Abstract
This review is intended to assess the state of current knowledge on the role of estrogen receptors (ER) in the neuroprotective effects of estrogens in models for acute neuronal injury and death. We evaluate the overall evidence that estrogens are neuroprotective in acute injury and critically assess the role of estrogen receptor (ER)α, ERβ, GPR 30 and non-receptor mediated mechanisms in these robust neuroprotective effects of this ovarian steroid hormone. We conclude that all three receptors, as well as non-receptor mediated mechanisms can be involved in neuroprotection, depending on the model used, the level of estrogen administrated and the mode of administration of the steroid. Also, the signaling pathways used by both ER-dependent and ER-independent mechanisms to exert neuroprotection are considered. Finally, further studies that are needed to parse out the relative contribution of receptor versus non-receptor-mediated signaling are discussed.
Keywords: Estradiol, estrogens, non-feminizing estrogens, estrogen receptor alpha, estrogen receptor beta, neuroprotection, traumatic brain injury, cerebral ischemia, stroke, spinal cord injury
1. Background and Other Recent Reviews on the Subject
Prior to 2002, the efficacy of estrogens was being assessed in preclinical studies in a large number of conditions caused by or associated with neurodegeneration. With the publication of the first results from the Women’s Health Initiative (WHI) Studies in 2002 [1], the field of estrogen and neuroprotection underwent a major change. In as much as there are about 450 reports of estrogen neuroprotection, we have chosen to limit our consideration to studies that assess the role of estrogen receptors (ERs) in acute neuronal loss from injury, either in vitro or in vivo. As such, in vivo studies that assess the effects of estrogens in chronic neurodegenerative disease models for Alzheimer’s disease (AD) or Parkinson’s disease (PD) are not herein considered. The reader is refereed to other excellent reviews on these topics [2, 3].
A PubMed search of reviews on ERs and neuroprotection since 2000 resulted in 58 hits. More than half of these (33) deal with specific disease states (AD, PD, schizophrenia, HIV dementia); specific mechanisms of interest to the authors, such as signaling pathways, mitochondria; astrocytes and very specific brain or eye regions. The remaining 25 dealt with acute neuroprotection, the major issue considered herein. Of these, we refer you to the following reviews that are recent and address the role of ER in neuroprotection [4–10]. Finally, this review focuses on the actions of estrogens on neurons, and does not discuss the neuroprotective effects of this steroid on oligodendrocytes or astrocytes. For consideration of these effects of estrogens in neuroprotection the reader is referred to several excellent recent reviews on the topic [11–14].
2. Overview of Evidence for Neuroprotection by Estrogens
Endogenous ovarian hormones were suspected to be neuroprotectants against global cerebral ischemia-reperfusion injury based upon studies demonstrating that gonadally intact adult female rodents sustain less neuronal damage compared to age-matched males [15]. Our laboratory first demonstrated that 17β-estradiol (17βE2) is a potent neuroprotectant in vitro [16] and is very effective against ischemia-induced brain damage [15]. There is now abundant evidence for neuroprotection by estrogens both in vitro and in vivo.
Multiple independent and lethal mechanisms are involved in cerebral ischemia-induced neuronal death, and estrogens have been identified as multi-faceted hormones that antagonize many aspects of damage-inducing cascades resulting from cerebral ischemia. Protective effects of estrogens have been widely reported in a variety of neurons against different toxicities, which mimic cerebral ischemia in vitro [17]. Antioxidant effects of the steroid [18] and attenuation of N-methyl-D-aspartate (NMDA) receptor activation [19] have been implicated as mechanisms for the neuroprotective effects of estrogens. Also, two major signaling pathways, ERK and PI-3K-Akt, have been well characterized as being able to mediate inhibition of apoptosis and support neuronal survival. Both signaling pathways can be activated by estrogens [20,21].
In in vivo studies, the neuroprotective effects of estrogens have been demonstrated in a variety of models of acute cerebral ischemia. These include transient and permanent middle cerebral artery occlusion models [15, 22, 23], global forebrain ischemia models [24], photothrombotic focal ischemia models [25], and glutamate-induced focal cerebral ischemia models [26]. The protective effects of estrogens have been described in rats, mice and gerbils [15, 27, 28]. Estrogen-induced neuroprotection has been demonstrated in adult female, middle-aged female as well as reproductively senescent female rats, in some [29], but not all [30, 31] studies. Similarly, these effects of estrogens can persist despite the presence of diabetes and hypertension [32, 33]. The neuroprotective effects of estrogens have also been demonstrated against subarachnoid hemorrhage, a highly prevalent form of stroke in females [34]. Finally, the neuroprotective action of estrogens is not limited to females, inasmuch as estrogen protection is also seen in males [35, 36]. Collectively, these results indicate that estrogens could be valuable candidates for brain protection during acute stroke in both males and females.
Estrogens in concentrations ranging from low physiological to high pharmacological produce protective effects in stroke models. Administration of a physiological level of 17βE2 at the onset of an ischemic event is not neuroprotective [29], but neuroprotective effects of pharmacological doses of 17βE2 were clearly demonstrated with acute treatment at the time of or just before an ischemic event, as well as after its onset [15, 37, 38]. The therapeutic window of 17βE2 at the dose of 100μg/kg lasts up to 3 hours after insult [39] and this therapeutic window can be extended up to 6 hours after ischemic insult with doses of 500 to 1,000 μg/kg [40]. This long post-event efficacy of estrogens is promising, since the therapeutic window for estrogen neuroprotection could be insult severity-dependent and may differ between species. For example, the infarct penumbra, which can be protected, develops over a longer period in human subjects than in rodents [41]. So, it is reasonable to predict that estrogens could have a longer therapeutic window in humans than the 6 hour window that we have described in rodents.
3. Clinical Studies of Estrogens in Neurodegenerative Conditions
Post-menopausal women are in an estrogen-deprived state and are at risk for stroke and other neurodegenerative diseases [42]. Epidemiological evidence suggests that post-menopausal estrogen therapy (ET) reduces the risk or delays the onset of AD [43, 44]. Estrogen loss from natural or surgical menopause is associated with a decline in cognitive function that is reversed by ET [45–47]. ET affects cognitive function during brain aging as well [48–50]. Clinical trials, however, have not shown a protective effect of estrogens on dementia or cognitive function in older women [51–54] or an alleviation of disease in women with mild to moderate AD [55]. Others find estrogen-amelioration of disease in Parkinson’s disease (PD) [56, 57] and recovery from neurotrauma such as stroke [4, 58] in the vast majority but not all cases [59]. Mortality from stroke is reduced in post-menopausal subjects who were taking ET at the time of stroke [60–62]. Finally, estrogen status plays role in recovery from brain injury [63, 64].
The WHI studies reported that Premarin® (and PremPro®) not only failed to positively affect conditions associated with neuroprotection, but that ET or combined hormone therapy (HT) may worsen cognition [51–55] and increase strokes [1, 65, 66] and cardiac arrests [1, 65]. It is only now that reanalysis of the WHI data indicates that early post-menopausal treatment with these compounds provides benefits [66, 67], as was reported previously in observational trials [43–49] and a great deal of animal model data as indicated above. Similarly, animal studies are completed [10] or ongoing to assess the “window of opportunity hypothesis”, which states that there is a limited period after the menopause during which ET or HT is effective. In view of these new studies, there is a need to re-assess the potential role of estrogens as a therapy for neurodegeneration.
4. Role of ERα in the Neuroprotective Effects of Estrogens
4.1. Focal ischemia.
Because there is intense interest in the possibility that estrogens can reduce stroke-induced neuronal injury, a large number of investigators have assessed the impact of estrogens on neuronal survival after middle cerebral artery occlusion (MCAO), the most well studied animal model of stroke (focal ischemia). More than a decade of research in these models points to classical ERs as the mediator of 17βE2-induced neuroprotective actions in both males and females when the hormone is administered at physiological doses prior to injury [29, 68, 69]. An early study [70] reported that the broad-spectrum ER antagonist ICI 182,780 (ICI) exacerbates the extent of MCAO-induced injury, thereby implicating classical ERs. ICI can also block the neuroprotective actions of acutely administered estradiol in this model [71].
When ER subtype-specific agonists are given to wild type animals prior to MCAO, or when estradiol is administered prior to MCAO in mice with selective ablation of ERα (ERKO) or ERβ (BERKO, the vast majority of studies show that ERα rather than ERβ is responsible for neuroprotection in this model of focal ischemia. The Wise laboratory was the first to use ERKO and BERKO mice, and they reported that 17βE2 was unable to reduce MCAO-induced injury in ERKOs but was as effective in BERKOs as in wild type mice [72]. This finding has since been replicated by other groups using knockout mice [69] or an ERα-selective agonist [73, 74]. A recent study of 17βE2’s neuroprotective efficacy after permanent MCAO in male and female mice with ERα selectively ablated in neurons or in myeloid cells (microglia) demonstrated that neuronal ERα is the mediator of neuroprotection [75]. Similarly, an ERβ agonist is unable to reduce infarct size or improve sensorimotor function in female rats undergoing permanent MCAO [76]. The finding that levels of ERα mRNA and protein are often upregulated in the injured brain after MCAO has also been interpreted as evidence that this receptor subtype is important for neuroprotection in focal ischemia. In contrast, Sampei [77] questioned the role of ERα because MCAO did not produce more damage in female ERKO mice than in wild type controls. However, these latter experiments were carried out in intact females, raising the possibility that ERα may not mediate the effects of endogenous estrogens. As discussed below, an ERβ agonist has also been reported to attenuate neuronal damage after transient MCAO and reperfusion [74].
4.2. Global ischemia
Global ischemia models the type of neuronal injury induced by transient cardiac arrest or other instances when the entire forebrain is temporarily subjected to loss of blood perfusion. It is produced experimentally in rodents (usually rats or gerbils) by transient (5-15 minute) blockade of the blood vessels supplying the forebrain and results in delayed, selective death of hippocampal pyramidal neurons, primarily in the CA1 subfield [78]. 17βE2 pretreatment for days or weeks is highly neuroprotective in this model, and ICI can abrogate the protective actions of estradiol [79–82]. Interestingly, ICI also blocks the ability of 17βE2 to reduce kainic acid-induced death of hilar neurons in the hippocampus [83]. There is strong evidence from studies with selective agonists that ERα can mediate neuroprotection after global ischemia [81, 84]. However, as described in the next section, several pharmacological studies also implicate ERβ as a mediator of 17βE2 neuroprotection in global ischemia. Most recently, the Brann laboratory showed that the hippocampus of aged and long-term hormone-deprived rats exhibits increased ubiquitination and degradation of ERα. ERβ was also reduced in the aged hippocampus. This reduction in hippocampal ERs was associated with a loss of estrogen neuroprotection after global ischemia [85].
4.3. β-amyloid toxicity, oxidative stress, and glutamate excitotoxicity
Many laboratories utilize cell cultures to address mechanistic questions about the ability of estrogens to protect neurons and astrocytes from cell death due to oxidative stress, excitotoxicity (predominantly in response to glutamate) and β-amyloid. Although a few studies with ER subtype selective agonists failed to document a role for classical ERs in protecting cultured neurons from excitotoxicity or oxidative stress (e.g., [86]), the bulk of empirical evidence indicates that ERα is an important (although not the exclusive) mediator of estrogen protection in cultured neurons and astrocytes. Early reports showed that 17βE2 protected immortalized HT22 hippocampal neurons [87] and a cholinergic cell line [88] from β-amyloid-induced toxicity and that transfection with ERα could confer this neuroprotection. A later study in primary neuronal cultures exposed to ER subtype-selective agonists also pointed to ERα as the primary mediator of protection from β-amyloid toxicity [89]. Subsequent work by the Marin group indicates that a membrane-bound form of ERα, which associates with caveolin and a voltage-dependent anion channel in lipid rafts, may be especially critical for conferring protection from β-amyloid [90]. In addition, 17βE2 is reported to reduce β-amyloid toxicity via ERα in fetal neuroepithelial cells [91]; these authors also implicated ERα in 17βE2-induced neuroprotection from oxidative stress. Most recently, an interaction between ERα and the metabotropic glutamate receptor mGluR1 was shown to mediate the neuroprotective effects of 17βE2 in primary mixed cultures of cortical neurons challenged with β-amyloid [92].
Similarly, studies in primary cultures of hippocampal neurons and in HT22 cells implicate ERα as a mediator of 17βE2 protection of glutamate toxicity [93–95]. 17βE2 also reduces glutamate-induced death of cultured astrocytes [96] and immortalized mouse hippocampal neurons [97] in an ICI-sensitive manner, although the ER subtype mediating this protection was not identified. There is also an interesting report that a brief pretreatment with 17βE2 or an ERα selective agonist can protect female but not male cortical neurons from glutamate toxicity [98]. Studies in human neuroblastoma cells (SK-NM-C) challenged with a calcium ionophore also indicate that transfection with ERα but not ERβ confers neuroprotection [99].
4.4. Other models
A contribution of ERα to neuroprotection has also been documented in several other models of neuronal injury. Studies using ER subtype-selective agonists or knockout mice in in vivo and in vitro models of neurotoxin-induced dopamine neuronal loss [100–104] implicate ERα. This classical ER subtype may also contribute to 17βE2 neuroprotection after traumatic brain injury [105], spinal cord injury [79], and experimental autoimmune encephalitis [106]. An ERα-selective agonist can mimic the ability of long-term pretreatment with 17βE2 to reduce death of immortalized mouse brain endothelial cells [107] subjected to oxygen-glucose deprivation. Studies with an ERα-selective antagonist show that this receptor may also contribute to estradiol reduction of lipopolysaccharide-induced death of cultured rat mesencephalic neurons [108] and TNFα-induced apoptosis in VSC4.1 motoneurons [79]. Finally, there is also evidence that classical ER antagonists can block the neuroprotective actions of 17βE2 in the retina [109–110], although the relevant ER subtype has not been identified.
5. Role of ERβ in the Neuroprotective Effects of Estrogens
5.1. Focal ischemia.
As noted above, most experimental studies in animal models subjected to MCAO suggest that ERα is the sole or primary mediator of 17βE2–induced protection when the hormone is provided at physiological levels prior to ischemia. However, there is a report that an ERβ-selective agonist can attenuate MCAO-induced autonomic dysfunction when administered 30 min prior to insult, but only if reperfusion is instituted after 30 min of MCAO. This same agent was ineffective if the MCAO was permanent, whereas pretreatment with an ERα agonist was effective after permanent MCAO [74]. Both ERα and ERβ may also be required to mediate 17βE2 enhancement of neurogenesis after focal ischemia [111]. In mice subjected to MCAO, 17βE2 is also reported to work via ERβ to attenuate neuroprotection induced by ischemic preconditioning [112].
5.2. Global ischemia.
There is more evidence that ERβ contributes to 17βE2’s ability to reduce neuronal loss induced by transient global ischemia than by focal ischemia. Two independent groups using different agonists showed that pretreatment with ERβ-selective agents can be neuroprotective. In mice, DPN reduced ischemic damage to the caudate nucleus [113], and in rats, WAY enhanced survival of hippocampal CA1 pyramidal neurons in some animals [81]. In the rat study, the ERα agonist PPT was also effective in a subset of animals, suggesting that either ER receptor subtype can mediate protection in this model. There is also a report that the phytoestrogen genistein can reduce ischemia-induced functional and histological damage in gerbils and that an ERβ-selective antagonist can reverse the beneficial effects of some doses of genistein [114].
5.3. β-amyloid toxicity, oxidative stress, and glutamate excitotoxicity
There is considerable evidence that ERβ participates in neuroprotective actions of 17βE2 in cultured cells. For example, many studies documenting a role for ERα also showed that transfection of cells with ERβ or treatment with ERβ-selective agonists was neuroprotective against glutamate [93–95] and β-amyloid toxicity [87]. In contrast, a recent study that employed both ER subtype-selective agonists and BERKO mice concluded that ERβ mediates 17βE2-induced protection against NMDA-induced toxicity in hippocampal slices [115]. Interestingly, estradiol increased protein levels of both ERα and ERβ in the latter study, whereas Cimarosti [116] reported selective up-regulation of ERβ in 17βE2-treated hippocampal explants subjected to oxygen-glucose deprivation.
5.4. Other models.
ERβ has been implicated as a mediator of neuroprotection in an acoustic trauma model of brain injury. This conclusion was based on studies of ERKO, BERKO and aromatase knockout mice treated with 17βE2 and on evaluation of ER subtype selective agonists [117]. Studies with an ERβ agonist also indicate that this receptor can attenuate TNFα-induced apoptosis in VSC4.1 motoneurons [79]. In a related study, genistein was shown to decrease apoptosis of cultured motoneurons exposed to supernatants containing microglial cytokines; this protection was reversed by ICI and was associated with increased expression of ERβ [118]. An ERβ agonist has also been reported to increase oligodendrocyte differentiation, improve myelination, and enhance axon conduction in a mouse model of chronic experimental autoimmune encephalomyelitis [119].
6. Role of GPR 30 and Other Non-Classical Estrogen Receptors in the Neuroprotective Effects of Estrogens
While most studies implicate either ERα or ERβ in the protective effects of estrogens, studies in which estradiol exerts positive effects in models that lack functional ERα and ERβ but express GPR30 [120–122] support the role of membrane associated estrogen receptors in the protective effects of estrogens. With respect to GPR30, not only has its activation been associated with neuroprotection [123], but also with improved spatial learning [124–127]. GPR30 is an important component of estrogen-mediated neuroprotection because, as G-protein coupled receptor, its activation can lead to rapid activation of intracellular signaling cascades (see examples of such cascades below) which in turn, could act in conjunction with the classical neuroprotective mechanisms associated with activation of the intracellular/nuclear receptors, ERα and ERβ. For example, activation of GPR30 can increase cAMP and calcium mobilization in neurons [128, 129]. Additionally, activation of this estrogen receptor in mouse cortical neurons can protect cells through a non-genomic mechanism in which ERK activation leads to increased Bcl-2 and decreased pro-caspase-3 [126]. Activation of the neuroprotection-associated PI3K-Akt pathway has also been implicated as a pathway elicited following GPR30 activation [130].
Other non-classical estrogen receptors have also been implicated in the protective effects of estrogens. For example, the Toran-Allerand laboratory proposed a novel estrogen receptor, termed ER-X [131], as a key mediator of estrogens’ actions on the brain. Interestingly, this receptor has also been proposed as the receptor for the endogenous diastereomer of 17βE2, 17α-estradiol [132]. And though its role in neuroprotection, per se, has not been specifically addressed, its expression increases in response to ischemic injury. This increase has been proposed as a potential compensatory mechanism to promote cell viability and function. Although other putative estrogen-binding proteins exist in the both nervous and non-nervous tissue, their roles in mediating estrogen-induced neuroprotection have yet to be determined.
7. Non-Receptor Mediated Effects of Estrogens in Neuroprotection
We made the seminal finding that the ERα/β ligand 17βE2 and its presumed inactive diastereomer, 17α-estradiol, which binds to these ER-isoforms with an approximately 40-fold lower affinity [133, 134], were equally potent in protecting neuronal cells from the effects were not blocked by an ER antagonist [135]. This observation prompted a closer evaluation of the structural requirements for neuroprotection among steroids [136, 137]; our data showed that estrogen analogues with little classical ER binding affinity were just as protective as 17βE2; therefore, we reasoned that a substantial portion of the neuroprotective activity of estrogens was ER-independent.
We [136] and others [137, 138] have determined that the phenolic nature of the 17βE2 molecule is essential for neuroprotection. Therefore, we synthesized estrogen analogues to perform structure-activity relationship (SAR) studies using rationally designed compounds in 17βE2-neuroprotection assays. Over 100 compounds were tested in HT-22 (murine hippocampal) cells for their ability to inhibit cytotoxicity against glutamate and iodoacetic acid. The EC50 (or IC50) values for neuroprotection, ER binding, and protection against lipid peroxidation were determined to ascertain potency in comparison with 17βE2 [133; Simpkins, unpublished observations]. Neuroprotective estratrienes that have electron donating constituents increase the redox potential of the phenoxy radical, providing better neuroprotective properties [133]. The donated hydrogen radical can quench free radicals formed in oxidative stress conditions. A-ring derivatives with electron donating constituents that stabilize the phenoxy radical were more potent than 17βE2 in protecting these cells from oxidative stress-induced toxicity. Our primary synthetic strategy was to replace the hydrogen with a bulky alkyl group at the 2- or 4-positions of the A-ring. For those compounds that included the addition of a single group to the 2-carbon position of the A-ring or 4-carbon position of the A-ring, there was an increase in potency by approximately ~2- to 170-fold as compared to the parent compounds, 17βE2 and estrone. When two groups flanked the 3-OH position, neuroprotection was enhanced, with approximately 9- and 4-fold decrease in EC50 values for protection against neurotoxicity, respectively. Preventing the ability of the phenolic A-ring to undergo redox chemistry by converting the phenolic OH to an OCH3 group eliminated the neuroprotection of the 2-substituted analogues. Switching the hydroxyl group to the 2-position and adding bulky groups (1-adamantyl or tert-buty) to the 3-position greatly improved neuroprotective potencies of the parent compound [133].
Introducing conjugated double bonds into the B- or C-rings of estratrienes is another way to increase the stability of the phenoxy radical. Compounds with these modifications were approximately 180- and 490-fold more potent than 17βE2 against glutamate and indoacetic acid toxicity, respectively. D-ring substituents alter lipophilicity; however, the addition of different alkyl side chains, esterification with benzoic acid and introduction of methyl ethers to the 17-position did not result in better neuroprotection, but instead decreased the potency of the parent compound.
In summary, we have determined critical features of the 17βE2 structure that dictate neuroprotective efficacy and thus advance our understanding of estrogen-mediated neuroprotection. A key observation is that stabilization of the phenoxy radical state increases potency against oxidative stress damage.
8. Signaling Pathways From ERs to Neuroprotection
In addition to promoting neuroprotection via classical genomic mechanisms (i.e., those which involve increased expression of genes and their associated cytoprotective proteins), activation of intracellular signaling pathways serves as another important mechanism by which estrogens promote brain cell health and viability. Given the multiplicity of estrogen receptors that can activate signaling pathways, however, the extent of overlap in the regulation of neuroprotection-associated signaling pathways triggered by these different receptors is still unclear. As stated above, GPR30 is a trans-membrane protein making it ideal for transducing an intracellular signaling cascade. However, because both ERα and ERβ can localize to the plasma membrane, they are not excluded as mediators of estrogen-induced rapid signaling [139–147]. Indeed ICI 182,780-sensitive estrogen receptors (ERα and ERβ) are responsible for neuroprotective signaling initiated by a membrane impermeable form of estrogen [148]. In other cases, GPR30 is the receptor that initiates protective signaling pathways 7]. Additionally, protein complexes consisting of B-Raf, hsp90, and the ERα are present in cortical tissue providing an intracellular junction by which ERs can converge with growth factor-associated signaling pathways 9].
Several groups have reported that activation of the ERK/MAP kinase pathway is essential for estrogen-induced neuroprotection [150–152]. Activation of the ERK/MAP kinase pathway is generally initiated with the activation of B-Raf by Ras, and through a series of sequential phosphorylation/activation events, leads to the phosphorylation and activation of ERK1/2. Activated ERK can, in turn, translocate to the nucleus and interact with various transcription factors to influence the expression of various cytoprotection-associated genes.
Estrogens may also activate the ERK/MAP kinase pathway through the interaction of ERα with metabotropic glutamate receptors at the plasma membrane, where such interaction has been reported to leads to the activation of a variety of second messengers [153]. In fact, membrane-associated estrogen receptors are able to activate metabotropic glutamate receptors even in the absence of glutamate, and in striatal neurons, can alter the phosphorylation of CREB, which can increase the expression of BDNF and other protective proteins [154].
Estrogens also activate the PI3K/Akt pathway in cortical neurons [155–157]. With respect to cell survival, activated Akt is known to decrease pro-apoptotic proteins within cells. Further, activation of the PI3K pathway by estrogen has been shown to increase the phosphorylation of 4E-binding protein 1 (4EBP1), which allows for the promotion of protein translation without altering mRNA levels [156].
Another signaling pathway affected by estrogens that has strong implications in maintaining brain function is the Rho A pathway. A recent study revealed that estrogen, acting through ERβ, is a positive modulator of the Rho A pathway, whose downstream signaling sequelae include alterations in synaptic physiology and long term potentiation [157–159].
While this is not a comprehensive list of all the cytoprotection-associated signaling pathways known to be activated by estrogen, and other pathways that include the Wnt/beta-catenin signaling pathway [160] or p53 [161] may be equally relevant, the examples provided are intended to support the fact that activation of intracellular signaling pathways, through one or more estrogen receptors, is a viable means by which estrogen promotes brain cell health, function and viability.
9. Role of ERs in Protective Effects of Other Hormones
Androgens, which include testosterone and its 5α- reduced metabolite, dihydrotestosterone (DHT), also exert protective effects on the brain. These effects of androgens can be mediated by the androgen receptor (AR) or, alternatively, through metabolites that bind to and activate ERs. For example, testosterone can be metabolized by the p450 enzyme aromatase to βE2. In contrast, DHT has long been considered the non-aromatizable androgen, and thus, used as a more “pure” AR-activating hormone. However, recent evidence suggests that the cytoprotective effects of DHT may also be mediated by the ERs, through prior conversion of DHT to 5α-androstane-3β, 17β-diol (3β-diol) by the enzyme, 3β-hydroxysteroid dehydrogenase [162]. 3β-diol does not bind appreciably to ARs but rather binds with nM affinity to the ERs [163]. These data suggest that androgens can have biologically meaningful effects through their metabolites, which elicit their effects through activation of ERs.
10. Future Research Needs and Conclusions
The discovery of selective and effective compounds for the treatment of estrogen deprivation symptoms, including vulnerability to neurodegeneration after the menopause, requires the determination of the receptor(s) involved in mediating there neuroprotective effects. As described above, there is strong evidence for the involvement of classical ERs, membrane ERs and non-ER mediated pathways in estrogen neuroprotection. This raises the possibility that estrogens either affect a multitude of receptors and/or pathways simultaneously, or that their effects are dose-, duration-, insult-, and/or animal model-specific. Resolving this issue is critical, not only in determining the targets for drug discovery programs for improved estrogens and for determining biomarkers of women who may benefit from estrogen therapy in the post-menopause, but also for our understanding of the mechanism(s) by which estrogen-deprivation leads to increased vulnerability of neurodegeneration following insult.
Intriguingly, estrogens are better studied for their neuroprotective effects than any other class of compounds, and despite this, and the rationale for clinical use of estrogens in injured patients [164] there is only one ongoing clinical trial of estrogen neuroprotection against traumatic brain Injury and hemorrhagic shock (Wigginton et al. unpublished observations). We submit that resolution of the ERs (or lack thereof) involved in estrogen-neuroprotection is critical to moving the field forward and to conducting appropriate clinical trails of selective estrogens for specific acute neurodegenerative insults.
References
- 1.Rossouw JE, Anderson GL, Prentice RL, LaCroix AZ, Kooperberg C, Stefanick ML, Jackson RD, Beresford SA, Howard BV, Johnson KC, Kotchen JM, Ockene J; Writing Group for the Women’s Health Initiative Investigators: Risks and benefits of estrogen plus progestin in healthy postmenopausal women: principal results From the Women’s Health Initiative randomized controlled trial. JAMA 2002; 288: 321–233. [DOI] [PubMed] [Google Scholar]
- 2.Henderson VW, Brinton RD: Menopause and mitochondria: windows into estrogen effects on Alzheimer’s disease risk and therapy. Prog Brain Res 2010;182: 77–96. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Bourque M, Dluzen DE, Di Paolo T: Neuroprotective actions of sex steroids in Parkinson’s disease. Front Neuroendocrinol 2009. July;30(2):142–57. [DOI] [PubMed] [Google Scholar]
- 4.Lebesgue D, Chevaleyre V, Zukin RS, Etgen AM: Estradiol rescues neurons from global ischemia-induced cell death: multiple cellular pathways of neuroprotection. Steroids 2009; 74: 555–561. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Morissette M, Le Saux M, D’Astous M, Jourdain S, Al Sweidi S, Morin N, Estrada-Camarena E, Mendez P, Garcia-Segura LM, Di Paolo T: Contribution of estrogen receptors alpha and beta to the effects of estradiol in the brain. J Steroid Biochem Mol Biol 2008; 108: 327–38. [DOI] [PubMed] [Google Scholar]
- 6.Brann DW, Dhandapani K, Wakade C, Mahesh VB, Khan MM: Neurotrophic and neuroprotective actions of estrogen: basic mechanisms and clinical implications. Steroids 2007; 72: 381–405. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Mendez P, Azcoitia I, Garcia-Segura LM: Interdependence of oestrogen and insulin-like growth factor-I in the brain: potential for analysing neuroprotective mechanisms. J Endocrinol 2005; 185: 11–17. [DOI] [PubMed] [Google Scholar]
- 8.Singh M, Dykens JA, Simpkins JW: Novel mechanisms for estrogen-induced neuroprotection. Exp Biol Med 2006; 231: 514–521. [DOI] [PubMed] [Google Scholar]
- 9.Wise PM, Dubal DB, Rau SW, Brown CM, Suzuki S: Are estrogens protective or risk factors in brain injury and neurodegeneration? Reevaluation after the Women’s health initiative. Endocr Rev 2005; 26: 308–312. [DOI] [PubMed] [Google Scholar]
- 10.Scott E, Zhang QG, Wang R, Vadlamudi R, Brann D: Estrogen neuroprotection and the critical period hypothesis. Front Neuroendocrinol 2011. November 4 [Epub ahead of print] [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Azcoitia I, Santos-Galindo M, Arevalo MA, Garcia-Segura LM: Role of astroglia in the neuroplastic and neuroprotective actions of estradiol. Eur J Neurosci 2010; 32: 1995–2002. [DOI] [PubMed] [Google Scholar]
- 12.Dhandapani KM, Brann DW: Role of astrocytes in estrogen-mediated neuroprotection. Exp Gerontol 2007; 42: 70–75. [DOI] [PubMed] [Google Scholar]
- 13.Jung-Testas I, Baulieu EE: Steroid hormone receptors and steroid action in rat glial cells of the central and peripheral nervous system. J Steroid Biochem Mol Biol 1998; 65: 243–251. [DOI] [PubMed] [Google Scholar]
- 14.Mong JA, Blutstein T: Estradiol modulation of astrocytic form and function: implications for hormonal control of synaptic communication. Neuroscience. 2006; 138: 967–975. [DOI] [PubMed] [Google Scholar]
- 15.Simpkins JW, Rajakumar G, Zhang YQ, Simpkins CE, Greenwald D, Yu CJ, Bodor N, Day AL: Estrogens may reduce mortality and ischemic damage caused by middle cerebral artery occlusion in the female rat. J Neurosurg 1997; 87: 724–730. [DOI] [PubMed] [Google Scholar]
- 16.Bishop J and Simpkins JW: Estradiol treatment increases viability of glioma and neuroblastoma cells in vitro. Mol Cell Neurosci 1994; 5: 303–308. [DOI] [PubMed] [Google Scholar]
- 17.Green PS and Simpkins JW: Neuroprotective effects of estrogens: potential mechanisms of action. Int J Dev Neurosci, 2000; 18: 347–358. [DOI] [PubMed] [Google Scholar]
- 18.Behl C, Skutella T, Lezoualc’h F, Post A, Widmann M, Newton CJ, Holsboer F: Neuroprotection against oxidative stress by estrogens: structure-activity relationship. Mol Pharmacol 1997; 51: 535–541. [PubMed] [Google Scholar]
- 19.Weaver CE Jr., Park-Chung M, Gibbs TT and Farb DH: 17beta-Estradiol protects against NMDA-induced excitotoxicity by direct inhibition of NMDA receptors. Brain Res 1997; 761: 338–341. [DOI] [PubMed] [Google Scholar]
- 20.Singh M, Setalo G Jr, Guan X, Frail DE and Toran-Allerand CD: Estrogen-induced activation of the mitogen-activated protein kinase cascade in the cerebral cortex of estrogen receptor-alpha knock-out mice. J Neurosci 2000; 20: 1694–1700. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Honda K, Sawada H, Kihara T, Urushitani M, Nakamizo T, Akaike A, Shimohama S: Phosphatidylinositol 3-kinase mediates neuroprotection by estrogen in cultured cortical neurons. J Neurosci Res 2000; 60: 321–327. [DOI] [PubMed] [Google Scholar]
- 22.Alkayed NJ, Harukuni I, Kimes AS, London ED, Traystman RJ and Hurn PD: Gender-linked brain injury in experimental stroke. Stroke 1998; 29: 159–165. [DOI] [PubMed] [Google Scholar]
- 23.Dubal DB, Kashon ML, Pettigrew LC, Ren JM, Finklestein SP, Rau SW, Wise PM: Estradiol protects against ischemic injury. J Cereb Blood Flow Metab 1998; 18: 1253–1258. [DOI] [PubMed] [Google Scholar]
- 24.Sudo S, Wen TC, Desaki J, Matsuda S, Tanaka J, Arai T, Maeda N, Sakanaka M: Beta-estradiol protects hippocampal CA1 neurons against transient forebrain ischemia in gerbil. Neurosci Res 1997; 29: 345–354. [DOI] [PubMed] [Google Scholar]
- 25.Fukuda K, Yao H, Ibayashi S, Nakahara T, Uchimura H, Fujishima M, Hall ED: Ovariectomy exacerbates and estrogen replacement attenuates photothrombotic focal ischemic brain injury in rats. Stroke 2000; 31: 155–160. [DOI] [PubMed] [Google Scholar]
- 26.Mendelowitsch A, Ritz MF, Ros J, Langemann H and Gratzl O: 17beta-Estradiol reduces cortical lesion size in the glutamate excitotoxicity model by enhancing extracellular lactate: a new neuroprotective pathway. Brain Res 200; 1901: 230–236. [DOI] [PubMed] [Google Scholar]
- 27.Chen J, Xu W and Jiang H: 17 beta-estradiol protects neurons from ischemic damage and attenuates accumulation of extracellular excitatory amino acids. Anesth Analg 2001; 92: 1520–1523. [DOI] [PubMed] [Google Scholar]
- 28.Culmsee C, Vedder H, Ravati A, Junker V, Otto D, Ahlemeyer B, Krieg JC, Krieglstein J: Neuroprotection by estrogens in a mouse model of focal cerebral ischemia and in cultured neurons: evidence for a receptor-independent antioxidative mechanism. J Cereb Blood Flow Metab 1999; 19: 1263–1269. [DOI] [PubMed] [Google Scholar]
- 29.Wise PM, Dubal DB, Wilson ME, Rau SW, Bottner M and Rosewell KL: Estradiol is a protective factor in the adult and aging brain: understanding of mechanisms derived from in vivo and in vitro studies. Brain Res Brain Res Rev 37: 2001; 313–319. [DOI] [PubMed] [Google Scholar]
- 30.Selvamani A, Sohrabji F: The neurotoxic effects of estrogens on ischemic stroke in older female rats is associated with age-dependent loss of insulin-like growth factor-1. J. Neurosci 2010; 30: 682–6861. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Lewis DK, Thomas KT, Selvamani A, Sohrabji F: Age-related severity of focal ischemia in female rats is associated with impaired astrocyte function, Neurobiology of Aging Electronic publication December 9, 2011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Toung TK, Hurn PD, Traystman RJ and Sieber FE: Estrogen decreases infarct size after temporary focal ischemia in a genetic model of type 1 diabetes mellitus. Stroke 31: 2000; 2701–2706. [DOI] [PubMed] [Google Scholar]
- 33.Carswell HV, Dominiczak AF and Macrae IM: Estrogen status affects sensitivity to focal cerebral ischemia in stroke-prone spontaneously hypertensive rats. Am J Physiol Heart Circ Physiol 278: 2000; H290–294. [DOI] [PubMed] [Google Scholar]
- 34.Yang SH, He Z, Wu SS, He YJ, Cutright J, Millard WJ, Day AL, Simpkins JW: 17-beta estradiol can reduce secondary ischemic damage and mortality of subarachnoid hemorrhage. J Cereb Blood Flow Metab 2001; 21: 174–181. [DOI] [PubMed] [Google Scholar]
- 35.Hawk T, Zhang YQ, Rajakumar G, Day AL and Simpkins JW: Testosterone increases and estradiol decreases middle cerebral artery occlusion lesion size in male rats. Brain Res 1998; 796: 296–298. [DOI] [PubMed] [Google Scholar]
- 36.Toung TJ, Traystman RJ and Hurn PD: Estrogen-mediated neuroprotection after experimental stroke in male rats. Stroke 1998; 29: 1666–1670. [DOI] [PubMed] [Google Scholar]
- 37.McCullough LD, Alkayed NJ, Traystman RJ, Williams MJ and Hurn PD: Postischemic estrogen reduces hypoperfusion and secondary ischemia after experimental stroke. Stroke 32: 2001; 796–802. [DOI] [PubMed] [Google Scholar]
- 38.Yang SH, Shi J, Day AL and Simpkins JW, Estradiol exerts neuroprotective effects when administered after ischemic insult. Stroke 31: 2000; 745–749. [DOI] [PubMed] [Google Scholar]
- 39.Yang SH, Liu R, Wu SS and Simpkins JW: The use of estrogens and related compounds in the treatment of damage from cerebral ischemia. Ann N Y Acad Sci 1007: 2003; 101–107. [DOI] [PubMed] [Google Scholar]
- 40.Liu R, Wang X, Liu Q, Yang SH, Simpkins JW: Dose dependence and therapeutic window for the neuroprotective effects of 17beta-estradiol when administered after cerebral ischemia. Neurosci Lett 2007; 415: 237–241. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.De Keyser J, Sulter G and Luiten PG: Clinical trials with neuroprotective drugs in acute ischaemic stroke: are we doing the right thing? Trends Neurosci 22: 1999; 535–540. [DOI] [PubMed] [Google Scholar]
- 42.Clark RA and Bessinger R: Clinical manifestations and predictors of survival in older women infected with HIV. J Acquir Immune Defic Syndr Hum Retrovirol 1997; 15: 341–345. [DOI] [PubMed] [Google Scholar]
- 43.Henderson VW, Paganini-Hill A, Emanuel CK, Dunn ME, and Buckwalter JG: Estrogen replacement therapy in older women. Comparisons between Alzheimer’s disease cases and nondemented control subjects. Arch Neurol 1994; 51: 896–900. [DOI] [PubMed] [Google Scholar]
- 44.Paganini-Hill A and Henderson VW: Estrogen deficiency and risk of Alzheimer’s disease in women. Am J Epidemiol 1994; 140: 256–261. [DOI] [PubMed] [Google Scholar]
- 45.Phillips SM and Sherwin BB: Effects of estrogen on memory function in surgically menopausal women. Psychoneuroendocrinology 1992; 17: 485–495. [DOI] [PubMed] [Google Scholar]
- 46.Sherwin BB: Estrogen and cognitive functioning in women. Proc Soc Exp Biol Med 1998; 217: 17–22. [DOI] [PubMed] [Google Scholar]
- 47.Sherwin BB: Estrogen effects on cognition in menopausal women. Neurology 1997; 48: S21–26. [DOI] [PubMed] [Google Scholar]
- 48.Maki PM, Zonderman AB, and Resnick SM: Enhanced verbal memory in nondemented elderly women receiving hormone-replacement therapy. Am J Psychiatry 2001; 158: 227–233. [DOI] [PubMed] [Google Scholar]
- 49.Resnick SM and Maki PM: Effects of hormone replacement therapy on cognitive and brain aging. Ann N Y Acad Sci 2001; 949: 203–214. [DOI] [PubMed] [Google Scholar]
- 50.Resnick SM, Maki PM, Golski S, Kraut MA, and Zonderman AB: Effects of estrogen replacement therapy on PET cerebral blood flow and neuropsychological performance. Horm Behav 1998; 34: 171–82. [DOI] [PubMed] [Google Scholar]
- 51.Espeland MA, Rapp SR, Shumaker SA, Brunner R, Manson JE, Sherwin BB, Hsia J, Margolis KL, Hogan PE, Wallace R, Dailey M, Freeman R, and Hays J: Conjugated equine estrogens and global cognitive function in postmenopausal women: Women’s Health Initiative Memory Study. JAMA, 2004; 291: 2959–2968. [DOI] [PubMed] [Google Scholar]
- 52.Rapp SR, Espeland MA, Shumaker SA, Henderson VW, Brunner RL, Manson JE, Gass ML, Stefanick ML, Lane DS, Hays J, Johnson KC, Coker LH, Dailey M, and Bowen D: Effect of estrogen plus progestin on global cognitive function in postmenopausal women: the Women’s Health Initiative Memory Study: a randomized controlled trial. JAMA 2003; 289: 2663–2672. [DOI] [PubMed] [Google Scholar]
- 53.Shumaker SA, Legault C, Kuller L, Rapp SR, Thal L, Lane DS, Fillit H, Stefanick ML: Hendrix SL, Lewis CE, Masaki K, and Coker LH: Conjugated equine estrogens and incidence of probable dementia and mild cognitive impairment in postmenopausal women: Women’s Health Initiative Memory Study. JAMA 2004; 291: 2947–2958. [DOI] [PubMed] [Google Scholar]
- 54.Shumaker SA, Legault C, Rapp SR, Thal L, Wallace RB, Ockene JK, Hendrix SL, Jones BN 3rd, Assaf AR, Jackson RD, Kotchen JM, Wassertheil-Smoller S, and Wactawski-Wende J: 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 2003; 289: 2651–2662. [DOI] [PubMed] [Google Scholar]
- 55.Mulnard RA, Cotman CW, Kawas C, van Dyck CH, Sano M, Doody R, Koss E, Pfeiffer E, Jin S, Gamst A, Grundman M, Thomas R, and Thal LJ: Estrogen replacement therapy for treatment of mild to moderate Alzheimer disease: a randomized controlled trial. Alzheimer’s Disease Cooperative Study. JAMA 2000; 283: 1007–1015. [DOI] [PubMed] [Google Scholar]
- 56.Marder K, Tang MX, Alfaro B, Mejia H, Cote L, Jacobs D, Stern Y, Sano M, and Mayeux R: Postmenopausal estrogen use and Parkinson’s disease with and without dementia. Neurology 1998; 50: 1141–1143. [DOI] [PubMed] [Google Scholar]
- 57.Horstink MW, Strijks E, and Dluzen DE: Estrogen and Parkinson’s disease. Adv Neurol 2003; 91: 107–114. [PubMed] [Google Scholar]
- 58.Paganini-Hill A: Estrogen replacement therapy and stroke. Prog Cardiovasc Dis 1995; 38: 223–242. [DOI] [PubMed] [Google Scholar]
- 59.Viscoli CM, Brass LM, Kernan WN, Sarrel PM, Suissa S, and Horwitz RI: A clinical trial of estrogen-replacement therapy after ischemic stroke. N Engl J Med 2001; 345: 1243–1249. [DOI] [PubMed] [Google Scholar]
- 60.Finucane FF, Madans JH, Bush TL, Wolf PH, and Kleinman JC: Decreased risk of stroke among postmenopausal hormone users. Results from a national cohort. Arch Intern Med 1993; 153: 73–79. [PubMed] [Google Scholar]
- 61.Henderson BE, Paganini-Hill A, and Ross RK: Decreased mortality in users of estrogen replacement therapy. Arch Intern Med 1991; 151: 75–78. [PubMed] [Google Scholar]
- 62.Falkeborn M, Persson I, Terent A, Adami HO, Lithell H, and Bergstrom R: Hormone replacement therapy and the risk of stroke. Follow-up of a population-based cohort in Sweden. Arch Intern Med 1993; 153: 1201–1209. [PubMed] [Google Scholar]
- 63.Groswasser Z, Cohen M, and Keren O: Female TBI patients recover better than males. Brain Inj 1998; 12: 805–808. [DOI] [PubMed] [Google Scholar]
- 64.Bayir H, Marion DW, Puccio AM, Wisniewski SR, Janesko KL, Clark RS, and Kochanek PM: Marked gender effect on lipid peroxidation after severe traumatic brain injury in adult patients. J Neurotrauma 2004; 21: 1–8. [DOI] [PubMed] [Google Scholar]
- 65.Manson JE, Hsia J, Johnson KC, Rossouw JE, Assaf AR, Lasser NL, Trevisan M, Black HR, Heckbert SR, Detrano R, Strickland OL, Wong ND, Crouse JR, Stein E, Cushman M; Women’s Health Initiative Investigators: Estrogen plus progestin and the risk of coronary heart disease. N Engl J Med 2003;349: 523–534. [DOI] [PubMed] [Google Scholar]
- 66.Anderson GL, Limacher M, Assaf AR, Bassford T, Beresford SA, Black H, Bonds D, Brunner R, Brzyski R, Caan B, Chlebowski R, Curb D, Gass M, Hays J, Heiss G, Hendrix S, Howard BV, Hsia J, Hubbell A, Jackson R, Johnson KC, Judd H, Kotchen JM, Kuller L, LaCroix AZ, Lane D, Langer RD, Lasser N, Lewis CE, Manson J, Margolis K, Ockene J, O’Sullivan MJ, Phillips L, Prentice RL, Ritenbaugh C, Robbins J, Rossouw JE, Sarto G, Stefanick ML, Van Horn L, Wactawski-Wende J, Wallace R, Wassertheil-Smoller S; Women’s Health Initiative Steering Committee. Effects of conjugated equine estrogen in postmenopausal women with hysterectomy: the Women’s Health Initiative randomized controlled trial: JAMA. 2004; 291: 1701–1712. [DOI] [PubMed] [Google Scholar]
- 67.Hsia J, Langer RD, Manson JE, Kuller L, Johnson KC, Hendrix SL, Pettinger M, Heckbert SR, Greep N, Crawford S, Eaton CB, Kostis JB, Caralis P, Prentice R; Women’s Health Initiative Investigators: Conjugated equine estrogens and coronary heart disease: the Women’s Health Initiative. Arch Intern Med 2006;166: 357–365. [DOI] [PubMed] [Google Scholar]
- 68.Hoffman GE, Merchenthaler I, and Zup SL: Neuroprotection by ovarian hormones in animal models of neurological disease. Endocrine 2006; 29: 217–231. [DOI] [PubMed] [Google Scholar]
- 69.Merchenthaler I, Dellovade TL, and Shughrue PJ: Neuroprotection by estrogen in animal models of global and focal ischemia. Ann N Y Acad Sci 2003; 1007: 89–100. [DOI] [PubMed] [Google Scholar]
- 70.Sawada M, Alkayed NJ, Goto S, Crain BJ, Traystman RJ, Shaivitz A, Nelson RJ, Hurn PD: Estrogen receptor antagonist ICI182,780 exacerbates ischemic injury in female mouse. J Cereb Blood Flow Metab 2000; 20: 112–118. [DOI] [PubMed] [Google Scholar]
- 71.Amantea D, Spagnuolo P, Bari M, Fezza F, Mazzei C, Tassorelli C, Morrone LA, Corasaniti MT, Maccarrone M, Bagetta G: Modulation of the endocannabinoid system by focal brain ischemia in the rat is involved in neuroprotection afforded by 17beta-estradiol. Febs J 2007; 274: 4464–4775. [DOI] [PubMed] [Google Scholar]
- 72.Dubal DB, Zhu H, Yu J, Rau SW, Shughrue PJ, Merchenthaler I, Kindy MS, Wise PM: Estrogen receptor alpha, not beta, is a critical link in estradiol-mediated protection against brain injury. Proc Natl Acad Sci U S A 2001; 98: 1952–1957. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Shimada K, Kitazato KT, Kinouchi T, Yagi K, Tada Y, Satomi J, Kageji T, Nagahiro S: Activation of estrogen receptor-alpha and of angiotensin-converting enzyme 2 suppresses ischemic brain damage in oophorectomized rats. Hypertension 2011; 57: 1161–1166. [DOI] [PubMed] [Google Scholar]
- 74.Connell B and Saleh TM: Differential Neuroprotection of Selective Estrogen Receptor Agonists against Autonomic Dysfunction and Ischemic Cell Death in Permanent versus Reperfusion Injury. Adv Pharmacol Sci 2011; 976951. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Elzer JG, Muhammad S, Wintermantel TM, Regnier-Vigouroux A, Ludwig J, Schütz G, Schwaninger M: Neuronal estrogen receptor-alpha mediates neuroprotection by 17beta-estradiol. J Cereb Blood Flow Metab 2010; 30: 935–942. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Farr TD, Carswell HV, Gsell W, Macrae IM: Estrogen receptor beta agonist diarylpropiolnitrile (DPN) does not mediate neuroprotection in a rat model of permanent focal ischemia. Brain Res 2007; 1185: 275–282. [DOI] [PubMed] [Google Scholar]
- 77.Sampei K, Goto S, Alkayed NJ, Crain BJ, Korach KS, Traystman RJ, Demas GE, Nelson RJ, Hurn PD: Stroke in estrogen receptor-alpha-deficient mice. Stroke 2000; 31: 738–743. [DOI] [PubMed] [Google Scholar]
- 78.Pulsinelli WA, Buchan AM: The four-vessel occlusion rat model: method for complete occlusion of vertebral arteries and control of collateral circulation. Stroke. 1988; 19:913–914. [DOI] [PubMed] [Google Scholar]
- 79.Das A, Smith JA, Gibson C, Varma AK, Ray SK, Banik NL: Estrogen receptor agonists and estrogen attenuate TNF-alpha-induced apoptosis in VSC4.1 motoneurons. J Endocrinol 2011; 208: 171–182. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Jover-Mengual T, Zukin RS, and Etgen AM: MAPK signaling is critical to estradiol protection of CA1 neurons in global ischemia. Endocrinology 2007; 148: 1131–1143. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Miller NR, Jover T, Cohen HW, Zukin RS, Etgen AM: Estrogen can act via estrogen receptor alpha and beta to protect hippocampal neurons against global ischemia-induced cell death. Endocrinology 2005; 146: 3070–3079. [DOI] [PubMed] [Google Scholar]
- 82.Zhang QG, Raz L, Wang R, Han D, De Sevilla L, Yang F, Vadlamudi RK, Brann DW: Estrogen attenuates ischemic oxidative damage via an estrogen receptor alpha-mediated inhibition of NADPH oxidase activation. J Neurosci 2009; 29: 13823–13836. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Azcoitia I, Sierra A, and Garcia-Segura LM: Neuroprotective effects of estradiol in the adult rat hippocampus: interaction with insulin-like growth factor-I signalling. J Neurosci Res 1999; 58: 815–822. [DOI] [PubMed] [Google Scholar]
- 84.Dai X, Chen L, and Sokabe M: Neurosteroid estradiol rescues ischemia-induced deficit in the long-term potentiation of rat hippocampal CA1 neurons. Neuropharmacology 2007; 52: 1124–1138. [DOI] [PubMed] [Google Scholar]
- 85.Zhang QG, Han D, Wang RM, Dong Y, Yang F, Vadlamudi RK, Brann DW: C terminus of Hsc70-interacting protein (CHIP)-mediated degradation of hippocampal estrogen receptor-alpha and the critical period hypothesis of estrogen neuroprotection. Proc Natl Acad Sci U S A 2011; 108: E617–E624. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86.Xia Y, Xing JZ, and Krukoff TL: Neuroprotective effects of R,R-tetrahydrochrysene against glutamate-induced cell death through anti-excitotoxic and antioxidant actions involving estrogen receptor-dependent and -independent pathways. Neuroscience 2009; 162: 292–306. [DOI] [PubMed] [Google Scholar]
- 87.Fitzpatrick JL, Mize AL, Wade CB, Harris JA, Shapiro RA, Dorsa DM: Estrogen-mediated neuroprotection against beta-amyloid toxicity requires expression of estrogen receptor alpha or beta and activation of the MAPK pathway. J Neurochem 2002; 82: 674–682. [DOI] [PubMed] [Google Scholar]
- 88.Marin R, Guerra B, Morales A, Diaz M, Alonso R: An ICI 182,780-sensitive, membrane-related estrogen receptor contributes to estrogenic neuroprotective actions against amyloid-beta toxicity. Ann N Y Acad Sci 2003; 1007: 108–116. [DOI] [PubMed] [Google Scholar]
- 89.Cordey M and Pike CJ: Neuroprotective properties of selective estrogen receptor agonists in cultured neurons. Brain Res 2005; 1045: 217–223. [DOI] [PubMed] [Google Scholar]
- 90.Marin R, Ramírez C, Morales A, González M, Alonso R, Díaz M: Modulation of Abeta-induced neurotoxicity by estrogen receptor alpha and other associated proteins in lipid rafts. Steroids 2008; 73: 992–996. [DOI] [PubMed] [Google Scholar]
- 91.Luciani P, Deledda C, Rosati F, Benvenuti S, Cellai I, Dichiara F, Morello M, Vannelli GB, Danza G, Serio M, Peri A: Seladin-1 is a fundamental mediator of the neuroprotective effects of estrogen in human neuroblast long-term cell cultures. Endocrinology 2008; 149: 4256–4266. [DOI] [PubMed] [Google Scholar]
- 92.Spampinato SF, Molinaro G, Merlo S, Iacovelli L, Caraci F, Battaglia G, Nicoletti F, Bruno V, Sortino MA Estrogen Receptors and Type-1 Metabotropic Glutamate Receptors are Interdependent in Protecting Cortical Neurons against {beta}-amyloid Toxicity. Mol Pharmacol 2012; 81: 12–20. [DOI] [PubMed] [Google Scholar]
- 93.Mize AL, Shapiro RA, and Dorsa DM: Estrogen receptor-mediated neuroprotection from oxidative stress requires activation of the mitogen-activated protein kinase pathway. Endocrinology 2003; 144: 306–312. [DOI] [PubMed] [Google Scholar]
- 94.Zhao L and Brinton RD: Estrogen receptor alpha and beta differentially regulate intracellular Ca(2+) dynamics leading to ERK phosphorylation and estrogen neuroprotection in hippocampal neurons. Brain Res 2007; 1172: 48–59. [DOI] [PubMed] [Google Scholar]
- 95.Zhao L, Wu TW, and Brinton RD: Estrogen receptor subtypes alpha and beta contribute to neuroprotection and increased Bcl-2 expression in primary hippocampal neurons. Brain Res 2004; 1010: 22–34. [DOI] [PubMed] [Google Scholar]
- 96.Sribnick EA, Ray SK, and Banik NL: Estrogen prevents glutamate-induced apoptosis in C6 glioma cells by a receptor-mediated mechanism. Neuroscience 2006; 137: 197–209. [DOI] [PubMed] [Google Scholar]
- 97.Gingerich S, Kim GL, Chalmers JA, Koletar MM, Wang X, Wang Y, Belsham DD: Estrogen receptor alpha and G-protein coupled receptor 30 mediate the neuroprotective effects of 17beta-estradiol in novel murine hippocampal cell models. Neuroscience 2010; 170: 54–66. [DOI] [PubMed] [Google Scholar]
- 98.Bryant DN and Dorsa DM: Roles of estrogen receptors alpha and beta in sexually dimorphic neuroprotection against glutamate toxicity. Neuroscience 2010; 170: 1261–1269. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99.Gamerdinger M, Manthey D, and Behl C: Oestrogen receptor subtype-specific repression of calpain expression and calpain enzymatic activity in neuronal cells--implications for neuroprotection against Ca-mediated excitotoxicity. J Neurochem 2006; 97: 57–68. [DOI] [PubMed] [Google Scholar]
- 100.Al Sweidi S, Sánchez MG, Bourque M, Morissette M, Dluzen D, Di Paolo T: Oestrogen receptors and signalling pathways: implications for neuroprotective effects of sex steroids in parkinson’s disease. J Neuroendocrinol 2011; 24: 48–61. [DOI] [PubMed] [Google Scholar]
- 101.Bains M, Cousins JC, and Roberts JL: Neuroprotection by estrogen against MPP+-induced dopamine neuron death is mediated by ERalpha in primary cultures of mouse mesencephalon. Exp Neurol 2007; 204: 767–776. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102.D’Astous M, Mendez P, Morissette M, Garcia-Segura LM, Di Paolo T: Implication of the phosphatidylinositol-3 kinase/protein kinase B signaling pathway in the neuroprotective effect of estradiol in the striatum of 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine mice. Mol Pharmacol 2006; 69: 1492–1498. [DOI] [PubMed] [Google Scholar]
- 103.D’Astous M, Morissette M, and Di Paolo T: Effect of estrogen receptor agonists treatment in MPTP mice: evidence of neuroprotection by an ER alpha agonist. Neuropharmacology 2004; 47: 1180–1188. [DOI] [PubMed] [Google Scholar]
- 104.Morissette M, Le Saux M, D’Astous M, Jourdain S, Al Sweidi S, Morin N, Estrada-Camarena E, Mendez P, Garcia-Segura LM, Di Paolo T: Contribution of estrogen receptors alpha and beta to the effects of estradiol in the brain. J Steroid Biochem Mol Biol 2008; 108: 327–338. [DOI] [PubMed] [Google Scholar]
- 105.Bao YJ, Li LZ, Li XG, Wang YJ: 17Beta-estradiol differentially protects cortical pericontusional zone from programmed cell death after traumatic cerebral contusion at distinct stages via non-genomic and genomic pathways. Mol Cell Neurosci 2011; 48: 185–194. [DOI] [PubMed] [Google Scholar]
- 106.Spence RD, Hamby ME, Umeda E, Itoh N, Du S, Wisdom AJ, Cao Y, Bondar G, Lam J, Ao Y, Sandoval F, Suriany MV, Voskuhl RR: Neuroprotection mediated through estrogen receptor-alpha in astrocytes. Proc Natl Acad Sci U S A 2011; 108: 8867–8872. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107.Guo J, Krause DN, Horne J, Weiss JH, Li X, Duckles SP: Estrogen-receptor-mediated protection of cerebral endothelial cell viability and mitochondrial function after ischemic insult in vitro. J Cereb Blood Flow Metab 2010; 30: 545–554. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 108.Liu X, Fan XL, Zhao Y, Luo GR, Li XP, Li R, Le WD: Estrogen provides neuroprotection against activated microglia-induced dopaminergic neuronal injury through both estrogen receptor-alpha and estrogen receptor-beta in microglia. J Neurosci Res 2005; 81: 653–665. [DOI] [PubMed] [Google Scholar]
- 109.Russo R, Cavaliere F, Watanabe C, Nucci C, Bagetta G, Corasaniti MT, Sakurada S, Morrone LA: 17Beta-estradiol prevents retinal ganglion cell loss induced by acute rise of intraocular pressure in rat. Prog Brain Res 2008; 173: 583–590. [DOI] [PubMed] [Google Scholar]
- 110.Zhou X, Li F, Ge J, Sarkisian SR Jr, Tomita H, Zaharia A, Chodosh J, Cao W: Retinal ganglion cell protection by 17-beta-estradiol in a mouse model of inherited glaucoma. Dev Neurobiol 2007; 67: 603–616. [DOI] [PubMed] [Google Scholar]
- 111.Suzuki S, Gerhold LM, Böttner M, Rau SW, Dela Cruz C, Yang E, Zhu H, Yu J, Cashion AB, Kindy MS, Merchenthaler I, Gage FH, Wise PM: Estradiol enhances neurogenesis following ischemic stroke through estrogen receptors alpha and beta. J Comp Neurol 2007; 500: 1064–1075. [DOI] [PubMed] [Google Scholar]
- 112.Wang L, Kitano H, Hurn PD, Murphy SJ: Estradiol attenuates neuroprotective benefits of isoflurane preconditioning in ischemic mouse brain. J Cereb Blood Flow Metab 2008; 28: 1824–1834. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 113.Carswell HV, Macrae IM, Gallagher L, Harrop E, Horsburgh KJ:Neuroprotection by a selective estrogen receptor beta agonist in a mouse model of global ischemia. Am J Physiol Heart Circ Physiol 2004; 287: H1501–H1504. [DOI] [PubMed] [Google Scholar]
- 114.Donzelli A, Braida D, Finardi A, Capurro V, Valsecchi AE, Colleoni M, Sala M: Neuroprotective effects of genistein in Mongolian gerbils: estrogen receptor-beta involvement. J Pharmacol Sci 2010; 114: 158–167. [DOI] [PubMed] [Google Scholar]
- 115.Aguirre C, Jayaraman A, Pike C, Baudry M: Progesterone inhibits estrogen-mediated neuroprotection against excitotoxicity by down-regulating estrogen receptor-beta. J Neurochem 2010; 115: 1277–87. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 116.Cimarosti H, O’Shea RD, Jones NM, Horn AP, Simão F, Zamin LL, Nassif M, Frozza R, Netto CA, Beart PM, Salbego C: The effects of estradiol on estrogen receptor and glutamate transporter expression in organotypic hippocampal cultures exposed to oxygen--glucose deprivation. Neurochem Res 2006; 31: 483–90. [DOI] [PubMed] [Google Scholar]
- 117.Meltser I, Tahera Y, Simpson E, Hultcrantz M, Charitidi K, Gustafsson JA, Canlon B: Estrogen receptor beta protects against acoustic trauma in mice . J Clin Invest 2008; 118: 1563–70. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 118.McDowell ML, Das A, Smith JA, Varma AK, Ray SK, Banik NL: Neuroprotective effects of genistein in VSC4.1 motoneurons exposed to activated microglial cytokines. Neurochem Int 2011; 59: 175–84. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 119.Crawford DK, Mangiardi M, Song B, Patel R, Du S, Sofroniew MV, Voskuhl RR, Tiwari-Woodruff SK:Oestrogen receptor beta ligand: a novel treatment to enhance endogenous functional remyelination. Brain 2010; 133: 2999–3016. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 120.Gu Q, Moss RL: Rapid action of 17beta-estradiol on kainate-induced currents in hippocampal neurons lacking intracellular estrogen receptors. Endocrinology 1999, 140: 660–666. [DOI] [PubMed] [Google Scholar]
- 121.Fugger HN, Kumar A, Lubahn DB, Korach KS, Foster TC.: Examination of estradiol effects on the rapid estradiol mediated increase in hippocampal synaptic transmission in estrogen receptor alpha knockout mice. Neurosci Lett 200; 309: 207–209. [DOI] [PubMed] [Google Scholar]
- 122.Bi R, Broutman G, Foy MR, Thompson RF, Baudry M. : The tyrosine kinase and mitogen-activated protein kinase pathways mediate multiple effects of estrogen in hippocampus. Proc Natl Acad Sci U S A. 2000; 97: 3602–3607. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 123.Lebesgue D, Traub M, De Butte-Smith M, Che C, Zukin RS, Kelly MJ, Etgen AM. Acute administration of non-classical estrogen receptor agonists attenuates ischemia-induced hippocampal neuron loss in middle-aged female rats. PLoS One 2010; 5:e8642. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 124.Abdelhamid R, Luo J, Vandevrede L, Kundu I, Michalsen B, Litosh VA, Schiefer IT, Gherezghiher T, Yao P, Qin Z, Thatcher GR: Benzothiophene Selective Estrogen Receptor Modulators Provide Neuroprotection by a novel GPR30-dependent Mechanism. ACS Chemical Neuroscience 2011; 2: 256–268. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 125.Gingerich S, Kim GL, Chalmers JA, Koletar MM, Wang X, Wang Y, Belsham DD: Estrogen receptor alpha and G-protein coupled receptor 30 mediate the neuroprotective effects of 17beta-estradiol in novel murine hippocampal cell models. Neuroscience 2010; 170: 54–66. [DOI] [PubMed] [Google Scholar]
- 126.Liu S HJ, Zhang N, Tian Z, Li X, Zhao M: Neuroprotective effects of oestrogen against oxidative toxicity through activation of G-protein-coupled receptor 30 receptor. Clinical and Experimental Pharmacology and Physiology 2011; 38: 577–585. [DOI] [PubMed] [Google Scholar]
- 127.Hammond R GR: GPR30 is positioned to mediate estrogen effects on basal forebrain cholinergic neurons and cognitive performance. Brain Research 2010; 1379: 53–60. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 128.Gu Q MR: Rapid action of 17 beta-estradiol on kainate-induced currents via activation of the cAMP cascade. Journal of Neuroscience 1996; 16: 3620–3629. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 129.Morely P WJ, Vanderhyden BC, Tsang BK, Schwartz JL: The rapid release of intracellular calcium. Endocrinology 1992; 131: 1305–1312. [DOI] [PubMed] [Google Scholar]
- 130.Revankar CM CD, Sklar LA, Arterburn JB, Prossnitz ER: A transmembrane intracellular estrogen receptor mediates rapid cell signaling. Science 2005; 3071625–1630. [DOI] [PubMed] [Google Scholar]
- 131.Toran-Allerand CD, Guan X, MacLusky NJ, Horvath TL, Diano S, Singh M, Connolly ES Jr., Nethrapalli IS, and Tinnikov AA: ER-X: a novel, plasma membrane-associated, putative estrogen receptor that is regulated during development and after ischemic brain injury. J Neurosci 2002; 22: 8391–8401. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 132.Toran-Allerand CD, Tinnikov AA, Singh RJ, and Nethrapalli IS: 17alpha-estradiol: a brain-active estrogen? Endocrinology 2005; 146: 3843–3850. [DOI] [PubMed] [Google Scholar]
- 133.Perez E, Liu R, Yang SH, Cai ZY, Covey DF, Simpkins JW: Neuroprotective effects of an estratriene analog are estrogen receptor independent in vitro and in vivo. Brain Res 2005; 1038: 216–222. [DOI] [PubMed] [Google Scholar]
- 134.Perez E, Cai Z Y, Covey D F, Simpkins J W: Neuroprotective effects of estratriene analogs: structure–activity relationships and molecular optimization. Drug Dev. Res 2006; 66: 78–92. [Google Scholar]
- 135.Green PS, Bishop J, Simpkins JW: 17 alpha-estradiol exerts neuroprotective effects on SK-N-SH cells. J. Neurosci 1997a; 17: 511–515. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 136.Richardson TE, Yang SH, Wen Y, Simpkins JW: Estrogen protection in Friedreich’s ataxia skin fibroblasts. Endocrinology 2011; 152: 2742–2749. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 137.Behl C, Skutella T, Lezoualc’h F, Post A, Widmann M, Newton CJ, Holsboer F: Neuroprotection againstoxidative stress by estrogens: structure–activity relationship. Mol. Pharmacol 1997; 51: 535–541. [PubMed] [Google Scholar]
- 138.Moosmann B, Behl C: The antioxidant neuroprotective effects of estrogens and phenolic compounds are independent from their estrogenic properties. Proc. Natl Acad. Sci. USA 1999; 96: 8867–8872. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 139.Nishio MY and Watanabe Y: Subcellular localization of estrogen receptor beta in mouse hippocampus. Neuroscience Letters 2004; 355:109–112. [DOI] [PubMed] [Google Scholar]
- 140.Milner TA, McEwen BS, Hayashi S, Li CJ, Reagan LP, Alves SE. : Ultrastructural evidence that hippocampal alpha estrogen receptors are located at extranuclear sites. Journal of Computational Neuroscience 2001; 429:355–371. [PubMed] [Google Scholar]
- 141.Marin R, Díaz M, Alonso R, Sanz A, Arévalo MA, Garcia-Segura LM.: Role of estrogen receptor alpha in membrane-initiated signaling in neural cells: interaction with IFG-1 receptor. Journal of Steroid Biochemistry 2009; 114: 2–7. [DOI] [PubMed] [Google Scholar]
- 142.Kelly MJ, Rønnekleiv OK: Control of CNS neuronal excitability by estrogens via membrane-initiated signaling. Molecular and Cellular Endocrinology 2009; 308: 17–25. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 143.Gorosito SV, Lorenzo AG, Cambiasso MJ: Estrogen receptor alpha is expressed on the cell-surface of embryonic hypothalamic neurons. Neuroscience 2008; 154: 1173–1177. [DOI] [PubMed] [Google Scholar]
- 144.Azcoitia I, Sierra A, Garcia-Segura LM.: Localization of estrogen receptor beta-immunoreactivity in astrocytes of the adult rat brain. Glia 1999; 26: 260–267. [PubMed] [Google Scholar]
- 145.Bondar G, Kuo J, Hamid N, Micevych P: Estradiol-induced estrogen receptor trafficking. J. Neuroscience 2009; 29:15323–30. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 146.Kuo J, Hamid N, Bondar G, Prossnitz ER, Micevych P: Membrane estrogen receptors stimuate intracellular calcium release and progesterone synthesis in hypothalamic astrocytes. J. Neuroscience 2010; 30:12950–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 147.Dominguez R and Micevych P: Estradiol rapidly regulates membrane estrogen receptor alpha levels in hypothalamic neurons. J. Neuroscience 2010; 30:12589–96. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 148.Bao YJ LL, Li XG, Wang YJ: 17 Beta-estradiol differentially protects cortical pericontusional zone from programmed cell death after traumatic cerebral contusion at distinct stages via non-genomic and genomic pathways. Molecular and Cellular Neuroscience 2011; 48: 185–194. [DOI] [PubMed] [Google Scholar]
- 149.Singh M, Sétáló G Jr, Guan X, Warren M, Toran-Allerand CD: Estrogen-Induced activation of mitogen-activated protein kinase in cerebral cortical explants: convergence of estrogen and neurotrophin signaling pathways. Journal of Neuroscience 1999; 191179–188. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 150.Singh M: Ovarian hormones elicit phosphorylation of Akt and extracellular-signal regulated kinase in explants of the cerebral cortex. Endocrine 2001; 14: 407–415. [DOI] [PubMed] [Google Scholar]
- 151.Singer CA, Figueroa-Masot XA, Batchelor RH, Dorsa DM: The mitogen activated protein kinase pathway mediates estrogen neuroprotection after glutamate toxicity in primary cortical neurons. Journal of Neuroscience 1999; 19: 2455–2463. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 152.Jover-Mengual T, Zukin RS, Etgen AM: MAPK signaling is critical to estradiol protection of CA1 neurons in global ischemia. Endocrinology 2007; 148: 1131–1143. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 153.Srivastava DP, Waters EM, Mermelstein PG, Kramár EA, Shors TJ, Liu F: Rapid Estrogen Signaling in the Brain: Implications for the Fine-Tuning of Neuronal Circuitry. Journal of Neuroscience 2011; 31: 4097–5011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 154.Grove-Strawser D, Boulware MI, Mermelstein PG: Membrane estrogen receptors activate the metabotropic glutamate receptors mGluR5 and mGluR3 to bidirectionally regulate CREB phosphorylation in female rat striatal neurons. Neuroscience 2010; 170: 1045–1055. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 155.Choi YC, Lee JH, Hong KW, Lee KS: 17 Beta-estradiol prevents focal cerebral ischemic damages via activation of Akt and CREB in association with reduced PTEN phosphorylation in rats. Fundamentals of Clinical Pharmacology 2004; 18: 547–557. [DOI] [PubMed] [Google Scholar]
- 156.Yang LC, Zhang QG, Zhou CF, Yang F, Zhang YD, Wang RM, Brann DW. Extranuclear estrogen receptors mediate the neuroprotective effects of estrogen in the rat hippocampus. PLoS One 2010. 5:e9851. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 157.Jover-Mengual T, Miyawaki T, Latuszek A, Alborch E, Zukin RS, Etgen AM: Acute estradiol protects CA1 neurons from ischemia-induced apoptotic cell death via the PI3K/Akt pathway. Brain Res 2010; 1321:1–12. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 158.Akama KT, McEwen BS: Estrogen stimulates postsynaptic density-95 rapid protein synthesis via the Akt/protein kinase B pathway. Journal of Neuroscience 2003; 23: 2333–2339. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 159.Kramár EA, Chen LY, Brandon NJ, Rex CS, Liu F, Gall CM, Lynch G: Cytoskeletal changes underlie estrogen’s acute effects on synaptic transmissions and plasticity. Journal of Neuroscience 2009; 29: 12982–12993. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 160.Zhang QG, Wang R, Khan M, Mahesh V, Brann DW. Role of Dickkopf-1, an antagonist of the Wnt/beta-catenin signaling pathway, in estrogen-induced neuroprotection and attenuation of tau phosphorylation. J. Neurosci 2008; 28:8430–41. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 161.Raz L, Zhang QG, Han D, Dong Y, De Sevilla L, Brann DW. Acetylation of the pro-apoptotic factor, p53 in the hippocampus following cerebral ischemia and modulation by estrogen. PLoS One 2011; 6:e27039. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 162.Mirshams N and Singh M: The role of the estrogen receptor in the protective effects of DHT Neuroscience Meeting Planner. Program No. 595.14, Washington D.C., Society for Neurosience, 2011. Online. [Google Scholar]
- 163.Picciarelli-Lima P, Oliveira AG, Reis A, Kalapothakis E, Mahecha G, Rex A, Hess E and Oliveira C: Effects of 3-beta-diol, an androgen metabolite with intrinsic estrogen-like effects, in modulating the aquaporin-9 expression in the rat efferent ductules. Reproductive Biology and Endocrinology 2006; 4:51–61. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 164.Wigginton JG, Pepe PE, Idris AH: Rationale for routine and immediate administration of intravenous estrogen for all critically ill and injured patients. Crit Care Med 2010; 38: S620–S629. [DOI] [PubMed] [Google Scholar]