404–410 (1995).
37. Watsona, C. S., Norfleet, A. M., Pappas, T. C. &
Gametchu, B. Rapid actions of estrogens in GH3/B6
pituitary tumor cells via a plasma membrane version
of estrogen receptor-α. Steroids 64, 5–13 (1999).
38. Razandi, M., Pedram, A., Greene, G. L. & Levin, E. R.
Cell membrane and nuclear estrogen receptors
(ERs) originate from a single transcript: studies of
ERα and ER β expressed in Chinese hamster ovary
cells. Mol. Endocrinol. 13, 307–319 (1999).
39. Clarke, C. H. etal. Perimembrane localization of the
estrogen receptor α protein in neuronal processes of
cultured hippocampal neurons. Neuroendocrinology
71, 34–42 (2000).
40. Gorosito, S. V., Lorenzo, A. G. & Cambiasso, M. J.
Estrogen receptor α is expressed on the cell-surface of
embryonic hypothalamic neurons. Neuroscience 154,
117 3 –11 7 7 (2 00 8 ).
41. Razandi, M., Pedram, A., Park, S. T. & Levin, E. R.
Proximal events in signaling by plasma membrane
estrogen receptors. J. Biol. Chem. 278, 2701–2712
(2003).
42. Ábrahám, I. M., Todman, M. G., Korach, K. S. &
Herbison, A. E. Critical invivo roles for classical
estrogen receptors in rapid estrogen actions on
intracellular signaling in mouse brain. Endocrinology
145, 3055–3061 (2004).
43. Blaustein, J. D. Cytoplasmic estrogen receptors in
rat brain: immunocytochemical evidence using three
antibodies with distinct epitopes. Endocrinology 131,
1336–1342 (1992).
44. Milner, T. A. etal. Ultrastructural evidence that
hippocampal alpha estrogen receptors are located
at extranuclear sites. J. Comp. Neurol. 429, 355–371
(2001).
45. Milner, T. A. etal. Ultrastructural localization of
estrogen receptor β immunoreactivity in the rat
hippocampal formation. J. Comp. Neurol. 491, 81–95
(2005).
46. Andersson, S. etal. Insufficient antibody validation
challenges oestrogen receptor beta research.
Nat. Commun. 8, 15840 (2017).
47. Zhang, Z., Kumar, R., Santen, R. J. & Song, R. X.-D.
The role of adapter protein Shc in estrogen
non-genomic action. Steroids 69, 523–529
(2004).
48. Russell, K. S., Haynes, M. P., Sinha, D., Clerisme, E. &
Bender, J. R. Human vascular endothelial cells contain
membrane binding sites for estradiol, which mediate
rapid intracellular signaling. Proc. Natl Acad. Sci. USA
97, 5930–5935 (2000).
49. Acconcia, F., Ascenzi, P., Fabozzi, G., Visca, P. &
Marino, M. S-palmitoylation modulates human
estrogen receptor-α function s. Biochem. Biophys.
Res. Commun. 316, 878–883 (2004).
50. Pedram, A. etal. A conserved mechanism for
steroid receptor translocation to the plasma
membrane. J. Biol. Chem. 282, 22278–22288
(2007).
51. Meitzen, J. etal. Palmitoylation of estrogen receptors
is essential for neuronal membrane signaling.
Endocrinology 154, 4293–4304 (2013).
52. Schlegel, A., Wang, C., Katzenellenbogen, B. S.,
Pestell, R. G. & Lisanti, M. P. Caveolin-1 potentiates
estrogen receptor α (ERα) signaling. J. Biol. Chem.
274, 33551–33556 (1999).
53. Razandi, M., Oh, P., Pedram, A., Schnitzer, J. &
Levin, E. R. ERs associate with and regulate the
production of caveolin: implications for signaling
and cellular actions. Mol. Endocrinol. 16, 100–115
(2002).
54. Acconcia, F. etal. Palmitoylation-dependent estrogen
receptor α membrane localization: regulation by
17β-estradiol. Mol. Biol. Cell 16, 231–237 (2004).
55. Boulware, M. I., Kordasiewicz, H. & Mermelstein, P. G.
Caveolin proteins are essential for distinct effects of
membrane estrogen receptors in neurons. J. Neurosci.
27, 9941–9950 (2007).
56. Boulware, M. I. etal. Estradiol activates group I
and II metabotropic glutamate receptor signaling,
leading to opposing influences on cAMP response
element-binding protein. J. Neurosci. 25, 5066–5078
(2005).
57. Boulware, M. I., Heisler, J. D. & Frick, K. M.
The memory-enhancing effects of hippocampal
estrogen receptor activation involve metabotropic
glutamate receptor signaling. J. Neurosci. 33,
15184–15194 (2013).
58. Martinez, L. A. etal. Estradiol facilitation of cocaine
self-administration in female rats requires activation
of mGluR5. eNeuro https://doi.org/10.1523/
ENEURO.0140-16.2016 (2016).
59. Dewing, P. etal. Membrane estrogen receptor-α
interactions with metabotropic glutamate receptor 1a
modulate female sexual receptivity in rats. J. Neurosci.
27, 9294–9300 (2007).
60. Kahlert, S. etal . Estrogen recepto r α rapidly activates
the IGF-1 receptor pathway. J. Biol. Chem. 275,
18447–18453 (2000).
61. Mendez, P., Azcoitia, I. & Garcia-Segura, L. M.
Estrogen receptor alpha forms estrogen-dependent
multimolecular complexes with insulin-like growth
factor receptor and phosphatidylinositol 3-kinase in
the adult rat brain. Mol. Brain. Res. 112 , 170–176
(2003).
62. Spencer-Segal, J. L. etal. Estradiol acts via estrogen
receptors alpha and beta on pathways important
for synaptic plasticity in the mouse hippocampal
formation. Neuroscience 202, 131–146 (2012).
63. Kramár, E. A., Babayan, A. H., Gall, C. M. & Lynch, G.
Estrogen promotes learning-related plasticity by
modifying the synaptic cytoskeleton. Neuroscience
239, 3–16 (2013).
64. Quesada, A. & Micevych, P. E. Estrogen interacts
with the IGF-1 system to protect nigrostriatal
dopamine and maintain motoric behavior after
6-hydroxdopamine lesions. J. Neurosci. Res. 75,
107–116 (20 04).
65. Selvamani, A. & Sohrabji, F. The neurotoxic effects
of estrogen on ischemic stroke in older female rats
is associated with age-dependent loss of insulin-like
growth factor-1. J. Neurosci. 30, 6852–6861
(2010).
66. Witty, C. F., Gardella, L. P., Perez, M. C. &
Daniel, J. M. Short-term estradiol administration in
aging ovariectomized rats provides lasting benefits for
memory and the hippocampus: a role for insulin-like
growth factor-I. Endocrinology 154, 842–852
(2013).
www.nature.com/nrn
REVIEWS
67. Cabodi, S. etal. p130Cas interacts with estrogen
receptor α and modulates non-genomic estrogen
signaling in breast cancer cells. J. Cell Sci. 117 ,
1603–1611 (2004).
68. Lu, Q. etal. Striatin assembles a membrane signaling
complex necessary for rapid, nongenomic activation
of endothelial NO synthase by estrogen receptor α.
Proc. Natl Acad. Sci. USA 101, 17126–17131
(2004).
69. Filardo, E. J., Quinn, J. A., Bland, K. I. & Frackelton, A. R.
Estrogen-induced activation of Erk-1 and Erk-2 requires
the G protein-coupled receptor homolog, GPR30, and
occurs via trans-activation of the epidermal growth factor
receptor th rough release of HB- EGF. Mol. Endocrinol.
14, 1649–1660 (2000).
70. Thomas, P., Pang, Y., Filardo, E. J. & Dong, J. Identity of
an estrogen membrane receptor coupled to a G protein
in human breast cancer cells. Endocrinology 146,
624–632 (2005).
This study describes GPER as a membrane-tethered
receptor that binds oestrogens, thereby establishing
the existence of a novel, non-nuclear ER.
71. Filardo, E. J., Quinn, J. A., Frackelton, A. R. &
Bland, K. I. Estrogen action via the G protein-coupled
receptor, GPR30: stimulation of adenylyl cyclase
and cAMP-mediated attenuation of the epidermal
growth factor receptor-to-MAPK signaling axis.
Mol. Endocrinol. 16, 70–84 (2002).
72. Revankar, C. M., Cimino, D. F., Sklar, L. A.,
Arterburn, J. B. & Prossnitz, E. R. A transmembrane
intracellular estrogen receptor mediates rapid cell
signaling. Science 307, 1625–1630 (2005).
73. Hazell, G. G. J. etal. Localisation of GPR30, a novel
G protein-coupled oestrogen receptor, suggests
multiple functions in rodent brain and peripheral
tissues. J. Endocrinol. 202, 223–236 (2009).
74. Brailoiu, E. etal. Distribution and characterization
of estrogen receptor G protein-coupled receptor 30
in the rat central nervous system. J. Endocrinol. 193,
311–321 (201 9).
75. Hammond, R., Nelson, D., Kline, E. & Gibbs, R. B.
Chronic treatment with a GPR30 antagonist impairs
acquisition of a spatial learning task in young female
rats. Horm. Behav. 62, 367–374 (2012).
76. Hawley, W. R., Grissom, E. M., Moody, N. M.,
Dohanich, G. P. & Vasudevan, N. Activation of
G-protein-coupled receptor 30 is sufficient to enhance
spatial recognition memory in ovariectomized rats.
Behav. Brain Res. 262, 68–73 (2014).
77. Gabor, C., Lymer, J., Phan, A. & Choleris, E. Rapid
effects of the G-protein coupled oestrogen receptor
(GPER) on learning and dorsal hippocampus dendritic
spines in female mice. Physiol. Behav. 149, 53–60
(2015).
78. Kim, J., Szinte, J. S., Boulware, M. I. & Frick, K. M.
17β-Estradiol and agonism of G-protein-coupled
estrogen receptor enhance hippocampal memory via
different cell-signaling mechanisms. J. Neurosci. 36,
3309–3321 (2016).
79. Ervin, K. S. J., Mulvale, E., Gallagher, N., Roussel, V.
& Choleris, E. Activation of the G protein-coupled
estrogen receptor, but not estrogen receptor
α or β, rapidly enhances social learning.
Psychoneuroendocrinology 58, 51–66 (2015).
80. Anchan, D., Gafur, A., Sano, K., Ogawa, S. &
Vasudevan, N. Activation of the GPR30 receptor
promotes lordosis in female mice. Neuroendocrinology
100, 71–80 (2014).
81. Long, N., Serey, C. & Sinchak, K. 17β-estradiol
rapidly facilitates lordosis through G protein-coupled
estrogen receptor 1 (GPER) via deactivation of
medial preoptic nucleus μ-opioid receptors in estradiol
primed female rats. Horm. Behav. 66, 663–666
(2014).
82. Briz, V. & Baudry, M. Estrogen regulates protein
synthesis and actin polymerization in hippocampal
neurons through different molecular mechanisms.
Neuroendocr. Sci. 5, 22 (2014).
83. Zhao, L., Chen, S., Ming Wang, J. & Brinton, R. D.
17β-Estradiol induces Ca2+ influx, dendritic and
nuclear Ca2+ rise and subsequent cyclic AMP response
element-binding protein activation in hippocampal
neurons: a potential initiation mechanism for
estrogen neurotrophism. Neuroscience 132,
299–311 (2005).
84. Wu, T.-W., Chen, S. & Brinton, R. D. Membrane
estrogen receptors mediate calcium signaling and
MAP kinase activation in individual hippocampal
neurons. Brain Res. 1379, 34–43 (2011).
85. Moss, R. L. & Gu, Q. Estrogen: mechanisms for a
rapid action in CA1 hippocampal neurons. Steroids
64, 14–21 (1999).
86. Kuroki, Y., Fukushima, K., Kanda, Y., Mizuno, K. &
Watanabe, Y. Putative membrane-bound estrogen
receptors possibly stimulate mitogen-activated protein
kinase in the rat hippocampus. Eur. J. Pharmacol.
400, 205–209 (2000).
87. Fernande z, S. M. etal . Oestra diol-in duced
enhancement of object memory consolidation involves
hippocampal extracellular signal-regulated kinase
activation and membrane-bound oestrogen receptors.
J. Neurosci. 28, 8660–8667 (2008).
This study reveals a crucial role for rapidly
activated cell signalling activity in the
memory-enhancing effects of E2.
88. Lee, S. J. etal. Estrogen induces phosphorylation
of cyclic AMP response element binding (pCREB)
in primary hippocampal cells in a time-dependent
manner. Neuroscience 124, 549–560 (2004).
89. Yoko m ak u, D . e t a l. E s tr og en en ha n ce s d ep ol a ri za t io n-
induced glutamate release through activation of
phosphatidylinositol 3-kinase and mitogen-activated
protein kinase in cultured hippocampal neurons.
Mol. Endocrinol. 17, 831–844 (2003).
90. Spencer, J. L., Waters, E. M., Milner, T. A. &
McEwen, B. S. Estrous cycle regulates activation of
hippocampal Akt, LIMK, and neurotrophin receptors
in C57BL6 mice. Neuroscience 155, 1106–1119
(2008).
91. Fan, L . etal. Estradi ol-indu ced obje ct memor y
consolidation in middle-aged female mice requires
dorsal hippocampal extracellular signal-regulated
kinase and phosphatidylinositol 3-kinase activation.
J. Neurosci. 30, 4390–4400 (2010).
92. Ruiz-Palmero, I., Hernando, M., Garcia-Segura, L. M.
& Arevalo, M.-A. G protein-coupled estrogen receptor
is required for the neuritogenic mechanism of
17β-estradiol in developing hippocampal neurons.
Mol. Cell. Endocrinol. 372, 105–115 (2013).
93. Lewis, M. C., Kerr, K. M., Orr, P. T. & Frick, K. M.
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. 122, 716–721
(2008).
94. Sato, K., Akaishi, T., Matsuki, N., Ohno, Y. &
Nakazawa, K. β-Estradiol induces synaptogenesis
in the hippocampus by enhancing brain-derived
neurotrophic factor release from dentate gyrus
granule cells. Brain Res. 1150 , 108–120 (2007).
95. Gu, Q., Korach, K. S. & Moss, R. L. Rapid action
of 17β-estradiol on kainate-induced currents in
hippocampal neurons lacking intracellular estrogen
receptors. Endocrinology 140, 660–666 (1999).
96. Hasegawa, Y. etal. Estradiol rapidly modulates
synaptic plasticity of hippocampal neurons:
Involvement of kinase networks. Brain Res. 1621,
147–161 (2015).
97. Fortress, A. M., Fan, L., Orr, P. T., Zhao, Z. &
Frick, K. M. Estradiol-induced object recognition
memory consolidation is dependent on activation
of mTOR signaling in the dorsal hippocampus.
Learn. Mem. 20, 147–155 (2013).
98. Sarkar, S. N., Smith, L. T., Logan, S. M. & Simpkins, J. W.
Estrogen-induced activation of extracellular signal-
regulated kinase signaling triggers dendritic resident
mRNA translation. Neuroscience 170, 1080–1085
(2010).
99. Tuscher, J. J., Luine, V., Frankfurt, M. & Frick, K. M.
Estradiol-mediated spine changes in the dorsal
hippocampus and medial prefrontal cortex of
ovariectomized female mice depend on ERK
and mTOR activation in the dorsal hippocampus.
J. Neurosci. 36, 1483–1489 (2016).
100. Yue n, G. S ., McE wen , B. S. & A kama , K . T. L IM
kinase mediates estrogen action on the actin
depolymerization factor cofilin. Brain Res. 1379,
44–52 (2011).
101. Zhao, Y. etal. Estrogen receptor alpha and beta
regulate actin polymerization and spatial memory
through an SRC-1/mTORC2-dependent pathway
in the hippocampus of female mice. J. Steroid
Biochem. Mol. Biol. 174, 96–113 (2017).
102. Yildirim, M. etal. Estrogen and aging affect synaptic
distribution of phosphorylated LIM Kinase (LIMK) in
CA1 region of female rat hippocampus. Neuroscience
152, 360–370 (2008).
103. Kim, J. etal. Dorsal hippocampal actin polymerization
is necessary for activation of G-protein-coupled
estrogen receptor (GPER) to increase CA1 dendritic
spine density and enhance memory consolidation.
J. Neurosci. 39, 9598–9610 (2019).
104. Zhou, Y., Watters, J. J. & Dorsa, D. M. Estrogen
rapidly induces the phosphorylation of the cAMP
response element binding protein in rat brain.
Endocrinology 137, 2163–2166 (1996).
105. Zhao, Z., Fan, L., Fortress, A. M., Boulware, M. I.
& Frick, K. M. Hippocampal histone acetylation
regulates object recognition and the estradiol-induced
enhancement of object recognition. J. Neurosci. 32,
2344–2351 (2012).
106. Fortress, A. M., Kim, J., Poole, R. L., Gould, T. J.
& Frick, K. M. 17β-Estradiol regulates histone
alterations associated with memory consolidation and
increases Bdnf promoter acetylation in middle-aged
female mice. Learn. Mem. 21, 457–467 (2014).
107. Carrer, H. F., Araque, A. & Buño, W. Estradiol
regulates the slow Ca2+-activated K+ current in
hippocampal pyramidal neurons. J. Neurosci. 23,
6338–6344 (2003).
108. Kuma r, A. & Fo ster, T. C. 17β-estradiol benzoate
decreases the AHP amplitude in CA1 pyramidal
neurons. J. Neurophysiol. 88, 621–626 (2002).
109. Foy, M. R . etal. 17β-estradiol enhances NMDA
receptor-mediated EPSPs and long-term potentiation.
J. Neurophysiol. 81, 925–929 (1999).
110 . Pozzo- Mille r, L. D., I noue, T. & M urph y, D. D . Est radi ol
increases spine density and NMDA-dependent Ca2+
transients in spines of CA1 pyramidal neurons from
hippocampal slices. J. Neurophysiol. 81, 1404–1411
(1999).
111. Oberlander, J. G. & Woolley, C. S. 17β-estradiol
acutely potentiates glutamatergic synaptic
transmission in the hippocampus through distinct
mechanisms in males and females. J. Neurosci. 37,
12314–12327 (2017).
This article provides a detailed example of
how E2 can work through sex-specific molecular
mechanisms to produce the same functional
outcomes in both males and females.
112 . Bi, R., Broutman, G., Foy, M. R., Thompson, R. F. &
Baudry, M. The tyrosine kinase and mitogen-activated
protein kinase pathways mediate multiple effects of
estrogen in hippocampus. Proc. Natl Acad. Sci. USA
97, 3602–3607 (2000).
113 . Smith, C. C. & McMahon, L. L. 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. 25,
7780–7791 (2005).
114 . Liu, F. etal. Activation of estrogen receptor-β regulates
hippocampal synaptic plasticity and improves memory.
Nat. Neurosci. 11, 334–343 (2008).
115 . Xu, X. etal. Bisphenol-A rapidly promotes dynamic
changes in hippocampal dendritic morphology
through estrogen receptor-mediated pathway by
concomitant phosphorylation of NMDA receptor
subunit NR2B. Tox i c o l . A p p l . P h a r m a c o l . 249,
188–196 (2010).
116 . Avila, J. A . etal. Est radio l rap idly incr ease s Glu A2-
mushroom spines and decreases GluA2-filopodia
spines in hippocampus CA1. Hippocampus 27,
1224–1229 (2017).
117 . Waters, E. M. etal. Effects of estrogen and aging
on synaptic morphology and distribution of
phosphorylated Tyr1472 NR2B in the female rat
hippocampus. Neurobiol. Aging 73, 200–210 (2019).
118 . Potier, M. etal. Temporal memory and its
enhancement by estradiol requires surface dynamics
of hippocampal CA1 N-methyl-D-aspartate receptors.
Biol. Psychiatry 79, 735–745 (2016).
119 . Phan, A. etal. Rapid increases in immature synapses
parallel estrogen-induced hippocampal learning
enhancements. Proc. Natl Acad. Sci. USA 112,
16018–16023 (2015).
This comprehensive study links rapid oestrogen-
induced hippocampal spine formation with
modulation of glutamatergic synapses and
enhancement of spatial and recognition memory.
120. Vedder, L. C., Smith, C. C., Flannigan, A. E. &
McMahon, L. L. Estradiol-induced increase in
novel object recognition requires hippocampal
NR2B-containing NMDA receptors. Hippocampus 23,
108– 115 (2 013).
121. Smith, C. C. & McMahon, L. L. Estradiol-induced
increase in the magnitude of long-term potentiation
is prevented by blocking NR2B-containing receptors.
J. Neurosci. 26, 8517–8522 (2006).
122. Huang, G. Z. & Woolley, C. S. Estradiol acutely
suppresses inhibition in the hippocampus through a
sex-specific endocannabinoid and mGluR-dependent
mechanism. Neuron 74, 801–808 (2012).
123. Tabatadze, N., Huang, G., May, R. M., Jain, A. &
Woolley, C. S. Sex differences in molecular signaling at
inhibitory synapses in the hippocampus. J. Neurosci.
35, 11252–11265 (2015).
NATURE REVIEWS
|
NEUROSCIENCE
REVIEWS
124. Fugger, H. N., Foster, T. C., Gustafsson, J. &
Rissman, E. F. Novel effects of estradiol and estrogen
receptor alpha and beta on cognitive function.
Brain Res. 883, 258–264 (2000).
125. Walf , A. A., Ko once, C. J . & F rye, C . A. E stra diol 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. 89,
513– 521 ( 2008) .
126. Rissman, E. F., Heck, A. L., Leonard, J. E.,
Shupnik, M. A. & Gustafsson, J.-A. Disruption of
estrogen receptor beta gene impairs spatial learning
in female mice. Proc. Natl Acad. Sci. USA 99,
3996–4001 (2002).
127. Witty, C. F., Foster, T. C., Semple-Rowland, S. L. &
Daniel, J. M. Increasing hippocampal estrogen
receptor alpha levels via viral vectors increases
MAP kinase activation and enhances memory in aging
rats in the absence of ovarian estrogens. PLoS ONE 7,
e51385 (2012).
128. Hanson, A. M. etal. A-C estrogens as potent and
selective estrogen receptor-beta agonists (SERBAs)
to enhance memory consolidation under low-estrogen
conditions. J. Med. Chem. 61, 4720–4738 (2018).
129. Frick, K. M. Estrogens and age-related memory
decline in rodents: what have we learned and where
do we go from here? Horm. Behav. 55, 2–23 (2009).
130. Boulware, M. I., Kent, B. A. & Frick, K. M. The impact
of age-related ovarian hormone loss on cognitive
and neural function. Curr. Top. Behav. Neurosci. 10,
165–184 (2012).
131. Mehra, R. D., Sharma, K., Nyakas, C. & Vij, U.
Estrogen receptor α and β immunoreactive neurons
in normal adult and aged female rat hippocampus:
a qualitative and quantitative study. Brain Res. 1056,
22–35 (2005).
132. Zhang, Q.-G. etal. Estrogen attenuates ischemic
oxida tive dam age via a n estrog en recep tor α-mediated
inhibition of NADPH oxidase activation. J. Neurosci.
29, 13823–13836 (2009).
133. Daniel, J. M. Estrogens, estrogen receptors,
and female cognitive aging: the impact of timing.
Horm. Behav. 63, 231–237 (2013).
134. Luine, V. & Frankfurt, M. Estrogenic regulation
of memory: the first 50 years. Horm. Behav. 121,
104711 (202 0).
135. Foster, T. C. Role of estrogen receptor alpha and beta
expression and signaling on cognitive function during
aging. Hippocampus 22, 656–669 (2012).
136. Warren, S. G. & Juraska, J. M. Spatial and nonspatial
learning across the rat estrous cycle. Behav. Neurosci.
111, 259–266 (1997).
137. Daniel, J. M., Roberts, S. L. & Dohanich, G. P.
Effects of ovarian hormones and environment on
radial maze and water maze performance of female
rats. Physiol. Behav. 66, 11–20 (1999).
138. Chesler, E. J. & Juraska, J. M. Acute administration of
estrogen and progesterone impairs the acquisition
of the spatial Morris water maze in ovariectomized
rats. Horm. Behav. 38, 234–242 (2000).
139. Frick, K. M. & Berger-Sweeney, J. Spatial reference
memory and neocortical neurochemistry vary with the
estrous cycle in C57BL/6 mice. Behav. Neurosci. 115 ,
229–237 (2001).
140. Sandstrom, N. J. & Williams, C. L. Memory retention
is modulated by acute estradiol and progesterone
replacement. Behav. Neurosci. 115 , 384–393 (2001).
141. Galea, L. A., Kavaliers, M., Ossenkopp, K. P. &
Hampson, E. Gonadal hormone levels and spatial
learning performance in the Morris water maze in male
and female meadow voles, Microtus pennsylvanicus.
Horm. Behav. 29, 106–125 (1995).
142. Packard, M. G. & Teather, L. A. Intra-hippocampal
estradiol infusion enhances memory in ovariectomized
rats. Neuroreport 8, 3009–3013 (1997).
This pioneering study demonstrates that
intrahippocampally administered E2 rapidly
enhances spatial memory consolidation within
just 2 h after training.
143. Daniel, J. M., Fader, A. J., Spencer, A. L. &
Dohanich, G. P. Estrogen enhances performance of
female rats during acquisition of a radial arm maze.
Horm. Behav. 32, 217–225 (1997).
144. Bimonte, H. A. & Denenberg, V. H. Estradiol facilitates
performance as working memory load increases.
Psychoneuroendocrinology 24, 161–173 (1999).
145. Luine, V. N., Richards, S. T., Wu, V. Y. & Beck, K. D.
Estradiol enhances learning and memory in a spatial
memory task and effects levels of monoaminergic
neurotransmitters. Horm. Behav. 34, 149–162
(1998).
146. Gibbs, R. B. & Johnson, D. A. Sex-specific effects of
gonadectomy and hormone treatment on acquisition
of a 12-arm radial maze task by Sprague Dawley rats.
Endocrinology 149, 3176–3183 (2008).
147. Nelson, B. S., Springer, R. C. & Daniel, J. M.
Antagonism of brain insulin-like growth factor-1
receptors blocks estradiol effects on memory
and levels of hippocampal synaptic proteins in
ovariectomized rats. Psychopharmacology 231,
899–907 (2014).
148. Sinopoli, K. J., Floresco, S. B. & Galea, L. A. M.
Systemic and local administration of estradiol into
the prefrontal cortex or hippocampus differentially
alters working memory. Neurobiol. Learn. Mem. 86,
293–304 (2006).
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. 112 6 , 176–182
(2006).
150. Fonseca, C. S. etal. Object recognition memory
and temporal lobe activation after delayed estrogen
replacement therapy. Neurobiol. Learn. Mem. 101,
19–25 (2013).
151. Inagaki, T., Gautreaux, C. & Luine, V. Acute estrogen
treatment facilitates recognition memory consolidation
and alters monoamine levels in memory-related brain
areas. Horm. Behav. 58, 415–426 (2010).
152. Luine, V. N., Jacome, L. F. & MacLusky, N. J.
Rapid enhancement of visual and place memory by
estrogens in rats. Endocrinology 144, 2836–2844
(2003).
153. Gresack, J. E. & Frick, K. M. Post-training estrogen
enhances spatial and object memory consolidation
in female mice. Pharmacol. Biochem. Behav. 84,
112 – 119 ( 2 00 6) .
154. Pereira, L. M., Bastos, C. P., de Souza, J. M.,
Ribeiro, F. M. & Pereira, G. S. Estradiol enhances
object recognition memory in Swiss female mice
by activating hippocampal estrogen receptor α.
Neurobiol. Learn. Mem. 114, 1–9 (2014).
155. Tuscher, J. J., Taxier, L. R., Schalk, J. C., Haertel, J. M.
& Frick, K. M. Chemogenetic suppression of medial
prefrontal-dorsal hippocampal interactions prevents
estrogenic enhancement of memory consolidation in
female mice. eNeuro https://doi.org/10.1523/
ENEURO.0451-18.2019 (2019).
This study leverages novel chemogenetic tools to
demonstrate that activity of multiple brain regions
in concert is required for oestrogenic enhancement
of memory consolidation.
156. Gervais, N. J., Jacob, S., Brake, W. G. & Mumby, D. G.
Systemic and intra-rhinal-cortical 17-β estradiol
administration modulate object-recognition memory
in ovariectomized female rats. Horm. Behav. 64,
642–652 (2013).
157. Gervais, N. J., Hamel, L. M., Brake, W. G. &
Mumby, D. G. Intra-perirhinal cortex administration
of estradiol, but not an ERβ agonist, mod ulates
object-recognition memory in ovariectomized rats.
Neurobiol. Learn. Mem. 133, 89–99 (2016).
158. Taxier, L. R., Philippi, S. M., Fortress, A. M. &
Frick, K. M. Dickkopf-1 blocks 17β-estradiol-enhanced
object memory consolidation in ovariectomized female
mice. Horm. Behav. 11 4, 104545 (2019).
159. Luine, V. & Frankfurt, M. Interactions between
estradiol, BDNF and dendritic spines in promoting
memory. Neuroscience 239, 34–45 (2013).
160. Warren, S. G., Humphreys, A. G., Juraska, J. M. &
Greenough, W. T. LTP varies across the estrous
cycle: enhanced synaptic plasticity in proestrus rats.
Brain Res. 703, 26–30 (1995).
161. Good, M., Day, M. & Muir, J. L. Cyclical changes
in endogenous levels of oestrogen modulate the
induction of LTD and LTP in the hippocampal CA1
region. Eur. J. Neurosci. 11, 4476–4480 (1999).
162. Vedder, L. C., Bredemann, T. M. & McMahon, L. L.
Estradiol replacement extends the window of
opportunity for hippocampal function. Neurobiol.
Aging 35, 2183–2192 (2014).
163. Ferguson, J. N., Young, L. J. & Insel, T. R.
The neuroendocrine basis of social recognition.
Front. Neuroendocrinol. 23, 200–224 (2002).
164. Gabor, C. S., Phan, A., Clipperton-Allen, A. E.,
Kavaliers, M. & Choleris, E. Interplay of oxytocin,
vasopressin, and sex hormones in the regulation of
social recognition. Behav. Neurosci. 126, 97–109
(2012).
165. Sánchez-Andrade, G. & Kendrick, K. M. Roles of
α- and β-estrogen receptors in mouse social
recognition memory: effects of gender and the estrous
cycle. Horm. Behav. 59, 114–122 (2011).
166. Hlinˇáck, Z. Social recognition in ovariectomized
and estradiol-treated female rats. Horm. Behav. 27,
159–166 (1993).
167. Ta n g , A . C . e t a l . E f f e c t s o f l o n g - t e r m e s t r o g e n
replacement on social investigation and social memory
in ovariectomized C57BL/6 mice. Horm. Behav. 47,
350–357 (2005).
168. Spiteri, T. & Ågmo, A. Ovarian hormones modulate
social recognition in female rats. Physiol. Behav. 98,
247–250 (2009).
169. Choleris, E. etal. An estrogen-dependent four-gene
micronet regulating social recognition: a study with
oxytocin a nd estro gen rece ptor-α and -β knockout
mice. Proc. Natl Acad. Sci. USA 100, 6192–6197
(2003).
170. Ferguson, J. N., Aldag, J. M., Insel, T. R. & Young, L. J.
Oxytocin in the medial amygdala is essential for
social recognition in the mouse. J. Neurosci. 21,
8278–8285 (2001).
171. Choleris, E. etal. Involvement of estrogen
receptor α, β and oxytocin in social discrimination:
a detailed behavioral analysis with knockout
female mice. Genes Brain Behav. 5, 528–539
(2006).
172. Spiteri, T. etal. The role of the estrogen receptor α
in the medial amygdala and ventromedial nucleus
of the hypothalamus in social recognition, anxiety
and aggression. Behav. Brain Res. 210, 211–220
(2010).
173. Phan, A., Lancaster, K. E., Armstrong, J. N.,
MacLusky, N. J. & Choleris, E. Rapid effects of
estrogen receptor α and β selective agonists on
learning and dendritic spines in female mice.
Endocrinology 152, 1492–1502 (2011).
174. Lymer, J., Robinson, A., Winters, B. D. & Choleris, E.
Rapid effects of dorsal hippocampal G-protein
coupled estrogen receptor on learning in female
mice. Psychoneuroendocrinology 77, 131–140
(2017).
175. Lymer, J. M. etal. Estrogens and their receptors in the
medial amygdala rapidly facilitate social recognition in
female mice. Psychoneuroendocrinology 89, 30–38
(2018).
176. Morgan, M. A. & Pfaff, D. W. Effects of estrogen
on activity and fear-related behaviors in mice.
Horm. Behav. 40, 472–482 (2001).
177. Jasnow, A. M., Schulkin, J. & Pfaff, D. W.
Estrogen facilitates fear conditioning and increases
corticotropin-releasing hormone mRNA expression
in the central amygdala in female mice. Horm. Behav.
49, 197–205 (2006).
178. Hiroi, R. & Neumaier, J. F. Differential effects of
ovarian steroids on anxiety versus fear as measured
by open field test and fear-potentiated startle.
Behav. Brain Res. 166, 93–100 (2006).
179. Markus, E. J. & Zecevic, M. Sex differences and
estrous cycle changes in hippocampus-dependent
fear conditioning. Psychobiology 25, 246–252
(1997).
180. Gupta, R. R., Sen, S., Diepenhorst, L. L., Rudick, C. N.
& Maren, S. Estrogen modulates sexually dimorphic
contextual fear conditioning and hippocampal
long-term potentiation (LTP) in rats. Brain Res. 888,
356–365 (2001).
181. McDermott, C. M., Liu, D., Ade, C. & Schrader, L. A.
Estradiol replacement enhances fear memory
formation, impairs extinction and reduces
COMT expression levels in the hippocampus of
ovariectomized female mice. Neurobiol. Learn. Mem.
118 , 167–177 (2015).
182. Matsumoto, Y. K., Kasai, M. & Tomihara, K. The
enhancement effect of estradiol on contextual fear
conditioning in female mice. PLoS ONE 13, e0197441
(2018).
183. Barha, C. K., Dalton, G. L. & Galea, L. A. Low doses
of 17α -estradiol a nd 17β-estradiol facilitate, whereas
higher doses of estrone and 17α- and 17β-estradiol
impair, contextual fear conditioning in adult female
rats. Neuropsychopharmacology 35, 547–559
(2010).
184. Rivas-Arancibia, S. & Vazquez-Pereyra, F. Hormonal
modulation of extinction responses induced by sexual
steroid hormones in rats. Life Sci. 54, PL363–PL367
(1994).
185. Yuan, D. L. & Chambers, K. C. Estradiol accelerates
extinction of a conditioned taste aversion in
female and male rats. Horm. Behav. 36, 1–16
(1999).
186. Chang, Y.-J. etal. Estrogen modulates sexually
dimorphic contextual fear extinction in rats through
estrogen receptor β. Hippocampus 19, 1142–1150
(2009).
www.nature.com/nrn
REVIEWS
187. Milad, M. R., Igoe, S. A., Lebron-Milad, K. &
Novales, J. E. Estrous cycle phase and
gonadal hormones influence conditioned fear
extin ction. Neuroscience 164, 887–895
(2009).
This article defines E2 as an important modulator
of fear extinction learning.
188. Zeida n, M. A. etal . Estrad iol modu lates media l
prefrontal cortex and amygdala activity during fear
extin ction in women an d femal e rats. Biol. Psychiatry
70, 920–927 (2011).
189. Graham, B. M. & Milad, M. R. Blockade of estrogen
by hormonal contraceptives impairs fear extinction
in female rats and women. Biol. Psychiatry 73,
371–378 (2013).
190. Graham, B. M. & Scott, E. Effects of systemic estradiol
on fear extinction in female rats are dependent on
interactions between dose, estrous phase, and
endogenous estradiol levels. Horm. Behav. 97,
67–74 (2018).
191. de Castilhos, J., Forti, C. D., Achaval, M. &
Rasia-Filho, A. A. Dendritic spine density of
posterodorsal medial amygdala neurons can
be affected by gonadectomy and sex steroid
manipulations in adult rats: a Golgi study. Brain Res.
1240, 73–81 (2008).
192. Ferri, S. L., Hildebrand, P. F., Way, S. E. &
Flanagan-Cato, L. M. Estradiol regulates markers of
synaptic plasticity in the hypothalamic ventromedial
nucleus and amygdala of female rats. Horm. Behav.
66, 409–420 (2014).
193. Amano, T., Unal, C. T. & Paré, D. Synaptic correlates
of fear extinction in the amygdala. Nat. Neurosci. 13,
489–494 (2010).
194. Shansky, R. M. etal. Estrogen promotes stress
sensitivity in a prefrontal cortex–amygdala
pathway. Cereb. Cortex 20, 2560–2567
(2010).
195. Maeng, L. Y. etal. Estradiol shifts interactions
between the infralimbic cortex and central amygdala
to enhance fear extinction memory in female rats.
J. Neurosci. Res. 95, 163–175 (2017).
196. Rey, C. D., Lipps, J. & Shansky, R. M. Dopamine D1
receptor activation rescues extinction impairments
in low-estrogen female rats and induces cortical
layer-specific activation changes in prefrontal–
amygdala circuits. Neuropsychopharmacology 39,
1282–1289 (2014).
197. Lynch, J., Cullen, P. K., Jasnow, A. M. & Riccio, D. C.
Sex differences in the generalization of fear as a
function of retention intervals. Learn. Mem. 20,
628–632 (2013).
198. Lynch, J. F., Winiecki, P., Vanderhoof, T., Riccio, D. C.
& Jasnow, A. M. Hippocampal cytosolic estrogen
receptors regulate fear generalization in females.
Neurobiol. Learn. Mem. 130, 83–92 (2016).
199. Ooishi, Y. etal. Modulation of synaptic plasticity in the
hippocampus by hippocampus-derived estrogen and
androgen. J. Steroid Biochem. Mol. Biol. 131, 37–51
(2012).
200. Hojo, Y. etal. Adult male rat hippocampus synthesizes
estradiol from pregnenolone by cytochromes
P45017α and P450 aromatase localized in
neurons. Proc. Natl Acad. Sci. USA 101, 865–870
(2004).
201. Wang, W. etal. Memory-related synaptic plasticity
is sexually dimorphic in rodent hippocampus.
J. Neurosci. 38, 7935–7951 (2018).
202. Jain, A., Huang, G. Z. & Woolley, C. S. Latent sex
differences in molecular signaling that underlies
excitatory synaptic potentiation in the hippocampus.
J. Neurosci. 39, 1552–1565 (2019).
203. Fester, L. etal. Control of aromatase in hippocampal
neurons. J. Steroid Biochem. Mol. Biol. 160, 9–14
(2016).
204. Vierk, R. etal. Aromatase inhibition abolishes LTP
generation in female but not in male mice. J. Neurosci.
32, 8116–8126 (2012).
205. Zhou, L. etal. Oestradiol-induced synapse formation
in the female hippocampus: roles of oestrogen
receptor subtypes. J. Neuroendocrinol. 26, 439–447
(2014).
206. Lu, Y. etal. Neuron-derived estrogen regulates
synaptic plasticity and memory. J. Neurosci. 39,
2792–2809 (2019).
207. Brandt, N. & Rune, G. M. Sex-dependency of
oestrogen-induced structural synaptic plasticity:
inhibition of aromatase versus application of estradiol
in rodents. Eur. J. Neurosci. https://doi.org/10.1111/
ejn.14541 (2019).
208. Jacome, L. F. etal. Gonadal hormones rapidly enhance
spatial memory and increase hippocampal spine
density in male rats. Endocrinology 157, 1357–1362
(2016).
This study shows that rapid E2 signalling produces
parallel effects on hippocampal structure and
function in males comparable to historical findings
in females.
209. Koss, W. A., Haertel, J. M., Philippi, S. M. & Frick, K. M.
Sex differences in the rapid cell signaling mechanisms
underlying the memory-enhancing effects of
17β-estradiol. eNeuro https://doi.org/10.1523/
ENEURO.0267-18.2018 (2018).
210. Frye, C. A., Rhodes, M. E. & Dudek, B. Estradiol to
aged female or male mice improves learning in
inhibitory avoidance and water maze tasks. Brain Res.
1036 , 101–108 (2005).
211. Packard, M. G. Posttraining estrogen and memory
modulation. Horm. Behav. 34, 126–139 (1998).
212. Heikkinen, T., Puoliväli, J., Liu, L., Rissanen, A.
& Tanila, H. Effects of ovariectomy and estrogen
treatment on learning and hippocampal
neurotransmitters in mice. Horm. Behav. 41, 22–32
(2002).
213. Pierman, S. etal. Activational effects of estradiol
and dihydrotestosterone on social recognition and
the arginine-vasopressin immunoreactive system in
male mice lacking a functional aromatase gene.
Horm. Behav. 54, 98–106 (2008).
214. Alejandre-Gomez, M., Garcia-Segura, L. M. &
Gonzalez-Burgos, I. Administration of an inhibitor
of estrogen biosynthesis facilitates working memory
acquisition in male rats. Neurosci. Res. 58, 272–277
(2007).
215. Koss, W. A. & Frick, K. M. Activation of androgen
receptors protects intact male mice from memory
impairments caused by aromatase inhibition.
Horm. Behav. 111, 96–104 (2019).
216. Koss, W. A. & Frick, K. M. Sex differences in
hippocampal function. J. Neurosci. Res. 95, 539–562
(2017).
217. Frick, K. M., Kim, J., Tuscher, J. J. & Fortress, A. M.
Sex steroid hormones matter for learning and
memory: estrogenic regulation of hippocampal
function in male and female rodents. Learn. Mem. 22,
472–493 (2015).
218. Prendergast, B. J., Onishi, K. G. & Zucker, I. Female
mice liberated for inclusion in neuroscience and
biomedical research. Neurosci. Biobehav. Rev. 40,
1–5 (2014).
219. Clayton, J. A. Studying both sexes: a guiding
principle for biomedicine. FASE B J . 30, 519–524
(2016).
220. Clayton, J. A. Applying the new SABV (sex as a
biological variable) policy to research and clinical care.
Physiol. Behav. 187, 2–5 (2018).
221. Brooks, C. E. & Clayton, J. A. Sex/gender influences
on the nervous system: basic steps toward clinical
progress. J. Neurosci. Res. 95, 14–16 (2017).
222. Woitowich, N. C. & Woodruff, T. K. Implementation of
the NIH sex-inclusion policy: Attitudes and opinions
of study section members. J. Womens Health 28,
9–16 (2018).
223. Sandoval, A., Elahi, H. & Ploski, J. E. Genetically
engineering the nervous system with CRISPR-Cas.
eNeuro https://doi.org/10.1523/ENEURO.0419-
19.2020 (2020) .
224. Mitchnick, K. A. etal. Dissociable involvement
of estrogen receptors in perirhinal cortex-
mediated object-place memory in male rats.
Psychoneuroendocrinology 107, 98–108 (2019).
225. Kim, J. & Frick, K. M. Distinct effects of estrogen
receptor antagonism on object recognition and
spatial memory consolidation in ovariectomized
mice. Psychoneuroendocrinology 85, 110–114
(2017).
226. Tuscher, J. J. etal. Inhibition of local estrogen
synthesis in the hippocampus impairs hippocampal
memory consolidation in ovariectomized female mice.
Horm. Behav. 83, 60–67 (2016).
227. Bayer, J. etal. The effect of estrogen
synthesis inhibition on hippocampal memory.
Psychoneuroendocrinology 56, 213–225 (2015).
228. Gervais, N. J. etal. Adverse effects of aromatase
inhibition on the brain and behavior in a
nonhuman primate. J. Neurosci. 39, 918–928
(2019).
229. Morris, R. Developments of a water-maze
procedure for studying spatial learning in the rat.
J. Neurosci. Methods 11, 47–60 (1984).
230. Vorhees, C. V. & Williams, M. T. Morris water maze:
procedures for assessing spatial and related forms
of learning and memory. Nat. Protoc. 1, 848–858
(2006).
231. Olton, D. S. The radial arm maze as a tool in
behavioral pharmacology. Physiol. Behav. 40,
793–797 (1987).
232. Olton, D. S. & Papas, B. C. Spatial memory and
hippocampal function. Neuropsychologia 17,
669–682 (1979).
233. Ennaceur, A. & Delacour, J. A new one-trial test
for neurobiological studies of memory in rats.
1: behavioral data. Behav. Brain Res. 31, 47–59
(1988).
234. Ennaceur, A. & Aggleton, J. P. Spontaneous
recognition of object configurations in rats: effects
of fornix lesions. Exp. Brain Res. 100, 85–92
(1994).
235. Phillips, R. G. & LeDoux, J. E. Differential contribution
of amygdala and hippocampus to cued and contextual
fear conditioning. Behav. Neurosci. 106, 274–285
(1992).
236. Frick, K. M., Fortress, A. M. in The Maze Book:
Theories, Practice, and Protocols for Testing Rodent
Cognition (ed Bimonte-Nelson, H.) 165–210
(Springer, 2015).
237. Naftolin, F., Ryan, K. J. & Petro, Z. Aromatization
of androstenedione by the anterior hypothalamus of
adult male and female rats. Endocrinology 90,
295–298 (1972).
238. Sato, S. M. & Woolley, C. S. Acute inhibition of
neurosteroid estrogen synthesis suppresses status
epilepticus in an animal model. eLife 5, e12917
(2016).
239. Prange-Kiel, J. etal. Inhibition of hippocampal
estrogen synthesis causes region-specific
downregulation of synaptic protein expression in
hippocampal neurons. Hippocampus 16, 464–471
(2006).
240. Zhou, L. etal. Aromatase inhibitors induce
spine synapse loss in the hippocampus of
ovariectomized mice. Endocrinology 151,
115 3 –11 6 0 (2 010 ) .
241. Bailey, D. J., Ma, C., Soma, K. K. & Saldanha, C. J.
Inhibition of hippocampal aromatization impairs
spatial memory performance in a male songbird.
Endocrinology 154, 4707–4714 (2013).
242. Blaustein, J. D. Treatments for breast cancer that
affect cognitive function in postmenopausal women.
Poli cy I nsigh ts B ehav. B rain Sci. 4, 170–177
(2017).
243. Bimonte-Nelson, H. A., Acosta, J. I. & Talboom, J. S.
Neuroscientists as cartographers: mapping the
crossroads of gonadal hormones, memory and age
using animal models. Molecules 15, 6050–6105
(2010).
244. Wise, P. M. Alterations in the proestrous pattern of
median eminence LHRH, serum LH, FSH, estradiol
and progesterone concentrations in middle-aged rats.
Life Sci. 31, 165–173 (1982).
245. Richardson, S. J. & Nelson, J. F. Follicular depletion
during the menopausal transition. Ann. N. Y. Acad. Sci.
592, 13–20 (1990).
246. Talboom, J. S., Williams, B. J., Baxley, E. R.,
West, S. G. & Bimonte-Nelson, H. A. Higher levels
of estradiol replacement correlate with better
spatial memory in surgically menopausal young
and middle-aged rats. Neurobiol. Learn. Mem. 90,
155–163 (2008).
247. Markham, J. A., Pych, J. C. & Juraska, J. M.
Ovarian hormone replacement to aged ovariectomized
female rats benefits acquisition of the Morris water
maze. Horm. Behav. 42, 284–293 (2002).
248. Gresack, J. E., Kerr, K. M. & Frick, K. M. Life-long
environmental enrichment differentially affects
the mnemonic response to estrogen in young,
middle-aged, and aged female mice. Neurobiol.
Learn. Mem. 88, 393–408 (2007).
249. Singh, M., Meyer, E. M., Millard, W. J. & Simpkins, J. W.
Ovarian steroid deprivation results in a reversible
learning impairment and compromised cholinergic
function in female Sprague-Dawley rats. Brain Res.
644, 305–312 (1994).
250. Foster, T. C., Sharrow, K. M., Kumar, A. & Masse, J.
Interaction of age and chronic estradiol replacement
on memory and markers of brain aging. Neurobiol.
Aging 24, 839–852 (2003).
251. Frick, K. M., Fernandez, S. M. & Bulinski, S. C.
Estrogen replacement improves spatial reference
memory and increases hippocampal synaptophysin
in aged female mice. Neuroscience 115, 547–558
(2002).
252. Vaucher, E. etal. Estrogen effects on object
memory and cholinergic receptors in young and
old female mice. Neurobiol. Aging 23, 87–95
(2002).
NATURE REVIEWS
|
NEUROSCIENCE
REVIEWS
253. Prakapenka, A. V. etal. Contrasting effects
of individual versus combined estrogen and
progestogen regimens as working memory load
increases in middle-aged ovariectomized rats: one plus
one does not equal two. Neurobiol. Aging 64, 1–14
(2018).
254. Gresack, J. E. & Frick, K. M. Effects of continuous
and intermittent estrogen treatments on memory
in aging female mice. Brain Res. 1115 , 135–147
(2006).
255. Markowska, A. L. & Savonenko, A. V. Effectiveness
of estrogen replacement in restoration of cognitive
function after long-term estrogen withdrawal in
aging rats. J. Neurosci. 22, 10985–10995
(2002).
256. Daniel, J. M., Hulst, J. L. & Berbling, J. L.
Estradiol replacement enhances working
memory in middle-aged rats when initiated
immediately after ovariectomy but not after
a long-term period of ovarian hormone
deprivation. Endocrinology 147, 607–614
(2006).
257. Gresack, J. E., Kerr, K. M. & Frick, K. M.
Short-term environmental enrichment decreases
the mnemonic response to estrogen in young,
but not aged, female mice. Brain Res. 116 0, 91–101
(2007).
258. Aenlle, K. K. & Foster, T. C. Aging alters the expression
of genes for neuroprotection and synaptic function
following acute estradiol treatment. Hippocampus 20,
1047 –1060 (2010) .
259. Adams, M. M. etal. Estrogen and aging affect the
subcellular distribution of estrogen receptor-α in
the hippocampus of female rats. J. Neurosci. 22,
3608–3614 (2002).
260. Waters, E. M. etal. Estrogen and aging affect
the synaptic distribution of estrogen receptor
beta-immunoreactivity in the CA1 region of female
rat hippocampus. Brain Res. 1379, 86–97
(2011).
Acknowledgements
The Frick laboratory is supported by the US National
Institutes of Health (R01MH107886, 2R15GM118304-02,
F31MH118822 and F32MH118782), the Alzheimer’s
Association (SAGA-17-419092), the University of Wisconsin
System, the University of Wisconsin-Milwaukee Research
Foundation, the University of Wisconsin-Milwaukee Office
of Undergraduate Research and the University of
Wisconsin-Milwaukee College of Letters and Science.
Author contributions
The authors contributed equally to all aspects of the article.
Competing interests
K.M.F. is a co-founder and Chief Scientific Officer of Estrigenix
Therapeutics Inc., and is listed as an inventor of a pending
patent held by the University of Wisconsin-Milwaukee,
Marquette University and Concordia University Wisconsin
entitled “Substituted (4-hydroxyphenyl)cycloalkane and
(4-hydroxyphenyl)cycloalkene compounds and uses thereof
as selective agonists of the estrogen receptor beta isoform
for enhanced memory consolidation”, inventors W. A.
Donaldson, D. S. Sem and K.M.F. (WO2018183800A1). The
other authors declare no competing interests.
Peer review inf ormat ion
Nature Reviews Neuroscience thanks H. Bimonte-Nelson, who
co-reviewed with V. Bernaud, and the other, anonymous,
reviewer(s) for their con tribution to the peer revi ew of this work.
Publisher’s note
Springer Nature remains neutral with regard to jurisdictional
claims in published maps and institutional affiliations.
© Sprin ger Nature Limit ed 2020
www.nature.com/nrn
REVIEWS
... Hippocampus is mainly involved in the formation of hippocampus-dependent contextual fear memory, executive functions, memory retrieval process, and cognitive abilities [10][11][12]. Estrogen modulates hippocampus-dependent learning and memory tasks in males and females [13][14][15] through various molecular mechanisms [17] ( Fig. 1; Table 1). In males, estrogen mediates the hippocampus-dependent memory consolidation process via CREB (cAMP response element binding protein) phosphorylation (ERK-independent mechanism), while in females, it takes the ERK (extracellular receptor kinase), Akt, and MAPK (mitogen activated protein kinase) signaling pathways [16,18]. ...
... The pituitary hormone prolactin (PRL) has traditionally been associated with lactogenesis; however, over the last few years, it has been associated with more than 300 functions in the organism, including neurogenesis, neuroprotection, and cognition [6]. While the literature on the procognitive effects of E2 is extensive (see Taxier et al. [7] for a review), reports regarding the effects of PRL on learning and memory are scarce and have yielded mixed results [8][9][10]. For instance, hyperprolactinemic male rats show impaired object recognition without modifications in spatial learning [11], while the lack of PRL receptors in both female and male mice results in learning and memory deficits [12]. ...
Article
Full-text available
The neuroprotective actions of the ovarian hormone 17β-estradiol (E2) against different brain lesions have been constantly confirmed in a variety of models including kainic acid (KA) lesions. Similarly, the pituitary hormone prolactin (PRL), traditionally associated with lactogenesis, has recently been linked to a large diversity of functions, including neurogenesis, neuroprotection, and cognitive processes. While the mechanisms of actions of E2 as regards its neuroprotective and behavioral effects have been extensively explored, the molecular mechanisms of PRL related to these roles remain under investigation. The current study aimed to investigate whether the simultaneous administration of PRL and a low dose of E2 prevents the KA-induced cognitive deficit and if this action is associated with changes in hippocampal neuronal density. Ovariectomized (OVX) rats were treated with saline, PRL, and/or E2 in the presence or absence of KA. Neuroprotection was assessed by Nissl staining and neuron counting. Memory was evaluated with the novel object recognition test (NOR). On their own, both PRL and E2 prevented short- and long-term memory deficits in lesioned animals and exerted neuroprotection against KA-induced excitotoxicity in the hippocampus. Interestingly, the combined hormonal treatment was superior to either of the treatments administered alone as regards improving both memory and neuronal survival. Taken together, these results point to a synergic effect of E2 and PRL in the hippocampus to produce their behavioral, proliferative, and neuroprotective effects.
... 17β-Estradiol (E2) is a critical sex steroid hormone secreted by the mammalian ovary (Frick et al., 2015), primarily in endometrium cells and follicular granulosa cells. Furthermore, considerable evidence supports that E2 is a neuromodulator of learning and memory in human brain (Taxier et al., 2020). As one of the potential environmental endogenous estrogens, human and mammal urine and feces contain E2 Tang et al., 2022b;Tang et al., 2022a). ...
Article
Full-text available
17β-Estradiol (E2) is a critical sex steroid hormone, which has significant effects on the endocrine systems of both humans and animals. E2 is also believed to play neurotrophic and neuroprotective roles in the brain. Biosensors present a powerful tool to detect E2 because of their small, efficient, and flexible design. Furthermore, Biosensors can quickly and accurately obtain detection results with only a small sampling amount, which greatly meets the detection of the environment, food safety, medicine safety, and human body. This review focuses on previous studies of biosensors for detecting E2 and divides them into non-biometric sensors, enzyme biosensors, antibody biosensors, and aptamer biosensors according to different bioreceptors. The advantages, disadvantages, and design points of various bioreceptors for E2 detection are analyzed and summarized. Additionally, applications of different bioreceptors of E2 detection are presented and highlight the field of environmental monitoring, food and medicine safety, and disease detection in recent years. Finally, the development of E2 detection by biosensor is prospected.
Article
Estrogens are believed to modulate cognitive functions in part through the modulation of synaptic transmission in the cortex and hippocampus. Administration of 17β‐estradiol (E2) can rapidly enhance excitatory synaptic transmission in the hippocampus and facilitate excitatory synaptic transmission in rat lateral entorhinal cortex via activation of the G protein‐coupled estrogen receptor‐1 (GPER1). To assess the mechanisms through which GPER1 activation facilitates synaptic transmission, we assessed the effects of acute 10 nM E2 administration on pharmacologically isolated evoked excitatory and inhibitory synaptic currents in layer II/III entorhinal neurons. Female Long‐Evans rats were ovariectomized between postnatal day (PD) 63 and 74 and implanted with a subdermal E2 capsule to maintain continuous low levels of E2. Electrophysiological recordings were obtained between 7 and 20 days after ovariectomy. Application of E2 for 20 min did not significantly affect AMPA or NMDA receptor‐mediated excitatory synaptic currents. However, GABA receptor‐mediated inhibitory synaptic currents (IPSCs) were markedly reduced by E2 and returned towards baseline levels during the 20‐min washout period. The inhibition of GABA‐mediated IPSCs was blocked in the presence of the GPER1 receptor antagonist G15. GPER1 can modulate protein kinase A (PKA), but blocking PKA with intracellular KT5720 did not prevent the E2‐induced reduction in IPSCs. GPER1 can also stimulate extracellular signal‐regulated kinase (ERK), a negative modulator of GABA A receptors, and blocking activation of ERK with PD90859 prevented the E2‐induced reduction of IPSCs. E2 can therefore result in a rapid GPER1 and ERK signaling‐mediated reduction in GABA‐mediated IPSCs. This provides a novel mechanism through which E2 can rapidly modulate synaptic excitability in entorhinal layer II/III neurons and may also contribute to E2 and ERK‐dependent alterations in synaptic transmission in other brain areas.
Article
The ability to spatial learning and the functionality of the spatial memory, and also the stressful reactivity of the hypothalamus-pituitary-adrenocortical axis (the HPA axis) were investigated in adult male and female rats, subjected to moderate acute hypoxia on the 2nd day of life, and then to chronic administration of the serotonin reuptake inhibitor fluoxetine. When testing the ability to spatial training in adult rats subjected to neonatal hypoxia, an increase in the latent period of reaching the platform in the Morris water maze in the first try in the first of five days of training was found. The results of memory analysis in the novel object recognition test and in the Morris water maze testify that hypoxia did not cause memory deficiency in adult animals. Moreover, hypoxia improved the memory indices on the first day in males and on the fourth day in females after removing the platform from the pool compared to the corresponding control values. The content of corticosterone in blood plasma of males in response to memory testing did not differ between control and experimental groups and was characterized by higher rates than in females of corresponding groups. Hypoxia increased the HPA axis reactivity in females, which was combined with a longer memory storage. Fluoxetine normalized the indicator of spatial learning, did not cause changes in control animals and did not change the identified improvement of memory in hypoxic rats without administration of this drug. The new data obtained expand the idea of the long-term effect of neonatal normobaric moderate hypoxia on the spatial memory and the HPA axis reactivity depending on sex and emphasize the absence of the harmful effect of fluoxetine on spatial memory in both control rats and in rats with the effects of hypoxia.
Article
Most of mammalian physiology is under the control of biological rhythms, including the endocrine system with time-varying hormone secretion. Precision neuroimaging studies provide unique insights into how the endocrine system dynamically regulates aspects of the human brain. Recently, we established estrogen's ability to drive widespread patterns of connectivity and enhance the global efficiency of large-scale brain networks in a woman sampled every 24 h across 30 consecutive days, capturing a complete menstrual cycle. Steroid hormone production also follows a pronounced sinusoidal pattern, with a peak in testosterone between 6 and 7 A.M. and nadir between 7 and 8 P.M. To capture the brain's response to diurnal changes in hormone production, we carried out a companion precision imaging study of a healthy adult man who completed MRI and venipuncture every 12–24 h across 30 consecutive days. Results confirmed robust diurnal fluctuations in testosterone, 17β-estradiol—the primary form of estrogen—and cortisol. Standardized regression analyses revealed widespread associations between testosterone, estradiol, and cortisol concentrations and whole-brain patterns of coherence. In particular, functional connectivity in the Dorsal Attention Network was coupled with diurnally fluctuating hormones. Further, comparing dense-sampling datasets between a man and a naturally cycling woman revealed that fluctuations in sex hormones are tied to patterns of whole-brain coherence in both sexes and to a heightened degree in the male. Together, these findings enhance our understanding of steroid hormones as rapid neuromodulators and provide evidence that diurnal changes in steroid hormones are associated with patterns of whole-brain functional connectivity.
Article
Full-text available
Estrogen regulates a wide range of neuronal functions in the brain, such as dendritic spine formation, remodeling of synaptic plasticity, cognition, neurotransmission, and neurodevelopment. Estrogen interacts with intracellular estrogen receptors (ERs) and membrane-bound ERs to produce its effect via genomic and non-genomic pathways. Any alterations in these pathways affect the number, size, and shape of dendritic spines in neurons associated with psychiatric diseases. Increasing evidence suggests that estrogen fluctuation causes changes in dendritic spine density, morphology, and synapse numbers of excitatory and inhibitory neurons differently in males and females. In this review, we discuss the role of estrogen hormone in rodents and humans based on sex differences. First, we explain estrogen role in learning and memory and show that a high estrogen level alleviates the deficits in learning and memory. Secondly, we point out that estrogen produces a striking difference in emotional memories in men and women, which leads them to display sex-specific differences in underlying neuronal signaling. Lastly, we discuss that fluctuations in estrogen levels in men and women are related to neuropsychiatric disorders, including schizophrenia, autism spectrum disorder (ASD), attention deficit hyperactivity disorder (ADHD), bipolar disorder (BPD), major depressive disorder (MDD), substance use disorder (SUD), and anxiety disorders.
Article
Synaptic plasticity is believed to be the cellular basis for experience-dependent learning and memory. Although long-term depression (LTD), a form of synaptic plasticity, is caused by the activity-dependent reduction of cell surface α-amino-3-hydroxy-5-methyl-4-isoxazole propionic acid (AMPA)-type glutamate receptors (AMPA receptors) at postsynaptic sites, its regulation by neuronal activity is not completely understood. In this study, we showed that the inhibition of toll-like receptor-9 (TLR9), an innate immune receptor, suppresses N-methyl-d-aspartate (NMDA)-induced reduction of cell surface AMPA receptors in cultured hippocampal neurons. We found that inhibition of TLR9 also blocked NMDA-induced activation of caspase-3, which plays an essential role in the induction of LTD. siRNA-based knockdown of TLR9 also suppressed the NMDA-induced reduction of cell surface AMPA receptors, although the scrambled RNA had no effect on the NMDA-induced trafficking of AMPA receptors. Overexpression of the siRNA-resistant form of TLR9 rescued the AMPA receptor trafficking abolished by siRNA. Furthermore, NMDA stimulation induced rapid mitochondrial morphological changes, mitophagy, and the binding of mitochondrial DNA (mtDNA) to TLR9. Treatment with dideoxycytidine and mitochondrial division inhibitor-1, which block mtDNA replication and mitophagy, respectively, inhibited NMDA-dependent AMPA receptor internalization. These results suggest that mitophagy induced by NMDA receptor activation releases mtDNA and activates TLR9, which plays an essential role in the trafficking of AMPA receptors during the induction of LTD.
Article
Full-text available
Aging-related memory impairment and pathological memory disorders such as Alzheimer’s disease differ between males and females, and yet little is known about how aging-related changes in the transcriptome and chromatin environment differ between sexes in the hippocampus. To investigate this question, we compared the chromatin accessibility landscape and gene expression/alternative splicing pattern of young adult and aged mouse hippocampus in both males and females using ATAC-seq and RNA-seq. We detected significant aging-dependent changes in the expression of genes involved in immune response and synaptic function and aging-dependent changes in the alternative splicing of myelin sheath genes. We found significant sex-bias in the expression and alternative splicing of hundreds of genes, including aging-dependent female-biased expression of myelin sheath genes and aging-dependent male-biased expression of genes involved in synaptic function. Aging was associated with increased chromatin accessibility in both male and female hippocampus, especially in repetitive elements, and with an increase in LINE-1 transcription. We detected significant sex-bias in chromatin accessibility in both autosomes and the X chromosome, with male-biased accessibility enriched at promoters and CpG-rich regions. Sex differences in gene expression and chromatin accessibility were amplified with aging, findings that may shed light on sex differences in aging-related and pathological memory loss.
Article
Full-text available
The multitude of neuronal subtypes and extensive interconnectivity of the mammalian brain presents a substantial challenge to those seeking to decipher its functions. While the molecular mechanisms of several neuronal functions remain poorly characterized, advances in Next-Generation Sequencing (NGS) and gene-editing technology have begun to close this gap. The Clustered Regularly Interspaced Palindromic Repeats - CRISPR Associated Protein (CRISPR-Cas) system has emerged as a powerful genetic tool capable of manipulating the genome of essentially any organism and cell type, an attribute which has advanced our understanding of complex neurological diseases by enabling the rapid generation of novel, disease-relevant in vitro and transgenic animal models. In this review, we discuss recent developments in the rapidly accelerating field of CRISPR-mediated genome engineering. We begin with an overview of the canonical function of the CRISPR platform, followed by a functional review of its many adaptations, with an emphasis on its applications for genetic interrogation of the normal and diseased nervous system. Additionally, we discuss limitations of the CRISPR editing system and suggest how future modifications to existing platforms may advance our understanding of the brain.
Article
Full-text available
Activation of the membrane estrogen receptor G-protein-coupled estrogen receptor (GPER) in ovariectomized mice via the GPER agonist G-1 mimics the beneficial effects of 17β-estradiol (E2) on hippocampal CA1 spine density and memory consolidation, yet the cell-signaling mechanisms mediating these effects remain unclear. The present study examined the role of actin polymerization and c-Jun N-terminal kinase (JNK) phosphorylation in mediating effects of dorsal hippocampally-infused G-1 on CA1 dendritic spine density and consolidation of object recognition and spatial memories in ovariectomized mice. We first showed that object learning increased apical CA1 spine density in the dorsal hippocampus (DH) within 40 min. We then found that DH infusion of G-1 increased both CA1 spine density and phosphorylation of the actin polymerization regulator cofilin, suggesting that activation of GPER may increase spine morphogenesis through actin polymerization. As with memory consolidation in our previous work (Kim et al., 2016), effects of G-1 on CA1 spine density and cofilin phosphorylation depended on JNK phosphorylation in the DH. Also consistent with our previous findings, E2-induced cofilin phosphorylation was not dependent on GPER activation. Finally, we found that infusion of the actin polymerization inhibitor, latrunculin A, into the DH prevented G-1 from increasing apical CA1 spine density and enhancing both object recognition and spatial memory consolidation. Collectively, these data demonstrate that GPER-mediated hippocampal spinogenesis and memory consolidation depend on JNK and cofilin signaling, supporting a critical role for actin polymerization in the GPER-induced regulation of hippocampal function in female mice.SIGNIFICANCE STATEMENTEmerging evidence suggests that GPER activation mimics effects of 17β-estradiol on hippocampal memory consolidation. Unlike canonical estrogen receptors, GPER activation is associated with reduced cancer cell proliferation and thus, understanding the molecular mechanisms through which GPER regulates hippocampal function may provide new avenues for the development of drugs that provide the cognitive benefits of estrogens without harmful side effects. Here, we demonstrate that GPER increases CA1 dendritic spine density and hippocampal memory consolidation in a manner dependent on actin polymerization and JNK phosphorylation. These findings provide novel insights into the role of GPER in mediating hippocampal morphology and memory consolidation, and may suggest first steps towards new therapeutics that more safely and effectively reduce memory decline in menopausal women.
Article
Full-text available
The memory-enhancing effects of 17β-estradiol (E2) depend upon rapid activation of several cell-signaling cascades within the dorsal hippocampus (DH). Among the many cell-signaling pathways that mediate memory processes, Wnt/β-catenin signaling has emerged as a potential key player because of its importance to hippocampal development and synaptic plasticity. However, whether E2 interacts with Wnt/β-catenin signaling to promote memory consolidation is unknown. Therefore, the present study examined whether Wnt/β-catenin signaling within the DH is necessary for E2-induced memory consolidation in ovariectomized mice tested in the object recognition and object placement tasks. Ovariectomized C57BL/6 mice received immediate post-training infusions of E2 or vehicle into the dorsal third ventricle plus the endogenous Wnt/β-catenin antagonist Dickkopf-1 (Dkk-1) or vehicle into the DH to assess whether the memory-enhancing effects of E2 depend on activation of Wnt/β-catenin signaling. Our results suggest that Dkk-1 blocks E2-induced memory enhancement as hypothesized, but may do so by only moderately blunting Wnt/β-catenin signaling while concurrently activating Wnt/JNK signaling. The current study provides novel insights into the mechanisms through which E2 enhances memory consolidation in the DH, as well as critical information about the mechanistic actions of Dkk-1.
Article
Full-text available
The importance of the dorsal hippocampus (DH) in mediating the memory-enhancing effects of the sex-steroid hormone 17β-estradiol (E2) is well established. However, estrogen receptors (ERs) are highly expressed in other brain regions that support memory formation, including the medial prefrontal cortex (mPFC). The mPFC and DH interact to mediate the formation of several types of memory, and behavioral tasks that recruit the mPFC are enhanced by systemic E2 administration, making this region a prime candidate for investigating circuit-level questions regarding the estrogenic regulation of memory. Further, infusion of E2 directly into the DH increases dendritic spine density in both the DH and mPFC, and this effect depends upon rapid activation of cell-signaling pathways in the DH, demonstrating a previously unexplored interaction between the DH and mPFC that led us to question the role of the mPFC in object memory consolidation and the necessity of DH-mPFC interactions in the memory-enhancing effects of E2. Here, we found that infusion of E2 directly into the mPFC of ovariectomized mice increased mPFC apical spine density and facilitated object recognition and spatial memory consolidation, demonstrating that E2 in the mPFC increases spinogenesis and enhances on memory consolidation. Next, chemogenetic suppression of the mPFC blocked the beneficial effects of DH-infused E2 on memory consolidation, indicating that systems-level DH-mPFC interactions are necessary for the memory-enhancing effects of E2. Together, these studies provide evidence that E2 in the mPFC mediates memory formation, and reveal that the DH and mPFC act in concert to support the memory-enhancing effects of E2 in female mice.
Article
Full-text available
17β-estradiol (E2) is produced from androgens via the action of the enzyme aromatase. E2 is known to be made in neurons in the brain, but its precise functions in the brain are unclear. Here, we used a forebrain-neuron-specific aromatase knock-out (FBN-ARO-KO) mouse model to deplete neuron-derived E2 in the forebrain of mice and thereby elucidate its functions. FBN-ARO-KO mice showed a 70-80% decrease in aromatase and forebrain E2 levels compared with FLOX controls. Male and female FBN-ARO-KO mice exhibited significant deficits in forebrain spine and synaptic density, as well as hippocampal-dependent spatial reference memory, recognition memory, and contextual fear memory, but had normal locomotor function and anxiety levels. Reinstating forebrain E2 levels via exogenous in vivo E2 administration was able to rescue both the molecular and behavioral defects in FBN-ARO-KO mice. Furthermore, in vitro studies using FBN-ARO-KO hippocampal slices revealed that, whereas induction of long-term potentiation (LTP) was normal, the amplitude was significantly decreased. Intriguingly, the LTP defect could be fully rescued by acute E2 treatment in vitro Mechanistic studies revealed that FBN-ARO-KO mice had compromised rapid kinase (AKT, ERK) and CREB-BDNF signaling in the hippocampus and cerebral cortex. In addition, acute E2 rescue of LTP in hippocampal FBN-ARO-KO slices could be blocked by administration of a MEK/ERK inhibitor, further suggesting a key role for rapid ERK signaling in neuronal E2 effects. In conclusion, the findings provide evidence of a critical role for neuron-derived E2 in regulating synaptic plasticity and cognitive function in the male and female brain.SIGNIFICANCE STATEMENT The steroid hormone 17β-estradiol (E2) is well known to be produced in the ovaries in females. Intriguingly, forebrain neurons also express aromatase, the E2 biosynthetic enzyme, but the precise functions of neuron-derived E2 is unclear. Using a novel forebrain-neuron-specific aromatase knock-out mouse model to deplete neuron-derived E2, the current study provides direct genetic evidence of a critical role for neuron-derived E2 in the regulation of rapid AKT-ERK and CREB-BDNF signaling in the mouse forebrain and demonstrates that neuron-derived E2 is essential for normal expression of LTP, synaptic plasticity, and cognitive function in both the male and female brain. These findings suggest that neuron-derived E2 functions as a novel neuromodulator in the forebrain to control synaptic plasticity and cognitive function.
Article
Full-text available
Although 17β-estradiol (E2) is known to regulate hippocampal function, the specific contributions of hippocampally-synthesized E2 remain unclear. Infusion of the aromatase inhibitor letrozole into the dorsal hippocampus (DH) of ovariectomized mice disrupts object recognition and object placement memory consolidation, suggesting that DH-synthesized E2 is essential for memory. However, the role of DH-synthesized E2 in memory among male rodents is unknown. Here, we examined effects of aromatase inhibition on memory consolidation in male mice. Intact and gonadectomized mice were infused with vehicle or letrozole into the DH immediately post-training in object placement and object recognition tasks. Letrozole blocked memory in both tasks among gonadectomized males only, suggesting that circulating androgens, or a rise in hippocampal androgens due to aromatase inhibition, may support memory consolidation in intact males. To test this hypothesis, intact males were infused with the androgen receptor antagonist flutamide into the DH after object training. A dose-dependent impairment was observed in both tasks, indicating that blocking androgen signaling can impair memory consolidation. To test if hippocampal androgen receptor activation protected intact males from the impairing effects of letrozole, a non-impairing dose of flutamide was co-infused with letrozole. Co-administration of both drugs blocked object placement and object recognition memory consolidation, demonstrating that letrozole impairs memory in intact males only if androgen receptors are blocked. Together, these data suggest that DH-synthesized E2 and androgen receptor activation may work in concert to mediate memory consolidation in intact males, such that androgen receptor activation protects against memory impairments caused by aromatase inhibition.
Article
This review highlights fifty years of progress in research on estradiol's role in regulating behavior(s). It was initially thought that estradiol was only involved in regulating estrus/menstrual cycles and concomitant sexual behavior, but it is now clear that estradiol also influences the higher order neural function of cognition. We provide a brief overview of estradiol's regulation of memory and some mechanisms which underlie its effects. Given systemically or directly into the hippocampus, to ovariectomized female rodents, estradiol or specific agonists, enhance learning and/or memory in a variety of rodent cognitive tasks. Acute (within minutes) or chronic (days) treatments enhance cognitive functions. Under the same treatment conditions, dendritic spine density on pyramidal neurons in the CA1 area of the hippocampus and medial prefrontal cortex increase which suggests that these changes are an important component of estrogen's ability to impact memory processes. Noradrenergic, dopaminergic and serotoninergic activity are also altered in these areas following estrogen treatments. Memory enhancements and increased spine density by estrogens are not limited to females but are also present in castrate males. In the next fifty years, neuroscientists need to determine how currently described neural changes mediate improved memory, how interactions among areas important for memory promote memory and the potential significance of neurally derived estrogens in normal cognitive processing. Answering these questions may provide significant advances for treatment of dementias as well as age and neuro-degenerative disease related memory loss.
Article
Sex dependent differences in learning and memory formation in humans have been frequently shown. The mechanisms underlying the formation and retention of memories are assumed to involve synaptic plasticity in the hippocampus. Estradiol was shown to effect synaptic plasticity in the hippocampus of rodents. The effects after exogenous application of estradiol to animals frequently produce inconsistent results, in particular if sex is not considered in the studies. Recently we provided evidence that locally synthesized estradiol, plays an essential role on synaptic connectivity in the hippocampus of females but not of male mice. In females inhibition of local estradiol synthesis leads to synapse loss, which results from impairment of long‐ term potentiation and dephosphorylation of cofilin, and thereby the destabilization of postsynaptic dendritic spines. This sex‐dependency was also seen in the classical aromatase knock‐out mouse. Intriguingly, no differences between sexes have been found in a conditional forebrain specific aromatase knock‐out mouse. Altogether, the findings underscore the necessity of including ‘Sex as Biological Variable' in studies of sex steroid‐induced synaptic plasticity. This article is protected by copyright. All rights reserved.
Article
Estrogens and the estrogen receptors (ER)- ERα, ERβ, and the G-protein coupled estrogen receptor (GPER)- are implicated in various forms of hippocampus (HPC)-dependent memory. However, the involvement of ER-related mechanisms in perirhinal cortex (PRh), which is necessary for object memory, remains much less clear. Moreover, there is a paucity of data assessing ER contributions to cognition in males,despite documented sex differences at the cellular level.We hypothesized that estrogens in PRh are important for object memory in males, assessingthe role of 17-βestradiol (E2), ERα, ERβ, GPER, and their downstream signaling pathways, in PRh-mediated object-in-place (OiP)memory in gonadally-intact male rats. Intra-PRh administration of E2 enhanced both long-term memory (LTM; 24 h)and short-term memory (STM; 20 min). Conversely, aromatase inhibition with letrozole impaired LTM and STM. The semi-selective ER inhibitor ICI 182780 impaired LTM, but not STM. This effect may be due to inhibition of ERβ, as the ERβagonist DPN, but not ERαagonist PPT, enhanced LTM. GPER was also found to be necessary in PRh, as the antagonist G15 impaired both LTM and STM. Western blot analyses demonstrated that phosphorylation levels of the extracellular signal-related kinase (ERK2 isoform), awell-establisheddownstream signaling pathway activated by estrogens through ERα/ERβ, was elevated in PRh 5 min following OiP learning.We also reportincreased levels of c-Jun N-terminal kinase (JNK; p46 and p54 isoforms)phosphorylation in PRh 5 min following learning,consistent with recent research linking GPER activation and JNK signaling in the HPC. This effect was abolished by intra-PRh administration of G15, but not letrozole, suggesting that JNK signaling is triggered via GPER activation during OiP learning, and is possibly E2-independent, similar to findings in the HPC. These results, therefore, reveal interesting dissociations between the roles of various ERs, possibly involving both estrogen-dependent and independent mechanisms, in PRh-mediated object-place learning in male rats.
Article
Breast cancer patients using aromatase inhibitors (AIs) as an adjuvant therapy often report side effects including hot flashes, mood changes, and cognitive impairment. Despite long-term use in humans, little is known about the effects of continuous AI administration on the brain and cognition. We used a primate model of human cognitive aging, the common marmoset, to examine the effects of a 4-week daily administration of the AI Letrozole (20 μg, p.o.) on cognition, anxiety, thermoregulation, brain estrogen content, and hippocampal pyramidal cell physiology. Letrozole treatment was administered to both male and female marmosets and reduced peripheral levels of estradiol (E2), but unexpectedly increased E2 levels in the hippocampus. Spatial working memory and intrinsic excitability of hippocampal neurons were negatively affected by the treatment possibly due to increased hippocampal E2. While no changes in hypothalamic E2 were observed, thermoregulation was disrupted by Letrozole in females only, indicating some impact on hypothalamic activity. These findings suggest adverse effects of AIs on the primate brain and call for new therapies that effectively prevent breast cancer recurrence while minimizing side effects that further compromise quality of life.