Alzheimer disease (AD) is the most common form of dementia among the elderly. A higher prevalence of AD in women than in men suggests a link between gonadal hormone levels and AD. Increasing evidence supports a role for estrogen in brain regions involved in learning and memory and in the protection and regulation of cholinergic neurons, which degenerate in AD. Despite the lack of consensus, many studies indicate that hormone replacement therapy may decrease the risk for or delay the onset of AD in postmenopausal women. Recent trials have suggested that estrogen treatment may have no significant effect on the clinical course of AD in elderly women with the disease. Thus, the role of estrogen therapy seems to be confined to primary rather than secondary prevention of AD. Ongoing clinical studies may help to determine the role of estrogen in the cognitive function of postmenopausal women and in the prevention of AD.
Alzheimer disease (AD) is a neurodegenerative disorder that progressively affects intellectual functions. Alzheimer disease is manifested primarily in the impairment of cognitive functions such as memory and language. In 2000, AD affected an estimated 2.5 million to 4.5 million Americans,1,2 resulting in a profound emotional, social, and economic burden. As life expectancy continues to increase in the United States, the delay or the prevention of this degenerative disorder will become even more pressing.
Alzheimer disease is more common in women,3 with the prevalence of AD among women in the United States double that among men.2 A recent meta-analysis of 7 sex-specific studies of incidence rates for AD concluded that AD is 1.5 times more likely to develop in women than in men.4 These data suggest that low estrogen levels may be linked to the decline in cognitive function associated with dementia of the Alzheimer type.5 Decreased estrogen levels after menopause is a risk factor for AD,6 and neurobiological studies have found a link between estrogen and learning and memory functions.7-13 For example, low estrogen levels negatively affect the performance of rodents on learning and memory tasks, whereas administration of estrogen reverses this effect.14 Clinical research has focused on various roles for estrogen replacement therapy (ERT) (consisting of unopposed estrogens) and hormone replacement therapy (HRT) (consisting of estrogens in combination with a progestin): ERT/HRT in the cognitive function of healthy postmenopausal women15-19; the effect of ERT/HRT on the cognitive decline of elderly women, some of whom have mild cognitive impairment20-24; the link between ERT/HRT and the risk for development of AD25-29; and the use of estrogen to treat AD.30-34
A recent meta-analysis35 found that, according to most studies, ERT/HRT has beneficial effects on learning and memory in postmenopausal women and is associated with a reduced risk for AD; a handful of studies, however, did not show significant effects.35 In the absence of large randomized studies, no definitive evidence or consensus exists regarding the use of estrogen to prevent or to delay AD. It is also unclear whether any beneficial effects of estrogen on cognitive function occur immediately after menopause or later in life, and whether estrogen is effective in preventing the cognitive decline observed in normal aging and/or in pathological conditions. Inconsistent findings in these areas may be attributed to variations among any of the following variables: the size of the study population; the participants' ages, lifestyles, and educational levels; demographic features; the method of obtaining information about estrogen use, which may depend on participant recall; the route of administration of the hormone; the duration of the treatment; and the approach used to evaluate cognitive decline. More multicenter studies with a larger number of participants and standardized methods of diagnosis and evaluation are necessary to settle these issues.
This article reviews the neuroprotective and neurotrophic effects of estrogen, focusing on brain regions involved in learning and memory. It then discusses evidence regarding the effectiveness of ERT/HRT in preventing or delaying the onset of AD and surveys the nascent research on the use of estrogen to treat the disease.
A number of underlying causes for the neuronal damage seen in AD have been proposed, including oxidative stress caused by free radicals, hormonal insufficiency, loss of trophic support, hypoxia, and trauma. Vascular disease, which diminishes regional cerebral blood flow, may also be a risk factor for AD.36 In addition, Panidis et al36 suggested that nearly 30% of cases of AD are attributable to genetic factors, particularly polymorphism of apolipoprotein E (ApoE). Of the 3 types of genes for ApoE (ϵ2, ϵ3, and ϵ4), the ϵ4 allele is a known risk factor for AD.37,38 The ϵ4 allele is responsible for the production of the ApoE4 isoform, which can interact with amyloid β-protein (Aβ) to form AD-associated neuritic plaques.39 Autopsy findings in patients with late-onset AD show increases in Aβ deposition in patients with ApoE ϵ4.40
Early AD mainly affects brain regions involved in learning and memory, such as the entorhinal cortex and the hippocampus.41-43 The 2 main signs of the pathologic changes of AD include neuritic plaques mainly containing fibrillar Aβ, and neurofibrillary tangles composed of phosphorylated tau molecules that form paired helical filaments.36,44-46 Amyloid β-protein is probably produced by the metabolism of the amyloid precursor protein (APP) at the β cleavage site.47 Mutations of APP have been implicated in early-onset AD and can lead to aggregations of Aβ plaques early in the development of the disease.48 Neurofibrillary tangles, which are found in aging brains in general, may mark a phase in neuronal degeneration, since they appear where neurons have died.44 Neurofibrillary tangles and Aβ plaques can occur independently.45 Other neurotoxic agents that may play a role in the degeneration associated with AD are hydrogen peroxide, a precursor for free radicals that have also been associated with the neuronal damage seen in AD,49,50 and glutamate, the principal excitatory neurotransmitter, which may contribute to AD through excitotoxicity.50
Patients with AD also exhibit profound, progressive loss of cholinergic neurons in the nuclei of the basal forebrain,45,51 which project to the hippocampus and the neocortex and are essential for learning and memory.52,53 The loss of these neurons in the nuclei of the basal forebrain, and a corresponding decrease in cholinergic innervation of the hippocampal formation and the neocortex, are hallmarks of AD.54-56 Whitehouse et al51 demonstrated that neurons in the nucleus basalis of Meynert, which project directly to the cerebral cortex, are decreased by as much as 80% in the brains of patients with AD or dementia of the Alzheimer type. The cortical neuronal atrophy and decline of synaptic density in the cortex and hippocampus are likely correlates of dementia.45
The best available marker for cholinergic neurons in the basal forebrain is choline acetyltransferase (ChAT) activity.51 Choline acetyltransferase synthesizes the neurotransmitter acetylcholine (ACh), which is involved in transmitting messages between the basal forebrain and the cortex, hippocampus, and amygdala.36 Choline acetyltransferase also inhibits the expression of acetylcholinesterase, an enzyme that is involved in the metabolism of ACh.57 Several studies have reported a significant decrease in ChAT activity in the postmortem brains of demented patients,56 and levels of ACh are 90% lower in patients with AD.36
Effects of estrogen on brain function
There are multiple pathways to neuronal injury, dysfunction, and ultimately death in AD, many of which are potentially modified by estrogen. Evidence suggests that estrogen protects against various neurotoxic events and has a neurotrophic, regulatory role in the cholinergic system (Table 1).12 New research supports additional protective and regulatory activities of estrogen on the expression of genes associated with AD.58,65,84-87
Neuroprotective effects of estrogen
Although more research is needed on the specific mechanisms, recent data from an in vitro study indicate that estrogen is highly neuroprotective against a wide range of neurologic insults associated with AD.50 Experimental evidence further suggests that the antioxidant potency of estrogen is inherent, independent of receptor binding.49,94 The neuroprotective effects of estrogen against the oxidative damage and lipid peroxidation caused by toxins could be a mechanism to explain the reduced risk for AD seen in women using estrogen therapy.50
Estrogen is particularly effective against neuronal injury induced by the toxins Aβ49,50,59-62,64 and glutamate.10,49,50 Using an animal model, Thomas and Rhodin64 recently found that low doses of conjugated equine estrogens prevented the abnormal deposition of Aβ in the cerebral vasculature and the adhesion and transmigration of leukocytes that mark an inflammatory reaction, which may have relevance for the chronic inflammation seen in AD. Estrogen has been shown to attenuate elevated calcium levels induced by Aβ and glutamate and to suppress lipid peroxidation induced by iron and Aβ.59 Xu et al63 recently found that 17β-estradiol reduced the generation of plaque-forming Aβ by rodent and human neurons.
Estrogen may also protect neurons from Aβ toxicity by stimulating the proteolysis of APP.58 Jaffe and colleagues58 reported that estrogen promotes the metabolism of APP into its nonamyloidogenic part. A more recent study of the effect of estrogen on neuronal Swedish-mutated APP found that although estradiol increased nonamyloidogenic APP secretion in primary cortical neurons, Aβ production was undiminished, possibly owing to the interference of astrocytes.65 Recent animal models of AD have also indicated a link between the excessive expression of APP and the loss of cholinergic function seen in AD.95-97 Interactions among estrogen, APP, and nerve growth factor have been suggested to protect against the degeneration of cholinergic neurons,98 but more research is needed to determine the mechanisms by which estrogen influences APP metabolism.
Estrogen also guards against intracellular hydrogen peroxide accumulation, preventing the degeneration of primary neurons and hippocampal cells.49,50 It appears that 17β-estradiol acts directly on synapses to prevent oxidative impairment of sodium and potassium ions in adenosine triphosphatase activity, glucose transport, and glutamate transport.61
Finally, a recent report73 that used magnetic resonance imaging to evaluate the effects of sex and estrogen use on hippocampal volume in 13 elderly women taking ERT, 46 women not taking any estrogen therapy, and 38 men found that the women taking ERT had significantly larger right anterior hippocampal volumes than the other 2 groups. Sex did not have a significant effect, supporting a neuroprotective effect of estrogen.
Neurotrophic effects of estrogen
In addition to the neuroprotective properties, estrogen exerts trophic and regulatory effects on basal forebrain cholinergic neurons.11 A number of studies have shown that the regulatory role of estrogen in the basal forebrain influences hippocampal morphology and function. Luine and colleagues70 have suggested that, in addition to the direct effects of estrogen on the hippocampus, estrogen initiates hippocampal effects that are mediated by areas projecting to the hippocampus. They found that the performance of rats on spatial memory tasks, which are dependent on hippocampal function, improved significantly after long-term estradiol treatment; however, increases in monoaminergic and amino acid neurotransmitter activity were seen in the frontal cortex and the basal forebrain rather than in the hippocampus.70 These results are consistent with those of other studies69,71 in which administration of estradiol to ovariectomized rats improved their performance on spatial memory tasks—in one study,71 after 3.5 and 12 months of continuous treatment—and reduced the cognitive-impairing effects of scopolamine hydrochloride. These experimental studies were supported by a recent randomized, placebo-controlled study72 among 15 postmenopausal women that showed that the effects of scopolamine hydrochloride (2.5 µg/kg given as a one-time dose) were blunted in subjects treated with 17β-estradiol (1 mg/d) for 3 months. Long-term benefits in hippocampal function may be due to the influence of short-term changes in cholinergic activity, induced by estrogen, that project to the hippocampus, and may help explain the reduction in the risk for and severity of AD in postmenopausal women who have taken ERT.71
Dendritic spines, the principal loci of neuronal interactions and communication in the central nervous system, are among the central targets of the mechanisms of action of estrogen. A dramatic decrease in dendritic spine density has been observed in the ventromedial hypothalamic neurons of ovariectomized rats that was reversed by administering estrogen.99 This is consistent with findings that the density of synapses and synaptic spines fluctuates during the estrous cycle, increasing in response to estrogen.7,8 More recent studies9,13 on rat hippocampal neurons in culture have confirmed that estrogen plays a critical role in a process that yields a 2-fold increase in dendritic spine density. This process may be mediated by brain-derived neurotrophic factor; however, the effects of estrogen on brain-derived neurotrophic factor regulation in this brain region are not yet fully understood.13,100 The functional consequence of increased dendritic spine density is reflected in the improvement of rodent performance in behaviors related to hippocampal function. Estrogen enhances the induction of long-term potentiation in awake animals, a model of synaptic plasticity in the hippocampus with potential relevance to learning and associative memory.101
Evidence of the local effects of estrogen in the hippocampus and the neocortex has also been established. Estrogen appears to interact closely with neurotrophins, which also promote neuronal growth and block apoptosis, in the basal forebrain. In the 1980s, O'Malley and colleagues74 proposed that estrogen modulated the production, release, and uptake of ACh by cholinergic neurons. Administering estrogen to ovariectomized rats induced the potassium-evoked release of ACh,68 which is inhibited by Aβ,102 in the hippocampus and the overlying cortex. The enhanced release of ACh in these areas is reflected in direct, trophic effects of estrogen on hippocampal neurons. Brinton66 reported filopodial growth in hippocampal neurons within 5 minutes of exposure to 17β-estradiol, and has since demonstrated with others significantly increased hippocampal and neocortical neurite outgrowth, viability, and survival after exposure to 17β-estradiol and conjugated equine estrogens.50,67 Effective protection of rodent neuronal cells in vitro from toxic complexes formed by the combination of acetylcholinesterase and Aβ by 17β-estradiol has also been demonstrated.62
Estradiol was also found to increase ChAT activity in certain basal forebrain cholinergic nuclei in female rats.57 A recent study, however, reported that although the administration of estradiol increased ChAT expression and high-affinity choline uptake in the cholinergic system of ovariectomized rats after 2 weeks, no changes were found after 4 weeks of continuous or repeated estrogen treatment.83 Furthermore, continuous estrogen therapy administered for 13 months led to a decrease in high-affinity choline uptake, especially in the hippocampus.83 Although these findings indicate that enhancement of cholinergic function by estrogen may be short-term, other data show that 4 weeks of estrogen treatment in rats produces consistent decreases in levels of the low-affinity nerve growth factor receptor p75NGFR, an effect of increased ChAT levels that plays a crucial role in regulating cholinergic activity.80
Toran-Allerand and colleagues103 consider estrogen to be a neuronal growth factor that shares many characteristics of neurotrophins, enabling convergence of estrogen- and neurotrophin-signaling pathways. The decline of gonadal steroid levels in both sexes with aging may thus contribute to the loss of neuronal systems integral to cognitive function.103
Effects of estrogen on genes associated with ad
Recent studies have investigated the effects of estrogen on the expression of ApoE4. Estradiol up-regulated ApoE gene expression by increasing levels of ApoE messenger RNA in an animal model of AD84; in a similar animal model, estradiol enhanced synaptogenesis, possibly through an ApoE-dependent mechanism.85 Teter and colleagues86 have recently confirmed these findings by studying the interaction of ApoE and estrogen in mouse hippocampal slice cultures. Neuronal sprouting increased in ApoE-dependent areas, possibly as a consequence of the up-regulation by estrogen of ApoE expression to enable the recycling of membrane lipids for use by sprouting neurons.86 A population-based case-control study investigating a possible link between estrogen and early-onset AD, which included 53% ApoE ϵ3/ϵ4 or ApoE ϵ4/ϵ4 carriers, found a stronger inverse correlation between estrogen use and early-onset AD in this group (odds ratio [OR], 0.37; 95% confidence interval [CI], 0.08-1.58) than in women with the ApoE3 ϵ3/ϵ3 genotype (OR, 0.60; 95% CI, 0.19-1.88).104 However, another study found no reduction in the risk for cognitive decline among women with the ApoE ϵ4 allele who used ERT/HRT, although hormone users without the ApoE ϵ4 allele exhibited less cognitive decline.38 Moreover, Lendon and Lambert105 recently reported that estrogen enhanced expression of the ϵ4 allele. The possible mechanisms involved in the interaction of estrogen and ApoE are the subject of ongoing research.
Mattson et al87 have shown that 17β-estradiol blocks the expression of mutant presenilin-1, a proapoptotic gene linked to early-onset AD that was found in a study of transgenic mice to have a synergistic effect with mutated APP, leading to decreased cholinergic function.96 In conjunction with findings that 17β-estradiol protects neurons against nip-2, another gene that promotes cell death,106 this research suggests a direct, receptor-independent role for estrogen in preventing neuronal loss associated with AD. More specifically, 17β-estradiol delayed cellular glucose deprivation induced by nip-2106 through a mechanism that may have relevance to AD, since glucose transport and metabolism are diminished in the disease.107 In their study of the antiapoptotic action of estrogen, Mattson and colleagues87 found an additional protective capacity of 17β-estradiol to stabilize mitochondrial function.
Other effects of estrogen on the brain
Studies of cognitive function in individuals have established that, during memory processing, estrogen increases glucose transport88 and regional cerebral blood flow,89-91 which are decreased in AD. Recently, Maki and Resnick91 examined longitudinal changes in regional cerebral blood flow in 12 ERT/HRT users and 16 nonusers during the performance of verbal and figural recognition memory tasks. In addition to obtaining higher scores than nonusers on a battery of standardized memory tests, the ERT/HRT users exhibited enhanced regional cerebral blood flow in the hippocampus, the parahippocampal gyrus, and the temporal lobe, regions fundamental to memory function that can reveal preclinical abnormalities in individuals at risk for AD. These results were similar to those found in 2 earlier studies,89,90 suggesting a key mechanism through which ERT/HRT may decrease the risk for AD. In their recent comparison of the effect of estradiol on middle-aged and young female rats, Dubal and Wise92 suggested that estrogen achieves neuroprotective effects by modulating regional blood flow, which may be maintained after menopause by ERT.
In summary, numerous studies confirm that estrogen exerts a wide range of neuroprotective and neurotrophic influences on brain regions and neuronal subtypes involved in memory and cognitive function that are negatively affected by AD. These studies have provided the basis for investigations of the effects of estrogen on cognitive impairment in the course of normal as well as pathologic aging of postmenopausal women.
Ert/hrt and the risk for and onset of ad
As mentioned earlier, AD is more likely to develop in women older than 65 years than in their male counterparts,3,4 possibly due to reduced estrogen levels.5 The association between ERT/HRT and the risk for AD remains controversial, although most investigations suggest that ERT/HRT reduces the risk for AD (Figure 1). A recent meta-analysis35 of 14 studies reported an OR of 0.56 (95% CI, 0.46-0.68) for the relative risk for the development of AD. The results of the studies analyzed were heterogeneous, and poor recall of ERT/HRT use may have confounded the results. A major 1998 meta-analysis116 of the effect of ERT/HRT on the risk for the development of AD in postmenopausal women, which examined 8 case-control studies6,108-114 and 2 prospective cohort studies,26,115 reported a summary OR of 0.71 (95% CI, 0.52-0.98) for the development of dementia among estrogen users. Both prospective cohort studies and 1 case-control study113 reported a significantly lower risk for dementia in women who had ever used estrogen. Of the remaining studies, 3 reported no significant increase among estrogen users,6,110,114 2 reported no difference in risk,112,113 and 2 found no significant increased risk for dementia among estrogen users compared with nonusers.108,109 Because of significant heterogeneity in the findings, which may be attributable to study design, a separate analysis was performed of the 2 study types. The summary OR for the case-control studies was 0.80 (95% CI, 0.56-1.16) for diagnosis of AD; for the prospective studies, the summary OR was 0.48 (95% CI, 0.29-0.81). The 3 studies that investigated the relationship between the duration of estrogen use and protection against dementia found inconsistent results,6,26,115 although in a follow-up investigation of their earlier study, Paganini-Hill and Henderson25 found a decreased risk among long-term users of ERT.
As the authors point out, observational studies are prone to confounding and compliance bias, which may influence their assessment for the risk of development of AD. In addition, despite the fact that HRT is prescribed more commonly than unopposed ERT in the United States, none of these studies included a significant proportion of HRT users. Two studies from the meta-analysis, however, merit more detailed consideration. The population-based investigations of Paganini-Hill and Henderson,6,25 which show a decreased risk for AD with estrogen treatment, included a cohort of 8877 women and the completion of health surveys by the individuals rather than by proxy informants, an approach that provides more accurate data about the use of estrogen. The risk for AD was found to be 35% lower in estrogen users.25 Tang and colleagues26 followed up 1124 older women for 1 to 5 years and identified 167 incident cases of AD. The risk for AD was reduced by 50% among subjects who had used estrogen (OR adjusted for education, ethnicity, and ApoE genotype). A direct relationship between the duration of hormone treatment and the risk for AD was also reported; the risk was lower among women who had used estrogen for 1 to 5 years than among subjects who had used estrogen for 1 year or less. When demented patients who had used and those who had never used estrogen were compared, the age of AD onset was significantly delayed among estrogen users.
One of the 2 case-control studies in the 1998 meta-analysis116 that reported no difference in risk111,112 raised the possibility that the route of administration may influence whether estrogen protects against AD. Brenner and colleagues112 compared the use of estrogen in 107 cases with AD and 120 age-matched control subjects and found no association between the use of estrogen and the risk for AD. However, the point estimate for AD was decreased by 30% when the results were analyzed for intake of oral estrogen alone. In contrast to these findings, the decrease in the risk for AD did not depend on the route of estrogen intake in the study by Paganini-Hill and Henderson.6
Two population-based, case-control studies published after the 1998 meta-analysis report conflicting findings. Results from a study by Waring and colleagues28 regarding the use of ERT/HRT and the risk for development of AD are consistent with the findings from Tang and associates.26 Among 222 women from the Rochester Epidemiology Project records-linkage system who were diagnosed as having AD from 1980 to 1984, the frequency of estrogen use was half that of the age-matched control group (n = 222) at 10% vs 5% (OR, 0.42; 95% CI, 0.18-0.96). In contrast, a nested case-control study by Seshadri et al29 found no reduced risk for development of AD among current ERT/HRT users. Starting with a large base cohort (n = 221 406) from the General Practice Research Database in the United Kingdom, 59 women were verified as having a new diagnosis of AD from 1992 to 1998 and matched to 221 controls. Fifteen (25%) of the 59 cases and 53 (24%) of the 221 controls were current hormone users, yielding an OR of 1.18 (95% CI, 0.59-2.37) for the risk for development of AD among current ERT/HRT users. The inconsistency in findings between these 2 studies indicates that this issue remains unresolved pending results from further studies.
Two recent studies suggest that ERT/HRT use is associated with a reduced risk for development of AD, but both have limitations due to study methods. Baldereschi and colleagues27 used results of the Mini-Mental State Examination for an initial screen in 2816 Italian women aged 65 to 84 years; those with positive findings underwent clinical assessment for dementia and AD. The frequency of hormone use was significantly higher among nondemented patients than those with AD after adjustment for age, education, age at menarche, age at menopause, cigarette and alcohol use, body weight at 50 years of age, and number of children (OR 0.28; 95% CI, 0.08-0.98). These findings were prone to recall bias regarding the elderly subjects' use of estrogen, especially among those who were cognitively impaired, although next of kin were questioned in these cases. In a longitudinal study of 3128 women who were outpatients at the California State Alzheimer's Disease Diagnostic and Treatment Centers, hormone users had significantly lower rates of diagnosis of AD at baseline and after 1 year, compared with nonusers.117 Moreover, patients who had not used estrogen showed increased cognitive deterioration from baseline to follow-up, whereas no significant change in cognitive function occurred among the estrogen users during this period. However, no significant difference was found between the performance of estrogen users and nonusers who had been diagnosed as having AD at baseline. Complete follow-up data were available for only a very small number (n = 16) of hormone users; nevertheless, these data suggest that estrogen may protect against cognitive decline in the earlier stages.
A recent population-based study in the Netherlands was, to our knowledge, the first to examine the effect of estrogen use on the risk for early-onset AD. In comparing patients with AD (n = 109) with age- and residence-matched controls (n = 119), Slooter and colleagues104 found a significant inverse correlation between estrogen treatment and early-onset AD (adjusted OR, 0.34; 95% CI, 0.12-0.94), suggesting that more research is needed in this specific area.
Treatment of ad with estrogen
Despite the significant neuroprotective and neurotrophic effect of estrogen, particularly in vitro, described already, human studies of the effect of estrogen as therapy for AD have been equivocal.5,30-35 Some studies, primarily short-term ones, have suggested that estrogen use results in short-term improvements in cognition.30-33,118,119 In contrast, 3 recent randomized, double-blind clinical trials analyzing the effects of estrogen on the clinical course of AD found no significant benefit with estrogen treatment.34,120,121 These findings may be surprising in view of the increasing evidence showing protective benefits of estrogen on neurons involved in learning and memory and studies showing a significantly reduced risk for AD among women using ERT or HRT. However, an agent that reduces the risk for AD will not necessarily influence the clinical course of the disease once it is established, since the mechanisms involved may be different. The administration of estrogen after neuronal injury has been initiated or has progressed (eg, after AD is expressed) may have no benefit, but estrogen may protect neurons at the initial phases or before the onset of the disease.
Thus, the initial timing of hormone treatment may be crucial. In the study by Mulnard and colleagues,34 ERT was not initiated immediately after hysterectomy, and the subjects were older than 60 years. The mean ages were 77 years for the estrogen group and 78 years for the placebo group in the study by Henderson et al120 and 73 years for the estrogen group and 71 years for the placebo group in the study by Wang et al.121 Estrogen may be most effective as a preventive agent immediately or shortly after menopause. Using an animal model, Gibbs122 determined that estrogen treatment initiated immediately after or 3 months after ovariectomy—but not after 10 months—improved performance on a spatial memory task. This research suggests that a key window of opportunity for the initiation of hormone treatment in the prevention of cognitive decline in humans may exist, a hypothesis that warrants further investigation. This situation is somewhat analogous to the prevention of osteoporosis, ie, the earlier estrogen treatment is initiated after menopause, the more beneficial its effect, since bone that is already lost cannot be replaced to any significant degree.123 Because the prevalence of osteoporosis rises dramatically with age, early intervention is necessary to forestall bone loss as long as possible.
Evidence supports a central role for estrogen in brain regions involved in memory and other cognitive tasks and in protection from AD-associated toxins and genes. A number of observational studies have suggested a significantly reduced risk for development of AD among women who have used ERT/HRT. The Women's Health Initiative Study of Cognitive Aging, a randomized, placebo-controlled trial among 2900 women that will assess cognitive outcomes with ERT/HRT use, is expected to provide substantial evidence regarding the influence of estrogen-based therapy on cognition and AD. That study is ancillary to the Women's Health Initiative Memory Study, which in turn is an arm of the Women's Health Initiative that is sponsored by the National Institutes of Health, Bethesda, Md. The Women's Health Initiative Memory Study will investigate the effect of conjugated equine estrogens on cognitive function and on the risk for the development of AD and other dementia among more than 8000 postmenopausal women, with a minimum follow-up of 6 years.50,124 The Women's International Study of Long-Duration Oestrogen After the Menopause, also under way, is not due to report results until 2010.125 These large clinical studies will provide valuable information regarding the potential effects of ERT/HRT on preserving cognitive function in postmenopausal women.
At present, most observational evidence, which is supported by neurobiological research findings on the action of estrogen, indicates that ERT/HRT mitigates the degeneration that may lead to AD. The lack of evidence of a role of estrogen in the treatment of AD suggests that ERT/HRT should be initiated as early as possible after menopause, before the onset or the progression of the disease. Thus, the relationship of postmenopausal hormone therapy to AD is somewhat parallel to its relationship to osteoporosis in that, in both cases, ERT/HRT seems to have a role in primary prevention.
Accepted for publication February 6, 2002.
1.Evans
DAScherr
PACook
NR
et al. Estimated prevalence of Alzheimer's disease in the United States.
Milbank Q. 1990;68267- 289
Google ScholarCrossref 2.Brookmeyer
RGray
SKawas
C Projections of Alzheimer's disease in the United States and the public health impact of delaying disease onset.
Am J Public Health. 1998;881337- 1342
Google ScholarCrossref 3.Molsa
PKMarttila
RJRinne
UK Epidemiology of dementia in a Finnish population.
Acta Neurol Scand. 1982;65541- 552
Google ScholarCrossref 4.Gao
SHendrie
HCHall
KSHui
S The relationships between age, sex, and the incidence of dementia and Alzheimer disease: a meta-analysis.
Arch Gen Psychiatry. 1998;55809- 815
Google ScholarCrossref 5.Fillit
HLuine
V The neurobiology of gonadal hormones and cognitive decline in late life.
Maturitas. 1997;26159- 164
Google ScholarCrossref 6.Paganini-Hill
AHenderson
VW Estrogen deficiency and risk of Alzheimer's disease in women.
Am J Epidemiol. 1994;140256- 261
Google Scholar 7.Woolley
CSGould
EFrankfurt
MMcEwen
BS Naturally occurring fluctuation in dendritic spine density on adult hippocampal pyramidal neurons.
J Neurosci. 1990;104035- 4039
Google Scholar 8.Woolley
CSMcEwen
BS Roles of estradiol and progesterone in regulation of hippocampal dendritic spine density during the estrous cycle in the rat.
J Comp Neurol. 1993;336293- 306
Google ScholarCrossref 9.Murphy
DDSegal
M Regulation of dendritic spine density in cultured rat hippocampal neurons by steroid hormones.
J Neurosci. 1996;164059- 4068
Google Scholar 10.Singer
CARogers
KLStrickland
TMDorsa
DM Estrogen protects primary cortical neurons from glutamate toxicity.
Neurosci Lett. 1996;21213- 16
Google ScholarCrossref 11.Gibbs
RBAggarwal
P Estrogen and basal forebrain cholinergic neurons: implications for brain aging and Alzheimer's disease-related cognitive decline.
Horm Behav. 1998;3498- 111
Google ScholarCrossref 12.Inestrosa
NCMarzolo
MPBonnefont
AB Cellular and molecular basis of estrogen's neuroprotection: potential relevance for Alzheimer's disease.
Mol Neurobiol. 1998;1773- 86
Google ScholarCrossref 13.Murphy
DDCole
NBSegal
M Brain-derived neurotrophic factor mediates estradiol-induced dendritic spine formation in hippocampal neurons.
Proc Natl Acad Sci U S A. 1998;9511412- 11417
Google ScholarCrossref 14.Singh
MMeyer
EMMillard
WJSimpkins
JW Ovarian steroid deprivation results in reversible learning impairment and compromised cholinergic function in Sprague-Dawley rats.
Brain Res. 1994;644305- 312
Google ScholarCrossref 15.Kampen
DLSherwin
BB Estrogen use and verbal memory in healthy postmenopausal women.
Obstet Gynecol. 1994;83979- 983
Google ScholarCrossref 16.Robinson
DFriedman
LMarcus
RTinklenberg
JYesavage
J Estrogen replacement therapy and memory in older women.
J Am Geriatr Soc. 1994;42919- 922
Google Scholar 17.Schmidt
RFazekas
FReinhart
B
et al. Estrogen replacement therapy in older women: a neuropsychological and brain MRI study.
J Am Geriatr Soc. 1996;441307- 1313
Google Scholar 18.Rice
MMGraves
ABMcCurry
SMLarson
EB Estrogen replacement therapy and cognitive function in postmenopausal women without dementia.
Am J Med. 1997;103(suppl 3A)26S- 35S
Google ScholarCrossref 19.Shaywitz
SEShaywitz
BAPugh
KR
et al. Effect of estrogen on brain activation patterns in postmenopausal women during working memory tasks.
JAMA. 1999;2811197- 1202
Google ScholarCrossref 20.Barrett-Connor
EKritz-Silverstein
D Estrogen replacement therapy and cognitive function in older women.
JAMA. 1993;2692637- 2641
Google ScholarCrossref 21.Resnick
SMMetter
EJZonderman
AB Estrogen replacement therapy and longitudinal decline in visual memory: a possible protective effect?
Neurology. 1997;491491- 1497
Google ScholarCrossref 22.Jacobs
DMTang
M-XStern
Y
et al. Cognitive function in nondemented older women who took estrogen after menopause.
Neurology. 1998;50368- 373
Google ScholarCrossref 23.Matthews
KCauley
JYaffe
KZmuda
JM Estrogen replacement therapy and cognitive decline in older community women.
J Am Geriatr Soc. 1999;47518- 523
Google Scholar 24.Steffens
DCNorton
MCPlassman
BL
et al. Enhanced cognitive performance with estrogen use in nondemented community-dwelling older women.
J Am Geriatr Soc. 1999;471171- 1175
Google Scholar 25.Paganini-Hill
AHenderson
VW Estrogen replacement therapy and risk of Alzheimer disease.
Arch Intern Med. 1996;1562213- 2217
Google ScholarCrossref 26.Tang
MXJacobs
DStern
Y
et al. Effect of oestrogen during menopause on risk and age at onset of Alzheimer's disease.
Lancet. 1996;348429- 432
Google ScholarCrossref 27.Baldereschi
MDi Carlo
ALepore
V
et al. Estrogen-replacement therapy and Alzheimer's disease in the Italian Longitudinal Study on Aging.
Neurology. 1998;50996- 1002
Google ScholarCrossref 28.Waring
SCRocca
WAPetersen
RCO'Brien
PCTangalos
EGKokmen
E Postmenopausal estrogen replacement therapy and risk of AD: a population-based study.
Neurology. 1999;52965- 970
Google ScholarCrossref 29.Seshadri
SZornberg
GLDerby
LEMyers
MWJick
HDrachman
DA Postmenopausal estrogen replacement therapy and the risk of Alzheimer disease.
Arch Neurol. 2001;58435- 440
Google ScholarCrossref 30.Fillit
HWeinreb
HCholst
I
et al. Observations in a preliminary open trial of estradiol therapy for senile dementia–Alzheimer's type.
Psychoneuroendocrinology. 1986;11337- 345
Google ScholarCrossref 31.Honjo
HOgino
YNaitoh
K
et al. In vivo effects by estrone sulfate on the central nervous system–senile dementia (Alzheimer's type).
J Steroid Biochem. 1989;34521- 525
Google ScholarCrossref 32.Ohkura
TIsse
KAkazawa
KHamamoto
MYaoi
YHagino
N Evaluation of estrogen treatment in female patients with dementia of the Alzheimer type.
Endocr J. 1994;41361- 371
Google ScholarCrossref 33.Asthana
SCraft
SBaker
LD
et al. Cognitive and neuroendocrine response to transdermal estrogen in postmenopausal women with Alzheimer's disease: results of a placebo-controlled, double-blind, pilot study.
Psychoneuroendocrinology. 1999;24657- 677
Google ScholarCrossref 34.Mulnard
RACotman
CWKawas
CW
et al. Estrogen replacement therapy for treatment of mild to moderate Alzheimer disease: a randomized controlled trial.
JAMA. 2000;2831007- 1015
Google ScholarCrossref 35.Hogervorst
EWilliams
JBudge
MRiedel
WJolles
J The nature of the effect of female gonadal hormone replacement therapy on cognitive function in post-menopausal women: a meta-analysis.
Neuroscience. 2000;101485- 512
Google ScholarCrossref 36.Panidis
DKMatalliotakis
IMRousso
DHKourtis
AIKoumantakis
EE The role of estrogen replacement therapy in Alzheimer's disease.
Eur J Obstet Gynecol Reprod Biol. 2001;9586- 91
Google ScholarCrossref 37.Petersen
RCSmith
GEIvnik
RJ
et al. Apolipoprotein E status as a predictor of the development of Alzheimer's disease in memory-impaired individuals.
JAMA. 1995;2731274- 1278
Google ScholarCrossref 38.Yaffe
KHaan
MByers
ATangen
CKuller
L Estrogen use,
APOE, and cognitive decline: evidence of gene-environment interaction.
Neurology. 2000;541949- 1954
Google ScholarCrossref 39.Naslund
JThyberg
JTjernberg
LO
et al. Characterization of stable complexes involving apolipoprotein E and the amyloid beta peptide in Alzheimer's disease brain.
Neuron. 1995;15219- 228
Google ScholarCrossref 40.Schmechel
DSaunders
AMTrittmatter
WJ
et al. Increased amyloid β-peptide deposition in cerebral cortex as a consequence of apolipoprotein E genotype in late-onset Alzheimer disease.
Proc Natl Acad Sci U S A. 1993;909649- 9653
Google ScholarCrossref 41.De Lacoste
MCWhite
CL The role of cortical connectivity in Alzheimer's disease pathogenesis: a review and model system.
Neurobiol Aging. 1993;141- 16
Google ScholarCrossref 42.Delacourte
ADavid
JPSergeant
N
et al. The biochemical pathway of neurofibrillary degeneration in aging and Alzheimer's disease.
Neurology. 1999;521158- 1165
Google ScholarCrossref 43.Yilmazer-Hanke
DMHanke
J Progression of Alzheimer-related neuritic plaque pathology in the entorhinal region, perirhinal cortex and hippocampal formation.
Dement Geriatr Cogn Disord. 1999;1070- 76
Google ScholarCrossref 45.Jellinger
K Morphology of Alzheimer's disease and related disorders. Maurer
KRiederer
PBeckman
H
Alzheimer's Disease Epidemiology, Neuropathology, Neurochemistry, and Clinics New York, NY Chapman & Hall1990;61- 77
Google Scholar 46.Grundke-Iqbal
IIqbal
KTung
YCQuinlan
MWisniewski
HMBinder
LI Abnormal phosphorylation of the microtubule-associated protein tau (tau) in Alzheimer cytoskeletal pathology.
Proc Natl Acad Sci U S A. 1986;834913- 4917
Google ScholarCrossref 48.Selkoe
DJ Amyloid β-protein and the genetics of Alzheimer's disease.
J Biol Chem. 1996;27118295- 18298
Google ScholarCrossref 49.Behl
CSkutella
TLezoualc'h
F
et al. Neuroprotection against oxidative stress by estrogens: structure-activity relationship.
Mol Pharmacol. 1997;51535- 541
Google Scholar 50.Brinton
RDChen
SMontoya
MHsieh
DMinaya
J The estrogen replacement therapy of the Women's Health Initiative promotes the cellular mechanisms of memory and neuronal survival in neurons vulnerable to Alzheimer's disease.
Maturitas. 2000;34(suppl 2)S35- S52
Google ScholarCrossref 51.Whitehouse
PJPrice
DLStruble
RGClark
AWCoyle
JTDeLong
MR Alzheimer's disease and senile dementia: loss of neurons in the basal forebrain.
Science. 1982;2151237- 1239
Google ScholarCrossref 52.Olton
DS Dementia: animal models of the cognitive impairments following damage to the basal forebrain cholinergic system.
Brain Res Bull. 1990;25499- 502
Google ScholarCrossref 53.McEntee
WJCrook
TH Cholinergic function in the aged brain: implications for treatment of memory impairments associated with aging.
Behav Pharmacol. 1992;3327- 336
Google ScholarCrossref 54.Sims
NRBowen
DMSmith
CCT
et al. Glucose metabolism and acetylcholine synthesis in relation to neuronal activity in Alzheimer's disease.
Lancet. 1980;1333- 336
Google ScholarCrossref 55.Fibiger
HC Cholinergic mechanisms in learning, memory and dementia: a review of recent evidence.
Trends Neurosci. 1991;14220- 223
Google ScholarCrossref 57.Luine
VN Estradiol increases choline acetyltransferase activity in specific basal forebrain nuclei and projection areas of female rats.
Exp Neurol. 1985;89484- 490
Google ScholarCrossref 58.Jaffe
ABToran-Allerand
CDGreengard
PGandy
SE Estrogen regulates metabolism of Alzheimer amyloid-β precursor protein.
J Biol Chem. 1994;26913065- 13068
Google Scholar 59.Goodman
YBruce
AJCheng
BMattson
MP Estrogens attenuate and corticosteroid exacerbates excitotoxicity, oxidative injury, and amyloid β-peptide toxicity in hippocampal neurons.
J Neurochem. 1996;661836- 1844
Google ScholarCrossref 60.Gridley
KEGreen
PSSimpkins
JW Low concentrations of estradiol reduce β-amyloid (25-35)–induced toxicity, lipid peroxidation and glucose utilization in human SK-N-SH neuroblastoma cells.
Brain Res. 1997;778158- 165
Google ScholarCrossref 61.Keller
JNGermeyer
ABegley
JGMattson
MP 17β-Estradiol attenuates oxidative impairment of synaptic Na+/K+-ATPase activity, glucose transport, and glutamate transport induced by amyloid β-peptide and iron.
J Neurosci Res. 1997;50522- 530
Google ScholarCrossref 62.Bonnefont
ABMunoz
FJInestrosa
NC Estrogen protects neuronal cells from the cytotoxicity induced by acetylcholinesterase-amyloid complexes.
FEBS Lett. 1998;441220- 224
Google ScholarCrossref 63.Xu
HGouras
GKGreenfield
JP
et al. Estrogen reduces neuronal generation of Alzheimer β-amyloid peptides.
Nat Med. 1998;4447- 451
Google ScholarCrossref 64.Thomas
TRhodin
J Vascular actions of estrogen and Alzheimer's disease.
Ann N Y Acad Sci. April2000;501- 509
Google Scholar 65.Vincent
BSmith
JD Effect of estradiol on neuronal Swedish-mutated β-amyloid precursor protein metabolism: reversal by astrocytic cells.
Biochem Biophys Res Commun. 2000;27182- 85
Google ScholarCrossref 66.Brinton
RD 17β-estradiol induction of filopodial growth in cultured hippocampal neurons within minutes of exposure.
Mol Cell Neurosci. 1993;436- 46
Google ScholarCrossref 67.Brinton
RDTran
JProffitt
PMontoya
M 17β-estradiol enhances the outgrowth and survival of neocortical neurons in culture.
Neurochem Res. 1997;221339- 1351
Google ScholarCrossref 68.Gibbs
RBHashash
AJohnson
DA Effects of estrogen on potassium-stimulated acetylcholine release in the hippocampus and overlying cortex of adult rats.
Brain Res. 1997;749143- 146
Google ScholarCrossref 69.Fader
AJHendricson
AWDohanich
GP Estrogen improves performance of reinforced T-maze alternation and prevents the amnestic effects of scopolamine administered systemically or intrahippocampally.
Neurobiol Learn Mem. 1998;69225- 240
Google ScholarCrossref 70.Luine
VNRichards
STWu
VYBeck
KD Estradiol enhances learning and memory in a spatial memory task and affects levels of monoaminergic neurotransmitters.
Horm Behav. 1998;34149- 162
Google ScholarCrossref 71.Gibbs
RB Estrogen replacement enhances acquisition of a spatial memory task and reduces deficits associated with hippocampal muscarinic receptor inhibition.
Horm Behav. 1999;36222- 233
Google ScholarCrossref 72.Newhouse
PAHancur
CKelton
MNaylor
M The effects of estrogen on anti–cholinergic-mediated cognitive impairment in post-menopausal women [abstract]. Programs and Abstracts From the 31st Annual Meeting of the Society for Neuroscience November 10-15, 2001 San Diego, Calif[book on CD-ROM] Washington, DC Society for Neuroscience2001;Program 73.13.
73.Eberling
JLWu
CHaan
MNMungas
DBuonocore
MJagust
WJ Gender differences in age-related hippocampal atrophy: the role of estrogen [abstract]. Programs and Abstracts From the 31st Annual Meeting of the Society for Neuroscience November 10-15, 2001 San Diego, Calif[book on CD-ROM] Washington, DC Society for Neuroscience2001;Program 550.2.
74.O'Malley
CAHautamaki
RDKelley
MMeyer
EM Effects of ovariectomy and estradiol benzoate on high affinity choline uptake, ACh synthesis, and release from rat cerebral cortical synaptosomes.
Brain Res. 1987;403389- 392
Google ScholarCrossref 75.Gibbs
RBPfaff
DW Effects of estrogen and fimbria/fornix transection on p75
NGFR and ChAT expression in the medial septum and diagonal band of Broca.
Exp Neurol. 1992;11623- 39
Google ScholarCrossref 76.Toran-Allerand
CDMiranda
RCBentham
WD
et al. Estrogen receptors colocalize with low-affinity nerve growth factor receptors in cholinergic neurons of the basal forebrain.
Proc Natl Acad Sci U S A. 1992;894668- 4672
Google ScholarCrossref 77.Miranda
RCSohrabji
FToran-Allerand
CD Presumptive estrogen target neurons express mRNAs for both the neurotrophins and neurotrophin receptors: a basis for potential developmental interactions of estrogen with the neurotrophins.
Mol Cell Neurosci. 1993;4510- 525
Google ScholarCrossref 78.Gibbs
RBWu
DHersh
LBPfaff
DW Effects of estrogen replacement on the relative levels of choline acetyltransferase, trkA, and nerve growth factor messenger RNAs in the basal forebrain and hippocampal formation of adult rats.
Exp Neurol. 1994;12970- 80
Google ScholarCrossref 79.McMillan
PJSinger
CADorsa
DM The effects of ovariectomy and estrogen replacement on trkA and choline acetyltransferase mRNA expression in the basal forebrain of the adult female Sprague-Dawley rat.
J Neurosci. 1996;161860- 1865
Google Scholar 80.Gibbs
RB Effects of estrogen on basal forebrain cholinergic neurons vary as a function of dose and duration of treatment.
Brain Res. 1997;75710- 16
Google ScholarCrossref 81.Gibbs
RB Levels of trkA and BDNF mRNA, but not NGF mRNA, fluctuate across the estrous cycle and increase in response to acute hormone replacement.
Brain Res. 1998;787259- 268
Google ScholarCrossref 82.Blurton-Jones
MMRoberts
JATuszynski
MH Estrogen receptor immunoreactivity in the adult primate brain: neuronal distribution and association with p75,
trkA, and choline acetyltransferase.
J Comp Neurol. 1999;405529- 542
Google ScholarCrossref 83.Gibbs
RB Effects of gonadal hormone replacement on measures of basal forebrain cholinergic function.
Neuroscience. 2000;101931- 938
Google ScholarCrossref 84.Srivastava
RAKSrivastava
NAverna
M
et al. Estrogen up-regulates apolipoprotein E (ApoE) gene expression by increasing ApoE mRNA in the translating pool via the estrogen receptor α–mediated pathway.
J Biol Chem. 1997;27233360- 33366
Google ScholarCrossref 85.Stone
DJRozovsky
IMorgan
TEAnderson
CPFinch
CE Increased synaptic sprouting in response to estrogen via an apolipoprotein E–dependent mechanism: implications for Alzheimer's disease.
J Neurosci. 1998;183180- 3185
Google Scholar 86.Teter
BHarris-White
MEFrautschy
SACole
GM Role of apolipoprotein E and estrogen in mossy fiber sprouting in hippocampal slice cultures.
Neuroscience. 1999;911009- 1016
Google ScholarCrossref 87.Mattson
MPRobinson
NGuo
Q Estrogens stabilize mitochondrial function and protect neural cells against the pro-apoptotic action of mutant presenilin-1.
Neuroreport. 1997;83817- 3821
Google ScholarCrossref 88.Bishop
JSimpkins
JW Estradiol enhances brain glucose uptake in ovariectomized rats.
Brain Res Bull. 1995;36315- 320
Google ScholarCrossref 89.Ohkura
TTeshima
YIsse
K
et al. Estrogen increases cerebral and cerebellar blood flows in postmenopausal women.
Menopause. 1995;213- 18
Google ScholarCrossref 90.Resnick
SMMaki
PMGolski
SKraut
MAZonderman
AB Effects of estrogen replacement therapy on PET cerebral blood flow and neuropsychological performance.
Horm Behav. 1998;34171- 182
Google ScholarCrossref 91.Maki
PMResnick
SM Longitudinal effects of estrogen replacement therapy on PET cerebral blood flow and cognition.
Neurobiol Aging. 2000;21373- 383
Google ScholarCrossref 92.Dubal
DBWise
PM Neuroprotective effects of estradiol in middle-aged female rats.
Endocrinology. 2001;14243- 48
Google Scholar 93.Österlund
MKHalldin
CHurd
YL Effects of chronic 17β-estradiol treatment on the serotonin 5-HT(1A) receptor mRNA and binding levels in the rat brain.
Synapse. 2000;3539- 44
Google ScholarCrossref 94.Bae
YHHwang
JYKim
YHKoh
JY Anti-oxidative neuroprotection by estrogens in mouse cortical cultures.
J Korean Med Sci. 2000;15327- 336
Google Scholar 95.Leanza
G Chronic elevation of amyloid precursor protein expression in the neocortex and hippocampus of rats with selective cholinergic lesions.
Neurosci Lett. 1998;25753- 56
Google ScholarCrossref 96.Wong
TPDebeir
TDuff
KCuello
AC Reorganization of cholinergic terminals in the cerebral cortex and hippocampus in transgenic mice carrying mutated presenilin-1 and amyloid precursor protein transgenes.
J Neurosci. 1999;192706- 2716
Google Scholar 97.Bronfman
FCMoechars
DVan Leuven
F Acetylcholinesterase-positive fiber deafferentation and cell shrinkage in the septohippocampal pathway of aged amyloid precursor protein London mutant transgenic mice.
Neurobiol Dis. 2000;7152- 168
Google ScholarCrossref 98.Granholm
AC Oestrogen and nerve growth factor: neuroprotection and repair in Alzheimer's disease.
Expert Opin Investig Drugs. 2000;9685- 694
Google ScholarCrossref 99.Frankfurt
MGould
EWoolley
CSMcEwen
BS Gonadal steroids modify dendritic spine density in ventromedial hypothalamic neurons: a Golgi study in the adult rat.
Neuroendocrinology. 1990;51530- 535
Google ScholarCrossref 100.Singh
MMeyer
EMSimpkins
JW The effect of ovariectomy and estradiol replacement on brain-derived neurotrophic factor messenger ribonucleic acid expression in cortical and hippocampal brain regions of female Sprague-Dawley rats.
Endocrinology. 1995;1362320- 2324
Google Scholar 101.Montoya
DACCarrer
HF Estrogen facilitates induction of long term potentiation in the hippocampus of awake rats.
Brain Res. 1997;778430- 438
Google ScholarCrossref 102.Kar
SSeto
DGaudreau
PQuirion
R β-Amyloid-related peptides inhibit potassium-evoked acetylcholine release from rat hippocampal slices.
J Neurosci. 1996;161034- 1040
Google Scholar 103.Toran-Allerand
CDSingh
MSétáló
G
Jr Novel mechanisms of estrogen action in the brain: new players in an old story.
Front Neuroendocrinol. 1999;2097- 121
Google ScholarCrossref 104.Slooter
AJBronzova
JWitteman
JCVan Broeckhoven
CHofman
Avan Duijn
CM Estrogen use and early onset Alzheimer's disease: a population-based study.
J Neurol Neurosurg Psychiatry. 1999;67779- 781
Google ScholarCrossref 105.Lendon
CLLambert
JC Polymorphisms associated with risk for Alzheimer's disease modulate the estrogen induced expression of the apolipoprotein E gene: possible implications for hormone replacement therapy [abstract]. Programs and Abstracts From the 31st Annual Meeting of the Society for Neuroscience November 10-15, 2001 San Diego, Calif Washington, DC Society for Neuroscience2001;Program 192.2.
106.Meda
CVegeto
EPollio
G
et al. Oestrogen prevention of neural cell death correlates with decreased expression of mRNA for the pro-apoptotic protein nip-2.
J Neuroendocrinol. 2000;121051- 1059
Google ScholarCrossref 107.Kalaria
RNHarik
SI Abnormalities of the glucose transporter at the blood-brain barrier and in brain in Alzheimer's disease.
Prog Clin Biol Res. 1989;317415- 421
Google Scholar 108.Heyman
AWilkinson
WEStafford
JAHelms
MJSigmon
AHWeinberg
T Alzheimer's disease: a study of epidemiological aspects.
Ann Neurol. 1984;15335- 341
Google ScholarCrossref 109.Amaducci
LAFratiglioni
LRocca
WA
et al. Risk factors for clinically diagnosed Alzheimer's disease: a case-control study of an Italian population.
Neurology. 1986;36922- 931
Google ScholarCrossref 110.Broe
GAHenderson
ASCreasey
H
et al. A case-control study of Alzheimer's disease in Australia.
Neurology. 1990;401698- 1707
Google ScholarCrossref 111.Graves
ABWhite
EKoepsell
TD
et al. A case-control study of Alzheimer's disease.
Ann Neurol. 1990;28766- 774
Google ScholarCrossref 112.Brenner
DEKukull
WAStergachis
A
et al. Postmenopausal estrogen replacement therapy and the risk of Alzheimer's disease: a population-based case-control study.
Am J Epidemiol. 1994;140262- 267
Google Scholar 113.Henderson
VWPaganini-Hill
AEmanuel
CKDunn
MEBuckwalter
JG Estrogen replacement therapy in older women: comparisons between Alzheimer's disease cases and nondemented control subjects.
Arch Neurol. 1994;51896- 900
Google ScholarCrossref 114.Mortel
KFMeyer
JS Lack of postmenopausal estrogen replacement therapy and the risk of dementia.
J Neuropsychiatry Clin Neurosci. 1995;7334- 337
Google Scholar 115.Kawas
CResnick
SMorrison
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;481517- 1521
Google ScholarCrossref 116.Yaffe
KSawaya
GLieberburg
IGrady
D Estrogen therapy in postmenopausal women: effects on cognitive function and dementia.
JAMA. 1998;279688- 695
Google ScholarCrossref 117.Costa
MMReus
VIWolkowitz
OMManfredi
FLieberman
M Estrogen replacement therapy and cognitive decline in memory-impaired post-menopausal women.
Biol Psychiatry. 1999;46182- 188
Google ScholarCrossref 118.Doraiswamy
PMBieber
FKaiser
LKrishnan
KRReuning-Scherer
JGulanski
B The Alzheimer's Disease Assessment Scale: patterns and predictors of baseline cognitive performance in multicenter Alzheimer's disease trials.
Neurology. 1997;481511- 1517
Google ScholarCrossref 119.Schneider
LSFarlow
MRHenderson
VWPogoda
JM Effects of estrogen replacement therapy on response to tacrine in patients with Alzheimer's disease.
Neurology. 1996;461580- 1584
Google ScholarCrossref 120.Henderson
VWPaganini-Hill
AMiller
BL
et al. Estrogen for Alzheimer's disease in women: randomized, double-blind, placebo-controlled trial.
Neurology. 2000;54295- 301
Google ScholarCrossref 121.Wang
PNLiao
SQLiu
RS
et al. Effects of estrogen on cognition, mood, and cerebral blood flow in AD: a controlled study.
Neurology. 2000;542061- 2066
Google ScholarCrossref 122.Gibbs
RB Long-term treatment with estrogen and progesterone enhances acquisition of a spatial memory task by ovariectomized aged rats.
Neurobiol Aging. 2000;21107- 116
Google ScholarCrossref 124.Shumaker
SAReboussin
BAEspeland
MA
et al. The Women's Health Initiative Memory Study (WHIMS): a trial of the effect of estrogen therapy in preventing and slowing the progression of dementia.
Control Clin Trials. 1998;19604- 621
Google ScholarCrossref