The female sex hormone estrogen has been ascribed potent neuroprotective properties. It signals by binding and activating estrogen receptors that, depending on receptor subtype and upstream or downstream effectors, can mediate gene transcription and rapid non-genomic actions. In this way, estrogen receptors in the brain participate in modulating neural differentiation, proliferation, neuroinflammation, cholesterol metabolism, synaptic plasticity, and behavior. Circulating sex hormones decrease in the course of aging, more rapidly at menopause in women, and slower in men. This review will discuss what this drop entails in terms of modulating neuroprotection and resilience in the aging brain downstream of spatiotemporal estrogen receptor alpha (ERα) and beta (ERβ) signaling, as well as in terms of the sex differences observed in Alzheimer’s disease (AD) and Parkinson’s disease (PD). In addition, controversies related to ER expression in the brain will be discussed. Understanding the spatiotemporal signaling of sex hormones in the brain can lead to more personalized prevention strategies or therapies combating neurodegenerative diseases.

The female sex hormone estrogen (E2, 17β-estradiol) is essential for reproduction. It is mainly produced in the ovary after puberty and by the placenta at pregnancy (in the form of estriol), and its levels drop sharply at menopause. Like all sex hormones, E2 is a lipophilic steroid that can diffuse through the blood–brain barrier (BBB) to reach the brain parenchyma, where it regulates various processes linked to development, reproduction, emotion, and cognition. Additionally, E2 can be synthesized locally in the brain of men and women through cholesterol metabolism. Both E2 and the male sex hormone testosterone (an androgen) have been ascribed neuroprotective properties. This is perhaps most evident in ischemic brain injury, where sex hormones elicit neuroprotection that most likely contribute to the sex differences observed in the stroke incidence and survival rate at all adult ages [1,2]. At reproductive age, the incidence and mortality rates of stroke is higher in men than in women; however, after menopause more women are affected by stroke than men, suggesting a neuroprotective contribution of sex hormones [3]. The molecular and pathophysiological mechanisms behind such neuroprotective events are still largely unknown, but clearly both E2 and androgens modulate several neuroprotective pathways including immune response, neurogenesis, glial cell functions, and response to excitotoxicity [2,4–8]. In this review, we will discuss the expression and roles of the estrogen receptors in the healthy aging brain, as well as in neurodegenerative diseases such as AD and PD. We will conclude by discussing if there is a clinical potential in using hormonal therapies to combat neurodegeneration.

Three estrogen receptor subtypes exist; Estrogen receptor alpha (ERα), estrogen receptor beta (ERβ), and G-protein-coupled estrogen receptor 1 (GPER1). ERα and ERβ belong to the steroid activated nuclear receptor family of transcription factors. Upon E2 binding, they homo- or hetero-dimerize, recruit coactivators, and bind to estrogen response element (ERE) regulatory sites on the DNA to mediate transcription of genes involved in proliferation, differentiation, and survival (Figure 1). In addition to this classical genomic transcriptional activation, ERs may also tether to other transcription factors and act independently of E2, e.g., downstream of growth factor signaling (Figure 1) [9]. There are also several splice variants of the ERs, of which the ERβ splice variants have been studied most. These can dimerize with both ERα and ERβ and bind DNA, but lack, or have reduced, ligand binding activity, thereby modulating ER activity [9,10].

While the classical ERα and ERβ belong to the nuclear receptor family of transcription factors, GPER1 is a membrane-bound G-protein-coupled receptor without transcriptional activity and is suggested to mediate E2's rapid non-genomic actions linked to e.g., calcium flux and kinase activations and can cross-talk with the classical ERs (Figure 1) [11,12]. The exact molecular mechanisms behind this crosstalk and its overall contribution to E2 signaling is still largely unknown.

Estrogen receptor expression in the brain

Both ERα and ERβ are overexpressed during neurodevelopment [13–15]. In rodents, ERs are responsible for masculinizing the brain during development since most of the androgens excreted by the developing testes from embryonic day 18 (E18) are converted into E2 by the aromatase enzyme (Cyp19a1) in the rodent brain [16]. In humans, however, the masculinization is mediated by brain androgen receptor (AR), although a role for ERs cannot be excluded [17,18]. Species differences in neuroendocrine control of adult brains also exist. For example, in the adult human brain, the highest expression of aromatase is found in thalamus, while rodents display the highest expression in amygdala [19]. There are also differences between humans and rodents in neuroendocrine control of gonadotropin-releasing hormone and in the distribution of ERs in different brain regions [20,21]. These important species differences should be taken into account when translating the influence of sex hormones between rodent and human brains.

Levels of ERs in the brain decrease after birth and their expression is confined to various brain regions. Studies mainly made on rodents show that, in the adult brain, both ERα and ERβ expression is overall low but with a wide distribution pattern [20,22–27]. However, this pattern of in particular ERβ, but also GPER1 and aromatase, is controversial due to issues related to antibody specificity to these proteins. This has resulted in confusion in the field of brain estrogen signaling. Several of the most widely used commercial ERβ antibodies have shown little or no specificity [28,29]. A recent study [28] could not detect any significant amounts of ERβ in the human brain using a validated antibody against human ERβ. In contrast, follow-up studies using the same antibody did detect ERβ in the mouse brain after optimizing antigen retrieval [30] or using frozen mouse brain sections [31]. In line with this, the expression of ERβ mRNA has previously been shown widely distributed in the rodent brain [32], which is similar to the expression pattern demonstrated by Merchenthaler and colleagues [23] using an in-house generated ERβ antibody. This latter study also detected different degree of overlap between ERβ and ERα immunoreactivity in different brain regions that correlated well with accumulation of 125I-labeled E2 in these regions [23]. In addition, a recent study using ERβ lineage tracing could detect significant amounts of ERβ in e.g., paraventricular nucleus (PVN), dorasal raphe nucles (DRN) and medial amygdala (MeA) of the mouse brain [33], while using multiple single cell sequencing data resources (i.e, Janelia portal https://hipposeq.janelia.org; Allen Brain Map https://portal.brain-map.org; Broad single cell portal https://singlecell.broadinstitute.org/single_cell) show varying but mostly low amounts of ER transcripts in both the rodent and human cortex and hippocampus. To date, the overall conclusion one can draw is that ERα is widely distributed at generally low levels in the rodent brain with highest expression in the amygdala and the hypothalamus, whereas ERβ expression is the main ER found in the cerebral cortex (although at low levels). ERβ is also expressed in the cerebellum, hippocampus, MeA, bed nucleus of the stria terminalis (BNST), PVN, anteroventral periventricular nucleus, substantia nigra, ventral tegmental area, and the DRN, which appears to be similar between rodents and humans [20,22–27]. Although there are no well-working commercial antibodies today for immunohistochemistry on GPER1 and aromatase, GPER1 is suggested to be expressed in most brain regions where either, or both, ERα or ERβ is expressed, including hippocampus, cerebral cortex, and the hypothalamus [11,34]. Only few studies have addressed aromatase mRNA distribution in the brain, showing that it is mainly expressed in the rat BNST and amygdala during development, and that its expression is much lower in adult brain areas, although this can be regulated by androgens and E2 [35,36].

Estrogen receptors in neuroendocrine signaling

A recent systematic review assessing the contribution of sex hormones on activation of neural circuits controlling emotion and cognition as measured by functional magnetic resonance imaging (fMRI) showed different activation pattern during the follicular and luteal phases in normally cycling healthy women in several brain regions such as amygdala, anterior hippocampus, and several cortical regions [37]. Thus, circulating E2 and progesterone (P4, which will not be discussed in this review) can influence neural circuits controlling emotional and cognitive processing in humans.

However, the underlying mechanisms behind sex hormonal modulation of neural circuits are not well understood. Several studies have demonstrated that E2 can rapidly influence synaptic transmission and dendritic spine formation [38–40], as well as long term potentiation (LTP) [1,39], likely through non-genomic and genomic actions, respectively. Both the mitogen-activated protein kinase (MAPK) family and the phosphatidylinositol 3 kinase (PI3K) are modulated by E2 to rapidly promote dendritic spine formation [41–43], which at least in part can be regulated through extranuclear ERα and ERβ [44–47], and their interaction with cell surface receptors such as insulin growth factor 1 receptor (IGF-1R) [48,49]. In addition, E2 enhances rat hippocampal N-methyl-D-aspartate (NMDA) receptor-mediated currents [50,51], and the loss of ERβ resulted in aberrant gamma-aminobutyric acid (GABA)ergic interneuron function in the mouse motor cortex [6], suggesting that ERs intricately modulate synaptic neurotransmission, LTP, and behavior. Studies on knockout (KO) mouse models have shown that mice lacking ERα have aberrant sexual behavior, linked to hypothalamic actions of this receptor [52], and that these mice have unaffected spatial learning but impaired performance in the passive avoidance tests, suggesting a role for ERα in non-hippocampal dependent (or non-spatial dependent) memory [53]. On the other hand, ERβ KO mice have normal sexual behavior (although female ERβ KO mice have reduced fertility [54]), impaired spatial learning [55], increased locomotor activity [6], and increased anxiety behavior [56], but not impaired performance in the passive avoidance test [53], suggesting that ERα and ERβ have some degree of complementary functions in cognition. Furthermore, selective ERβ activation can modulate microglia-mediated neuroinflammation [57] and protects oligodendrocytes against myelination damage in a mouse model of multiple sclerosis [58]. Similar effects were also seen upon selective ERα activation, although this neuroprotection appears mediated explicitly via astrocytes [59]. Since ERs have important functions in neurodevelopment it is important to discriminate between their developmental effects and their functional effects in the adult brain, nevertheless, ERs function clearly beyond controlling sexual behavior and reproduction.

In the course of healthy ageing a magnitude of brain-specific and systemic changes occur that have an impact on the resilience against adverse events. It has been demonstrated that the levels of both ERα and ERβ, found in synapses of CA1 neurons in the hippocampus of female rats, decrease with age [22,44,60]. If ER expression is linked to resilience in the brain, then this would imply that one level of neuroprotection is lost in the course of healthy ageing. However, since ERs function mainly as E2 binding proteins, the largest risk of their decreased activity in the brain would be when gonadal E2 levels drop. In women, a sharp drop in E2 levels occurs naturally at menopause. An increasing body of evidence suggest that the menopausal E2 drop in fact increases the vulnerability to adverse neurological events. This will be discussed in the following sections.

Menopausal loss of gonadal estrogen—loss of resilience?

Women experience a drastic decrease in E2 levels at menopause. In contrast, aged men have a slow decrease in testosterone levels. Although testosterone has also been ascribed neuroprotective functions [61,62], it is still debated whether testosterone acts through brain AR or via its conversion to E2 by brain aromatase to elicit neuroprotective functions in humans [63]. Likely both are true to varying extents in different brain regions and under different pathological conditions, which implies that there may be sex differences between the female and male brain with respect to neuroprotection mediated by circulating sex hormones. Perhaps the most convincing evidence that links systemic loss of estrogen during menopause to neurological changes is the increased risk of depression associated with the menopausal transition (reviewed in [64]). This risk is significantly reduced in perimenopausal women treated with estrogen [65–67], lending further support to the hypothesis that systemic E2 levels do influence behavior. A similar reduction in risk has not been demonstrated in postmenopausal women who are older when initiating estrogen therapy [68], which is in line with the ‘critical time period hypothesis’ stating that estrogen treatment must be initiated early in relation to menopause onset to provide neuroprotection [69]. In addition, the type of hormone treatment (HT; E2 or in combination with progestins) and its duration may also have a bearing on the risk of depression, but knowledge of this is limited. Similarly, increasing evidence put forward a link between menopause and risk of subjective cognitive decline [70], which is often reported by menopausal women [71], and is associated with increased risk of developing AD later in life [72]. A recent systematic review demonstrated that the menopausal transition associates negatively with attention, and verbal and working memory [71]. Additionally, premenopausal women that have undergone oophorectomy have an increased risk of developing depression [73–75] and dementia later in life [74]. Similar findings have been found in premenopausal women that have undergone hysterectomy [76,77], suggesting that early removal of both E2 and P4 in premenopausal women affects mental health.

It would therefore be predicted that HT use in these women may not only alleviate symptoms of depression and cognitive decline but also prevent a possible excess risk of dementia. However, data on HT use as a modulator of present and future cognitive decline in postmenopausal women is controversial, owing to misinterpretations and inconsistencies, that in turn is a result of limited study population and not taking crucial biases into account, such as age, education, socioeconomic factors, and comorbid diseases. Further, factors related to HT type and its initiation in relation to menopause onset (e.g., the ‘critical time period hypothesis’) have not been sufficiently taken into account. Interestingly, HT treatment including both E2 and progestins appears to not elicit the same neuroprotective effects on cognitive decline as with E2 alone, in fact progestins may antagonize the protective effects of E2 [64,78,79].

In the brain, E2 can be synthesized de novo, mainly in neurons, in a process called neurosteroidogenesis [80]. Therefore, in order to present an overview on estrogen signaling in the brain, it is warranted to include a discussion on neurosteroidogenesis and how aberrant control of this process may modulate local E2 production.

Cholesterol acts as precursor of sex-hormone synthesis, and therefore an interplay between brain cholesterol metabolism and E2 signaling in the brain can be assumed. Cholesterol homeostasis is essential for healthy brain functioning, and cholesterol imbalance can trigger pathological processes underlying neurodegenerative diseases. Homeostasis of cholesterol is maintained by continuous effluxes of BBB-permeable oxysterols (sidechain-oxidized cholesterol metabolites) between the circulation and the brain [81–83]. 24-S-hydroxycholesterol (24SOH) and 27-hydroxycholesterol (27OH) are key players in maintaining such balance [82], by e.g., activating Liver X Receptors (LXRs) [84]. LXR signaling is involved in multiple pathways in the brain, including regulation of cholesterol synthesis and neurosteroidogenesis [85]. Upon activation, LXR can dimerize with retinoic acid receptor (RAR) [86] and promote transcription of genes involved in neurosteroidogenesis [87]. In the periphery, LXR has been found to decrease breast cancer growth through an ER-dependent mechanism [88] and AR down-regulates LXR signaling, leading to cholesterol accumulation in prostate cancer cells [89]. Such cross-talk between steroid hormone receptors, LXR signaling, and cholesterol metabolism can be suggested to occur in a variety of cellular contexts, including brain cells, and could differ between men and women. Conditions as hypercholesterolemia or BBB disruption may also affect this interplay. It is well known that cardiovascular risk factors such as hypercholesterolemia increase the risk of developing cognitive decline and AD, and the flux of 27OH into the brain has been considered as the missing link between hypercholesterolemia and AD [90–92]. 27OH levels are linked to cardiovascular disease and, importantly, it has been described as an endogenous selective estrogen receptor modulator (SERM) [93]. As a SERM, 27OH has different ER affinity and cell type selectivity [93], likely having different effects in periphery and brain. It still remains unclear how oxysterols affect E2 mediated neuroprotection, and to what extent hypercholesterolemia (with high 27OH) corresponds to AD risk. In addition to 27OH, the enzyme CYP46A1 converting brain cholesterol into excretable 24SOH has received increasing attention in relation to aging and neurodegeneration [94–96]. CYP46A1 overexpressing mice showed sex-specific differences in cognitive function, with females displaying positive memory effects, higher resilience against neural aging, as well as increased aromatase levels and increased ERβ signaling in hippocampus. On the opposite, males were found to develop anxiety-like behavior and memory deterioration, accompanied by high levels of dihydrotestosterone (DHT) [97]. In a memory clinic cohort, higher 24OH in the cerebrospinal fluid was found to correlate with lower neurofilament-light chain (a marker of axonal damage) and phospho-Tau only in women [97]. Taken together, these evidence highlights the need to further study brain cholesterol metabolism in relation to E2 signaling and the sex differences observed in neurodegenerative diseases.

Clear sex differences exist in the risk and pathophysiological progression of several neurodegenerative diseases. These sex differences are perhaps most apparent in AD and PD, and ERs have been ascribed important roles in the risk and progression of these neurodegenerative diseases.

Estrogen receptors in Alzheimer’s disease

AD is the most common neurodegenerative disease, characterized by amyloid plaques of aggregated amyloid β peptide (Aβ), neurofibrillary tangles of hyperphosphorylated protein Tau, and neuroinflammation, leading to progressive neuronal atrophy, dementia, and death. Middle-aged women have a 2- to 3-fold increased risk compared with men in developing AD [98]. Although women on average live longer than men, it is clear that other factors also contribute to the increased AD risk in women, including sex hormones, genetic factors, and environment including geographic location [99–103]. In addition, women converts faster from mild cognitive impairment (MCI) to AD and present a more aggravated AD pathophysiology than men [104,105]. Women homozygous for APOE-ε4 run a greater risk of developing late-onset AD, while homozygous men run a greater risk of early-onset AD [106]. ApoE is involved in maintaining lipid/cholesterol metabolism in the brain, which in turn crosstalks with estrogen signaling (as discussed above). Taken together, this proposes a very complex interaction between sex hormones, cholesterol metabolism, genes and environment, which has restrained our understanding on the mechanisms behind the sex difference in AD. Nevertheless, an increasing body of evidence put forward E2 signaling through ERs as one important contributing factor to these sex differences.

E2 has been proposed to exhibit neuroprotective effects in experimental models of the AD brain [107,108]. However, the actual ER subtype contribution to the neuroprotection in AD is not well understood. Regarding ERα, studies have shown that this receptor is overexpressed in several neuronal nuclei of the basal forebrain, mammillary body, and hypothalamus in AD patients compared with sex- and age- matched healthy brains [109–112], while it is decreased in hippocampal neurons [113]. Some studies have also shown that ERα overexpression leads to increased Tau phosphorylation, while others show the opposite [114], and that ERα has protective effects on glutamate-mediated trauma in cultured rat neurons of female, but not male, origin [115]. The conflicting results have raised controversies to the actual contribution of ERα to neuroprotection in AD. However, it has been demonstrated that selective ERα activation in ovariectomized female transgenic AD mice (APP/PS1 and 3xTg AD mouse models) results in improved cognitive performance [116] and reduced Aβ levels in hippocampus, subiculum, and amygdala [117]. In addition, ERα gene polymorphisms in humans are associated with cognitive decline after menopause [118,119] and have been found in subsets of AD patients [120,121]. These data suggest that ERα has some role in AD risk and progression, although the molecular mechanisms and its overall neuroprotective contributions are still not clear.

In contrast, data on a neuroprotective role of ERβ in AD seems more consistent. A study on female AD brains detected decreased ERβ levels in the frontal cortex compared to age-matched controls, and they showed that ERβ was associated with mitochondria in these brains [122]. It appears that ligand binding to ERβ promotes its association with neuronal mitochondria and that loss of ERβ impairs the mitochondrial membrane potential and function [123–125]. These data could point toward neuronal exhaustion upon loss of ERβ. In line with this, ERβ KO mice display gradual progression of neurological deficits as they age, with significant loss of neuronal cell bodies throughout the brain in both sexes compared to wild-type littermates, in particular in substantia nigra [126]. Furthermore, studies using the 3xTg mouse model of AD demonstrated that selective ERβ activation is more effective than ERα activation in lowering Aβ levels in the frontal cortex and equally effective in the amygdala [117], and that long-term ERβ treatment improved spatial recognition memory, attenuated Aβ plaque formation, prolonged survival, and promoted physical health in these mice [127]. Although not well understood, molecular studies have suggested that, in addition to modulating mitochondrial health, ERβ can promote Aβ degradation through directly interacting with the autophagy machinery, thereby enhancing the clearance [128] and intracellular trafficking of Aβ [107], as well as by modulating microglia-mediated neuroinflammation [57] and interacting with cholesterol metabolism [97].

Estrogen receptors in Parkinson’s disease

Parkinson’s disease is caused by age-related neurodegeneration in substantia nigra and/or loss of dopaminergic neurons, leading to motor dysfunction. Oppositely to AD, PD is more common in men than in women, and the age of PD onset is generally also earlier in men [129]. Men also display a more aggravated disease progression [103], which is mirrored in animal models of PD where neurotoxic hydroxydopamine administration results in increased motor deficits and nigrostriatal dopamine (DA) loss in male compared with female animals [130]. Additionally, the expression of genes in substantia nigra and striatum involved in PD pathogenesis and dopamine machinery is different between male and female PD brains [131–133].

E2 is proposed to have a modulatory effect on PD [134], and observational studies of postmenopausal women show a decreased risk of PD in women taking HT [135]. Of great interest is also a study linking HT to lower severity during the early phase of PD in women, prior to L-DOPA treatment [136], suggesting that E2 treatment could be useful in patients that already have developed PD. Similar disease-modifying effects of E2 has not been observed in AD. The beneficial effects of E2 may be associated with its facilitation of rapid DA release in the striatum and nucleus accumbens shown in rats [137,138], as well as with the neuroprotective properties of physiological E2 levels discussed above. Interestingly a role for testosterone in mediating neuroprotection in PD has not been convincingly demonstrated. In contrast with E2 treatment, administration of testosterone or DHT (which cannot be aromatized to E2) does not protect against neurotoxicity in the 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) model of PD but rather worsens pathology [134]. This implies that testosterone cannot be aromatized efficiently enough to exhibit neuroprotection in mouse models of PD, or that it has effects opposite of E2 in the brain. In line with this, aromatase knockout mice that lack the capacity of synthesizing E2 had increased pathology in the MPTP model compared to wild-type mice [139]. It was also shown that ERs are necessary for the neuroprotective effects of E2 in the MPTP model since, in contrast with 17β-estradiol (E2), 17α-estradiol, which cannot bind to ERs, does not protect against neurotoxicity [140]. Both ERα and ERβ are suggested to participate in this protection. In two studies, selective ERα activation was demonstrated to exhibit neuroprotection by preventing DA depletion and loss of dopamine transporter in the striatum [141] and preventing nigral dopaminergic cell death [142] in rodent models of PD. Similar effects could not be observed with a selective synthetic ERβ agonist in these studies. However, another study found that the endogenously produced 5-androstene-3β, 17β-diol, an ERβ agonist, protects against neuroinflammation in the rotenone-induced rat model of PD [143], suggesting that type of PD model and type of selective ER modulators may influence the neuroprotective readouts. Furthermore, a polymorphism in the human ERβ gene was more common in both sexes of early-onset PD patients compared with late-onset patients [144], whereas ERα polymorphisms appear to not be associated with PD [145].

As reviewed above, there is compelling evidence that E2 has neuroprotective functions, and that this is mediated by different ERs in different brain regions. It is therefore warranted to ask if E2 or a selective ER agonist treatment could be a clinical option in combating neurodegenerative diseases. Observational studies point towards a decreased risk of AD and PD in women taking at least menopausal estrogenic replacement therapy (ERT, without progestins) compared with never HT users [78,79,135]. However, the results and interpretations of these observational studies varies [135], which has impeded any clinical recommendations on HT use with regards to dementia risks. In addition, the use of HT is controversial since it can increase the risk of breast cancer, thromboembolism, and stroke. Lately, reassessment of old studies and addition of new knowledge has convincingly demonstrated that the benefits of HT can outweigh these risks. In particular, a large study demonstrated no increased risk of stroke exist when HT is initiated early after menopause [146]. Additionally, new data show that when adjusting for confounding factors, there is no increased breast cancer associated with HT, and even a lower risk in women only taking ERT [147,148]. Clearly, larger and more detailed epidemiological studies are needed to assess the contribution of menopausal E2 loss on PD, AD and non-AD dementia risk, and which individuals that may particularly benefit from estrogenic treatment or preventive interventions.

The preclinical data that exist on ERβ as a neuroprotective factor is very interesting from a therapeutic point of view. Since ERβ is mainly expressed in the brain, intestine and the gonads [23,28,30], it may be a promising target avoiding the side effects of systemic ERα activation. BBB permeable ERβ selective ligands are already in clinical trials for alleviating menopausal mood symptoms, and it would be interesting to explore their clinical potential in preventing neurodegeneration.

Despite that E2 has been proposed as a neuroprotective factor in both AD and PD, it is interesting that women have an increased risk in developing AD, while men have an increased risk in being diagnosed with PD. How could loss of neuroprotective E2 at menopause increase the risk of AD but not of PD in women? And why does testosterone, which has also been ascribed neuroprotective roles, and which levels decrease much slower in aged men, not protect men from PD? Based on data described in this review, one can speculate that testosterone has, in the case of PD, the potential to be anti-neuroprotective, while in the case of AD be neuroprotective (possibly via its conversion to E2), and that brain-region specific differences exist in this neuroprotection. Thus, it is also tempting to speculate that, while testosterone is neuroprotective in regions affected in AD, such as cortex and hippocampus, it is not protective, or is not efficiently converted into E2, in PD affected regions, such as the midbrain, which in turn could be due to different spatial expression of aromatase between the two diseases. In addition, all aged women experience menopause, but why do only some develop AD? What other resilience factors cooperate with E2, and can local E2 production compensate postmenopausal drop of circulating sex hormones? Clearly, many factors interact with the risk of developing multifactorial AD and PD, including sex hormone levels, genes, comorbid diseases (such as cardiovascular disease), and the environment. In addition, interactions with neurodevelopmental processes should be taken into consideration, such as that microglia appears to display a more neuroprotective phenotype in females than males, and that this may originate from the surge in testosterone during the male brain masculinization [149,150]. Deciphering these interactions and the spatiotemporal involvement of sex hormone receptors will be tedious, warranting more accurate methods of detecting ERs and aromatase expression, and importantly requiring new integrative approaches combining preclinical, clinical, and detailed epidemiological data. Clearly, such integration can truly contribute to increase our knowledge on ER signaling in the ageing brain and whether this signaling impacts on resilience against neurodegenerative diseases, offering new clinical, more personalized, recommendations or treatment options for both men and women.

  • Understanding the spatiotemporal expression and signaling of estrogen receptors in the brain may offer new understanding to the normal and pathophysiological aging of the brain, and the sex differences observed in these processes.

  • Although great achievements have been made on the topic of sex hormone signaling in the brain, a deeper understanding of the molecular mechanisms involving the different estrogen receptor subtypes and their neuroprotective responses in the course of human healthy and pathological ageing is largely missing.

  • Integration of preclinical, clinical and epidemiological studies are needed to address the impact of estrogen receptor signaling in the ageing brain and their potential as therapeutic targets in neurodegeneration.

The authors declare that there are no competing interests associated with the manuscript.

This work was supported by the National Institute On Aging of the National Institutes of Health under award number [grant number R01AG065209]. The content is solely the responsibility of the authors and does not necessarily represent the official views of the National Institutes of Health.

The manuscript was written by S.M, K.L, P.N., and I.N. We thank Aphrodite Demetriou for ideas on graphic representations.

27OH

27-hydroxycholesterol

24SOH

24-S-hydroxycholesterol

DHT

dihydrotestosterone

ERT

estrogenic replacement therapy

LXR

Liver X Receptor

MCI

mild cognitive impairment

MPTP

1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine

RAR

retinoic acid receptor

SERM

selective estrogen receptor modulator

1.
Galea
L.A.
,
Frick
K.M.
,
Hampson
E.
,
Sohrabji
F.
and
Choleris
E.
(
2017
)
Why estrogens matter for behavior and brain health
.
Neurosci. Biobehav. Rev.
76
,
363
379
[PubMed]
2.
Céspedes Rubio Á
E.
,
Pérez-Alvarez
M.J.
,
Lapuente Chala
C.
and
Wandosell
F.
(
2018
)
Sex steroid hormones as neuroprotective elements in ischemia models
.
J. Endocrinol.
237
,
R65
R81
[PubMed]
3.
Meyer
M.R.
and
Barton
M.
(
2016
)
Chapter Ten - Estrogens and Coronary Artery Disease: New Clinical Perspectives
. In
Advances in Pharmacology
vol.
77
, (
Khalil
R.A.
, ed.), pp.
307
360
,
Academic Press
4.
Liu
M.
,
Kelley
M.H.
,
Herson
P.S.
and
Hurn
P.D.
(
2010
)
Neuroprotection of sex steroids
.
Minerva Endocrinol.
35
,
127
143
[PubMed]
5.
Suzuki
S.
,
Gerhold
L.M.
,
Böttner
M.
,
Rau
S.W.
,
Dela Cruz
C.
,
Yang
E.
et al.
(
2007
)
Estradiol enhances neurogenesis following ischemic stroke through estrogen receptors alpha and beta
.
J. Comp. Neurol.
500
,
1064
1075
[PubMed]
6.
Varshney
M.K.
,
Yu
N.Y.
,
Katayama
S.
,
Li
X.
,
Liu
T.
,
Wu
W.F.
et al.
(
2020
)
Motor function deficits in the estrogen receptor beta knockout mouse: Role on excitatory neurotransmission and myelination in the motor cortex
.
Neuroendocrinology
27
44
[PubMed]
7.
Villa
A.
,
Vegeto
E.
,
Poletti
A.
and
Maggi
A.
(
2016
)
Estrogens, neuroinflammation, and neurodegeneration
.
Endocr. Rev.
37
,
372
402
[PubMed]
8.
Lan
Y.L.
,
Zhao
J.
and
Li
S.
(
2014
)
Estrogen receptors’ neuroprotective effect against glutamate-induced neurotoxicity
.
Neurol. Sci.
35
,
1657
1662
[PubMed]
9.
Heldring
N.
,
Pike
A.
,
Andersson
S.
,
Matthews
J.
,
Cheng
G.
,
Hartman
J.
et al.
(
2007
)
Estrogen receptors: how do they signal and what are their targets
.
Physiol. Rev.
87
,
905
931
[PubMed]
10.
Kim
C.K.
,
Torcaso
A.
,
Asimes
A.
,
Chung
W.C.J.
and
Pak
T.R.
(
2018
)
Structural and functional characteristics of oestrogen receptor β splice variants: Implications for the ageing brain
.
J. Neuroendocrinol.
30
,
11.
Hadjimarkou
M.M.
and
Vasudevan
N.
(
2018
)
GPER1/GPR30 in the brain: crosstalk with classical estrogen receptors and implications for behavior
.
J. Steroid Biochem. Mol. Biol.
176
,
57
64
[PubMed]
12.
Revankar
C.M.
,
Cimino
D.F.
,
Sklar
L.A.
,
Arterburn
J.B.
and
Prossnitz
E.R.
(
2005
)
A transmembrane intracellular estrogen receptor mediates rapid cell signaling
.
Science
307
,
1625
1630
[PubMed]
13.
Fan
X.
,
Xu
H.
,
Warner
M.
and
Gustafsson
J.A.
(
2010
)
ERbeta in CNS: new roles in development and function
.
Prog. Brain Res.
181
,
233
250
[PubMed]
14.
Lemmen
J.G.
,
Broekhof
J.L.
,
Kuiper
G.G.
,
Gustafsson
J.A.
,
van der Saag
P.T.
and
van der Burg
B.
(
1999
)
Expression of estrogen receptor alpha and beta during mouse embryogenesis
.
Mech. Dev.
81
,
163
167
[PubMed]
15.
Varshney
M.K.
,
Inzunza
J.
,
Lupu
D.
,
Ganapathy
V.
,
Antonson
P.
,
Ruegg
J.
et al.
(
2017
)
Role of estrogen receptor beta in neural differentiation of mouse embryonic stem cells
.
Proc. Natl. Acad. Sci. USA
114
,
E10428
E10437
[PubMed]
16.
Simpson
E.R.
,
Clyne
C.
,
Rubin
G.
,
Boon
W.C.
,
Robertson
K.
,
Britt
K.
et al.
(
2002
)
Aromatase—a brief overview
.
Annu. Rev. Physiol.
64
,
93
127
[PubMed]
17.
Varshney
M.
and
Nalvarte
I.
(
2017
)
Genes, gender, environment, and novel functions of estrogen receptor beta in the susceptibility to neurodevelopmental disorders
.
Brain Sciences
7
,
[PubMed]
18.
Zuloaga
D.G.
,
Puts
D.A.
,
Jordan
C.L.
and
Breedlove
S.M.
(
2008
)
The role of androgen receptors in the masculinization of brain and behavior: what we've learned from the testicular feminization mutation
.
Horm. Behav.
53
,
613
626
[PubMed]
19.
Biegon
A.
(
2016
)
In vivo visualization of aromatase in animals and humans
.
Front. Neuroendocrinol.
40
,
42
51
[PubMed]
20.
Osterlund
M.K.
and
Hurd
Y.L.
(
2001
)
Estrogen receptors in the human forebrain and the relation to neuropsychiatric disorders
.
Prog. Neurobiol.
64
,
251
267
[PubMed]
21.
Goodman
R.L.
and
Lehman
M.N.
(
2012
)
Kisspeptin neurons from mice to men: similarities and differences
.
Endocrinology
153
,
5105
5118
[PubMed]
22.
Mehra
R.D.
,
Sharma
K.
,
Nyakas
C.
and
Vij
U.
(
2005
)
Estrogen receptor alpha and beta immunoreactive neurons in normal adult and aged female rat hippocampus: a qualitative and quantitative study
.
Brain Res.
1056
,
22
35
[PubMed]
23.
Merchenthaler
I.
,
Lane
M.V.
,
Numan
S.
and
Dellovade
T.L.
(
2004
)
Distribution of estrogen receptor alpha and beta in the mouse central nervous system: in vivo autoradiographic and immunocytochemical analyses
.
J. Comp. Neurol.
473
,
270
291
[PubMed]
24.
Mitra
S.W.
,
Hoskin
E.
,
Yudkovitz
J.
,
Pear
L.
,
Wilkinson
H.A.
,
Hayashi
S.
et al.
(
2003
)
Immunolocalization of estrogen receptor beta in the mouse brain: comparison with estrogen receptor alpha
.
Endocrinology
144
,
2055
2067
[PubMed]
25.
Gonzalez
M.
,
Cabrera-Socorro
A.
,
Perez-Garcia
C.G.
,
Fraser
J.D.
,
Lopez
F.J.
,
Alonso
R.
et al.
(
2007
)
Distribution patterns of estrogen receptor alpha and beta in the human cortex and hippocampus during development and adulthood
.
J. Comp. Neurol.
503
,
790
802
[PubMed]
26.
Dietrich
A.K.
,
Humphreys
G.I.
and
Nardulli
A.M.
(
2015
)
Expression of estrogen receptor α in the mouse cerebral cortex
.
Mol. Cell. Endocrinol.
406
,
19
26
[PubMed]
27.
Kanaya
M.
,
Higo
S.
and
Ozawa
H.
(
2019
)
Neurochemical characterization of neurons expressing estrogen receptor β in the hypothalamic nuclei of rats using in situ hybridization and immunofluorescence
.
Int. J. Mol. Sci.
21
[PubMed]
28.
Andersson
S.
,
Sundberg
M.
,
Pristovsek
N.
,
Ibrahim
A.
,
Jonsson
P.
,
Katona
B.
et al.
(
2017
)
Insufficient antibody validation challenges oestrogen receptor beta research
.
Nat Commun.
8
,
15840
[PubMed]
29.
Nelson
A.W.
,
Groen
A.J.
,
Miller
J.L.
,
Warren
A.Y.
,
Holmes
K.A.
,
Tarulli
G.A.
et al.
(
2017
)
Comprehensive assessment of estrogen receptor beta antibodies in cancer cell line models and tissue reveals critical limitations in reagent specificity
.
Mol. Cell. Endocrinol.
440
,
138
150
[PubMed]
30.
Hattori
Y.
,
Ishii
H.
,
Higo
S.
,
Otsuka
M.
,
Kanaya
M.
,
Matsumoto
K.
et al.
(
2021
)
Optimization of immunohistochemical detection of rat ESR2 proteins with well-validated monoclonal antibody PPZ0506
.
Mol. Cell. Endocrinol.
523
,
111145
[PubMed]
31.
Ishii
H.
,
Otsuka
M.
,
Kanaya
M.
,
Higo
S.
,
Hattori
Y.
and
Ozawa
H.
(
2019
)
Applicability of anti-human estrogen receptor β antibody PPZ0506 for the immunodetection of rodent estrogen receptor β proteins
.
Int. J. Mol. Sci.
20
32.
Shima
N.
,
Yamaguchi
Y.
and
Yuri
K.
(
2003
)
Distribution of estrogen receptor beta mRNA-containing cells in ovariectomized and estrogen-treated female rat brain
.
Anat. Sci. Int.
78
,
85
97
[PubMed]
33.
Sagoshi
S.
,
Maejima
S.
,
Morishita
M.
,
Takenawa
S.
,
Otubo
A.
,
Takanami
K.
et al.
(
2020
)
Detection and characterization of estrogen receptor beta expression in the brain with newly developed transgenic mice
.
Neuroscience
438
,
182
197
[PubMed]
34.
Hazell
G.G.
,
Yao
S.T.
,
Roper
J.A.
,
Prossnitz
E.R.
,
O'Carroll
A.M.
and
Lolait
S.J.
(
2009
)
Localisation of GPR30, a novel G protein-coupled oestrogen receptor, suggests multiple functions in rodent brain and peripheral tissues
.
J. Endocrinol.
202
,
223
236
[PubMed]
35.
Zhao
C.
,
Fujinaga
R.
,
Tanaka
M.
,
Yanai
A.
,
Nakahama
K.
and
Shinoda
K.
(
2007
)
Region-specific expression and sex-steroidal regulation on aromatase and its mRNA in the male rat brain: immunohistochemical and in situ hybridization analyses
.
J. Comp. Neurol.
500
,
557
573
[PubMed]
36.
Zhao
C.
,
Fujinaga
R.
,
Yanai
A.
,
Kokubu
K.
,
Takeshita
Y.
,
Watanabe
Y.
et al.
(
2008
)
Sex-steroidal regulation of aromatase mRNA expression in adult male rat brain: a quantitative non-radioactive in situ hybridization study
.
Cell Tissue Res.
332
,
381
391
[PubMed]
37.
Toffoletto
S.
,
Lanzenberger
R.
,
Gingnell
M.
,
Sundström-Poromaa
I.
and
Comasco
E.
(
2014
)
Emotional and cognitive functional imaging of estrogen and progesterone effects in the female human brain: a systematic review
.
Psychoneuroendocrinology
50
,
28
52
[PubMed]
38.
Smith
C.C.
,
Vedder
L.C.
and
McMahon
L.L.
(
2009
)
Estradiol and the relationship between dendritic spines, NR2B containing NMDA receptors, and the magnitude of long-term potentiation at hippocampal CA3-CA1 synapses
.
Psychoneuroendocrinology
34
,
S130
S142
[PubMed]
39.
Barha
C.K.
and
Galea
L.A.
(
2010
)
Influence of different estrogens on neuroplasticity and cognition in the hippocampus
.
Biochim. Biophys. Acta
1800
,
1056
1067
[PubMed]
40.
Frankfurt
M.
and
Luine
V.
(
2015
)
The evolving role of dendritic spines and memory: Interaction(s) with estradiol
.
Horm. Behav.
74
,
28
36
[PubMed]
41.
Akama
K.T.
and
McEwen
B.S.
(
2003
)
Estrogen stimulates postsynaptic density-95 rapid protein synthesis via the Akt/protein kinase B pathway
.
J. Neurosci: Off. J. Soc. Neurosci.
23
,
2333
2339
[PubMed]
42.
Znamensky
V.
,
Akama
K.T.
,
McEwen
B.S.
and
Milner
T.A.
(
2003
)
Estrogen levels regulate the subcellular distribution of phosphorylated Akt in hippocampal CA1 dendrites
.
J. Neurosci.: Off. J. Soc. Neurosci.
23
,
2340
2347
[PubMed]
43.
Kuroki
Y.
,
Fukushima
K.
,
Kanda
Y.
,
Mizuno
K.
and
Watanabe
Y.
(
2001
)
Neuroprotection by estrogen via extracellular signal-regulated kinase against quinolinic acid-induced cell death in the rat hippocampus
.
Eur. J. Neurosci.
13
,
472
476
[PubMed]
44.
Waters
E.M.
,
Yildirim
M.
,
Janssen
W.G.
,
Lou
W.Y.
,
McEwen
B.S.
,
Morrison
J.H.
et al.
(
2011
)
Estrogen and aging affect the synaptic distribution of estrogen receptor β-immunoreactivity in the CA1 region of female rat hippocampus
.
Brain Res.
1379
,
86
97
[PubMed]
45.
Milner
T.A.
,
McEwen
B.S.
,
Hayashi
S.
,
Li
C.J.
,
Reagan
L.P.
and
Alves
S.E.
(
2001
)
Ultrastructural evidence that hippocampal alpha estrogen receptors are located at extranuclear sites
.
J. Comp. Neurol.
429
,
355
371
[PubMed]
46.
Milner
T.A.
,
Ayoola
K.
,
Drake
C.T.
,
Herrick
S.P.
,
Tabori
N.E.
,
McEwen
B.S.
et al.
(
2005
)
Ultrastructural localization of estrogen receptor beta immunoreactivity in the rat hippocampal formation
.
J. Comp. Neurol.
491
,
81
95
[PubMed]
47.
Zhao
L.
and
Brinton
R.D.
(
2007
)
Estrogen receptor α and β differentially regulate intracellular Ca2+ dynamics leading to ERK phosphorylation and estrogen neuroprotection in hippocampal neurons
.
Brain Res.
1172
,
48
59
[PubMed]
48.
Jiang
K.
,
Yang
Z.
,
Cheng
L.
,
Wang
S.
,
Ning
K.
,
Zhou
L.
et al.
(
2013
)
Mediator of ERBB2-driven cell motility (MEMO) promotes extranuclear estrogen receptor signaling involving the growth factor receptors IGF1R and ERBB2
.
J. Biol. Chem.
288
,
24590
24599
[PubMed]
49.
Mendez
P.
,
Azcoitia
I.
and
Garcia-Segura
L.M.
(
2003
)
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
50.
Foy
M.R.
,
Xu
J.
,
Xie
X.
,
Brinton
R.D.
,
Thompson
R.F.
and
Berger
T.W.
(
1999
)
17beta-estradiol enhances NMDA receptor-mediated EPSPs and long-term potentiation
.
J. Neurophysiol.
81
,
925
929
[PubMed]
51.
Tozzi
A.
,
Bellingacci
L.
and
Pettorossi
V.E.
(
2020
)
Rapid estrogenic and androgenic neurosteroids effects in the induction of long-term synaptic changes: implication for early memory formation
.
Front. Neurosci.
14
,
572511
[PubMed]
52.
Lubahn
D.B.
,
Moyer
J.S.
,
Golding
T.S.
,
Couse
J.F.
,
Korach
K.S.
and
Smithies
O.
(
1993
)
Alteration of reproductive function but not prenatal sexual development after insertional disruption of the mouse estrogen receptor gene
.
Proc. Natl. Acad. Sci. USA
90
,
11162
11166
[PubMed]
53.
Fugger
H.N.
,
Foster
T.C.
,
Gustafsson
J.
and
Rissman
E.F.
(
2000
)
Novel effects of estradiol and estrogen receptor alpha and beta on cognitive function
.
Brain Res.
883
,
258
264
[PubMed]
54.
Antonson
P.
,
Apolinário
L.M.
,
Shamekh
M.M.
,
Humire
P.
,
Poutanen
M.
,
Ohlsson
C.
et al.
(
2020
)
Generation of an all-exon Esr2 deleted mouse line: Effects on fertility
.
Biochem. Biophys. Res. Commun.
529
,
231
237
[PubMed]
55.
Rissman
E.F.
,
Heck
A.L.
,
Leonard
J.E.
,
Shupnik
M.A.
and
Gustafsson
J.A.
(
2002
)
Disruption of estrogen receptor beta gene impairs spatial learning in female mice
.
Proc. Natl. Acad. Sci. USA
99
,
3996
4001
[PubMed]
56.
Imwalle
D.B.
,
Gustafsson
J.A.
and
Rissman
E.F.
(
2005
)
Lack of functional estrogen receptor beta influences anxiety behavior and serotonin content in female mice
.
Physiol. Behavior
84
,
157
163
57.
Wu
W.F.
,
Tan
X.J.
,
Dai
Y.B.
,
Krishnan
V.
,
Warner
M.
and
Gustafsson
J.A.
(
2013
)
Targeting estrogen receptor beta in microglia and T cells to treat experimental autoimmune encephalomyelitis
.
Proc. Natl. Acad. Sci. USA
110
,
3543
3548
[PubMed]
58.
Khalaj
A.J.
,
Yoon
J.
,
Nakai
J.
,
Winchester
Z.
,
Moore
S.M.
,
Yoo
T.
et al.
(
2013
)
Estrogen receptor (ER) beta expression in oligodendrocytes is required for attenuation of clinical disease by an ERbeta ligand
.
Proc. Natl. Acad. Sci. USA
110
,
19125
19130
[PubMed]
59.
Spence
R.D.
,
Hamby
M.E.
,
Umeda
E.
,
Itoh
N.
,
Du
S.
,
Wisdom
A.J.
et al.
(
2011
)
Neuroprotection mediated through estrogen receptor-alpha in astrocytes
.
Proc. Natl. Acad. Sci. USA
108
,
8867
8872
[PubMed]
60.
Adams
M.M.
,
Fink
S.E.
,
Shah
R.A.
,
Janssen
W.G.
,
Hayashi
S.
,
Milner
T.A.
et al.
(
2002
)
Estrogen and aging affect the subcellular distribution of estrogen receptor-alpha in the hippocampus of female rats
.
J. Neurosci.: Off. J. Soc. Neurosci.
22
,
3608
3614
[PubMed]
61.
Spence
R.D.
and
Voskuhl
R.R.
(
2012
)
Neuroprotective effects of estrogens and androgens in CNS inflammation and neurodegeneration
.
Front. Neuroendocrinol.
33
,
105
115
[PubMed]
62.
Sarchielli
E.
,
Comeglio
P.
,
Filippi
S.
,
Cellai
I.
,
Guarnieri
G.
,
Marzoppi
A.
et al.
(
2021
)
Neuroprotective effects of testosterone in the hypothalamus of an animal model of metabolic syndrome
.
Int. J. Mol. Sci.
22
,
[PubMed]
63.
Saldanha
C.J.
,
Duncan
K.A.
and
Walters
B.J.
(
2009
)
Neuroprotective actions of brain aromatase
.
Front. Neuroendocrinol.
30
,
106
118
[PubMed]
64.
Morrison
J.H.
,
Brinton
R.D.
,
Schmidt
P.J.
and
Gore
A.C.
(
2006
)
Estrogen, menopause, and the aging brain: how basic neuroscience can inform hormone therapy in women
.
J. Neurosci.: Off. J. Soc. Neurosci.
26
,
10332
10348
[PubMed]
65.
Schmidt
P.J.
,
Nieman
L.
,
Danaceau
M.A.
,
Tobin
M.B.
,
Roca
C.A.
,
Murphy
J.H.
et al.
(
2000
)
Estrogen replacement in perimenopause-related depression: a preliminary report
.
Am. J. Obstet. Gynecol.
183
,
414
420
[PubMed]
66.
Soares
C.N.
,
Almeida
O.P.
,
Joffe
H.
and
Cohen
L.S.
(
2001
)
Efficacy of estradiol for the treatment of depressive disorders in perimenopausal women: a double-blind, randomized, placebo-controlled trial
.
Arch. Gen. Psychiatry
58
,
529
534
[PubMed]
67.
Dwyer
J.B.
,
Aftab
A.
,
Radhakrishnan
R.
,
Widge
A.
,
Rodriguez
C.I.
,
Carpenter
L.L.
et al.
(
2020
)
Hormonal treatments for major depressive disorder: state of the art
.
Am. J. Psychiatry
177
,
686
705
[PubMed]
68.
Morrison
M.F.
,
Kallan
M.J.
,
Ten Have
T.
,
Katz
I.
,
Tweedy
K.
and
Battistini
M.
(
2004
)
Lack of efficacy of estradiol for depression in postmenopausal women: a randomized, controlled trial
.
Biol. Psychiatry
55
,
406
412
[PubMed]
69.
Guo
H.
,
Liu
M.
,
Zhang
L.
,
Wang
L.
,
Hou
W.
,
Ma
Y.
et al.
(
2020
)
The critical period for neuroprotection by estrogen replacement therapy and the potential underlying mechanisms
.
Curr. Neuropharmacol.
18
,
485
500
[PubMed]
70.
Reuben
R.
,
Karkaby
L.
,
McNamee
C.
,
Phillips
N.A.
and
Einstein
G.
(
2021
)
Menopause and cognitive complaints: are ovarian hormones linked with subjective cognitive decline?
Climacteric
24
,
321
332
[PubMed]
71.
Weber
M.T.
,
Maki
P.M.
and
McDermott
M.P.
(
2014
)
Cognition and mood in perimenopause: a systematic review and meta-analysis
.
J. Steroid Biochem. Mol. Biol.
142
,
90
98
[PubMed]
72.
Jessen
F.
,
Wiese
B.
,
Bachmann
C.
,
Eifflaender-Gorfer
S.
,
Haller
F.
,
Kölsch
H.
et al.
(
2010
)
Prediction of dementia by subjective memory impairment: effects of severity and temporal association with cognitive impairment
.
Arch. Gen. Psychiatry
67
,
414
422
[PubMed]
73.
Chen
X.
,
Guo
T.
and
Li
B.
(
2013
)
Influence of prophylactic oophorectomy on mood and sexual function in women of menopausal transition or postmenopausal period
.
Arch. Gynecol. Obstet.
288
,
1101
1106
[PubMed]
74.
Rocca
W.A.
,
Grossardt
B.R.
,
Geda
Y.E.
,
Gostout
B.S.
,
Bower
J.H.
,
Maraganore
D.M.
et al.
(
2008
)
Long-term risk of depressive and anxiety symptoms after early bilateral oophorectomy
.
Menopause
15
,
1050
1059
[PubMed]
75.
Parker
W.H.
(
2010
)
Bilateral oophorectomy versus ovarian conservation: effects on long-term women’s health
.
J. Minim. Invasive Gynecol.
17
,
161
166
[PubMed]
76.
Phung
T.K.
,
Waltoft
B.L.
,
Laursen
T.M.
,
Settnes
A.
,
Kessing
L.V.
,
Mortensen
P.B.
et al.
(
2010
)
Hysterectomy, oophorectomy and risk of dementia: a nationwide historical cohort study
.
Dement. Geriatr. Cogn. Disord.
30
,
43
50
[PubMed]
77.
Laughlin-Tommaso
S.K.
,
Satish
A.
,
Khan
Z.
,
Smith
C.Y.
,
Rocca
W.A.
and
Stewart
E.A.
(
2020
)
Long-term risk of de novo mental health conditions after hysterectomy with ovarian conservation: a cohort study
.
Menopause
27
,
33
42
[PubMed]
78.
Nilsen
J.
,
Deng
J.
and
Brinton
R.D.
(
2005
)
Impact of clinically relevant progestins on the neural effects of estradiol and the signaling pathways involved
.
Drug News Perspect.
18
,
545
553
[PubMed]
79.
Shumaker
S.A.
,
Legault
C.
,
Rapp
S.R.
,
Thal
L.
,
Wallace
R.B.
,
Ockene
J.K.
et al.
(
2003
)
Estrogen plus progestin and the incidence of dementia and mild cognitive impairment in postmenopausal women: the Women’s Health Initiative Memory Study: a randomized controlled trial
.
JAMA
289
,
2651
2662
[PubMed]
80.
Giatti
S.
,
Garcia-Segura
L.M.
,
Barreto
G.E.
and
Melcangi
R.C.
(
2019
)
Neuroactive steroids, neurosteroidogenesis and sex
.
Prog. Neurobiol.
176
,
1
17
[PubMed]
81.
Bjorkhem
I.
(
2006
)
Crossing the barrier: oxysterols as cholesterol transporters and metabolic modulators in the brain
.
J. Intern. Med.
260
,
493
508
[PubMed]
82.
Loera-Valencia
R.
,
Goikolea
J.
,
Parrado-Fernandez
C.
,
Merino-Serrais
P.
and
Maioli
S.
(
2019
)
Alterations in cholesterol metabolism as a risk factor for developing Alzheimer's disease: potential novel targets for treatment
.
J. Steroid Biochem. Mol. Biol.
190
,
104
114
[PubMed]
83.
Bjorkhem
I.
,
Lutjohann
D.
,
Diczfalusy
U.
,
Stahle
L.
,
Ahlborg
G.
and
Wahren
J.
(
1998
)
Cholesterol homeostasis in human brain: turnover of 24S-hydroxycholesterol and evidence for a cerebral origin of most of this oxysterol in the circulation
.
J. Lipid Res.
39
,
1594
1600
[PubMed]
84.
Janowski
B.A.
,
Grogan
M.J.
,
Jones
S.A.
,
Wisely
G.B.
,
Kliewer
S.A.
,
Corey
E.J.
et al.
(
1999
)
Structural requirements of ligands for the oxysterol liver X receptors LXRalpha and LXRbeta
.
Proc. Natl. Acad. Sci. USA
96
,
266
271
[PubMed]
85.
Cermenati
G.
,
Giatti
S.
,
Cavaletti
G.
,
Bianchi
R.
,
Maschi
O.
,
Pesaresi
M.
et al.
(
2010
)
Activation of the liver X receptor increases neuroactive steroid levels and protects from diabetes-induced peripheral neuropathy
.
J. Neurosci.: Off. J. Soc. Neurosci.
30
,
11896
11901
[PubMed]
86.
Huq
M.D.
,
Tsai
N.P.
,
Gupta
P.
and
Wei
L.N.
(
2006
)
Regulation of retinal dehydrogenases and retinoic acid synthesis by cholesterol metabolites
.
EMBO J.
25
,
3203
3213
[PubMed]
87.
Damdimopoulou
P.
,
Chiang
C.
and
Flaws
J.A.
(
2019
)
Retinoic acid signaling in ovarian folliculogenesis and steroidogenesis
.
Reprod. Toxicol.
87
,
32
41
[PubMed]
88.
Vedin
L.L.
,
Lewandowski
S.A.
,
Parini
P.
,
Gustafsson
J.A.
and
Steffensen
K.R.
(
2009
)
The oxysterol receptor LXR inhibits proliferation of human breast cancer cells
.
Carcinogenesis
30
,
575
579
[PubMed]
89.
Krycer
J.R.
and
Brown
A.J.
(
2011
)
Cross-talk between the androgen receptor and the liver X receptor: implications for cholesterol homeostasis
.
J. Biol. Chem.
286
,
20637
20647
[PubMed]
90.
Merino-Serrais
P.
,
Loera-Valencia
R.
,
Rodriguez-Rodriguez
P.
,
Parrado-Fernandez
C.
,
Ismail
M.A.
,
Maioli
S.
et al.
(
2019
)
27-hydroxycholesterol induces aberrant morphology and synaptic dysfunction in hippocampal neurons
.
Cereb. Cortex
29
,
429
446
[PubMed]
91.
Ismail
M.A.
,
Mateos
L.
,
Maioli
S.
,
Merino-Serrais
P.
,
Ali
Z.
,
Lodeiro
M.
et al.
(
2017
)
27-Hydroxycholesterol impairs neuronal glucose uptake through an IRAP/GLUT4 system dysregulation
.
J. Exp. Med.
214
,
699
717
[PubMed]
92.
Heverin
M.
,
Maioli
S.
,
Pham
T.
,
Mateos
L.
,
Camporesi
E.
,
Ali
Z.
et al.
(
2015
)
27-Hydroxycholesterol mediates negative effects of dietary cholesterol on cognition in mice
.
Behav. Brain Res.
278
,
356
359
[PubMed]
93.
Umetani
M.
and
Shaul
P.W.
(
2011
)
27-Hydroxycholesterol: the first identified endogenous SERM
.
Trends Endocrinol. Metab.
22
,
130
135
[PubMed]
94.
Maioli
S.
,
Bavner
A.
,
Ali
Z.
,
Heverin
M.
,
Ismail
M.A.
,
Puerta
E.
et al.
(
2013
)
Is it possible to improve memory function by upregulation of the cholesterol 24S-hydroxylase (CYP46A1) in the brain?
PLoS ONE
8
,
e68534
[PubMed]
95.
Djelti
F.
,
Braudeau
J.
,
Hudry
E.
,
Dhenain
M.
,
Varin
J.
,
Bieche
I.
et al.
(
2015
)
CYP46A1 inhibition, brain cholesterol accumulation and neurodegeneration pave the way for Alzheimer's disease
.
Brain
138
,
2383
2398
[PubMed]
96.
van der Kant
R.
,
Langness
V.F.
,
Herrera
C.M.
,
Williams
D.A.
,
Fong
L.K.
,
Leestemaker
Y.
et al.
(
2019
)
Cholesterol metabolism is a druggable axis that independently regulates tau and amyloid-beta in iPSC-derived Alzheimer's disease neurons
.
Cell Stem Cell
24
,
363e9
375e9
97.
Latorre-Leal
M.
,
Rodriguez-Rodriguez
P.
,
Franchini
L.
,
Daniilidou
M.
,
Eroli
F.
,
Winblad
B.
et al.
(
2021
)
Sex-dependent effects of CYP46A1 overexpression on cognitive function during aging
.
bioRxiv
98.
Alzheimer's Association
(
2019
)
2019 Alzheimer's disease facts and figures
.
Alzheimer’s Dementia
15
,
321
387
99.
Rocca
W.A.
(
2017
)
Time, sex, gender, history, and dementia
.
Alzheimer Dis. Assoc. Disord.
31
,
76
79
[PubMed]
100.
Gurvich
C.
,
Le
J.
,
Thomas
N.
,
Thomas
E.H.X.
and
Kulkarni
J.
(
2021
)
Sex hormones and cognition in aging
.
Vitam. Horm.
115
,
511
533
[PubMed]
101.
Duarte-Guterman
P.
,
Albert
A.Y.
,
Inkster
A.M.
,
Barha
C.K.
and
Galea
L.A.M.
(
2020
)
Inflammation in Alzheimer’s disease: do sex and APOE matter?
J. Alzheimer's Dis.: JAD
78
,
627
641
102.
Plassman
B.L.
,
Langa
K.M.
,
Fisher
G.G.
,
Heeringa
S.G.
,
Weir
D.R.
,
Ofstedal
M.B.
et al.
(
2007
)
Prevalence of dementia in the United States: the aging, demographics, and memory study
.
Neuroepidemiology
29
,
125
132
[PubMed]
103.
Pinares-Garcia
P.
,
Stratikopoulos
M.
,
Zagato
A.
,
Loke
H.
and
Lee
J.
(
2018
)
Sex: a significant risk factor for neurodevelopmental and neurodegenerative disorders
.
Brain Sci.
8
,
[PubMed]
104.
Henderson
V.W.
and
Buckwalter
J.G.
(
1994
)
Cognitive deficits of men and women with Alzheimer's disease
.
Neurology
44
,
90
96
[PubMed]
105.
Barnes
L.L.
,
Wilson
R.S.
,
Bienias
J.L.
,
Schneider
J.A.
,
Evans
D.A.
and
Bennett
D.A.
(
2005
)
Sex differences in the clinical manifestations of Alzheimer disease pathology
.
Arch. Gen. Psychiatry
62
,
685
691
[PubMed]
106.
Akwa
Y.
(
2020
)
Steroids and Alzheimer’s disease: changes associated with pathology and therapeutic potential
.
Int. J. Mol. Sci.
21
,
[PubMed]
107.
Xu
H.
,
Wang
R.
,
Zhang
Y.W.
and
Zhang
X.
(
2006
)
Estrogen, beta-amyloid metabolism/trafficking, and Alzheimer’s disease
.
Ann. N. Y. Acad. Sci.
1089
,
324
342
[PubMed]
108.
Li
R.
,
He
P.
,
Cui
J.
,
Staufenbiel
M.
,
Harada
N.
and
Shen
Y.
(
2013
)
Brain endogenous estrogen levels determine responses to estrogen replacement therapy via regulation of BACE1 and NEP in female Alzheimer’s transgenic mice
.
Mol. Neurobiol.
47
,
857
867
[PubMed]
109.
Ishunina
T.A.
and
Swaab
D.F.
(
2001
)
Increased expression of estrogen receptor alpha and beta in the nucleus basalis of Meynert in Alzheimer’s disease
.
Neurobiol. Aging
22
,
417
426
[PubMed]
110.
Ishunina
T.A.
and
Swaab
D.F.
(
2003
)
Increased neuronal metabolic activity and estrogen receptors in the vertical limb of the diagonal band of Broca in Alzheimer’s disease: relation to sex and aging
.
Exp. Neurol.
183
,
159
172
[PubMed]
111.
Ishunina
T.A.
,
Kamphorst
W.
and
Swaab
D.F.
(
2003
)
Changes in metabolic activity and estrogen receptors in the human medial mamillary nucleus: relation to sex, aging and Alzheimer’s disease
.
Neurobiol. Aging
24
,
817
828
[PubMed]
112.
Hestiantoro
A.
and
Swaab
D.F.
(
2004
)
Changes in estrogen receptor-alpha and -beta in the infundibular nucleus of the human hypothalamus are related to the occurrence of Alzheimer’s disease neuropathology
.
J. Clin. Endocrinol. Metab.
89
,
1912
1925
[PubMed]
113.
Hu
X.Y.
,
Qin
S.
,
Lu
Y.P.
,
Ravid
R.
,
Swaab
D.F.
and
Zhou
J.N.
(
2003
)
Decreased estrogen receptor-alpha expression in hippocampal neurons in relation to hyperphosphorylated tau in Alzheimer patients
.
Acta Neuropathol.
106
,
213
220
[PubMed]
114.
Wang
C.
,
Zhang
F.
,
Jiang
S.
,
Siedlak
S.L.
,
Shen
L.
,
Perry
G.
et al.
(
2016
)
Estrogen receptor-α is localized to neurofibrillary tangles in Alzheimer’s disease
.
Sci. Rep.
6
,
20352
[PubMed]
115.
Bryant
D.N.
and
Dorsa
D.M.
(
2010
)
Roles of estrogen receptors alpha and beta in sexually dimorphic neuroprotection against glutamate toxicity
.
Neuroscience
170
,
1261
1269
[PubMed]
116.
Lai
Y.J.
,
Zhu
B.L.
,
Sun
F.
,
Luo
D.
,
Ma
Y.L.
,
Luo
B.
et al.
(
2019
)
Estrogen receptor α promotes Cav1.2 ubiquitination and degradation in neuronal cells and in APP/PS1 mice
.
Aging Cell.
18
,
e12961
[PubMed]
117.
Carroll
J.C.
and
Pike
C.J.
(
2008
)
Selective estrogen receptor modulators differentially regulate Alzheimer-like changes in female 3xTg-AD mice
.
Endocrinology
149
,
2607
2611
[PubMed]
118.
Yaffe
K.
,
Lui
L.Y.
,
Grady
D.
,
Stone
K.
and
Morin
P.
(
2002
)
Estrogen receptor 1 polymorphisms and risk of cognitive impairment in older women
.
Biol. Psychiatry
51
,
677
682
[PubMed]
119.
Olsen
L.
,
Rasmussen
H.B.
,
Hansen
T.
,
Bagger
Y.Z.
,
Tankó
L.B.
,
Qin
G.
et al.
(
2006
)
Estrogen receptor alpha and risk for cognitive impairment in postmenopausal women
.
Psychiatr. Genet.
16
,
85
88
[PubMed]
120.
Ji
Y.
,
Urakami
K.
,
Wada-Isoe
K.
,
Adachi
Y.
and
Nakashima
K.
(
2000
)
Estrogen receptor gene polymorphisms in patients with Alzheimer’s disease, vascular dementia and alcohol-associated dementia
.
Dement. Geriatr. Cogn. Disord.
11
,
119
122
[PubMed]
121.
Cheng
D.
,
Liang
B.
,
Hao
Y.
and
Zhou
W.
(
2014
)
Estrogen receptor α gene polymorphisms and risk of Alzheimer’s disease: evidence from a meta-analysis
.
Clin. Interv. Aging
9
,
1031
1038
[PubMed]
122.
Long
J.
,
He
P.
,
Shen
Y.
and
Li
R.
(
2012
)
New evidence of mitochondria dysfunction in the female Alzheimer’s disease brain: deficiency of estrogen receptor-β
.
J. Alzheimers Dis.
30
,
545
558
[PubMed]
123.
Yang
S.H.
,
Sarkar
S.N.
,
Liu
R.
,
Perez
E.J.
,
Wang
X.
,
Wen
Y.
et al.
(
2009
)
Estrogen receptor beta as a mitochondrial vulnerability factor
.
J. Biol. Chem.
284
,
9540
9548
[PubMed]
124.
Burstein
S.R.
,
Kim
H.J.
,
Fels
J.A.
,
Qian
L.
,
Zhang
S.
,
Zhou
P.
et al.
(
2018
)
Estrogen receptor beta modulates permeability transition in brain mitochondria
.
Biochim. Biophys. Acta Bioenerg.
1859
,
423
433
[PubMed]
125.
Irwin
R.W.
,
Yao
J.
,
To
J.
,
Hamilton
R.T.
,
Cadenas
E.
and
Brinton
R.D.
(
2012
)
Selective oestrogen receptor modulators differentially potentiate brain mitochondrial function
.
J. Neuroendocrinol.
24
,
236
248
[PubMed]
126.
Wang
L.
,
Andersson
S.
,
Warner
M.
and
Gustafsson
J.A.
(
2001
)
Morphological abnormalities in the brains of estrogen receptor beta knockout mice
.
Proc. Natl. Acad. Sci. USA
98
,
2792
2796
[PubMed]
127.
Zhao
L.
,
Mao
Z.
,
Chen
S.
,
Schneider
L.S.
and
Brinton
R.D.
(
2013
)
Early intervention with an estrogen receptor β-selective phytoestrogenic formulation prolongs survival, improves spatial recognition memory, and slows progression of amyloid pathology in a female mouse model of Alzheimer's disease
.
J. Alzheimer's Dis.: JAD
37
,
403
419
128.
Wei
Y.
,
Zhou
J.
,
Wu
J.
and
Huang
J.
(
2019
)
ERβ promotes Aβ degradation via the modulation of autophagy
.
Cell Death Dis.
10
,
565
[PubMed]
129.
Sánchez
M.G.
,
Bourque
M.
,
Morissette
M.
and
Di Paolo
T.
(
2010
)
Steroids-dopamine interactions in the pathophysiology and treatment of CNS disorders
.
CNS Neurosci. Ther.
16
,
e43
e71
[PubMed]
130.
Murray
H.E.
,
Pillai
A.V.
,
McArthur
S.R.
,
Razvi
N.
,
Datla
K.P.
,
Dexter
D.T.
et al.
(
2003
)
Dose- and sex-dependent effects of the neurotoxin 6-hydroxydopamine on the nigrostriatal dopaminergic pathway of adult rats: differential actions of estrogen in males and females
.
Neuroscience
116
,
213
222
[PubMed]
131.
Pohjalainen
T.
,
Rinne
J.O.
,
Någren
K.
,
Syvälahti
E.
and
Hietala
J.
(
1998
)
Sex differences in the striatal dopamine D2 receptor binding characteristics in vivo
.
Am. J. Psychiatry
155
,
768
773
[PubMed]
132.
Cantuti-Castelvetri
I.
,
Keller-McGandy
C.
,
Bouzou
B.
,
Asteris
G.
,
Clark
T.W.
,
Frosch
M.P.
et al.
(
2007
)
Effects of gender on nigral gene expression and parkinson disease
.
Neurobiol. Dis.
26
,
606
614
[PubMed]
133.
Lee
A.M.
,
Mansuri
M.S.
,
Wilson
R.S.
,
Lam
T.T.
,
Nairn
A.C.
and
Picciotto
M.R.
(
2021
)
Sex differences in the ventral tegmental area and nucleus accumbens proteome at baseline and following nicotine exposure
.
Front. Mol. Neurosci.
14
,
657064
[PubMed]
134.
Bourque
M.
,
Dluzen
D.E.
and
Di Paolo
T.
(
2009
)
Neuroprotective actions of sex steroids in Parkinson’s disease
.
Front. Neuroendocrinol.
30
,
142
157
[PubMed]
135.
Song
Y.-j
,
Li
S.-r
,
Li
X.-w
,
Chen
X.
,
Wei
Z.-x
,
Liu
Q.-s
et al.
(
2020
)
The effect of estrogen replacement therapy on Alzheimer’s disease and Parkinson’s disease in postmenopausal women: a meta-analysis
.
Front. Neurosci.
14
,
1
13
136.
Saunders-Pullman
R.
,
Gordon-Elliott
J.
,
Parides
M.
,
Fahn
S.
,
Saunders
H.R.
and
Bressman
S.
(
1999
)
The effect of estrogen replacement on early Parkinson's disease
.
Neurology
52
,
1417
1421
[PubMed]
137.
Becker
J.B.
and
Rudick
C.N.
(
1999
)
Rapid effects of estrogen or progesterone on the amphetamine-induced increase in striatal dopamine are enhanced by estrogen priming: a microdialysis study
.
Pharmacol. Biochem. Behav.
64
,
53
57
[PubMed]
138.
Yoest
K.E.
,
Cummings
J.A.
and
Becker
J.B.
(
2019
)
Oestradiol influences on dopamine release from the nucleus accumbens shell: sex differences and the role of selective oestradiol receptor subtypes
.
Br. J. Pharmacol.
176
,
4136
4148
[PubMed]
139.
Morale
M.C.
,
L'Episcopo
F.
,
Tirolo
C.
,
Giaquinta
G.
,
Caniglia
S.
,
Testa
N.
et al.
(
2008
)
Loss of aromatase cytochrome P450 function as a risk factor for Parkinson’s disease?
Brain Res. Rev.
57
,
431
443
[PubMed]
140.
Callier
S.
,
Morissette
M.
,
Grandbois
M.
and
Di Paolo
T.
(
2000
)
Stereospecific prevention by 17beta-estradiol of MPTP-induced dopamine depletion in mice
.
Synapse
37
,
245
251
[PubMed]
141.
D'Astous
M.
,
Morissette
M.
and
Di Paolo
T.
(
2004
)
Effect of estrogen receptor agonists treatment in MPTP mice: evidence of neuroprotection by an ER alpha agonist
.
Neuropharmacology
47
,
1180
1188
[PubMed]
142.
Baraka
A.M.
,
Korish
A.A.
,
Soliman
G.A.
and
Kamal
H.
(
2011
)
The possible role of estrogen and selective estrogen receptor modulators in a rat model of Parkinson’s disease
.
Life Sci.
88
,
879
885
[PubMed]
143.
Salama
R.M.
,
Tadros
M.G.
,
Schaalan
M.F.
,
Bahaa
N.
,
Abdel-Tawab
A.M.
and
Khalifa
A.E.
(
2018
)
Potential neuroprotective effect of androst-5-ene-3β, 17β-diol (ADIOL) on the striatum, and substantia nigra in Parkinson's disease rat model
.
J. Cell. Physiol.
233
,
5981
6000
[PubMed]
144.
Westberg
L.
,
Håkansson
A.
,
Melke
J.
,
Shahabi
H.N.
,
Nilsson
S.
,
Buervenich
S.
et al.
(
2004
)
Association between the estrogen receptor beta gene and age of onset of Parkinson's disease
.
Psychoneuroendocrinology
29
,
993
998
[PubMed]
145.
Li
X.L.
,
Cheng
W.D.
,
Li
J.
,
Zheng
Y.F.
,
Guo
C.J.
,
Sun
S.G.
et al.
(
2009
)
Does estrogen receptor gene polymorphism play a role in Parkinson’s disease?
Biomed. Pharmacother.
63
,
599
602
[PubMed]
146.
Carrasquilla
G.D.
,
Frumento
P.
,
Berglund
A.
,
Borgfeldt
C.
,
Bottai
M.
,
Chiavenna
C.
et al.
(
2017
)
Postmenopausal hormone therapy and risk of stroke: a pooled analysis of data from population-based cohort studies
.
PLoS Med.
14
,
e1002445
[PubMed]
147.
Anderson
G.L.
,
Chlebowski
R.T.
,
Aragaki
A.K.
,
Kuller
L.H.
,
Manson
J.E.
,
Gass
M.
et al.
(
2012
)
Conjugated equine oestrogen and breast cancer incidence and mortality in postmenopausal women with hysterectomy: extended follow-up of the Women’s Health Initiative randomised placebo-controlled trial
.
Lancet Oncol.
13
,
476
486
[PubMed]
148.
Lobo
R.A.
(
2017
)
Hormone-replacement therapy: current thinking
.
Nat. Rev. Endocrinol.
13
,
220
231
[PubMed]
149.
Villa
A.
,
Della Torre
S.
and
Maggi
A.
(
2019
)
Sexual differentiation of microglia
.
Front. Neuroendocrinol.
52
,
156
164
[PubMed]
150.
Villa
A.
,
Gelosa
P.
,
Castiglioni
L.
,
Cimino
M.
,
Rizzi
N.
,
Pepe
G.
et al.
(
2018
)
Sex-specific features of microglia from adult mice
.
Cell Rep.
23
,
3501
3511
[PubMed]
This is an open access article published by Portland Press Limited on behalf of the Biochemical Society and distributed under the Creative Commons Attribution License 4.0 (CC BY).