Skip to main content

REVIEW article

Front. Endocrinol., 25 March 2015
Sec. Neuroendocrine Science
This article is part of the Research Topic Trends in Comparative Endocrinology and Neurobiology View all 23 articles

Hypothalamic–Pituitary–Gonadal Axis Involvement in Learning and Memory and Alzheimer’s Disease: More than “Just” Estrogen

\r\n      Jeffrey A. BlairJeffrey A. Blair1Henry McGeeHenry McGee1Sabina BhattaSabina Bhatta1Russell PalmRussell Palm2Gemma Casadesus*Gemma Casadesus1*
  • 1Department of Biological Sciences, Kent State University, Kent, OH, USA
  • 2University of Toledo School of Medicine, Toledo, OH, USA

Accumulating studies affirm the effects of age-related endocrine dysfunction on cognitive decline and increasing risk of neurodegenerative diseases. It is well known that estrogens can be protective for cognitive function, and more recently androgens and luteinizing hormone have also been shown to modulate learning and memory. Understanding the mechanisms underlying hypothalamic–pituitary–gonadal axis-associated cognitive dysfunction is crucial for therapeutic advancement. Here, we emphasize that reproductive hormones are influential in maintaining neuronal health and enhancing signaling cascades that lead to cognitive impairment. We summarize and critically evaluate age-related changes in the endocrine system, their implications in the development of Alzheimer’s disease, and the therapeutic potential of endocrine modulation in the prevention of age-related cognitive decline.

Introduction

As the baby boomer generation continues to age and advances in medicine have resulted in longer life expectancy, it is vital to consider the eminent risk of age-associated neurodegenerative diseases, such as Alzheimer’s disease (AD) and their effects on our society. The number of people diagnosed with AD is estimated to be over 13 million by 2050 (1). Furthermore, expense estimates for AD and dementia patient care in the United States alone are over $180 billion per year, which makes dementia one of the most costly diseases (2). Not only will more individuals continue to be diagnosed and threatened with neurodegenerative disease, but their caregivers will also suffer more physical, mental, and economic burdens.

Senile plaques containing amyloid-β, neurofibrillary tangles comprised of aggregated tau and cell death in the hippocampal formation are the hallmark pathologies of AD (3). Clinically, AD is characterized by prominent deficits in memory and attention leading to deterioration of judgment, language skills, and spatial orientation as the disease progresses (4). While AD pathogenesis has been linked to oxidative stress, inflammation, and neuronal dysfunction, concrete evidence explaining disease pathogenesis is absent, compelling us and others to search beyond the hallmark pathologies for alternative molecular cues underlying AD pathogenesis (5).

Hormones are known to impact central nervous system (CNS) function and stability (6), and gonadal hormones have been extensively studied for their effect on cognition in elderly men and women (7). We believe that age-associated endocrine system dysfunction is a major factor in the onset and progression of neurodegenerative diseases. This is supported by research showing changes in gender and age-dependent reproductive hormones increases the risk of AD, therefore implicating the hypothalamic–pituitary–gonadal (HPG) axis (814).

Hypothalamic–Pituitary–Gonadal Axis

The HPG axis is controlled by a negative feedback loop. In the healthy brain, the hypothalamus releases gonadotropin-releasing hormone (GnRH) into the median eminence, and then GnRH is transported via the hypophyseal portal system to the anterior pituitary where it acts on its receptor (GnRHR). Signaling from GnRHR leads to the production and secretion of the gonadotropins, including luteinizing hormone (LH) and follicle-stimulating hormone (FSH). Once the gonadotropins are secreted into the blood stream, LH acts on its receptor in the gonads, which in turn stimulates the release of the sex steroids, androgens and estrogens. These sex steroids complete a negative feedback loop by inhibiting the release of GnRH.

Peripheral hormone levels have long been implicated in changes of behavior. Studies of the modulatory effects of hormones on cognition have led to the discovery that hormone receptors are expressed in the CNS (15, 16). Importantly, many of these hormone receptors are present in areas of the brain associated with learning and memory, such as the hippocampus. Crucial to HPG axis dysfunction and its connection to AD is that the rate of hormone synthesis varies throughout one’s life. With age, androgen production decreases gradually in men, while there is an abrupt diminution in gonadal secretion of estrogens in women. Besides, the dysregulation of the HPG axis caused by menopause may be due to a diminished ability of estrogens to inhibit the hypothalamus (17, 18). Interestingly, in addition to the negative feedback estrogens have on the HPG axis, estrogens can also produce positive feedback, which is necessary for fertility (19) and is attenuated with age (20). This body of work strongly suggests that cognitive function may rely on the levels of sex steroids and gonadotropins, implicating HPG axis dysfunction as a factor in the strong correlation between aging and dementia.

Estrogens

Gender has been associated as an independent risk factor for AD. Females have a higher risk of developing AD as shown by age-adjusted odds ratios regardless whether familial or sporadic AD was acquired (21, 22). The higher prevalence and incidence of AD in women has been linked to the changes in reproductive hormones that occur during menopause. Therefore, women have become a main focus of many aging studies including the Women’s Health Initiative Memory Study (9, 10). Endocrinological evaluations show lower levels of estrogens in women with AD (12, 23), thus encouraging hormone replacement therapy (HRT) to improve cognition and decrease the risk of AD in post-menopausal women (2427). This theory is based on an abundance of evidence displaying the neuroprotective effects of estrogens observed in healthy cells. Estrogens signal through a multitude of mechanisms to induce axonal sprouting, regeneration, synaptic transmission, and the prevention of cell death (28). Estrogen receptors (ER) can activate production of brain-derived neurotrophic factor (BDNF), which has been shown to protect against ischemic injury in vitro and retain cognitive function as measured with passive avoidance in mice in vivo (29). Additionally, other in vitro studies have shown protective effects of estrogens from excitotoxicity by increasing the apoptosis regulator Bcl-2 (30). Moreover, estrogens have been shown to protect against oxidative stress induced by amyloid-β fibrils alone or in a complex with acetylcholinesterase, making estrogens a target for AD therapeutics (31). This hypothesis is influential in that the only current AD treatment is cholinesterase inhibitors, with which meta-analyses show a modest benefit (32).

Estrogen signaling has long been known to occur through nuclear receptor activity, but can also arise through membrane-associated ERs. The classic signaling pathway relies on nuclear ERα or ERβ binding to estrogen response elements (ERE) in order to instigate transcription, therefore controlling gene expression. These long-term effects on gene expression are implicated in development as well as changes later in life. However, non-ERE-dependent signaling has been shown to rapidly produce effects of estrogens on neural processes. Membrane-associated ERs can activate PI3K/PLC, MAPK/ERK, and cAMP/PKA signaling pathways, which have been associated with neuroprotection (16). Importantly for the HPG axis, ERα mediates the negative feedback estrogens have on the hypothalamus without ERE activation to inhibit LH secretion through p21-activated kinase (33).

The neuroprotection evoked by estrogens was the basis of clinical trials looking for benefits on cognition. A randomized double-blind study found that 17β-estradiol (E2) treatment and a consequent increase in serum estrogens provided no significant improvement in cognition (34). Furthermore, the Women’s Health Initiative funded a study of 16,000 women that indicated HRT, with E2 and progestin, in fact increased risk of dementia. However, after controlling for treatment onset, it was found that initiation of HRT 10 or more years after menopause increases the risk of AD, while initiation of HRT during menopause lowers risk of AD (9, 10). Supplemental research showed the treatment group in the Women’s Health Initiative Memory Study had a higher incidence of breast cancer (35). This off target effect was pronounced with a combined E2 and progestin treatment, but is a major concern in HRT despite cognitive benefits observed. Overall, the clinical trials suggest that a critical period between menopause and HRT onset exists (36).

The results of E2 treatment have led to the proposal of the critical period hypothesis, which states that HRT is beneficial immediately after menopause, but benefits are lost the longer post-menopause is endured (36). The critical period hypothesis has also been demonstrated in ovariectomized rodents. The immediate post-operational treatment with E2 produces the well-characterized cognitive improvements, but a latency period of several months before E2 treatment ablates all benefits (37, 38). Along with the critical period hypothesis is the idea of the healthy cell bias. Estrogens may only be beneficial when administered to healthy neurons, and in order to potentially protect from neurological damage estrogens would need to be preemptively administered (39, 40). Conversely, once an unhealthy neurological state prevails estrogens signaling produces deleterious effects. Current hypotheses on the negative effects of estrogens in unhealthy neurological conditions include aberrant calcium signaling and mitochondrial dysfunction (41).

The reduced efficacy of estrogens downregulation of gonadotropins is evident in rodents after ovariectomy (42) and in post-menopausal women (43). Therefore, an alternate hypothesis that explains the lack of efficacy of HRT in older post-menopausal women is the inability of HRT to provide efficient negative feedback on gonadotropins. In this regard, LH is now known to influence cognition as well (4448). Therefore, methodologically separating the roles of estrogens and LH on cognition is imperative to fully understand the impact of these hormones on cognitive decline and AD (37).

Luteinizing Hormone

In conjunction with estrogens function in cognition, the loss of sex steroids leads to large increases in peripheral levels of LH. In aging women, a threefold increase in LH and a fourfold increase in FSH levels occur (49) while in aging men both hormones increase two to threefold (50). Until recently, the impact of changing levels of gonadotropins during reproductive aging was virtually ignored given the primarily peripheral role of gonadotropins on reproduction. However, mounting studies in humans, rodents, and in vitro demonstrate that LH and activation of its receptor may have an important role on cognitive function and neuronal plasticity.

In humans, increases in peripheral LH levels are correlated with decreased cognition in both healthy women (51) and men (52), and levels of LH and FSH have been shown to be significantly increased in AD patients compared to control (8, 13) in some but not all studies. For example, serum LH and FSH levels have been shown to remain consistent in female (12) and male (53) subjects regardless of dementia status, but by increasing the sample size, Hogervorst et al. (11) observed a trend toward high serum LH correlating with AD status in males. In a recent study, LH levels were correlated with amyloid-β levels, further implicating LH in development and progression of AD (14).

Human chorionic gonadotropin (hCG) and LH share a receptor (LHCGR) crucial for reproductive functions, such as testosterone production by Leydig cells and follicular maturation. A large body of literature describes the physiological and molecular role of LHCGR in reproduction (54). The isolation and purification of LHCGR revealed that it is a member of the G-protein-coupled receptor (GPCR) family (55, 56), and part of rhodopsin-like class A GPCRs expressed as several splice variants, all of which have leucine-rich repeats in the extracellular domain (54). Upon phosphorylation of LHCGR, Gαs protein activates adenylyl cyclase and ultimately stimulates the cAMP/PKA and ERK pathways. Although LHCGR primarily signals through the Gs/cAMP/PKA pathway, it can independently mediate the activation of phospholipase C [PLC; (15, 57)].

Accumulating research shows LHCGR is atypically expressed in the CNS (5860). LHCGR transcripts of 2.5 and 4.3 kb have been shown in fetal rat brain neurons and glial cells (61, 62). An 80 kDa band as seen by western immunoblot is evidence of protein expression in cultured rat neurons. Furthermore, time course studies show that day in vitro (DIV) 3 neurons have the highest levels of expression of LHCGR (61). A similar 80 kDa band is seen in immunoblots from glial cells which increase with an increase glial proliferation (62). Similarly, in situ hybridization has shown an abundance of LHCGR in the rat brain including the hippocampal formation, hypothalamus, cerebellum, choroid plexus, and ependymal cells of the ventricles and cortex (63). LHCGR expression has also been shown in the CNS of Xenopus laevis, where it plays a functional role in the reproductive behavior of courtship songs, therefore implicating a non-gonadal function of LHCGR in the CNS (64). Although many laboratories have researched LHCGR mechanisms in the gonadal organs, extra-gonadal receptor processes in rodents are just now beginning to be elucidated.

For example, early work shows that direct activation of the LHCGR with hCG drives cellular and functional changes. To this end, rat neurons cultured in the presence of hCG appeared to have increased numbers of neurite-bearing cells (61). The same treatment paradigm for glial cells showed an increase in prostaglandin D2 and a decrease in prostaglandin E2 (62). Importantly, prostaglandin D2 is known to curb the proliferation of glial cells; therefore, LHCGR activation in the CNS may be an important modulator of glial cell populations (62). Cell viability was increased with hCG treatment as observed through increased total protein content and decreased DNA fragmentation (61).

At a functional level, open-field behavior on rats injected with hCG either intraperitoneally (IP) or intracerebroventricularly showed a decrease in locomotor activity, but no increase in anxiety (65). Taste neophobia was overcome by intraperitoneal injections and intracerebroventricular injections of hCG, and hCG treatment did not evoke differences on T-maze behavior for the goal box, but a decrease was observed in the stereotypic behavior (65). This study shows that at a high dose hCG affects overall activity, which will supersede any findings related to learning and memory or fear. It has been proposed that at these high doses, hCG may be responsible for changes in sleep that occur during pregnancy since hCG is present in cerebrospinal fluid during the first trimester and correlates with serum levels during the first and third trimester (6567). IP injection of hCG in rats showed increases in both high and low amplitude sleep and a decrease in active awake phase via electroencephalography [EEG; (68)]. Similarly, peripheral hCG administration has been shown to decrease walking and increase resting in rats (68). It is important to note that during sleep LH pulses decrease in frequency but increase in amplitude (68).

Furthermore, studies using therapies aimed at downregulating peripheral LH show significant improvements in cognition and AD pathogenesis (44, 45). For example, in AD mice, downregulation of serum LH improves function in Y-maze and Morris water maze cognitive tasks and reduces amyloid-β immunoreactivity in the Tg2576 AD mouse (44). Animal studies utilizing GnRHR antagonists, antide and Cetrorelix, which also lowers serum levels of LH, also show cognitive improvements in spatial memory tasks in rats (46) and in non-transgenic models of AD (47, 69).

At a signaling level, studies show that downregulation of LH leads to activation of memory-associated cascades. CaMKII auto-phosphorylation, which is associated with hippocampal long-term potentiation (LTP), is downregulated after ovariectomy, but rescued with leuprolide acetate treatment (70). Furthermore, downstream targets of CaMKII, such as the phosphorylation of GluR1 subunit of AMPA are also activated by leuprolide acetate treatment (70). These changes in LTP-related cascades are one potential mechanism underlying the behavioral improvements observed with leuprolide acetate treatments in vivo (44, 48, 70). Interestingly, leuprolide acetate can also affect the synthesis of E2 from testosterone by modulating the transcription of p450 aromatase (70) suggesting functional benefits may be secondary to up-regulation of endogenous estrogens and downstream modulation of CREB and GSK3β (48, 71, 72). Taken together, how LHCGR activation influences these cascades and leads to functional changes is a critical area of future study.

Interestingly, LH immunoreactivity has been demonstrated by radioimmunoassay as well as immunocytochemistry in the hypothalamus, amygdala, septal area, preoptic area, thalamus caudate nucleus, and hippocampus (73). Extracts from rat brain have shown that brain-derived LH has a similar chromatographic profile of pituitary LH and is active in the testis LH radio-ligand receptor assay as well as the interstitial cell testosterone secretion bioassay (74). Additionally, an adsorption significantly reduced rat brain LH immunoreactivity (74). Therefore, LH is both present and biologically active in the CNS. In support of these earlier studies, we have shown LH immunoreactivity in cognition-related brain regions in the 3xTg AD mouse model. Importantly, levels of brain-derived LH are decreased by ovariectomy, a state of high peripheral LH, and positively correlated with improvements in the Morris water maze (48). This suggests an inverse correlation between serum (peripheral) LH levels and CNS LH levels. Importantly, this inverse relationship between peripheral and brain-derived LH may explain the beneficial effects of hCG seen by Al-Hader et al. (61, 62) in vitro as well as the functional benefits and activation of LHCGR-dependent cascades by leuprolide acetate (48, 70).

Androgens

Hypogonadism, a common feature of aging, may significantly contribute to both physical and cognitive decline in men, including the development of AD (75, 76). It has been shown that men with AD have lower levels of testosterone compared to non-AD patients (53). However, results have been mixed concerning testosterone replacement in healthy men. Testosterone replacement in non-demented patients with healthy gonadal function increased spatial cognition, verbal memory, and working memory (7779). In contrast, testosterone replacement studies focusing on upper extremity strength or self-perceived health did not observe changes in cognition after 1 year of treatment (80, 81). Nevertheless, patients with mild cognitive impairment or AD have benefited from testosterone replacement. Spatial memory, constructional abilities, and verbal memory have all been shown to improve in cognitively impaired patients with short- (82) and long-term (83, 84) testosterone treatment. Meanwhile, one study suggests no difference between placebo and testosterone treatment (85), but the behavioral measures used may not have been sensitive to testosterone treatment (86), and, importantly, the testosterone injections may have been too interspersed to improve cognition. Trends in the literature suggest positive correlations between testosterone treatment and cognitive improvement warranting further investigation as a viable option for treating AD.

Essential to direct effects of testosterone on cognition, the androgen receptor (AR) is expressed in neurons of the hippocampus and amygdala (87, 88). Primary neuron cultures treated with testosterone show decreases in secretion of amyloid protein (89). In vivo studies show increased testosterone corresponds to decreases in β-secretase, an enzyme responsible for the cleavage of the amyloid-β precursor protein, and improves cognitive function in male double-transgenic mice (90). Effects of testosterone on CA1 spine synapses are independent from the AR since similar effects are seen in normal rats and AR mutants (91). In rats, rapid acting cytoplasmic androgen signaling in axons of the dorsal CA1 hippocampal neurons innervating the amygdala may be the most likely route for learning, especially in relation to environmental cues (88).

The AR binds testosterone or dihydrotestosterone (DHT) and translocates from the cytoplasm to the nucleus. Ultimately, AR signaling modifies gene expression, but like ERs, the ARs also have effects on intracellular signaling such as DHT’s induction of CREB activation (92, 93). Extranuclear ARs are localized exclusively in asymmetric synapses (94) while ERs are found in both asymmetric and symmetric synapses (95). Redundancy may exist given the location of the ERs and ARs, but a unique function may exist for ER in symmetric synapses, which are typically inhibitory. Interestingly, for the action of ARs and ERs, testosterone can be converted into E2, which requires the experimental delineation of androgen and aromatized androgen metabolite action (96).

The conversion of testosterone into E2 by p450 aromatase (97) affords testosterone the ability to activate both ARs and ERs within the brain. Testosterone conversion can be controlled by aromatase inhibitors, therefore allowing experimental separation of testosterone pathways and the pathways of its aromatized metabolites (96). In a study of 60 healthy men aged 50–90, men treated with testosterone alone, and which consequently increases E2 levels from aromatase activity, displayed improvements in verbal memory. In contrast, when an aromatase inhibitor was administered along with testosterone, the improvement was no longer observed, suggesting that aromatization of testosterone is necessary for improvements in verbal memory (77). The individual actions of the sex steroids have separate roles in cognition; however, it is necessary to take neurological health into consideration during treatment. Testosterone metabolites improve cognition in healthy individuals, but as is evident from studies of estrogens, the neurological health of the patient may be a confounding factor.

Overall, a decrease in testosterone levels is associated with a higher risk of AD (11, 13, 14), and improvements in cognition have been observed with testosterone treatment (8284). Testosterone signaling can occur directly through AR action or through the aromatization products acting on ERs but the bioactivity of the sex hormones may be a confounding factor (96). Increased levels of sex hormone-binding globulin (SHBG), which binds to sex steroids and reduces their bioactivity, have been linked to an increased risk of dementia in both men and women (98).

Sex Hormone-Binding Globulin

Confounding the age-dependent attenuation of sex steroid signaling in the HPG axis is the ability of SHBG to reduce the bioactivity of sex steroids through binding and inhibiting action on their respective receptors. AD patients have increased levels of SHBG and, therefore, lower serum levels of bioactive sex steroids. The ensuing inverse correlation between cognition and SHBG is observed in both male and female AD patients (11, 98101). SHBG levels and its inverse correlation with cognitive decline are important to consider for determining the efficacy of E2 and testosterone treatment. Importantly, SHBG levels may confound previous studies by deactivating the sex steroid treatments. Although further research is necessary, therapeutic control of SHBG would allow endogenous estrogens and androgens to remain bioactive and potentially reduce the risk of cognitive decline.

Conclusion

A myriad of research on the role of hormones in cognition has been undertaken and has led to many outlets for therapeutic potential to combat the cognitive loss associated with aging and AD. Epidemiological studies of the aging population show not only a gender difference in risk for AD, but also that HRT can have cognitive benefits. Prospective studies in humans and animals show that E2 treatment provides benefits to cognition when treatment onset is within the critical period, temporally close to menopause or hormone dysfunction. On a mechanistic level, it is well known that the levels of estrogens positively correlate with dendritic spine density in rodents as well as axonal sprouting, lending evidence for E2 treatment benefits being mediated through increasing plasticity. However, the diminished and deleterious effects of E2 treatment implemented outside of the critical period and the implication of this hormone in the development of breast cancer begets the search for hormone-based therapeutic strategies that go beyond E2 treatment. In this regard, one potential area of research is testosterone replacement. Work presented in this review demonstrates the beneficial effects of this hormone on learning and memory. However, effects of testosterone on cognitive function are primarily related to the aromatization of testosterone into E2. As such, while testosterone may provide advantages through its ability to signal through AR and E2 receptors after its aromatization, this also poses issues at the level of dosing and off target effects. Taken together, while the actions of sex steroids have adverse effects, the improvements observed highlight the feasibility of HRT as a pharmaceutical treatment for cognitive decline throughout aging. However, aspects such as timing of replacement and clinical follow-up to mitigate important side effects must be carefully watched. Also important is the fact that secondary players, such as SHBG and p450 aromatase, that regulate bioavailability or production of steroids are key in determining the magnitude of effect and specificity of steroid hormone treatment. Whether these secondary players have a direct impact on cognition and neuroplasticity is unclear, and their determination is particularly important when interpreting the results of clinical trials using sex steroid replacement.

Lastly, we provide evidence that LH, a relatively unknown player in cognitive function and neuronal plasticity, is gaining support as a therapeutic target for age-related cognitive decline. Support for a role of LH in cognitive processes is highlighted not only by clinical data demonstrating high levels of peripheral LH are associated with cognitive deficits and AD, but also in pre-clinical studies. To this end, several investigators have now shown that lowering peripheral LH rescues cognitive decline in various aging and AD rodent models, and these treatments are associated with signaling important to synaptic plasticity. Importantly, a recent clinical trial in AD female patients shows benefits of downregulating peripheral LH (102).

Here, we propose the hypothesis that brain levels of LH become downregulated by increases in peripheral LH observed during ovarian function loss and cause cognitive dysfunction; this aspect, we hypothesize to be mediated through loss of LHR signaling in the brain. While the mechanisms in charge of downregulating brain LH production are unknown, support for this hypothesis is emerging based on data showing LHR expression and functionality in the brain as well as receptor actions associated with cognition-related signaling and neuroplasticity. Importantly, in our recent study, we observed a positive correlation between LH levels in the brain and learning and memory in the Morris water maze task. Studies directed at elucidating the specific role of the LHR in the brain are likely to shed light on the role of LH in cognition and neuroplasticity.

While in the last 30 years primary focus has been placed on studying the role of gonadal steroids on neuronal plasticity and function, we show here that aspects thought to be secondary or irrelevant, such as LH signaling, are important to fully evaluate the effects or lack thereof of gonadal steroids, and may potentially have direct roles and drive processes previously assigned to loss of steroid function. As such, a comprehensive study of HPG axis hormones is necessary in clinical and pre-clinical work and likely to be more effective in providing novel therapeutic targets and strategies for cognitive dysfunction and AD.

Conflict of Interest Statement

The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

Acknowledgments

This work was supported by grants from the National Institute on Aging (R01 AG032325).

References

1. Hebert LE, Scherr PA, Bienias JL, Bennett DA, Evans DA. Alzheimer disease in the US population: prevalence estimates using the 2000 census. Arch Neurol (2003) 60(8):1119–22. doi: 10.1001/archneur.60.8.1119

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text | Google Scholar

2. Stefanacci RG. The costs of Alzheimer’s disease and the value of effective therapies. Am J Manag Care (2011) 17(S13):S356–62.

Pubmed Abstract | Pubmed Full Text | Google Scholar

3. Hampel H, Lista S, Khachaturian ZS. Development of biomarkers to chart all Alzheimer’s disease stages: the royal road to cutting the therapeutic Gordian knot. Alzheimers Dement (2012) 8(4):312–36. doi:10.1016/j.jalz.2012.05.2116

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text | Google Scholar

4. Cummings JL, Vinters HV, Cole GM, Khachaturian ZS. Alzheimer’s disease etiologies, pathophysiology, cognitive reserve and treatment opportunities. Neurology (1998) 51(S1):S2–17. doi:10.1212/WNL.51.1_Suppl_1.S2

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text | Google Scholar

5. Nunomura A, Perry G, Aliev G, Hirai K, Takeda A, Balraj EK, et al. Oxidative damage is the earliest event in Alzheimer disease. J Neuropathol Exp Neurol (2001) 60(8):759–67.

Pubmed Abstract | Pubmed Full Text | Google Scholar

6. Marshall KM. Introduction to the interaction between gonadal steroids and the central nervous system. Curr Top Behav Neurosci (2011) 8:1–13. doi:10.1007/7854_2011_136

CrossRef Full Text | Google Scholar

7. Hogervorst E. Effects of gonadal hormones on cognitive behaviour in elderly men and women. J Neuroendocrinol (2013) 25(11):1182–95. doi:10.1111/jne.12080

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text | Google Scholar

8. Short RA, O’Brien PC, Graff-Radford NR, Bowen RL. Elevated gonadotropin levels in patients with Alzheimer disease. Mayo Clin Proc (2001) 76(9):906–9. doi:10.1016/S0025-6196(11)62109-5

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text | Google Scholar

9. Zandi PP, Carlson MC, Plassman BL, Welsh-Bohmer KA, Mayer LS, Steffens DC, et al. Hormone replacement therapy and incidence of Alzheimer disease in older women. JAMA (2002) 288(17):2123–9. doi:10.1001/jama.288.17.2123

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text | Google Scholar

10. Rapp SR, Espeland MA, Shumaker SA, Henderson VW, Brunner RL, Manson JE, et al. Effect of estrogen plus progestin on global cognitive function in postmenopausal women. JAMA (2003) 289(20):2663–72. doi:10.1001/jama.289.20.2663

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text | Google Scholar

11. Hogervorst E, Bandelow S, Combrinck M, Smith AD. Low free testosterone is an independent risk factor for Alzheimer’s disease. Exp Gerontol (2004) 39(11):1633–9. doi:10.1016/j.exger.2004.06.019

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text | Google Scholar

12. Tsolaki M, Grammaticos P, Karanasou C, Balaris V, Kapoukranidou D, Kalpidis I, et al. Serum estradiol, progesterone, testosterone, FSH and LH levels in postmenopausal women with Alzheimer’s dementia. Hell J Nucl Med (2005) 8(1):39–42.

Pubmed Abstract | Pubmed Full Text | Google Scholar

13. Butchart J, Birch B, Bassily R, Wolfe L, Holmes C. Male sex hormones and systemic inflammation in Alzheimer disease. Alzheimer Dis Assoc Disord (2012) 27(2):153–6. doi:10.1097/WAD.0b013e318258cd63

CrossRef Full Text | Google Scholar

14. Verdile G, Laws SM, Henley D, Ames D, Bush AI, Ellis KA, et al. Associations between gonadotropins, testosterone and β amyloid in men at risk of Alzheimer’s disease. Mol Psychiatry (2014) 19:1–7. doi:10.1038/mp.2012.147

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text | Google Scholar

15. Ascoli M, Fanelli F, Segaloff DL. The lutropin/choriogonadotropin receptor, a 2002 perspective. Endocr Rev (2002) 23(2):141–74. doi:10.1210/edrv.23.2.0462

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text | Google Scholar

16. Roepke TA, Ronnekleiv OK, Kelly MJ. Physiological consequences of membrane-initiated estrogen signaling in the brain. Front Biosci (Landmark Ed) (2011) 16:1560–73. doi:10.2741/3805

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text | Google Scholar

17. Lloyd JM, Hoffman GE, Wise PM. Decline in immediate early gene expression in gonadotropin-releasing hormone neurons during proestrus in regularly cycling, middle-aged rats. Endocrinology (1994) 134(4):1800–5. doi:10.1210/en.134.4.1800

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text | Google Scholar

18. Wise PM, Smith MJ, Dubal DB, Wilson ME, Rau SW, Cashion AB, et al. Neuroendocrine modulation and repercussions of female reproductive aging. Recent Prog Horm Res (2002) 57(1):235–56. doi:10.1210/rp.57.1.235

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text | Google Scholar

19. Wintermantel TM, Campbell RE, Porteous R, Bock D, Gröne HJ, Todman MG, et al. Definition of estrogen receptor pathway critical for estrogen positive feedback to gonadotropin-releasing hormone neurons and fertility. Neuron (2006) 52(2):271–80. doi:10.1016/j.neuron.2006.07.023

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text | Google Scholar

20. Dimitraki M, Koutlaki N, Gioka T, Messini CI, Dafopoulos K, Anifandis G, et al. Attenuation of the estrogen positive feedback mechanism with the age in postmenopausal women. Clin Endocrinol (2015). doi:10.1111/cen.12735

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text | Google Scholar

21. Payami H, Montee K, Grimslid H, Shattuc S, Kaye J. Increased risk of familial late-onset Alzheimer’s disease in women. Neurology (1996) 46(1):126–9. doi:10.1212/WNL.46.1.126

CrossRef Full Text | Google Scholar

22. Gao S, Hendrie HC, Hall KS, Hui S. The relationships between age, sex, and the incidence of dementia and Alzheimer disease: a meta-analysis. Arch Gen Psychiatry (1998) 55(9):809–15. doi:10.1001/archpsyc.55.9.809

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text | Google Scholar

23. Robusto-Leitao O, Ferreira H. Hormones and dementia – a comparative study of hormonal impairment in post-menopausal women, with and without dementia. Neuropsychiatr Dis Treat (2006) 2(2):199–206. doi:10.2147/nedt.2006.2.2.199

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text | Google Scholar

24. Phillips SM, Sherwin BB. Effects of estrogen on memory function in surgically menopausal women. Psychoneuroendocrinology (1992) 17(5):485–95. doi:10.1016/0306-4530(92)90007-T

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text | Google Scholar

25. Henderson VW, Paganini-Hill A, Emanuel CK, Dunn ME, Buckwalter JG. Estrogen replacement therapy in older women: comparisons between Alzheimer’s disease cases and nondemented control subjects. Arch Neurol (1994) 51(9):896–900. doi:10.1001/archneur.1994.00540210068014

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text | Google Scholar

26. Kawas C, Resnick S, Morrison A, Brookmeyer R, Corrada M, Zonderman A, et al. A prospective study of estrogen replacement therapy and the risk of developing Alzheimer’s disease: the Baltimore longitudinal study of aging. Neurology (1997) 48(6):1517–21. doi:10.1212/WNL.48.6.1517

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text | Google Scholar

27. Sherwin BB. Estrogen and cognitive functioning in women. Proc Soc Exp Biol Med (1998) 217(1):17–22. doi:10.3181/00379727-217-44200

CrossRef Full Text | Google Scholar

28. Garcia-Segura LM, Azcoitia I, DonCarlos LL. Neuroprotection by estradiol. Prog Neurobiol (2001) 63:29–60. doi:10.1016/S0301-0082(00)00025-3

CrossRef Full Text | Google Scholar

29. Sudo S, Wen T, Desaki J, Matsuda S, Tanaka J, Arai T, et al. Beta-estradiol protects hippocampal CA1 neurons against transient forebrain ischemia in gerbil. Neurosci Res (1997) 29:345–54. doi:10.1016/S0168-0102(97)00106-5

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text | Google Scholar

30. Singer CA, Rogers KL, Dorsa DM. Modulation of Bcl-2 expression: a potential component of estrogen protection in NT2 neurons. Neuroreport (1998) 9(11):2565–8. doi:10.1097/00001756-199808030-00025

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text | Google Scholar

31. Bonnefont AB, Muñoz FJ, Inestrosa NC. Estrogen protects neuronal cells from the cytotoxicity induced by acetylcholinesterase-amyloid complexes. FEBS Lett (1998) 441(2):220–4. doi:10.1016/S0014-5793(98)01552-X

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text | Google Scholar

32. Trinh NH, Hoblyn J, Mohanty S, Yaffe K. Efficacy of cholinesterase inhibitors in the treatment of neuropsychiatric symptoms and functional impairment in Alzheimer disease: a meta-analysis. JAMA (2003) 289(2):210–6. doi:10.1001/jama.289.2.210

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text | Google Scholar

33. Zhao Z, Park C, McDevitt MA, Glidewell-Kenney C, Chambon P, Weiss J, et al. p21-Activated kinase mediates rapid estradiol-negative feedback actions in the reproductive axis. Proc Natl Acad Sci U S A (2009) 106(17):7221–6. doi:10.1073/pnas.0812597106

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text | Google Scholar

34. Polo-Kantola P, Portin R, Polo O, Helenius H, Irjala K, Erkkola R. The effect of short-term estrogen replacement therapy on cognition: a randomized, double-blind, cross-over trial in postmenopausal women. Obstet Gynecol (1998) 91(3):459–66. doi:10.1016/S0029-7844(97)00700-X

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text | Google Scholar

35. Chlebowski RT, Anderson GL, Gass M, Lane DS, Aragaki AK, Kuller LH, et al. Estrogen plus progestin and breast cancer incidence and mortality in postmenopausal women. JAMA (2010) 304(15):1684–92. doi:10.1001/jama.2010.1500

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text | Google Scholar

36. Sherwin BB. Estrogen and cognitive functioning in women. Endocr Rev (2003) 24(2):133–51. doi:10.1210/er.2001-0016

CrossRef Full Text | Google Scholar

37. Daniel JM, Hulst JL, Berbling JL. Estradiol replacement enhances working memory in middle-aged rats when initiated immediately after ovariectomy but not after a long-term period of ovarian hormone deprivation. Endocrinology (2006) 147(1):607–14. doi:10.1210/en.2005-0998

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text | Google Scholar

38. Bohacek J, Daniel JM. The beneficial effects of estradiol on attentional processes are dependent on timing of treatment initiation following ovariectomy in middle-aged rats. Psychoneuroendocrinology (2010) 35(5):694–705. doi:10.1016/j.psyneuen.2009.10.010

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text | Google Scholar

39. Brinton RD. Investigative models for determining hormone therapy-induced outcomes in brain: evidence in support of a healthy cell bias of estrogen action. Ann N Y Acad Sci (2005) 1052(1):57–74. doi:10.1196/annals.1347.005

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text | Google Scholar

40. Sohrabji F. Estrogen: a neuroprotective or proinflammatory hormone? Emerging evidence from reproductive aging models. Ann N Y Acad Sci (2006) 1052(1):75–90. doi:10.1196/annals.1347.006

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text | Google Scholar

41. Brinton RD. The healthy cell bias of estrogen action: mitochondrial bioenergetics and neurological implications. Trends Neurosci (2008) 31(10):529. doi:10.1016/j.tins.2008.07.003

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text | Google Scholar

42. King JC, Anthony ELP, Damassa DA, Elkind-Hirsch KE. Morphological evidence that luteinizing hormone-releasing hormone neurons participate in the suppression by estradiol of pituitary luteinizing hormone secretion in ovariectomized rats. Neuroendocrinology (1987) 45(1):1–13. doi:10.1159/000124698

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text | Google Scholar

43. Rossmanith WG, Reichelt C, Scherbaum WA. Neuroendocrinology of aging in humans: attenuated sensitivity to sex steroid feedback in elderly postmenopausal women. Neuroendocrinology (1994) 59(4):355–62. doi:10.1159/000126678

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text | Google Scholar

44. Casadesus G, Webber KM, Atwood CS, Pappolla MA, Perry G, Bowen RL, et al. Luteinizing hormone modulates cognition and amyloid-β deposition in Alzheimer APP transgenic mice. Biochim Biophys Acta (2006) 1762(4):447–52. doi:10.1016/j.bbadis.2006.01.009

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text | Google Scholar

45. Bowen RL, Verdile G, Liu T, Parlow AF, Perry G, Smith MA. Luteinizing hormone, a reproductive regulator that modulates the processing of amyloid-β precursor protein and amyloid-β deposition. J Biol Chem (2004) 279(19):20539–45. doi:10.1074/jbc.M311993200

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text | Google Scholar

46. Ziegler SG, Thornton JE. Low luteinizing hormone enhances spatial memory and has protective effects on memory loss in rats. Horm Behav (2010) 58(5):705–13. doi:10.1016/j.yhbeh.2010.07.002

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text | Google Scholar

47. Kovacs M, Schally AV, Csernus B, Rekasi Z. Luteinizing hormone-releasing hormone (LH-RH) antagonist cetrorelix down-regulates the mRNA expression of pituitary receptors for LH-RH by counteracting the stimulatory effect of endogenous LH-RH. Proc Natl Acad Sci U S A (2001) 98(4):1829–34. doi:10.1073/pnas.98.4.1829

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text | Google Scholar

48. Palm R, Chang J, Blair J, Garcia-Mesa Y, Lee HG, Castellani RJ, et al. Down-regulation of serum gonadotropins but not estrogen replacement improves cognition in aged-ovariectomized 3xTg AD female mice. J Neurochem (2014) 130:115–25. doi:10.1111/jnc.12706

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text | Google Scholar

49. Chakravarti S, Collins WP, Forecast JD, Newton JR, Oram DH, Studd JW. Hormonal profiles after the menopause. Br Med J (1976) 2(6039):784–7. doi:10.1136/bmj.2.6039.784

CrossRef Full Text | Google Scholar

50. Neaves WB, Johnson L, Porter JC, Parker CR, Petty CS. Leydig cell numbers, daily sperm production, and serum gonadotropin levels in aging men. J Clin Endocrinol Metab (1984) 59(4):756–63. doi:10.1210/jcem-59-4-756

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text | Google Scholar

51. Rodrigues MA, Verdile G, Foster JK, Hogervorst E, Joesbury K, Dhaliwal S, et al. Gonadotropins and cognition in older women. J Alzheimers Dis (2008) 13(3):267–74.

Pubmed Abstract | Pubmed Full Text | Google Scholar

52. Hyde Z, Flicker L, Almeida OP, McCaul KA, Jamrozik K, Hankey GJ, et al. Higher luteinizing hormone is associated with poor memory recall: the health in men study. J Alzheimers Dis (2010) 19(3):943–51. doi:10.3233/JAD-2010-1342

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text | Google Scholar

53. Hogervorst E, Combrinck M, Smith AD. Testosterone and gonadotropin levels in men with dementia. Neuro Endocrinol Lett (2003) 24(3–4):203–8.

Pubmed Abstract | Pubmed Full Text | Google Scholar

54. Menon KMJ, Menon B. Structure, function and regulation of gonadotropin receptors–a perspective. Mol Cell Endocrinol (2012) 356(1):88–97. doi:10.1016/j.mce.2012.01.021

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text | Google Scholar

55. McFarland KC, Sprengel R, Phillips HS, Köhler M, Rosemblit N, Nikolics K, et al. Lutropin-choriogonadotropin receptor: an unusual member of the G protein-coupled receptor family. Science (1989) 245(4917):494–9. doi:10.1126/science.2502842

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text | Google Scholar

56. Loosfelt H, Misrahi M, Atger M, Salesse R, Vu HLTM, Jolivet A, et al. Cloning and sequencing of porcine LH-hCG receptor cDNA: variants lacking transmembrane domain. Science (1989) 245(4917):525–8. doi:10.1126/science.2502844

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text | Google Scholar

57. Gudermann T, Birnbaumer M, Birnbaumer L. Evidence for dual coupling of the murine luteinizing hormone receptor to adenylyl cyclase and phosphoinositide breakdown and Ca2+ mobilization. Studies with the cloned murine luteinizing hormone receptor expressed in L cells. J Biol Chem (1992) 267(7):4479–88.

Pubmed Abstract | Pubmed Full Text | Google Scholar

58. Oliver C, Mical RS, Porter JC. Hypothalamic-pituitary vasculature: evidence for retrograde blood flow in the pituitary stalk. Endocrinology (1977) 101(2):598–604. doi:10.1210/endo-101-2-598

CrossRef Full Text | Google Scholar

59. Apaja PM, Harju KT, Aatsinki JT, Petäjä-Repo UE, Rajaniemi HJ. Identification and structural characterization of the neuronal luteinizing hormone receptor associated with sensory systems. J Biol Chem (2004) 279(3):1899–906. doi:10.1074/jbc.M311395200

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text | Google Scholar

60. Hämäläinen T, Poutanen M, Huhtaniemi I. Age-and sex-specific promoter function of a 2-kilobase 5’-flanking sequence of the murine luteinizing hormone receptor gene in transgenic mice. Endocrinology (1999) 140(11):5322–9. doi:10.1210/endo.140.11.7115

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text | Google Scholar

61. Al-Hader AA, Lei ZM, Rao CV. Neurons from fetal rat brains contain functional luteinizing hormone/chorionic gonadotropin receptors. Biol Reprod (1997) 56(5):1071–6. doi:10.1095/biolreprod56.5.1071

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text | Google Scholar

62. Al-Hader AA, Lei ZM, Rao CV. Novel expression of functional luteinizing hormone/chorionic gonadotropin receptors in cultured glial cells from neonatal rat brains. Biol Reprod (1997) 56(2):501–7. doi:10.1095/biolreprod56.5.1071

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text | Google Scholar

63. Lei ZM, Rao CV, Kornyei JL, Licht P, Hiatt ES. Novel expression of human chorionic gonadotropin/luteinizing hormone receptor gene in brain. Endocrinology (1993) 132(5):2262–70. doi:10.1210/en.132.5.2262

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text | Google Scholar

64. Yang EJ, Nasipak BT, Kelley DB. Direct action of gonadotropin in brain integrates behavioral and reproductive functions. Proc Natl Acad Sci U S A (2007) 104(7):2477–82. doi:10.1073/pnas.0608391104

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text | Google Scholar

65. Lukacs H, Hiatt ES, Lei ZM, Rao CV. Peripheral and intracerebroventricular administration of human chorionic gonadotropin alters several hippocampus-associated behaviors in cycling female rats. Horm Behav (1995) 29(1):42–58. doi:10.1006/hbeh.1995.1004

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text | Google Scholar

66. Bagshawe KD, Orr AH, Rushworth AGJ. Relationship between concentrations of human chorionic gonadotropin in plasma and cerebrospinal fluid. Nature (1968) 217(5132):950–1. doi:10.1038/217950a0

CrossRef Full Text | Google Scholar

67. Berkowitz RS, Osathanondh R, Goldstein DP, Martin PM, Mallampati SR, Datta S. Cerebrospinal fluid human chorionic gonadotropin levels in normal pregnancy and choriocarcinoma. Surg Gynecol Obstet (1981) 153(5):687–9.

Pubmed Abstract | Pubmed Full Text | Google Scholar

68. Toth P, Lukacs H, Hiatt ES, Reid KH, Iyer V, Rao CV. Administration of human chorionic gonadotropin affects sleep-wake phases and other associated behaviors in cycling female rats. Brain Res (1994) 654(2):181–90. doi:10.1016/0006-8993(94)90478-2

CrossRef Full Text | Google Scholar

69. Telegdy G, Tanaka M, Schally AV. Effects of the LHRH antagonist cetrorelix on the brain function in mice. Neuropeptides (2009) 43(3):229–34. doi:10.1016/j.npep.2009.03.001

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text | Google Scholar

70. Bryan KJ, Mudd JC, Richardson SL, Chang J, Lee HG, Zhu X, et al. Down-regulation of serum gonadotropins is as effective as estrogen replacement at improving menopause-associated cognitive deficits. J Neurochem (2010) 112(4):870–81. doi:10.1111/j.1471-4159.2009.06502.x

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text | Google Scholar

71. Palaniappan M, Menon KMJ. Luteinizing hormone/human chorionic gonadotropin-mediated activation of mTORC1 signaling is required for androgen synthesis by theca-interstitial cells. Mol Endocrinol (2012) 26(10):1732–42. doi:10.1210/me.2012-1106

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text | Google Scholar

72. Flynn MP, Maizels ET, Karlsson AB, McAvoy T, Ahn JH, Nairn AC, et al. Luteinizing hormone receptor activation in ovarian granulosa cells promotes protein kinase A-dependent dephosphorylation of microtubule-associated protein 2D. Mol Endocrinol (2008) 22(7):1695–710. doi:10.1210/me.2007-0457

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text | Google Scholar

73. Hostetter G, Gallo RV, Brownfield MS. Presence of immunoreactive luteinizing hormone in the rat forebrain. Neuroendocrinology (1981) 33(4):241–5. doi:10.1159/000123238

CrossRef Full Text | Google Scholar

74. Emanuele NV, Anderson J, Andersen E, Connick E, Baker G, Kirsteins L, et al. Extrahypothalamic brain luteinizing hormone: characterization by radioimmunoassay, chromatography, radioligand assay and bioassay. Neuroendocrinology (1983) 36(4):254–60. doi:10.1159/000123464

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text | Google Scholar

75. Pike CJ, Rosario ER, Nguyen T-VV. Androgens, aging, and Alzheimer’s disease. Endocrine (2006) 29:233–42. doi:10.1385/ENDO:29:2:233

CrossRef Full Text | Google Scholar

76. Holland J, Bandelow S, Hogervorst E. Testosterone levels and cognition in elderly men: a review. Maturitas (2011) 69:322–37. doi:10.1016/j.maturitas.2011.05.012

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text | Google Scholar

77. Cherrier MM, Matsumoto AM, Amory JK, Ahmed S, Bremner W, Peskind ER, et al. The role of aromatization in testosterone supplementation effects on cognition in older men. Neurology (2005) 64(2):290–6. doi:10.1212/01.WNL.0000149639.25136.CA

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text | Google Scholar

78. Janowsky JS, Chavez B, Orwoll E. Sex steroids modify working memory. J Cogn Neurosci (2000) 12(3):407–14. doi:10.1162/089892900562228

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text | Google Scholar

79. Janowsky JS, Oviatt SK, Orwoll ES. Testosterone influences spatial cognition in older men. Behav Neurosci (1994) 108(2):325–32. doi:10.1037/0735-7044.108.2.325

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text | Google Scholar

80. Sih R, Morley JE, Kaiser FE, Perry HM, Patrick P, Ross C. Testosterone replacement in older hypogonadal men: a 12-month randomized controlled trial. J Clin Endocrinol Metab (1997) 82(6):1661–7. doi:10.1210/jcem.82.6.3988

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text | Google Scholar

81. Kenny AM, Bellantonio S, Gruman CA, Acosta RD, Prestwood KM. Effects of transdermal testosterone on cognitive function and health perception in older men with low bioavailable testosterone levels. J Gerontol A Biol Sci Med Sci (2002) 57(5):M321–5. doi:10.1093/gerona/57.5.M321

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text | Google Scholar

82. Cherrier MM, Matsumoto AM, Amory JK, Asthana S, Bremner W, Peskind ER, et al. Testosterone improves spatial memory in men with Alzheimer disease and mild cognitive impairment. Neurology (2005) 64(12):2063–8. doi:10.1212/01.WNL.0000165995.98986.F1

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text | Google Scholar

83. Tan RS, Pu SJ. A pilot study on the effects of testosterone in hypogonadal aging male patients with Alzheimer’s disease. Aging Male (2003) 6(1):13–7. doi:10.1080/tam.6.1.13.17

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text | Google Scholar

84. Lu PH, Masterman DA, Mulnard R, Cotman C, Miller B, Yaffe K, et al. Effects of testosterone on cognition and mood in male patients with mild Alzheimer disease and healthy elderly men. Arch Neurol (2006) 63(2):177–85. doi:10.1001/archneur.63.2.nct50002

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text | Google Scholar

85. Kenny AM, Fabregas G, Song C, Biskup B, Bellantonio S. Effects of testosterone on behavior, depression, and cognitive function in older men with mild cognitive loss. J Gerontol A Biol Sci Med Sci (2004) 59(1):M75–8. doi:10.1093/gerona/59.1.M75

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text | Google Scholar

86. Beauchet O. Testosterone and cognitive function: current clinical evidence of a relationship. Eur J Endocrinol (2006) 155(6):773–81. doi:10.1530/eje.1.02306

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text | Google Scholar

87. Tohgi H, Utsugisawa K, Yamagata M, Yoshimura M. Effects of age on messenger RNA expression of glucocorticoid, thyroid hormone, androgen, and estrogen receptors in postmortem human hippocampus. Brain Res (1995) 700:245–53. doi:10.1016/0006-8993(95)00971-R

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text | Google Scholar

88. Sarkey S, Azcoitia I, Garcia-Segura LM, Garcia-Ovejero D, DonCarlos LL. Classical androgen receptors in non-classical sites in the brain. Horm Behav (2008) 53(5):753–64. doi:10.1016/j.yhbeh.2008.02.015

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text | Google Scholar

89. Gouras GK, Xu H, Gross RS, Greenfield JP, Hai B, Wang R, et al. Testosterone reduces neuronal secretion of Alzheimer’s β-amyloid peptides. Proc Natl Acad Sci U S A (2000) 97(3):1202–5. doi:10.1073/pnas.97.3.1202

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text | Google Scholar

90. McAllister C, Long J, Bowers A, Walker A, Cao P, Honda SI, et al. Genetic targeting aromatase in male amyloid precursor protein transgenic mice down-regulates β-secretase (BACE1) and prevents Alzheimer-like pathology and cognitive impairment. J Neurosci (2010) 30(21):7326–34. doi:10.1523/JNEUROSCI.1180-10.2010

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text | Google Scholar

91. MacLusky NJ, Hajszan T, Johansen JA, Jordan CL, Leranth C. Androgen effects on hippocampal CA1 spine synapse numbers are retained in Tfm male rats with defective androgen receptors. Endocrinology (2006) 147(5):2392–8. doi:10.1210/en.2005-0673

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text | Google Scholar

92. Nguyen TVV, Yao M, Pike CJ. Dihydrotestosterone activates CREB signaling in cultured hippocampal neurons. Brain Res (2009) 1298:1–12. doi:10.1016/j.brainres.2009.08.066

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text | Google Scholar

93. Schmidt BM, Gerdes D, Feuring M, Falkenstein E, Christ M, Wehling M. Rapid, nongenomic steroid actions: a new age? Front Neuroendocrinol (2000) 21(1):57–94. doi:10.1006/frne.1999.0189

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text | Google Scholar

94. Tabori NE, Stewart LS, Znamensky V, Romeo RD, Alves SE, McEwen BS, et al. Ultrastructural evidence that androgen receptors are located at extranuclear sites in the rat hippocampal formation. Neuroscience (2005) 130(1):151–63. doi:10.1016/j.neuroscience.2004.08.048

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text | Google Scholar

95. Milner TA, McEwen BS, Hayashi S, Li CJ, Reagan LP, Alves SE. Ultrastructural evidence that hippocampal alpha estrogen receptors are located at extranuclear sites. J Comp Neurol (2000) 429(3):355–71. doi:10.1002/1096-9861(20010115)429:3<355::AID-CNE1>3.3.CO;2-R

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text | Google Scholar

96. Rosario ER, Carroll J, Pike CJ. Testosterone regulation of Alzheimer-like neuropathology in male 3xTg-AD mice involves both estrogen and androgen pathways. Brain Res (2010) 1359:281–90. doi:10.1016/j.brainres.2010.08.068

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text | Google Scholar

97. Roselli CE, Horton LE, Resko JA. Distribution and regulation of aromatase activity in the rat hypothalamus and limbic system. Endocrinology (1985) 117:2471–7. doi:10.1210/endo-117-6-2471

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text | Google Scholar

98. Muller M, Schupf N, Manly JJ, Mayeux R, Luchsinger JA. Sex hormone binding globulin and incident Alzheimer’s disease in elderly men and women. Neurobiol Aging (2010) 31(10):1758–65. doi:10.1016/j.neurobiolaging.2008.10.001

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text | Google Scholar

99. Yaffe K, Lui LY, Zmuda J, Cauley J. Sex hormones and cognitive function in older men. J Am Geriatr Soc (2002) 50(4):707–12. doi:10.1046/j.1532-5415.2002.50166.x

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text | Google Scholar

100. Hoskin EK, Tang MX, Manly JJ, Mayeux R. Elevated sex-hormone binding globulin in elderly women with Alzheimer’s disease. Neurobiol Aging (2004) 25(2):141–7. doi:10.1016/S0197-4580(03)00046-0

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text | Google Scholar

101. Paoletti AM, Congia S, Lello S, Tedde D, Orru M, Pistis M, et al. Low androgenization index in elderly women and elderly men with Alzheimer’s disease. Neurology (2004) 62(2):301–3. doi:10.1212/01.WNL.0000094199.60829.F5

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text | Google Scholar

102. Bowen RL, Perry G, Xiong C, Smith MA, Atwood CS. A clinical study of lupron depot in the treatment of women with Alzheimer’s disease: preservation of cognitive function in patients taking an acetylcholinesterase inhibitor and treated with high dose lupron over 48 weeks. J Alzheimers Dis (2015) 44(2):549–60. doi:10.3233/JAD-141626

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text | Google Scholar

Keywords: luteinizing hormone, menopause, ovariectomy, estrogen, testosterone, memory, Alzheimer’s disease

Citation: Blair JA, McGee H, Bhatta S, Palm R and Casadesus G (2015) Hypothalamic–pituitary–gonadal axis involvement in learning and memory and Alzheimer’s disease: more than “just” estrogen. Front. Endocrinol. 6:45. doi: 10.3389/fendo.2015.00045

Received: 12 December 2014; Accepted: 12 March 2015;
Published online: 25 March 2015.

Edited by:

Hubert Vaudry, University of Rouen, France

Reviewed by:

Jacques Epelbaum, INSERM, France
Vance Trudeau, University of Ottawa, Canada

Copyright: © 2015 Blair, McGee, Bhatta, Palm and Casadesus. This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) or licensor are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.

*Correspondence: Gemma Casadesus, Department of Biological Sciences, Kent State University, 256 Cunningham Hall, Kent, OH 44242, USA e-mail: gcasades@kent.edu

Disclaimer: All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article or claim that may be made by its manufacturer is not guaranteed or endorsed by the publisher.