Abstract
Clinical studies indicate that Alzheimer’s disease (AD) disproportionately affects women in both disease prevalence and rate of symptom progression, but the mechanisms underlying this sexual divergence are unknown. Although some have suggested this difference in risk is a reflection of the known differences in longevity between men and women, mounting clinical and preclinical evidence supports women also having intrinsic susceptibilities toward the disease. Although a number of potential risk factors have been hypothesized to mediate these differences, none have been definitively verified. In this review, we first summarize the epidemiologic studies of prevalence and incidence of AD among the sexes. Next, we discuss the most likely risk factors to date that interact with biological sex, including (1) genetic factors, (2) sex hormones (3) deviations in brain structure, (4) inflammation and microglia, and (5) and psychosocial stress responses. Overall, though differences in life span are likely to account for part of the divide between the sexes in AD prevalence, the abundance of preclinical and clinical evidence presented here suggests an increase in intrinsic AD risk for women. Therefore, future studies focusing on the underlying biological mechanisms for this phenomenon are needed to better understand AD pathogenesis in both sexes, with the eventual goal of sex-specific prevention and treatment strategies.
Keywords: Sex difference, Cognition, APOE, Brain structure, Hormones, Stress, Alzheimer’s disease
1. Introduction
Alzheimer’s disease (AD) is a progressive neurodegenerative disease that affects 5.4 million Americans aged 65 or older and is the fifth leading cause of death in the US. Of these 5.4 million patients, 3.3 million (nearly 70%) were women (Alzheimer’s Association, 2015, Hebert et al., 2013), but the biological basis of the sex-based differences in AD onset and progression remain elusive (Hua et al., 2010).
While a definitive cause has yet to be proven, scientists and physicians have developed both epidemiologic and biological explanations for this apparent increase in risk (Lin and Doraiswamy, 2014; Mielke et al., 2014; Riedel et al., 2016). Clinicians and researchers have noted that in most populations around the world, women tend to outlive men so that the gap between the number of men and women in a population widens with advancing age (Brookmeyer et al., 1998; Hebert et al., 2001; Plassman et al., 2007; Seshadri et al., 1997). Although prevalence and incidence are rates, and so the frequency of women or men would normally not impact these measures, advancing age is the strongest risk factor for AD, and so the increased frequency of women at the highest age groups increases the likelihood of AD being diagnosed. Thus, the greater frequency of women of the oldest ages also impacts women’s risk profiles, since AD diagnoses double roughly every 5 years (Kukull et al., 2002). Combined with hypotheses integrating the improved cardiovascular health profiles of men who live to greatly advanced ages (i.e., 85 years or older) (Carmelli et al., 1998), many experts have concluded that the apparent differences in AD prevalence between men and women are due to the indirect consequences of greater female longevity.
While this view still remains widely held, many recent studies have demonstrated that there are age- and risk factor-matched increases in the incidence of developing AD in women, though previous results have been inconsistent (Bachman et al., 1993; Edland et al., 2002; Fiest et al., 2016; Hebert et al., 2001; Rocca et al., 1998). These recent clinical (human) and preclinical studies (animal models) have led to some new hypotheses predicting a female-specific increase in AD risk based on intrinsic biological differences, including sex-specific genetic interactions (Altmann et al., 2014; Janicki et al., 2014; Ungar et al., 2014), hormones and associated endocrinological changes with age (Morrison et al., 2006; Rocca et al., 2011), sexual dimorphism in brain structures (Elbejjani et al., 2015; Mielke et al., 2014; Sampedro et al., 2015), heightened inflammatory responses in women (Hanamsagar and Bilbo, 2016), and sex by stress interactions (Bangasser et al., 2017). Still, the collection of evidence necessary to validate one or more of these hypotheses is in its infancy, and so the perspectives of individual experts on this subject remain varied.
In this review, we summarize and evaluate both the epidemiologic and biological evidence explaining the sex difference in AD risk to better inform both researchers and clinicians on the issue. After reviewing the available evidence, we believe this review will demonstrate that while some of the sex differences observed in AD prevalence are due to differences in longevity, it is very plausible that other distinct biological mechanisms increase the risk and disease progression of AD in women. We hope to present the most recent data shaping expert perspectives, inspiring researchers to go in new directions, and driving development of future diagnostic tools and treatments for AD.
2. Epidemiologic perspective
Age remains the greatest risk factor for AD, and the prevalence of AD in populations aged 85 years and older is vastly increased when compared to other age ranges (Riedel et al., 2016), with multiple studies showing that the incidence of AD rises exponentially after the sixth decade of life (Kukull et al., 2002; Masters et al., 2015). Similar to this increase in disease diagnoses, there are more women aged 85 years or older than men in most global subpopulations (Fratiglioni et al., 2000), meaning there are more women at greatest risk based on age. In addition, women outlive men by an average of 4.5 years (World Population Ageing, 2015). Therefore, while the prevalence of AD is often reported to be higher for women compared to men due to the increase in risk by greater age, the relative increase in women living to more advanced ages makes it difficult to assess whether women also have an increased aged-matched risk of the disease (Brookmeyer et al., 1998; Hebert et al., 2001; Plassman et al., 2007; Seshadri et al., 1997).
Reflecting this, the literature examining incidence has also yielded conflicting results, as some studies report higher age-adjusted numbers for women (Irvine et al., 2012; Seshadri et al., 1997). In addition, some evidence suggests that disease progression is accelerated in women, such as imaging studies indicating that women with mild cognitive impairment (MCI) and AD show greater memory impairment compared to males with the same diagnoses (Gale et al., 2016; Lin et al., 2015). Still, other studies fail to find an association between incidence, progression, and sex (Bachman et al., 1993; Edland et al., 2002; Hebert et al., 2001; Rocca et al., 1998) or remain inconclusive (Fiest et al., 2016).
The inconsistent findings between studies may be explained by many factors, such as (1) type of study (i.e., prospective cohort, retrospective cohort, cross-sectional analysis), (2) cultural differences affecting diet, exercise, and stress response strategies, (3) differences in inclusion/exclusion criteria, especially for comorbid diseases, (4) sample size and statistical power, and (5) differing methods of classifying and confirming AD diagnoses. This last factor is of particular importance as many different criteria have been used across studies. According to the NINCDS-ADRDA diagnostic criteria, which a majority of current AD studies use, a definitive diagnosis of AD can only be made postmortem while probable AD is routinely diagnosed clinically through the presentation of certain symptoms, such as memory loss, and the exclusion of other possible diagnoses, including vascular dementia, other neurodegenerative diseases, other neuropsychiatric diseases, or acute causes of dementia such as traumatic injury or stroke (Trojanowski and Revesz, 2007). While the diagnoses of AD are verified with autopsies in some studies (Gustafson, 1987; Matyi et al., 2017; Miech et al., 2002; Zandi et al., 2002), many others (Barnes et al., 2005, Hebert et al., 2001, Galasko et al., 1998) use a clinical diagnosis, which may categorize other dementia presentations incorrectly as being AD. In addition, the pre-AD diagnosis of MCI can complicate the timing of when the later diagnosis of AD is made, making the process of defining the age-of-onset for AD more variable. Furthermore, aging women without dementia and with early MCI and sex-matched quantities of Aβ seem to have better verbal recall memory than their respective age-matched male counterparts, and as the diagnosis of later stage MCI and AD are partially dependent on this cognitive process, women receive the diagnosis of AD later than men and may not be classified as having MCI as readily; (Sundermann et al., 2016, 2017). Finally, one would think that a higher age-specific incidence of AD dementia among women would translate to a higher incidence of MCI and subclinical AD. Yet, some data have found the opposite (Roberts et al., 2012), and although this may be explained by the increased verbal memory reserve in women, possibly leading to later MCI diagnoses (Sundermann et al., 2016, 2017), this paradox deserves careful investigation.
The culmination of these classification difficulties is apparent in a study looking at subjects from 2 longitudinal assessments on community dwelling elderly patients by Schneider et al. (Schneider et al., 2009). When comparing autopsy results of 483 participants with a diagnosis of probable AD, MCI, or no cognitive impairment, they found that only 41.8% of patients had pathology consistent with an AD-only diagnosis. Of the remaining patients, 45.8% had mixed pathology with AD and vascular dementia or FTD-like findings, 6.2% were misclassified, and 6.2% had no pathologic markers of any of the studied dementias. Likewise, 29.4% of patients without cognitive impairment had signs of AD pathology, 8.8% had signs of mixed pathology with AD, 16.5% had infarcts or Lewy bodies, whereas only 45.3% had no significant pathologic findings (Schneider et al., 2009). The findings in the cognitively unimpaired patients are generally concerning for our understanding of AD pathogenesis, but the results suggesting that a large portion of clinical diagnoses are not corroborated by pathology makes quantifying the actual incidence and prevalence of AD even more challenging. It is also interesting to note that the most often-cited study reporting statistically significant differences in incidence between men and women, the Cache County Memory Study, verified many of their probable AD diagnoses with autopsy (Matyi et al., 2017, Miech et al., 2002, Zandi et al., 2002).
Further supporting the classification bias as an issue in determining the real incidence of AD in men and women, many studies have shown that vascular dementia, as well as Lewy body dementia, is more common in men (Ferencz and Gerritsen, 2015, Whitmer et al., 2005), and because of the high occurrence of mixed pathology in clinical AD diagnoses, it could be suggested that more men with dementia are being incorrectly classified as having AD as the precipitating factor for their cognitive decline, thus overestimating the total population of men with AD, especially at younger ages where the sex difference in prevalence is not as apparent. Indeed, it has been suggested that one aspect of the enlarged gap in AD prevalence between sexes in patients aged 85 years and older is because men with vascular complications tend to die earlier, so that the remaining healthy males have reduced cardiovascular risk factors for AD (Whitmer et al., 2005). In addition, it has been reported that men have better overall health at older ages than their female counterparts in terms of morbidity but not mortality (Graves et al., 2006). Although these differences have been shown reproducibly, this argument for improved male health with older age is a double-edged sword, as it becomes unclear if AD diagnoses are overestimated in younger men due to increasing adverse cardiovascular events and possible misclassification of the resultant dementia.
For all these stated reasons, it is unsurprising that there remain large differences in the reported incidence of AD between men and women across studies. Perhaps, the single most pertinent example of this discrepancy was found in a recent meta-analysis, where inclusion of 22 studies on sex differences revealed that “all estimates of incidence and prevalence were higher for females compared to males, though the differences were not statistically different” (Fiest et al., 2016). Still, the relative narrowing of the incidence rates compared to prevalence in studies that have found a difference, such as the Cache County Memory Study, suggest that longevity remains an important reason why there are more women with AD than men. The complications in estimating incidence and the increasing body of clinical and preclinical evidence supporting sex-specific biological mechanisms in diverging AD risk remain an important adjunct explanation to the epidemiologic perspective and should be investigated more closely in future studies.
Table 1 summarizes most representative studies investigating sex differences in humans collected from 2000 to date.
Table 1.
Article | Author (s) | Year | Journal | Control |
MIC |
AD |
Study design | Length of follow-up or study period | Diagnostic criteria for AD | Main result/conclusion | |||
---|---|---|---|---|---|---|---|---|---|---|---|---|---|
M | F | M | F | M | F | ||||||||
Sex Differences in Risk for Alzheimer’s Disease Related to Neurotrophin Gene Polymorphisms: The Cache County Memory Study. | Matyi et al. | 2017 | J Gerontology: Biological Sciences | 1476 | 2023 | Prospective | 12 y | NINCDS-ADRDA | Female carriers of the minor T allele for rs2072446 and rs56164415 had a 60% and 93% higher hazard for AD, respectively, than male noncarriers of the T allele. Furthermore, male carriers of the T allele of rs2072446 had a 61% lower hazard than male noncarriers at trend-level significance. | ||||
Female-specific effect of the BDNF gene on Alzheimer’s disease. | Li et al. | 2017 | Neurobiology of Aging | 113/121 (A/G) | 262/314 (A/G) | 164/182 (A/G) | 208/188 (A/G) | Association Studies and meta-analysis | Variable | NINCDS-ADRDA | Genetic association between BDNF and AD in females but not in males. BDNF mRNA was significantly decreased in brain tissues of AD patients, especially in females. Female-specific effect of BDNF SNP rs6265 on AD endophenotypes | ||
Apolipoprotein E Genotype and Sex Risk Factors for Alzheimer Disease | Neu et al. | 2017 | JAMA Neurology | Variable depending on comparison group (31,430 total analyzed patients of 57,979 potential patients identified) | Meta-analysis | Variable | Not stated | Women heterozygous for Women heterozygous for ApoE-ε4 had increased risk of AD from ages 65 e75 year old | |||||
Analysis of the MIRIAD Data Shows Sex Differences in Hippocampal Atrophy Progression | Ardekani et al. | 2016 | J Alzheimer’s | 18 | 25 | Prospective Cohort | 1y | NINCDS-ADRDA | In patients with probable AD, the hippocampus atrophies at a significantly faster rate in women compared to men. | ||||
Sex differences in the association between AD biomarkers and cognitive decline | Koran et al. | 2017 | Brain Imaging Behav. | 164 | 184 | 327 | 238 | 100 | 85 | Prospective Cohort | 2.5 y | NINCDS-ADRDA | Women with Ab-42 and total tau levels indicative of worse pathologic changes showed more rapid hippocampal atrophy and cognitive decline |
Sex-specific patterns and differences in dementia and Alzheimer’s disease using informatics approaches | Ronquillo et al. | 2016 | J Women & aging | 4110 | 4028 | 7423 | 8709 | Retrospective Analysis of 3 longitudinal studies (ADNI, CAMD, NACC) | Variable | NINCDS-ADRDA | Females were 1.5 times more likely than males to have a documented diagnosis of probable Alzheimer’s disease | ||
Impact of sex and ApoE 4 on cerebral amyloid angiopathy in Alzheimer’s disease | Shinohara et al. | 2016 | Acta Neuropathologica | 68 | 103 | 114 | 143 | Postmortem pathologic assessment | N/A | NINCDS-ADRDA; Definitive AD only | The associations between APOE4 and average cerebral amyloid angiopathy score were statistically significant only in females but not males. | ||
Gender differences in white matter pathology and mitochondrial dysfunction in Alzheimer’s disease with cerebrovascular disease | Gallart-Palau et al. | 2016 | Mol Brain | 7 | 10 | Postmortem Proteomic Study | N/A | NINCDS-ADRDA; Definitive AD only | Hypercitrullination and hyperdeamidation of MBP were prevalent in female patients with AD. | ||||
Greater memory impairment in dementing females than males relative to sex-matched healthy controls. | Gale et al. | 2016 | J Clin Exp Neuropsychol. | 64 | 113 | 46 | 30 | 63 | 38 | Cross-Sectional Analysis | N/A | Clinical Dementia Rating Scores | Performance gap between healthy and dementing women was consistently larger than the gap between healthy and dementing men for both MCI and AD groups. |
Sex Differences in Neuropsychiatric Symptoms of Alzheimer’s Disease: The Modifying Effect of Apolipoprotein E ε4 Status | Xing et al. | 2015 | Behav Neurol. | 74 | 84 | 65 | 92 | Cross-Sectional Analysis of AD patients | N/A | NINCDS-ADRDA | Female patients with at least 1 copy of the APOE- ε4 allele were significantly more likely to have some neuropsychiatric symptoms in moderate to severe AD, such as disinhibition and irritability. | ||
Gender differences in risk factors for transition from mild cognitive impairment to Alzheimer’s disease: A CREDOS study | Ferencz and Gerritsen | 2015 | Comprehensive Psychiatry | 101 | 193 | Prospective Cohort | 3 y | NINCDS-ADRDA | In men, the significant risk factors for transition from MCI to AD were severe periventricular white matter hyperintensities and poorer global cognitive function. In women, risk factors were older age, clinically significant depressive symptoms at baseline, and positive APOE ε4 alleles. | ||||
Marked Gender differences in progression of mild cognitive impairment over 8 y. | Lin et al. | 2015 | Alzheimers Dement (NY) | 257 | 141 | Prospective Cohort | 8 y | N/A; MCI diagnostic criteria | Women with MCI had greater longitudinal rates of cognitive and functional decline than men. | ||||
APOE-by-sex interactions on brain structure and metabolism in healthy elderly controls. | Sampedro et al. | 2015 | Oncotarget | 78 (MRI) 166 (FED- PET) 136 (CSF) | 90 (MRI) 162 (FED- PET) 138 (CSF) | Cross-Sectional Analysis | N/A | N/A: Healthy non- demented patients | Female APOE-ε4 carriers show greater brain hypometabolism and atrophy than male carriers. | ||||
Sex modifies the APOE- related risk of developing Alzheimer disease | Altmann et al. | 2014 | Ann Neurol. | 1844 | 3652 | 1331 | 1275 | Retrospective Cohort | 8 y | NINCDS-ADRDA | APOE-ε4 confered greater AD risk in women. Biomarker results suggest that increased ApoE- related risk in women may be associated with tau pathology. | ||
Higher Rates of Decline for Women and Apolipoprotein E ε4 Carriers | Holland et al. | 2013 | American Journal of Neuroradiology | ε4– 70 ε4þ 26 | ε4– 67 ε4þ 25 | ε4– 74 ε4þ 102 | ε4– 42 ε4þ 55 | ε4– 36 ε4þ 87 | ε4– 25 ε4þ 67 | Prospective Cohort | 3 y | NINCDS-ADRDA | APOE-s4 significantly accelerated rates of the brain regional specific atrophy abd clinical decline, and women in all cohorts had higher rates of decline than men. |
Gender modulates the APOE epsilon4 effect in healthy older adults: convergent evidence from functional brain connectivity and spinal fluid tau levels | Damoiseaux et al. | 2012 | J Neurosci. | ε3þ 66 ε4þ 33 | ε3þ 85 ε4þ 26 | Cross-Sectional Analysis | N/A | N/A; Healthy non- demented patients | Higher prevalence of the APOEE-ε4 allele among women with AD and interaction between ApoE genotype and gender was detectable in the preclinical period | ||||
Brain levels of sex steroid hormones in men and women during normal aging and in Alzheimer’s disease | Rosario et al. | 2011 | Neurobiology of Aging | 17 | 12 | 17 | 33 | 32 | Postmortem pathologic assessment | N/A | NINCDS-ADRDA; | Within the defined age ranges, AD in women is associated with low estrogens, whereas AD in men is associated with low androgens. | |
Sex and age differences in atrophic rates: an ADNI study with n¼1368 MRI scans. | Hua et al. | 2010 | Neurobiology of Aging | 202 | 338 | 144 | Prospective Cohort | 1 y | NINCDS-ADRDA | Brain atrophy rates were faster in women than men. | |||
Sexually dimorphic effect of the Val66Met polymorphism of BDNF on susceptibility to Alzheimer’s disease | Fukumoto et al. | 2010 | American Journal of Medical Genetics | 220 | 305 | 230 | 427 | Case-Control study and meta-analysis | N/A | NINCDS-ADRDA | Significant allelic association between the Val66Met polymorphism of BDNF and AD in Japenese women but not in man | ||
Estrogen receptor beta gene variants are associated with increased risk of Alzheimer’s disease in women | Pirskanen et al. | 2005 | European Journal of Human Genetics | 185 | 282 | 116 | 271 | Case-Control | N/A | NINCDS-ADRDA | Women carrying 2 T alleles were over 3 times more prone to develop AD when compared to subjects with no T alleles in the SNP2 or SNP3 loci of the ESR2 but not in man | ||
Sex differences in the clinical manifestations of Alzheimer disease pthology | Barnes et al. | 2005 | Archives of General Psychiatry | 28 | 25 | 13 | 18 | 23 | 34 | Prospective Cohort with pathological follow-up | Variable (yearly follow-up until death) | NINCDS-ADRDA | AD pathology was more likely to be clinically expressed as dementia in women than in men. |
Sex, apolipoprotein E epsilon 4 status, and hippocampal volume in mild cognitive impairment. | Fleisher et al. | 2005 | Arch Neurol. | 107 | 86 | Cross-sectional Analysis | N/A | N/A; MCI diagnostic criteria | The APOE-ε4 genotype status had a greater deleterious effect on gross hippocampal pathology and memory performance in women than in men. | ||||
Incidence of AD may decline in the early 90s for men, later for women: The Cache County study | Miech et al. | 2002 | Neurology | 1384 | 1924 | 62 | 123 | Prospective Cohort | 3 y | NINCDS-ADRDA with 83.3% PPV of consensus AD diagnosis by autopsy | A statistical interaction of sex and age indicated greater incidence of AD in women than in men after age 85 y. | ||
Hormone Replacement Therapy and Incidence of Alzheimer Disease in Older Women | Zandi et al. | 2002 | JAMA | 1352 | 1901 | 35 | 88 | Prospective Cohort | 3 y | NINCDS-ADRDA | AD risk was more common in women than men but less common in women with previous hormone replacement therapy use compared to those without | ||
Increased glucocorticoid production and altered cortisol metabolism in women with mild to moderate Alzheimer’s disease | Rasmuson et al. | 2001 | Biol Psychiatry | 7 | 10 | Case-Control | N/A | NINCDS-ADRDA | Increased daily glucocorticoid production and altered cortisol metabolism in women in the early stages of AD | ||||
A gender difference in the association between APOE genotype and age-related cognitive decline | Mortensen and Hogh | 2001 | Neurology | 79 | 84 | Prospectivy Cohort | 30 y | N/A; Healthy non- demented patients | The APOE-ε4 allele is associated with normal age-related decline in cognitive functions in women only. | ||||
The Prevalence and Incidence of Dementia Due to Alzheimer’s Disease: a Systematic Review and Meta- Analysis | Fiest et al. | 2016 | Canadian Journal of Neurological Sciences | Not described for meta-regression by sex | Meta-analysis and Systemic Review | Variable | Predominantly NINCDS-ADRDA | Meta-regression of 22 studies determining incidence or prevalence of AD found that “all estimates of incidence and prevalence were higher for females compared to males, though the differences were not statistically different.” | |||||
Gender, cognitive decline, and risk of AD in older persons | Barnes et al. | 2005 | Neurology | 271 | 577 | Prospective Cohort | 8y | NINCDS-ADRDA | The results suggest that patterns of cognitive decline and incidence of AD are similar in older men and women. | ||||
Is the risk of developing Alzheimer’s disease greater for women than for men? | Hebert et al. | 2001 | Am J Epidemiol. | 280 | 362 | 28 | 57 | Prospetive Cohort | 11 y | NINCDS-ADRDA | These findings suggest that the excess number of women with AD is due to the longer life expectancy of women rather than sex-specific risk factors for the disease. | ||
Gender and age modify the association between APOE and AD-related neuropathology | Ghebremedhin et al. | 2001 | Neurology | 359 | 370 | Postmortem pathologic assessment | N/A | N/A; no clinical assessment | An association between the ε4 allele and senile plaques for women was found at ages 60e79 y (p < 0.0001) but not at ::80 y of age (p ¼ 0.063). By comparison, men showed an association in both age categories (p ¼ 0.001 and p ¼ 0.001). |
The articles in black color indicate studies supporting women more vulunable to neuropathgenesis of AD and those in shaded region oppose those findings.
Key: AD, Alzheimer’s Disease
3. Biological perspective
It is interesting to note that women tend to develop diseases associated with senescence--defined as reduced function due to aging--at earlier ages than men, especially for diseases that are notorious for resulting in increased morbidity but rarely lead directly to mortality (Graves et al., 2006). In addition, women consistently have a higher age-specific prevalence of disability globally (Buttery et al., 2016; Crimmins et al., 2011; Freedman et al., 2016; Jagger et al., 2016; Wahrendorf et al., 2013; Whiteford et al., 2015). It is therefore very likely that similar trajectories are present for the development of cognitive deficits due to diseases with low- or late-stage mortality. The potential for a real increase in AD incidence among women may be part of this trend toward rapid senescence and disability, and many of the biological mechanisms proposed to explain this global, age-related reduction in functionality also extend to the development of AD. In the following sections, we review some of the prevailing biological mechanisms that may explain the increased incidence of AD in women reported in some studies, drawing on both clinical and preclinical studies from the literature.
3.1. Sex and genetics interaction in AD: focus on ApoE and BDNF
3.1.1. ApoE
As more genomic studies uncover risk genes for various diseases, there have been more instances of differing risk profiles for a particular genomic variant between sexes (Gatz et al., 2006; Riedel et al., 2016). In line with this, recent epidemiologic studies demonstrated that the APOE allele confers different AD risk profiles based on sex. These studies indicate that women with APOE-ε4 are at greater risk for developing AD than age-matched men, especially when comparing heterozygous individuals carrying the APOE-ε4 allele (Altmann et al., 2014; Damoiseaux et al., 2012). In a large meta-analysis (Farrer et al., 1997) and in a large population study of AD (Bremner, 1999), women who were heterozygous for the APOE-ε4 allele had AD diagnosed 5 years earlier than heterozygous men (Poirier et al., 1993). Similarly, the odds ratio for AD in women with one copy of the APOE-ε4 allele is 4-fold greater than in men (Altmann et al., 2014; Farrer et al., 1997; Payami et al., 1994). In addition, women carrying ε3/ε4 often show faster age-related decline and greater cognitive deterioration than age-matched ε3/ ε4 men (Beydoun et al., 2012; Farrer et al., 1997). However, one study found that the association between the APOE genotype and cognitive decline is significant only in women aged greater than 70 years (Mortensen and Hogh, 2001), whereas a recent meta-analysis suggested women with 1 copy of APOE-ε4 only had higher risk in the age range of 65–75 years (Neu et al., 2017), and therefore, more research is needed to determine the impact of this sex-by-gene interaction.
In support of these clinical findings, preclinical investigations of AD mouse models have revealed similar sex-by-APOE interactions for AD-like pathology and cognitive deficits. Notably, 1 study demonstrated that female 3xTg-AD mice with neural expression of the human APOE-ε4 showed increased spatial memory deficits compared to APOE-ε4 male mice or APOE-ε3 males and females starting at 6 months of age (Raber et al., 1998).
Paradoxically, men homozygous for APOE-ε4 are reported to be at greater risk for MCI and AD in some studies than homozygous women, with men in these studies demonstrating a shift to lower episodic memory scores (Farrer et al., 1997; Lehmann et al., 2006). In conjunction, one report noted that men who are APOE-ε4 homozygotes with MCI have smaller hippocampal volumes than homozygous women (Fleisher et al., 2005). These discrepancies between sexes for AD risk in the homozygous and heterozygous states complicate the interpretation of the sex-by-gene interactions concerning the APOE-ε4 allele.
APOE’s influence on AD pathogenesis is multifaceted and likely includes regulation of Aβ clearance, tau phosphorylation, cerebrovascular permeability, blood-brain barrier integrity, inflammatory responses, and synaptic maintenance (Bu, 2009, Tai et al., 2016). It is tempting to speculate that the heterozygotes may suffer more from abnormal Aβ production and clearance, whereas the homozygotes have increased dementia pathogenesis from impaired cardiovascular function (Liu et al., 2013) and, thus, may affect men and women differently. However, a more detailed understanding of APOE’s role in AD pathogenesis is essential to discovering why such divergent risk profiles by sex are apparent in APOE-ε4 carriers.
3.1.2. BDNF
The neurotrophin BDNF is an important factor controlling synaptic plasticity in the CNS (Kowian’ski et al., 2017), and many neuropsychiatric diseases have demonstrated changes in BDNF in their pathogenesis or treatment, especially mood disorders (Yeh et al., 2015). Recently, certain BDNF polymorphisms in humans have been shown to affect the risk of AD development and cognitive decline (Li et al., 2017a,b, Chen et al., 2014; Fukumoto et al., 2010; Lim et al., 2015; Matyi et al., 2017), and AD patients have been reported to have decreased BDNF in postmortem brain samples (Li et al., 2017a,b) and peripherally in blood (Qin et al., 2017). Similar to APOE, many of these association studies discovered increased risk of AD for women over men. For instance, the BDNF Val66Met polymorphism has been shown to increase AD risk only in women but not men (Chen et al., 2014; Fukumoto et al., 2010; Li et al., 2017a,b), whereas similar findings of increased risk in only women were found for 2 other SNPs, rs2072446 and rs56164415 (Matyi et al., 2017). In addition, one of these studies reported that while BDNF was decreased in many cortical areas in both sexes, only females demonstrated BDNF downregulation in the entorhinal cortex, a main input and output area of the hippocampus (Li et al., 2017a,b). Preclinically, it is interesting to note that in a prominent AD mouse model, APPsw/PS1dE9, female mice had higher levels of soluble Aβ and mature BDNF (mBDNF) at baseline but saw a precipitous drop in mBDNF and the mBDNF/proBDNF ratio with stress. In contrast, the male counterparts had an increase in mBDNF with chronic stress, suggesting that female mice may suffer more from a stress-by-genotype interaction in terms of BDNF loss than males.
In addition, both genetic and biochemical studies have demonstrated a link between APOE and BDNF in AD pathogenesis (Lim et al., 2015; Sen et al., 2015, 2017; Ward et al., 2014). Multiple studies have suggested that in the presence of Aβ, APOE-ε4, and BDNF Val66Met worsen cognitive decline more than either allele in isolation, especially for episodic memory (Lim et al., 2015; Ward et al., 2014). Mechanistically, it has been suggested that APOE directly regulates BDNF expression and secretion (Sen et al., 2015, 2017). It has also been shown that the APOE-ε4 causes HDAC4 and HDAC6 to translocate into the nucleus, where these deacetylases reduce BDNF expression through transcriptional repression (Sen et al., 2015), leading to decreased BDNF creation and secretion (Sen et al., 2017). In contrast, APOE-ε3 has little or no effect on BDNF levels (Sen et al., 2015). Given the increasing evidence that APOE status can affect BDNF and that certain APOE and BDNF alleles may predispose women to develop AD more easily than men, a sexually divergent interaction between these AD effectors is quite possible. In general, it is also interesting to note that estrogen can influence the expression of BDNF and that many of the pathways supported by BDNF signaling are also influenced by estrogen (Scharfman and MacLusky, 2006). However, whether estrogen plays a mechanistic role in the cross-talk between APOE and BDNF remains to be seen.
While evidence demonstrating BDNF’s role in AD pathogenesis continues to build, it remains unclear how BDNF alone or combined with APOE influences AD. As BDNF is a dynamic regulator of neuronal survival and synaptic plasticity (Kowianski et al., 2017), a simple hypothesis may suggest that loss of BDNF makes neurons more susceptible to AD-related toxicity. In addition, it is possible that BDNF directly acts on AD-causing intermediates, as BDNF has been shown to reduce Aβ−42 in primary neurons from AD mice through the SORLA-MEK/ERK pathway (Rohe et al., 2009), another pathway regulated by APOE (Bu, 2009). Although progress has been made, future investigations will be needed to define this mechanism more clearly. Because of the ample evidence suggesting greater AD susceptibility for women with BDNF mutations, investigating sex differences should be an important part of these investigations.
3.2. Sex-specific brain structure changes and biomarkers in AD
One explanation for the sexual dimorphism in AD risk and severity centers on the divergent changes in brain structures that men and women have in response to disease or disease-causing insults. It is posited that women may be more sensitive to pathologic agents for AD and experience more rapid structural loss when compared to their male counterparts, who are posited to have a larger cognitive reserve based on increased brain volume (Mielke et al., 2014). Many examples of this are apparent in the literature.
As part of the Alzheimer’s Disease Neuroimaging Initiative, a study examined 1-year atrophy rates using 3D-tensorebased MRI morphometry in 1368 MRI scans (144 subjects with AD, 338 subjects with MCI, and 202 controls), it was discovered that annual atrophy rates were faster in women (Hua et al., 2010). A follow-up study further showed that women with MCI had greater atrophy in numerous brain regions and a steeper decline in certain cognitive tasks compared to men, as well as greater atrophy in numerous regions once an AD diagnosis had been made (Holland et al., 2013). In the Minimal Interval Resonance Imaging in AD study, women with probable AD demonstrated significantly faster rates of hippocampal atrophy than their male counterparts (Ardekani et al., 2016). On a molecular level, another study used discovery driven quantitative proteomics to detect modulation of several redox proteins in the temporal lobe of patients with combined AD and cerebrovascular disease (AD + CVD subjects) and observed sex-specific alterations in the white matter and mitochondrial proteomes in women. Female patients also displayed downregulation of certain ATP synthase and cytochrome oxidase subunits, suggesting increased severity of mitochondrial impairment in women with AD + CVD (Gallart-Palau et al., 2016). These studies suggest that when degeneration begins, it may affect women more rapidly than men, resulting in the observed increase in pathophysiology.
In concert, a clinicopathological study observed a large sex difference in the association of AD pathology with the odds of AD dementia in men and women. Specifically, it was reported that each additional unit of AD pathology was associated with a nearly 3-fold increase in the odds of AD dementia in men compared with a more than 22-fold increase in the odds of AD dementia in women (Barnes et al., 2005), again suggesting that women might possibly be more sensitive to the pathologic agents that cause AD. In agreement, a recent study reported that women with higher CSF Aβ−42 and total tau levels, indicative of worse pathologic changes, showed more rapid hippocampal atrophy and cognitive decline than men, furthering the notion that divergent responses in structural integrity to pathologic insults are based on sex in AD (Koran et al., 2017).
Although the current studies seem to be in agreement in regard to divergent structural responses to pro-AD agents, it is difficult to conclude whether the observed sex difference is driven by differences in the etiology and pathophysiology of the AD cascade or the late-life risk factors of poor brain aging. Furthermore, although these structural changes are readily observed, the underlying molecular mechanism leading to this divergence is still far from clear.
3.3. Impact of differing sex hormones in AD
Changes in sex steroid hormones (i.e. estrogen and progesterone in females and testosterone in males) have been associated with AD onset and progression, representing an endocrinological explanation for AD risk (Grimm et al., 2016). Among these sex hormones, estrogen has received the most attention in clinical and preclinical investigations. Although much of the body’s estrogen is synthesized in the ovaries, a significant amount is also produced in the CNS, where 1 of the 3 isoforms of estrogen, E2 and its receptor (ER), has been observed in high levels in limbic system regions, such as the hypothalamus and hippocampus (Cui et al., 2013). E2 has been shown to mediate sex-specific behaviors, regulate synaptic plasticity, and promote neural survival in a number of studies (Green and Simpkins, 2000). Because of its neuroprotective role, the rapid decrease in endogenous estrogen levels during menopause has been proposed as a trigger for the development of AD in females (Paganini-Hill and Henderson, 1994).
However, only a few studies have shown a direct link between estrogen and AD. One clinical study from the Mayo Clinic, the Cohort Study of Oophorectomy and Aging, showed an almost 2-fold increase in dementia risk in women who underwent bilateral oophorectomy before menopause (Rocca et al., 2011). In the Cache County Memory Study, women who used hormone replacement therapy in the perimenopausal period had fewer cases of AD later on life (Zandi et al., 2002). Another study found that surgical menopause was associated with a faster rate of cognitive decline and more pHF-tau pathology (Bove et al., 2014). In support of this, 1 preclinical study demonstrated that in young APPswe mice with ovariectomies, absolute Aβ levels remain similar to controls, but the Aβ/sAPPα ratio significantly increases due to decreased APP. However, supplemental estrogen administered to these mice paradoxically reduced Aβ levels but did not alter the total levels of APP. These results suggest that estrogen might enhance α-secretase activity leading to increased formation of sAPPα, although the uncoupling between APP and Aβ metabolism with ovarectomy and subsequent estrogen replacement complicates this conclusion (Levin-Allerhand and Smith, 2002). Another study showed that ovariectomies in APP transgenic mice (Tg2576) increased the levels of Aβ levels in the brain and that estrogen replacement therapy blocked this effect (Zheng et al., 2002). However, the same experiments in Tg2576+PS1 double mutant mice demonstrated a much smaller effect, suggesting that differences in transgenes or strains might mediate the relative impact of estrogen in these models (Wang et al., 2003).
The downstream mechanisms of estrogen’s effect on AD neuropathology are still poorly understood, and hypotheses centering on transcriptional regulation and downstream kinase cascades have both been presented (Hwang et al., 2016; Sarkar et al., 2015). One probable mechanism whereby estrogen is neuroprotective in AD is through shifting APP processing toward the nonamyloidogenic pathway via α-secretase through activation of extracellular-regulated kinase 1 and 2 (ERK1 and 2) and protein kinase C (Cordey et al., 2003; Wang et al., 2016). Furthermore, estrogen may act through activation of the MARK/ERK pathway to reduce Aβ levels, potentially through BACE1 inhibition and increased Aβ clearance (Tamagno et al., 2009).
But although the preclinical evidence seems to suggest a protective role for estrogen in AD pathogenesis, clinical trials concerning estrogen replacement therapy (ERT) have resulted in inconclusive data, with some even showing decreased cognitive function with estrogen-only therapies (Asthana et al., 2001; Henderson et al., 2000; Mulnard et al., 2000; Shumaker et al., 2003; Tang et al., 1996; Wang et al., 2000). The results of these studies are surprising, although it is possible that ERT confers other risks that interact with AD pathogenesis to worsen cognition, such as those regarding cardiovascular health. Furthermore, it remains unclear if estrogen + progesterone or progesterone alone may have different neuroprotective properties in postmenopausal women compared to those who took ERT. Finally, some preclinical studies have suggested that estrogen may be an important factor in preventing AD during certain “critical windows,” which is supported by association data in the Cache County Memory Study (Zandi et al., 2002). One preclinical study suggested that ERT had a higher likelihood of being neuroprotective in the perimenopausal but not postmenopausal period (Christensen and Pike, 2017). Intriguingly, another study noticed an increase in Aβ and poorer working memory in female AD mice compared to males that could be reversed by supplementing neonatal females with testosterone propionate, resulting in brain masculinization (Carroll et al., 2011) and suggesting that early exposure to sex hormones during development may have a large impact on later life AD risk. Still, whatever the reason for the disconnect between preclinical and clinical studies, the role of estrogen in AD pathogenesis is likely to be more complex than originally perceived.
Interestingly, studies in human patients and animal models suggest that androgen deprivation may also represent a risk factor for AD pathogenesis in males (Pike et al., 2009; Rosario et al., 2010; Verdile et al., 2014). One study demonstrated that brain testosterone levels were inversely correlated to soluble Aβ concentration in men (Rosario et al., 2011). Another preclinical study indicated that increased testosterone in aged male 3xTg-AD mice was correlated with reduced Aβ plaque pathology (Overk et al., 2013), suggesting a role for testosterone in amyloid processing or clearance. These data were supported by findings where testosterone altered APP processing (Goodenough et al., 2000). Despite these interesting data points, there still remains only a few studies to date that have investigated androgens’ role in AD pathogenesis, and so closer investigations are likely warranted.
In summary, while an endocrinological basis of AD risk is likely to exist, the significance of these declining sex hormones with age is relatively unclear. For instance, although estrogen seems to be neuroprotective in rodent models, ERT in humans was unable to improve cognition or delay AD onset. Regardless of the impact of sex hormones on AD, it is very likely that subtle second messenger system alterations between sexes underlie and likely unify many of the observed increases in AD risk for women when viewed at the genetic (APOE, BDNF), structural, or hormonal levels. In agreement with this, it has been observed that sexually dimorphic second messenger signaling is evident for certain GPCRs and may increase AD risk in a sex-specific manner (Bangasser et al., 2017). One of the most salient examples of this is discussed in Section 3.5.
3.4. Sexual dimorphism in immune function and the link with AD
While Aβ and hyperphosphorylated-tau are considered to be the pathogenic intermediates underlying AD, their interaction with the immune system has garnered a lot of attention. Recently, many immune function genes that are expressed in microglia have been identified in GWAS studies investigating genetic causes of AD risk, including TREM2 (Guerreiro et al., 2013; Wang et al., 2015), CR1 (Lambert et al., 2009), CLU (Harold et al., 2009; Lambert et al., 2009), and CD33 (Bradshaw et al., 2013; Griciuc et al., 2013). In addition, a recent gene-network analysis implicated immune and microglial dysfunction as the leading perturbed pathway in AD in human patients, a result which was specific to AD and not found in other neurodegenerative disorders, such as Huntington’s Disease (Zhang et al., 2013). In line with this, it has been found that elevation of certain cytokines in plasma are correlated with dementia development or conversion from MCI to AD, and imaging studies have found that increased microglial activation is a better predictor of cognitive decline than Aβ load (Buchhave et al., 2010; Cagnin et al., 2001; Edison et al., 2008; Engelhart et al., 2004; Holmes et al., 2009; Jin et al., 2008; Laurin et al., 2009; Patel et al., 2005).
Microglia play an active role in both Aβ regulation and immunologic response (Heneka et al., 2015). As evidence accumulates showing that persistent immune dysregulation affects AD development, numerous studies have demonstrated specific mechanisms whereby microglia may mediate neural degeneration and cognitive dysfunction (Hong et al., 2016; Ji et al., 2013; Lenz and McCarthy, 2015). Of these mechanisms, microglial regulation of synapses may be the most important for AD. It has been shown that the degree of synaptic degeneration is a much better predictor of cognitive decline in AD than Aβ levels, as degeneration of these synaptic structures syncs up more closely with symptom development and precedes neuronal loss (DeKosky and Scheff, 1990; Hong et al., 2016; Lue et al., 1996; Mucke and Selkoe, 2012; Terry et al., 1991). These findings are in line with the hypothesis that chronic Aβ toxicity primes microglia, causing them to respond more readily to local environmental changes with cytokine release and enhanced pruning of synapses (Heneka et al., 2015; Holmes, 2013; Perry and Holmes, 2014).
Given the role for the immune system in AD development, it is important to note that females have a propensity toward greater immune reactions than men. First, males tend to develop infections more readily than females (Klein, 2000), and females tend to have a more robust immune response in the form of greater antibody production and a greater propensity toward TH1 responses (Amadori et al., 1995; Hanamsagar and Bilbo, 2016; Kivity and Ehrenfeld, 2010; Kovacs et al., 2002). In addition, estrogen generally enhances the immune system at low doses while androgens tend to be more immunosuppressive (Hanamsagar and Bilbo, 2016; McClelland and Smith, 2011). At the level of the brain, males tend to have more microglia early in development while females tend to have many more microglia from early adulthood onward (Lenz and McCarthy, 2015; Schwarz et al., 2012).
Perhaps most interestingly, however, is the fact that about 80% of patients with autoimmune disorders are women (Cooper and Stroehla, 2003; Gleicher and Barad, 2007; Kivity and Ehrenfeld, 2010), (Hanamsagar and Bilbo, 2016), suggesting that women may have more primed immune systems and thus more readily exhibit heightened immune responses. In support of this, one study found that while children of women with autoimmune disorders often developed autoimmune diseases, they were rarely the same disease exhibited by the mother (Gleicher and Barad, 2007). This is an important point, as it may explain why AD is not necessarily associated with other autoimmune diseases, since AD patients may manifest disease due to common immunological priming while expressing divergent pathophysiology to specific insults.
Finally, the interaction between stress and the immune system is different between men and women. Specifically, it was shown that while glucocorticoid signaling, which is a main mediator of the vertebrate stress response, induced apoptotic pathway upregulation in male hepatic cells, female hepatic cells tended to upregulate IL-6-related pathways, which are proinflammatory (Deak et al., 2015). Furthermore, glucocorticoid signaling tends to be higher in females at baseline, and estrogen has been shown to increase glucocorticoid release while androgens decrease it (Gaillard and Spinedi, 1998). Interestingly, when challenged with a lethal dose of the immunogen LPS, the corticosteroid dexamethasone was able to prevent death in male rats at low doses while much higher doses were needed for females (Deak et al., 2015), demonstrating that the female immunosuppressive response to glucocorticoids is less potent than males.
Still, while AD is likely to be affected by immune system function and while ample evidence suggests very different inflammatory responses between sexes, whether these divergent responses lead to a difference in AD susceptibility is unclear. For one, even the role of sex in autoimmune disease pathogenesis is unknown, with hypotheses describing differences in sex hormones, X-inactivation patterns, and gene dosing, and microchimerism all having evidence supporting and diminishing their likelihood as being the underlying mechanism (Gleicher and Barad, 2007; Kivity and Ehrenfeld, 2010; Selmi, 2008). Future studies demonstrating an immune system and sex interaction in AD pathogenesis would be needed, and rigorous hypothesis-based testing would be essential to confirm if any of the mechanisms potentially leading to increased autoimmunity also lead to increased AD risk in women.
3.5. Stress and sex-specific CRF signaling in AD
Clinical evidence has suggested a link between psychosocial stress and AD going back at least 2 decades. Stress-related increases in plasma cortisol levels (Swanwick et al., 1998; Rehman, 2002; Umegaki et al., 2000) and correlations between increased cortisol levels and the severity of cognitive decline (Csernansky et al., 2006; Pedersen et al., 2001) have been reported in AD. These changes in the hypothalamus-pituitary-adrenal (HPA) axis do not appear to be secondary to MDD, as AD patients with and without MDD have higher cerebrospinal fluid cortisol levels compared to controls (Hoogendijk et al., 2006). In support of this, the centrally active stress response receptor corticotropin releasing factor 1 (CRF1) is also increased in the hippocampi of AD patients (Behan et al., 1995, De Souza, 1995), and HPA axis changes have been associated with hippocampal atrophy (O’Brien et al., 1996). Furthermore, many abnormal behaviors, termed Behavioral and Psychosocial Symptoms in Dementia, occur at an alarmingly high rate in AD patients, and at least some of these abnormal behaviors can be attributed to alterations in stress hormone signaling (Reisberg et al., 1987). In line with these human studies, transgenic mouse models have recapitulated at least some of the neuropathological and behavioral changes associated with AD and thus provide an opportunity to investigate how the different biochemical markers underlying the psychosocial stress response and AD pathogenesis interact in vivo (Campbell et al., 2015; Carroll et al., 2011; Cuadrado-Tejedor et al., 2012; Dong et al., 2004, 2008; Green et al., 2006; Jeong et al., 2006; Kang et al., 2007; Rissman et al., 2012).
Women are more susceptible to stress-influenced diseases, and it is possible that sexually dimorphic stress responses could greatly influence the difference between men and women in the rate of AD conversion and progression. Although clinical evidence demonstrating an interaction between sex and stress in AD development remains scarce, an intriguing study demonstrated that women with mild-to-moderate AD have significantly increased levels of cortisol production compared to their male counterparts (Rasmuson et al., 2011), although these data could not establish causation.
Although studies in humans are limited, preclinical studies have also shown a sex-specific difference in AD pathogenesis under psychosocial stress (Sierksma et al., 2012). For instance, 5-day restraint stress (6 h/d) in 5XFAD transgenic mice leads to increased levels of neurotoxic Aβ42 peptides, β-secretase-cleaved C-terminal fragment (C99), and plaque burden in the hippocampus of female but not male mice at a time point before severe AD-like behavioral deficits are observed. Moreover, sex- and brain region-specific accelerations in β-amyloidosis are accompanied by elevations in BACE1, APP, and phosphorylated EIF2a in response to psychosocial stress (Devi et al., 2010). Also, environmental stress preferentially triggers memory impairments in female but not in male P301L-tau mice (Sotiropoulos et al., 2015). Furthermore, in this model, stress-related increases in caspase-3-truncated tau and insoluble tau have been found to aggregate exclusively in the female hippocampus (Sotiropoulos et al., 2015). Potentially driving this effect, the expression of the molecular chaperones Hsp90, Hsp70, and Hsp105 were altered to favor accumulation of tau aggregates (Sotiropoulos et al., 2015). Although some ambiguity exists between these studies regarding the interaction between stress and sex, it seems likely that both are impacting disease pathogenesis.
As mentioned, the exact stress-response molecules that mediate increased AD pathogenesis are still unknown, though data confirming an important hypothesis implicating central CRF1 signaling in this process is emerging. Increased CRF1 signaling has been associated with multiple stages of APP proteolysis, regulation of Aβ generation, and Aβ-mediated toxicity (Thathiah and De Strooper, 2011; Thathiah et al., 2013), and GPCR signaling in general has been suggested as a novel target for AD drug development (Femminella et al., 2013; Wolfe, 2013). Importantly, CRF overexpression in the forebrain can lead to accumulation of Aβ and tau phosphorylation through CRF1-Gs-PKA signaling, consistent with Gs-PKA signaling’s role in influencing the amyloid production cascade through modulation of α-, β-, and γ-secretases (Park et al., 2015; Robert et al., 2001; Thathiah and De Strooper, 2011; Thathiah et al., 2013; Xu et al., 1996). Specifically, while transient activation of these signaling cascades shifts APP metabolism toward the α-secretase-mediated pathway that results in nonpathogenic amyloids, chronic activation shifts APP metabolism to the β-, γ-, and perhaps also the η-secretase-mediated pathways that result in increased pathogenic Aβ generation (APP-CTF99, A-beta 40, and A-beta 42). (da Cruz e Silva et al., 2009, Willem et al., 2015). In addition, PKA signaling is associated with tau phosphorylation, another molecular pathway that is highly implicated in AD pathogenesis (Blanchard et al., 1994, Sanchez-Mut et al., 2014). Thus, there is much evidence implicating CRF1 signaling as the causative pathway facilitating psychosocial stress’ detrimental effects on AD pathogenesis.
When taken together with the evidence that prolonged CRF signaling through CRF1 results in sexually dimorphic second messenger activation, a potential mechanism begins to take shape: Because Gs-PKA signaling is central to the formation of AD-causative amyloids and phospho-tau, and there exists a bias toward CRF1-Gs-PKA signaling in females, this signaling pathway may provide a functional link between these 2 processes and begins to create a unified, biologically driven explanation for why women develop AD more readily than men. Still, this hypothesis is based on evidence from a combination of isolated experiments that only suggest this mechanism, but rigorous and direct testing of this hypothesis will need to be specifically undertaken to assess whether it is a suitable explanation for what is observed in AD patients. In addition, how CRF1 signaling and peripheral glucocorticoid signaling interact is still unknown in terms of increased AD risk, and experiments specifically targeting one or the other are needed to see if an interaction between the 2 is necessary for the increased risk of AD.
Table 2 summarizes the studies on sex difference in AD with animal models, collected from 2000 to date.
Table 2.
Article | Author(s) | Year | Journal | Genotype/strain | Age (mo) | Measurements | Main results/conclusion |
---|---|---|---|---|---|---|---|
Sex Dimorphism Profile of Alzheimer’s Disease-Type Pathologies in an APP/ PS1 Mouse Model | Jiao et al. | 2016 | Neurotox Res. | APP/PS1 mice | 12 | Neuropathology | Female APP/PS1 mice had more severe cerebral amyloid angiopathy, subsequent microhemorrhage, higher levels of phosphorylated tau, proinflammatory cytokines, more severe astrocytosis and microgliosis, and greater neuronal and synaptic degenerations than male counterparts. |
A TgCRND8 Mouse Model of Alzheimer’s Disease Exhibits Sexual Dimorphisms in Behavioral Indices of Cognitive Reserve. | Granger et al. | 2016 | J Alzheimers Dis. | TgCRND8 mice | 2,4,6,8 | Neuropathology | AbPP females and males exhibit comparable Ab burden at all ages, yet Tg females show enhanced vulnerability to Aβ pathology and Ab- associated stereotypy, phenocovert in the MWM maze earlier, and exhibit fewer behavioral indices of cognitive reserve than males. |
Sex differences between APPswe/ PS1dE9 mice in A-beta accumulation and pancreatic islet function during the development of Alzheimer’s disease. | Li et al. | 2016 | Lab Anim. | APP/PS1mice | 4, 6, 9 | Neuropathology Memory function pancreatic islet function | Aβ levels and plaques, in the female mice were significantly higher than in the males. Cognitive function of female APP/PS1 mice was attenuated earlier than in the males Impairment of glucose tolerance and insulin tolerance occurred earlier in male APP/PS1 mice than in female mice |
Peripheral amyloid levels present gender differences associated with aging in AβPP/PS1 mice. | Ordonez-Gutierrez et al. | 2015 | J Alzheimers Dis. | APP/PS1 mice | 3,15 | Amyloid levels in plasma | After 9 mo of age, there is an increase in amyloid levels in plasma among females and a decrease among males. 3xTgAD mice show increased cognitive deficits in females and increased levels of novelty- induced behavioral inhibition in males. |
Cognitive and emotional profiles of aged Alzheimer’s disease (3xTgAD) mice: effects of environmental enrichment and sexual dimorphism. | Blazquez et al. | 2014 | Behav Brain Res. | 3xTgAD mice | 12e15 | Behaviors Neuropathology | 3xTgAD mice show increased cognitive deficits in females and increased levels of novelty- induced behavioral inhibition in males. |
Cumulative effects of the ApoE genotype and gender on the synaptic proteome and oxidative stress in the mouse brain. | Shi et al. | 2014 | Int J Neuropsychopharmacol. | Human-ApoE4 mice | N/A | Isolated synaptosomes Label-free quantitative proteomics | The lack of estrogen-mediated protection regulated by the ApoE genotype led to synaptic mitochondrial dysfunction and increased oxidative stress |
Effects of prenatal stress exposure on soluble Aβ and brain-derived neurotrophic factor signaling in male and female APPswe/PS1dE9 mice. | Sierksma et al. | 2012 | Neurochem Int. | APP/PS1 mouse | Prenatal, 8 | Stress Neuropathology | Female APPswe/PS1dE9 mice have higher levels of hippocampal proBDNF and soluble Aβ as compared to their male littermates. |
Gender-dependent transthyretin modulation of brain amyloid-β levels: evidence from a mouse model of Alzheimer’s disease. | Oliveira et al. | 2011 | J Alzheimers Dis. | AbPPswe/PS1A246E/TT-/- AbPPswe/PS1A246E/TTRþ/-AbPPswe/PS1A246E/ | 3,6,10 | Neuropathology Brain levels of testosterone and 17b-estradiol | Reduced levels of brain testosterone and 17β- estradiol in female mice with TTR genetic reduction might associated with increased AD- like neuropathology |
Sex differences in β-amyloid accumulation in 3xTg-AD mice: Role of neonatal sex steroid hormone exposure | Carroll 2010 | 2010 | Brain Res. | TTRþ/þ 3xTg-AD mice | Sex steroid hormone | Male 3xTg-AD mice demasculinized during early development exhibit significantly increased Aβ accumulation in adulthood. In contrast, female mice defeminized during early development exhibit a more male-like pattern of Aβ pathology in adulthood. | |
Sex- and brain region-specific acceleration of β-amyloidogenesis following behavioral stress in a mouse model of Alzheimer’s disease | Devi et al. | 2010 | Mol Brain. | 5xFAD mice | 3 | Stress, Neuropathology | Exposure to the relatively brief behavioral stress increased levels of neurotoxic Aβ42 peptides, the β-secretase-cleaved C-terminal fragment (C99) and plaque burden in the hippocampus of female 5XFAD mice but not in that of male 5XFAD mice. |
Females exhibit more extensive amyloid, but not tau, pathology in an Alzheimer transgenic model. | Hirata-Fukae et al. | 2008 | Brain Res. | 3xTg-AD transgenic mice | 3, 6, 9, 12, 16, 20, 23 | Amyloid, Tau. | Increase in beta-secretase activity and a reduction of neprilysin but not tau in female mice compared to males |
Age-dependent sexual dimorphism in | Clinton et al. | 2007 | Neurobiol Dis. | 3xTg-AD mice | 4, 12 | Stress Cognition Neuropathology | 3xTg-AD females performed worse than males at 4 mo of age, which correlated with the enhanced corticosterone response of the stressful tasks. |
Progesterone and Estrogen Regulate Alzheimer-Like Neuropathology in Female 3xTg-AD Mice | Carroll et al. | 2007 | J Neurosci. | 3xTg-AD mice | 3, 12 | Progesterone and Estrogen intervention Neuropathology | Ovariectomy-induced depletion of sex steroid hormones in adult female 3xTg-AD mice signifcantly increased Abeta accumulation and worsened memory performance. Treatment of ovariectomized 3xTg-AD mice with estrogen, but not progesterone, prevented these effects. |
Gender differences in the amount and deposition of amyloidbeta in APPswe and PS1 double transgenic mice | Wang et al. | 2003 | Neurobiol Dis. | APP þ PS1 mice | 4, 12, 17 | Neuropathology | Female APP/PS1 mice accumulate amyloid at an earlier age and develop more amyloid deposits in the hippocampus than age-matched male mice |
Modulation of A (beta) peptides by estrogen in mouse models | Zheng et al. | 2002 | J Neurochem | Tg2576 mice | 4e7 | Ovariectomy and estradiol supplementation | The (beta) levels were higher in estrogen- deprived mice than intact mice, and this effect could be reversed through the administration of estradiol. |
Effects of BACE1 haploinsufficiency on APP processing and Aβ concentrations in male and female 5XFAD Alzheimer mice at different disease stages. | Devi and Ohno | 2015Neuroscience | Neuroscience | BACE1 (þ/-) BACE1 (þ/-)$5XFAD mice | 6e7 12e14 | Neuropathology | BACE1 haploinsufficiency rescues memory defcits in 5XFAD mice irrespective of sex but only in the younger age group. |
Olfactory delayed matching-to-sample performance in mice: sex differences in the 5XFAD mouse model of Alzheimer’s disease. | Roddick et al. | 2014 | Behav Brain Res. | 5XFAD mice | 6e7 | Olfactory delayed matching-to-sample | 5XFAD female mice showed higher levels of performance on the delayed matching-to- sample task than males, indicative of better working memory. |
Sex differences in β-amyloid accumulation in 3xTg-AD mice: Role of neonatal sex steroid hormone exposure | Carroll et al. | 2010 | Brain Res. | 3xTg-AD mice | Sex steroid hormone | Male 3xTg-AD mice demasculinized during early development exhibit significantly increased Aβ accumulation in adulthood. In contrast, female mice defeminized during early development exhibit a more male-like pattern of Aβ pathology in adulthood. | |
Testosterone regulation of Alzheimer- like neuropathology in male 3xTg- AD mice involves both estrogen and androgen pathways | Rosario et al. | 2010 | Brain Res. | 3xTg-AD mice | 3e7 | Androgen and estrogen | Testosterone regulated Aβ pathology through androgen and estrogen pathways and reduced tau pathology largely through estrogen pathways. |
Androgens Regulate the Development of Neuropathology in a Triple Transgenic Mouse Model of Alzheimer’s Disease | Rosario et al. | 2006 | J Neurosci. | 3xTg-AD mice | 3, 7, 13 | Gonadectomized (GDX) androgen | Androgen depletion accelerates the development of AD-like neuropathology |
Testosterone stimulates rapid secretory amyloid precursor protein release from rat hypothalamic cells via the activation of the mitogen-activated protein kinase pathway. | Goodenough et al. | 2000 | Neurosci Lett. | Immortalized rat hypothalamic cell line (GT1-7) | N/A | Androgens | Testosterone was found to increase the amount of APP secretion rapidly after treatment without effecting the overall amount of cellular APP. The species of APP secreted was found to be predominantly the product of the nonamyloidogenic alpha-secretory pathway. Furthermore, this event is regulated via aromatase-mediated conversion of testosterone to estrogen and the mitogen-activated protein kinase (MAP kinase) signaling pathway. |
The articles in black color indicate studies supporting females more vulunable to neuropathgenesis of AD and those in shaded region oppose those findings
4. Conclusion
An explanation for the apparent difference in the rate of AD diagnoses between men and women remains elusive, but more and more evidence is beginning to suggest that a true increase in risk is complementing the epidemiologic explanations describing the increased female prevalence for the disease. Importantly, biological mechanisms that are intrinsic to women are increasingly prominent in the literature. Specifically, those involving age-related changes in sex hormone signaling, sexual dimorphism in neural structures, gene-by-sex interactions, female-specific increases in immune responses, and the interaction between sex and stress have been gaining traction as important determinants of AD. Still, much remains to be described before adopting any of these potential explanations as drivers of disease, and certainly a greater understanding of the biology underlying these mechanisms is warranted before therapeutic development should be undertaken. The ERT trials stand as a sobering testament to that fact. It seems clear that more detailed studies using preclinical animal models to explore these molecular explanations will begin to unravel the mystery concerning the increased risk of AD in women, and the promise of future therapeutics through this line of investigation remains high.
One of the most important outstanding questions--and perhaps the largest “elephant in the room”--is why so many studies have suggested the presence of female-specific, biological risk factors and why so few studies have uncovered a difference in incidence between sexes. As described in length previously, difficulties in correct classification of AD remain one potential source of ambiguity, but this alone is unlikely to be the sole source of bias. What remains a possibility is that prospective cohort studies and meta-analyses have been insufficiently powered to detect a difference, as seen in the Fiest et al., 2016 meta-analysis which consistently detected higher prevalence and incidence rates for women without reaching statistical significance. Although a significant undertaking, a large, prospective cohort study with postmortem confirmation of AD would be immensely helpful in resolving this issue.
Finally, as we move toward “personalized medicine,” improving our understanding of sex-specific disease mechanisms will be one of the first areas to yield substantial improvements in prevention and treatment outcomes across all branches of medicine. But, it is important to note that investigations of sex-specific risk are just as likely to reveal central mechanisms of disease pathogenesis that are independent of sex. By careful comparison and inclusion of both sexes in preclinical and clinical research, upstream and downstream pathways that converge in both sexes will point the way toward shared mechanisms resulting in disease pathogenesis and severity. Exploiting these commonalities is more likely to lead to successful interventions and treatments than ignoring sex as a potential risk factor, as careful consideration of disease pathogenesis by sex will reduce sex-specific motifs that are mistaken as being causative but are in truth epiphenomenal. Therefore, although improving our understanding of underlying sexual dimorphism in AD risk will be critical for developing treatments that are especially effective in women, this line of research also has the potential to reveal new targets for the treatment of AD and other neurodegenerative disorders in both sexes.
Acknowledgements
This work was supported by the National Institute of Health (1R56AG053491–01, HD) and (1 R01 AG057884–01, HD).
Footnotes
Disclosure statement
The authors have no actual or potential conflicts of interest.
References
- Alzheimer Association, 2015. Alzheimer’s disease facts and figures. Alzheimers Dement 11, 332e384. [DOI] [PubMed] [Google Scholar]
- Altmann A, Tian L, Henderson VW, Greicius MD, 2014. Sex modifies the APOE-related risk of developing Alzheimer disease. Ann. Neurol 75, 563e573. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Amadori A, Zamarchi R, De Silvestro G, Forza G, Cavatton G, Danieli GA, Clementi M, Chieco-Bianchi L, 1995. Genetic control of the CD4/CD8 T-cell ratio in humans. Nat. Med 1, 1279e1283. [DOI] [PubMed] [Google Scholar]
- Ardekani BA, Convit A, Bachman AH, 2016. Analysis of the MIRIAD data shows sex differences in hippocampal atrophy progression. J. Alzheimers Dis 50, 847e857. [DOI] [PubMed] [Google Scholar]
- Asthana S, Baker LD, Craft S, Stanczyk FZ, Veith RC, Raskind MA, Plymate SR, 2001. High-dose estradiol improves cognition for women with AD: results of a randomized study. Neurology 57, 605e612. [DOI] [PubMed] [Google Scholar]
- Bachman DL, Wolf PA, Linn RT, Knoefel JE, Cobb JL, Belanger AJ, White LR, D’Agostino RB, 1993. Incidence of dementia and probable Alzheimer’s disease in a general population: the Framingham Study. Neurology 43 (3 Pt 1), 515e519. [DOI] [PubMed] [Google Scholar]
- Bangasser DA, Dong H, Carroll J, Plona Z, Ding H, Rodriguez L, McKennan C, Csernansky JG, Seeholzer SH, Valentino RJ, 2017. Corticotropin-releasing factor overexpression gives rise to sex differences in Alzheimer’s disease-related signaling. Mol. Psychiatry 22, 1126e1133. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Barnes LL, Wilson RS, Bienias JL, Schneider JA, Evans DA, Bennett DA, 2005. Sex differences in the clinical manifestations of Alzheimer disease pathology. Arch. Gen. Psychiatry 62, 685e691. [DOI] [PubMed] [Google Scholar]
- Behan DP, Heinrichs SC, Troncoso JC, Liu XJ, Kawas CH, Ling N, De Souza EB, 1995. Displacement of corticotropin releasing factor from its binding protein as a possible treatment for Alzheimer’s disease. Nature 378, 284e287. [DOI] [PubMed] [Google Scholar]
- Beydoun MA, Boueiz A, Abougergi MS, Kitner-Triolo MH, Beydoun HA, Resnick SM, O’Brien R, Zonderman AB, 2012. Sex differences in the association of the apolipoprotein E epsilon 4 allele with incidence of dementia, cognitive impairment and decline. Neurobiol. Aging 33, 720. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Blanchard BJ, Devi Raghunandan R, Roder HM, Ingram VM, 1994. Hyperphosphorylation of human TAU by brain kinase PK40erk beyond phosphorylation by cAMP-dependent PKA: relation to Alzheimer’s disease. Biochem. Biophys. Res. Commun 200, 187e194. [DOI] [PubMed] [Google Scholar]
- Bove R, Secor E, Chibnik LB, Barnes LL, Schneider JA, Bennett DA, De Jager PL, 2014. Age at surgical menopause influences cognitive decline and Alzheimer pathology in older women. Neurology 82, 222e229. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bradshaw EM, Chibnik LB, Keenan BT, Ottoboni L, Raj T, Tang A, Rosenkrantz LL, Imboywa S, Lee M, Von Korff A, Morris MC, Evans DA, Johnson K, Sperling RA, Schneider JA, Bennett DA, De Jager PL, 2013. CD33 Alzheimer’s disease locus: altered monocyte function and amyloid biology. Nat. Neurosci 16, 848e850. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bremner JD, 1999. Does stress damage the brain? Biol. Psychiatry 45, 797e805. [DOI] [PubMed] [Google Scholar]
- Brookmeyer R, Gray S, Kawas C, 1998. Projections of Alzheimer’s disease in the United States and the public health impact of delaying disease onset. Am. J. Public Health 88, 1337e1342. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bu G, 2009. Apolipoprotein E and its receptors in Alzheimer’s disease: pathways, pathogenesis and therapy. Nat. Rev. Neurosci 10, 333e344. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Buchhave P, Zetterberg H, Blennow K, Minthon L, Janciauskiene S, Hansson O, 2010. Soluble TNF receptors are associated with Aβ metabolism and conversion to dementia in subjects with mild cognitive impairment. Neurobiol. Aging 31, 1877e1884. [DOI] [PubMed] [Google Scholar]
- Buttery AK, Du Y, Busch MA, Fuchs J, Gaertner B, Knopf H, Scheidt-Nave C, 2016. Changes in physical functioning among men and women aged 50e79 years in Germany: an analysis of National Health Interview and Examination Surveys, 1997e1999 and 2008e2011. BMC Geriatr. 16, 205. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Cagnin A, Brooks DJ, Kennedy AM, Gunn RN, Myers R, Turkheimer FE, Jones T, Banati RB, 2001. In-vivo measurement of activated microglia in dementia. Lancet 358, 461e467. [DOI] [PubMed] [Google Scholar]
- Campbell SN, Zhang C, Monte L, Roe AD, Rice KC, Tache Y, Masliah E, Rissman RA, 2015. Increased tau phosphorylation and aggregation in the hippocampus of mice overexpressing corticotropin-releasing factor. J. Alzheimers Dis 43, 967e976. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Carmelli D, Swan GE, Reed T, Miller B, Wolf PA, Jarvik GP, Schellenberg GD, 1998. Midlife cardiovascular risk factors, ApoE, and cognitive decline in elderly male twins. Neurology 50, 1580e1585. [DOI] [PubMed] [Google Scholar]
- Carroll JC, Iba M, Bangasser DA, Valentino RJ, James MJ, Brunden KR, Lee VM, Trojanowski JQ, 2011. Chronic stress exacerbates tau pathology, neurodegeneration, and cognitive performance through a corticotropin-releasing factor receptor-dependent mechanism in a transgenic mouse model of tauopathy. J. Neurosci 31, 14436e14449. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Chen J, Liang X, Li B, Jiang X, Xu Z, 2014. Gender-related association of brain-derived neurotrophic factor gene 196A/G polymorphism with Alzheimer’s disease: a meta-analysis including 6854 cases and 6868 controls. Int. J. Neurosci 124, 724e733. [DOI] [PubMed] [Google Scholar]
- Christensen A, Pike CJ, 2017. Age-dependent regulation of obesity and Alzheimer-related outcomes by hormone therapy in female 3xTg-AD mice. PLoS One 12, e0178490. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Cooper GS, Stroehla BC, 2003. The epidemiology of autoimmune diseases. Autoimmun. Rev 2, 119e125. [DOI] [PubMed] [Google Scholar]
- Cordey M, Gundimeda U, Gopalakrishna R, Pike CJ, 2003. Estrogen activates protein kinase C in neurons: role in neuroprotection. J. Neurochem 84, 1340e1348. [DOI] [PubMed] [Google Scholar]
- Crimmins EM, Kim JK, Solé-Auró A, 2011. Gender differences in health: results from SHARE, ELSA and HRS. Eur. J. Public Health 21, 81. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Csernansky JG, Dong H, Fagan AM, Wang L, Xiong C, Holtzman DM, Morris JC, 2006. Plasma cortisol and progression of dementia in subjects with Alzheimer-type dementia. Am. J. Psychiatry 163, 2164e2169. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Cuadrado-Tejedor M, Ricobaraza A, Frechilla D, Franco R, Perez-Mediavilla A, Garcia-Osta A, 2012. Chronic mild stress accelerates the onset and progression of the Alzheimer’s disease phenotype in Tg2576 mice. J. Alzheimers Dis 28, 567e578. [DOI] [PubMed] [Google Scholar]
- Cui J, Shen Y, Li R, 2013. Estrogen synthesis and signaling pathways during aging: from periphery to brain. Trends Mol. Med 19, 197e209. [DOI] [PMC free article] [PubMed] [Google Scholar]
- da Cruz e Silva OA, Rebelo S, Vieira SI, Gandy S, da Cruz e Silva EF, Greengard P, 2009. Enhanced generation of Alzheimer’s amyloid-beta following chronic exposure to phorbol ester correlates with differential effects on alpha and epsilon isozymes of protein kinase C. J. Neurochem 108, 319e330. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Damoiseaux JS, Seeley WW, Zhou J, Shirer WR, Coppola G, Karydas A, Rosen HJ, Miller BL, Kramer JH, Greicius MD, 2012. Gender modulates the APOE epsilon4 effect in healthy older adults: convergent evidence from functional brain connectivity and spinal fluid tau levels. J. Neurosci 32, 8254e8262. [DOI] [PMC free article] [PubMed] [Google Scholar]
- De Souza EB, 1995. Corticotropin-releasing factor receptors: physiology, pharmacology, biochemistry and role in central nervous system and immune disorders. Psychoneuroendocrinology 20, 789e819. [DOI] [PubMed] [Google Scholar]
- Deak T, Quinn M, Cidlowski JA, Victoria NC, Murphy AZ, Sheridan JF, 2015. Neuroimmune mechanisms of stress: sex differences, developmental plasticity, and implications for pharmacotherapy of stress-related disease. Stress 18, 367. [DOI] [PMC free article] [PubMed] [Google Scholar]
- DeKosky ST, Scheff SW, 1990. Synapse loss in frontal cortex biopsies in Alzheimer’s disease: correlation with cognitive severity. Ann. Neurol 27, 457e464. [DOI] [PubMed] [Google Scholar]
- Devi L, Alldred MJ, Ginsberg SD, Ohno M, 2010. Sex- and brain region-specific acceleration of beta-amyloidogenesis following behavioral stress in a mouse model of Alzheimer’s disease. Mol. Brain 3, 34. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Dong H, Goico B, Martin M, Csernansky CA, Bertchume A, Csernansky JG, 2004. Modulation of hippocampal cell proliferation, memory, and amyloid plaque deposition in APPsw (Tg2576) mutant mice by isolation stress. Neuroscience 127, 601e609. [DOI] [PubMed] [Google Scholar]
- Dong H, Yuede CM, Coughlan C, Lewis B, Csernansky JG, 2008. Effects of memantine on neuronal structure and conditioned fear in the Tg2576 mouse model of Alzheimer’s disease. Neuropsychopharmacology 33, 3226e3236. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Edison P, Archer HA, Gerhard A, Hinz R, Pavese N, Turkheimer FE, Hammers A, Tai YF, Fox N, Kennedy A, Rossor M, Brooks DJ, 2008. Microglia, amyloid, and cognition in Alzheimer’s disease: an [11C](R)PK11195-PET and [11C]PIB-PET study. Neurobiol. Dis 32, 412e419. [DOI] [PubMed] [Google Scholar]
- Edland SD, Rocca WA, Petersen RC, Cha RH, Kokmen E, 2002. Dementia and Alzheimer disease incidence rates do not vary by sex in Rochester, Minn. Arch. Neurol 59, 1589e1593. [DOI] [PubMed] [Google Scholar]
- Elbejjani M, Fuhrer R, Abrahamowicz M, Mazoyer B, Crivello F, Tzourio C, Dufouil C, 2015. Depression, depressive symptoms, and rate of hippocampal atrophy in a longitudinal cohort of older men and women. Psychol. Med 45, 1931e1944. [DOI] [PubMed] [Google Scholar]
- Engelhart MJ, Geerlings MI, Meijer J, Kiliaan A, Ruitenberg A, Swieten JC, Stijnen T, Hofman A, Witteman JC, Breteler MM, 2004. Inflammatory Proteins in Plasma and the Risk of Dementia: The Rotterdam Study. Arch. Neurol 61, 668e672. [DOI] [PubMed] [Google Scholar]
- Farrer LA, Cupples LA, Haines JL, Hyman B, Kukull WA, Mayeux R, Myers RH, Pericak-Vance MA, Risch N, van Duijn CM, 1997. Effects of age, sex, and ethnicity on the association between apolipoprotein E genotype and Alzheimer disease. A meta-analysis. APOE and Alzheimer Disease Meta Analysis Consortium. JAMA 278, 1349e1356. [PubMed] [Google Scholar]
- Femminella GD, Rengo G, Pagano G, de Lucia C, Komici K, Parisi V, Cannavo A, Liccardo D, Vigorito C, Filardi PP, Ferrara N, Leosco D, 2013. beta-adrenergic receptors and G protein-coupled receptor kinase-2 in Alzheimer’s disease: a new paradigm for prognosis and therapy? J. Alzheimers Dis 34, 341e347. [DOI] [PubMed] [Google Scholar]
- Ferencz B, Gerritsen L, 2015. Genetics and underlying pathology of dementia. Neuropsychol. Rev 25, 113e124. [DOI] [PubMed] [Google Scholar]
- Fiest KM, Roberts JI, Maxwell CJ, Hogan DB, Smith EE, Frolkis A, Cohen A, Kirk A, Pearson D, Pringsheim T, Venegas-Torres A, Jetté N, 2016. The prevalence and incidence of dementia due to Alzheimer’s disease: a systematic review and meta-analysis. Can. J. Neurol. Sci 43, S51eS82. [DOI] [PubMed] [Google Scholar]
- Fleisher A, Grundman M, Jack CR Jr., Petersen RC, Taylor C, Kim HT, Schiller DH, Bagwell V, Sencakova D, Weiner MF, DeCarli C, DeKosky ST, van Dyck CH, Thal LJ, 2005. Sex, apolipoprotein E epsilon 4 status, and hippocampal volume in mild cognitive impairment. Arch. Neurol 62, 953e957. [DOI] [PubMed] [Google Scholar]
- Fratiglioni L, Launer LJ, Andersen K, Breteler MM, Copeland JR, Dartigues JF, Lobo A, Martinez-Lage J, Soininen H, Hofman A, 2000. Incidence of dementia and major subtypes in Europe: a collaborative study of population-based cohorts. Neurologic Diseases in the Elderly Research Group. Neurology 54 (11 Suppl 5), S10eS15. [PubMed] [Google Scholar]
- Freedman VA, Wolf DA, Spillman BC, 2016. Disability-Free life expectancy over 30 Years: a growing female disadvantage in the US population. Am. J. Public Health 106, 1079e1085. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Fukumoto N, Fujii T, Combarros O, Kamboh MI, Tsai S-J, Matsushita S, Nacmias B, Comings DE, Arboleda H, Ingelsson M, Hyman BT, Akatsu H, Grupe A, Nishimura AL, Zatz M, Mattila KM, Rinne J, Goto Y. i., Asada T, Nakamura S, Kunugi H, 2010. Sexually dimorphic effect of the Val66Met polymorphism of BDNF on susceptibility to Alzheimer’s disease: new data and meta-analysis. Am. J. Med. Genet. B Neuropsychiatr. Genet 153B, 235e242. [DOI] [PubMed] [Google Scholar]
- Gaillard RC, Spinedi E, 1998. Sex- and stress-steroids interactions and the immune system: evidence for a neuroendocrine-immunological sexual dimorphism. Domest. Anim. Endocrinol 15, 345e352. [DOI] [PubMed] [Google Scholar]
- Galasko D, Chang L, Motter R, Clark CM, Kaye J, Knopman D, Thomas R, Kholodenko D, Schenk D, Lieberburg I, Miller B, Green R, Basherad R, Kertiles L, Boss MA, Seubert P, 1998. High cerebrospinal fluid tau and low amyloid beta42 levels in the clinical diagnosis of Alzheimer disease and relation to apolipoprotein E genotype. Arch. Neurol 55, 937e945. [DOI] [PubMed] [Google Scholar]
- Gale SD, Baxter L, Thompson J, 2016. Greater memory impairment in dementing females than males relative to sex-matched healthy controls. J. Clin. Exp. Neuropsychol 6, 1e7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gallart-Palau X, Lee BS, Adav SS, Qian J, Serra A, Park JE, Lai MK, Chen CP, Kalaria RN, Sze SK, 2016. Gender differences in white matter pathology and mitochondrial dysfunction in Alzheimer’s disease with cerebrovascular disease. Mol. Brain 9, 27. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gatz M, Reynolds CA, Fratiglioni L, Johansson B, Mortimer JA, Berg S, Fiske A, Pedersen NL, 2006. Role of genes and environments for explaining Alzheimer disease. Arch. Gen. Psychiatry 63, 168e174. [DOI] [PubMed] [Google Scholar]
- Gleicher N, Barad DH, 2007. Gender as risk factor for autoimmune diseases. J. Autoimmun 28, 1e6. [DOI] [PubMed] [Google Scholar]
- Goodenough S, Engert S, Behl C, 2000. Testosterone stimulates rapid secretory amyloid precursor protein release from rat hypothalamic cells via the activation of the mitogen-activated protein kinase pathway. Neurosci. Lett 296, 49e52. [DOI] [PubMed] [Google Scholar]
- Graves BM, Strand M, Lindsay AR, 2006. A reassessment of sexual dimorphism in human senescence: theory, evidence, and causation. Am. J. Hum. Biol 18, 161e168. [DOI] [PubMed] [Google Scholar]
- Green KN, Billings LM, Roozendaal B, McGaugh JL, LaFerla FM, 2006. Glucocorticoids increase amyloid-beta and tau pathology in a mouse model of Alzheimer’s disease. J. Neurosci 26, 9047e9056. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Green PS, Simpkins JW, 2000. Neuroprotective effects of estrogens: potential mechanisms of action. Int. J. Dev. Neurosci 18, 347e358. [DOI] [PubMed] [Google Scholar]
- Griciuc A, Serrano-Pozo A, Parrado AR, Lesinski AN, Asselin CN, Mullin K, Hooli B, Choi SH, Hyman BT, Tanzi RE, 2013. Alzheimer’s disease risk gene CD33 inhibits microglial uptake of amyloid beta. Neuron 78, 631e643. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Grimm A, Biliouris EE, Lang UE, Gotz J, Mensah-Nyagan AG, Eckert A, 2016. Sex hormone-related neurosteroids differentially rescue bioenergetic deficits induced by amyloid-beta or hyperphosphorylated tau protein. Cell Mol. Life Sci 73, 201e215. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Guerreiro R, Wojtas A, Bras J, Carrasquillo M, Rogaeva E, Majounie E, Cruchaga C, Sassi C, Kauwe JS, Younkin S, Hazrati L, Collinge J, Pocock J, Lashley T, Williams J, Lambert JC, Amouyel P, Goate A, Rademakers R, Morgan K, Powell J, St George-Hyslop P, Singleton A, Hardy J, 2013. TREM2 variants in Alzheimer’s disease. N. Engl. J. Med 368, 117e127. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gustafson L, 1987. Frontal lobe degeneration of non-Alzheimer type. II. Clinical picture and differential diagnosis. Arch. Gerontol. Geriatr 6, 209e223. [DOI] [PubMed] [Google Scholar]
- Hanamsagar R, Bilbo SD, 2016. Sex differences in neurodevelopmental and neurodegenerative disorders: focus on microglial function and neuroinflammation during development. J. Steroid Biochem. Mol. Biol 160, 127. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Harold D, Abraham R, Hollingworth P, Sims R, Gerrish A, Hamshere ML, Pahwa JS, Moskvina V, Dowzell K, Williams A, Jones N, Thomas C, Stretton A, Morgan AR, Lovestone S, Powell J, Proitsi P, Lupton MK, Brayne C, Rubinsztein DC, Gill M, Lawlor B, Lynch A, Morgan K, Brown KS, Passmore PA, Craig D, McGuinness B, Todd S, Holmes C, Mann D, Smith AD, Love S, Kehoe PG, Hardy J, Mead S, Fox N, Rossor M, Collinge J, Maier W, Jessen F, Schurmann B, Heun R, van den Bussche H, Heuser I, Kornhuber J, Wiltfang J, Dichgans M, Frolich L, Hampel H, Hull M, Rujescu D, Goate AM, Kauwe JS, Cruchaga C, Nowotny P, Morris JC, Mayo K, Sleegers K, Bettens K, Engelborghs S, De Deyn PP, Van Broeckhoven C, Livingston G, Bass NJ, Gurling H, McQuillin A, Gwilliam R, Deloukas P, Al-Chalabi A, Shaw CE, Tsolaki M, Singleton AB, Guerreiro R, Muhleisen TW, Nothen MM, Moebus S, Jockel KH, Klopp N, Wichmann HE, Carrasquillo MM, Pankratz VS, Younkin SG, Holmans PA, O’Donovan M, Owen MJ, Williams J, 2009. Genome-wide association study identifies variants at CLU and PICALM associated with Alzheimer’s disease. Nat. Genet 41, 1088e1093. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hebert LE, Scherr PA, McCann JJ, Beckett LA, Evans DA, 2001. Is the risk of developing Alzheimer’s disease greater for women than for men? Am. J. Epidemiol 153, 132e136. [DOI] [PubMed] [Google Scholar]
- Hebert LE, Weuve J, Scherr PA, Evans DA, 2013. Alzheimer disease in the United States (2010–2050) estimated using the 2010 census. Neurology 80, 1778e1783. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Henderson VW, Paganini-Hill A, Miller BL, Elble RJ, Reyes PF, Shoupe D, McCleary CA, Klein RA, Hake AM, Farlow MR, 2000. Estrogen for Alzheimer’s disease in women: randomized, double-blind, placebo-controlled trial. Neurology 54, 295e301. [DOI] [PubMed] [Google Scholar]
- Heneka MT, Carson MJ, Khoury JE, Landreth GE, Brosseron F, Feinstein DL, Jacobs AH, Wyss-Coray T, Vitorica J, Ransohoff RM, Herrup K, Frautschy SA, Finsen B, Brown GC, Verkhratsky A, Yamanaka K, Koistinaho J, Latz E, Halle A, Petzold GC, Town T, Morgan D, Shinohara ML, Perry VH, Holmes C, Bazan NG, Brooks DJ, Hunot S, Joseph B, Deigendesch N, Garaschuk O, Boddeke E, Dinarello CA, Breitner JC, Cole GM, Golenbock DT, Kummer MP, 2015. Neuroinflammation in Alzheimer’s disease. Lancet Neurol. 14, 388e405. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Holland D, Desikan RS, Dale AM, McEvoy LK, 2013. Higher rates of decline for women and APOE ε4 carriers. AJNR Am. J. Neuroradiol 34, 2287. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Holmes C, 2013. Review: systemic inflammation and Alzheimer’s disease. Neuropathol. Appl. Neurobiol 39, 51e68. [DOI] [PubMed] [Google Scholar]
- Holmes C, Cunningham C, Zotova E, Woolford J, Dean C, Kerr S, Culliford D, Perry VH, 2009. Systemic inflammation and disease progression in Alzheimer disease. Neurology 73, 768e774. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hong S, Beja-Glasser VF, Nfonoyim BM, Frouin A, Li S, Ramakrishnan S, Merry KM, Shi Q, Rosenthal A, Barres BA, Lemere CA, Selkoe DJ, Stevens B, 2016. Complement and microglia mediate early synapse loss in Alzheimer mouse models. Science 352, 712. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hoogendijk WJ, Meynen G, Endert E, Hofman MA, Swaab DF, 2006. Increased cerebrospinal fluid cortisol level in Alzheimer’s disease is not related to depression. Neurobiol. Aging 27, 780.e1e780.e2. [DOI] [PubMed] [Google Scholar]
- Hua X, Hibar DP, Lee S, Toga AW, Jack CR Jr., Weiner MW, Thompson PM, 2010. Sex and age differences in atrophic rates: an ADNI study with n¼1368 MRI scans. Neurobiol. Aging 31, 1463e1480. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hwang CJ, Park MH, Choi MK, Choi JS, Oh KW, Hwang DY, Han SB,Hong JT, 2016. Acceleration of amyloidogenesis and memory impairment by estrogen deficiency through NF-kappaΒ dependent beta-secretase activation in presenilin 2 mutant mice. Brain Behav. Immun 53, 113e122. [DOI] [PubMed] [Google Scholar]
- Irvine K, Laws KR, Gale TM, Kondel TK, 2012. Greater cognitive deterioration in women than men with Alzheimer’s disease: a meta analysis. J. Clin. Exp. Neuropsychol 34, 989e998. [DOI] [PubMed] [Google Scholar]
- Jagger C, Matthews FE, Wohland P, Fouweather T, Stephan BCM, Robinson L, Arthur A, Brayne C Medical Research Council Cognitive Function and Ageing Collaboration, 2016. A comparison of health expectancies over two decades in England: results of the Cognitive Function and Ageing Study I and II. Lancet 387, 779e786. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Janicki SC, Park N, Cheng R, Clark LN, Lee JH, Schupf N, 2014. Estrogen receptor alpha variants affect age at onset of Alzheimer’s disease in a multiethnic female cohort. Dement. Geriatr. Cogn. Disord 38, 200e213. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Jeong YH, Park CH, Yoo J, Shin KY, Ahn SM, Kim HS, Lee SH, Emson PC,Suh YH, 2006. Chronic stress accelerates learning and memory impairments and increases amyloid deposition in APPV717I-CT100 transgenic mice, an Alzheimer’s disease model. FASEB J. 20, 729e731. [DOI] [PubMed] [Google Scholar]
- Ji K, Akgul G, Wollmuth LP, Tsirka SE, 2013. Microglia actively regulate the number of functional synapses. PLoS One 8, e56293. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Jin J-J, Kim H-D, Maxwell JA, Li L, Fukuchi K. i., 2008. Toll-like receptor 4-dependent upregulation of cytokines in a transgenic mouse model of Alzheimer’s disease. J. Neuroinflammation 5, 23. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kang JE, Cirrito JR, Dong H, Csernansky JG, Holtzman DM, 2007. Acute stress increases interstitial fluid amyloid-beta via corticotropin-releasing factor and neuronal activity. Proc. Natl. Acad. Sci. U. S. A 104, 10673e10678. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kivity S, Ehrenfeld M, 2010. Can we explain the higher prevalence of autoimmune disease in women? Expert Rev. Clin. Immunol 6, 691e694. [DOI] [PubMed] [Google Scholar]
- Klein SL, 2000. The effects of hormones on sex differences in infection: from genes to behavior. Neurosci. Biobehav. Rev 24, 627e638. [DOI] [PubMed] [Google Scholar]
- Koran ME, Wagener M, Hohman TJ, 2017. Sex differences in the association between AD biomarkers and cognitive decline. Brain Imaging Behav. 11, 205e213. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kovacs EJ, Messingham KAN, Gregory MS, 2002. Estrogen regulation of immune responses after injury. Mol. Cell Endocrinol 193, 129e135. [DOI] [PubMed] [Google Scholar]
- Kowian’ski P, Lietzau G, Czuba E, Wa’skow M, Steliga A, Mory’s J, 2017. BDNF: a key factor with multipotent impact on brain signaling and synaptic plasticity. Cell Mol. Neurobiol 38 (3), 579e593. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kukull WA, Higdon R, Bowen JD, McCormick WC, Teri L, Schellenberg GD, Belle GV, Jolley L, Larson EB, 2002. Dementia and Alzheimer disease incidence: a prospective cohort study. Arch. Neurol 59, 1737e1746. [DOI] [PubMed] [Google Scholar]
- Lambert JC, Heath S, Even G, Campion D, Sleegers K, Hiltunen M, Combarros O, Zelenika D, Bullido MJ, Tavernier B, Letenneur L, Bettens K, Berr C, Pasquier F, Fievet N, Barberger-Gateau P, Engelborghs S, De Deyn P, Mateo I, Franck A, Helisalmi S, Porcellini E, Hanon O, de Pancorbo MM, Lendon C, Dufouil C, Jaillard C, Leveillard T, Alvarez V, Bosco P, Mancuso M, Panza F, Nacmias B, Bossu P, Piccardi P, Annoni G, Seripa D, Galimberti D, Hannequin D, Licastro F, Soininen H, Ritchie K, Blanche H, Dartigues JF, Tzourio C, Gut I, Van Broeckhoven C, Alperovitch A, Lathrop M, Amouyel P, 2009. Genome-wide association study identifies variants at CLU and CR1 associated with Alzheimer’s disease. Nat. Genet 41, 1094e1099. [DOI] [PubMed] [Google Scholar]
- Laurin D, David Curb J, Masaki KH, White LR, Launer LJ, 2009. Midlife C-reactive protein and risk of cognitive decline: a 31-year follow-up. Neurobiol. Aging 30, 1724e1727. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lehmann DJ, Refsum H, Nurk E, Warden DR, Tell GS, Vollset SE, Engedal K, Nygaard HA, Smith AD, 2006. Apolipoprotein E epsilon4 and impaired episodic memory in community-dwelling elderly people: a marked sex difference. The Hordaland Health Study. J. Neurol. Neurosurg. Psychiatry 77, 902e908. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lenz KM, McCarthy MM, 2015. A starring role for microglia in brain sex differences. Neuroscientist 21, 306e321. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Levin-Allerhand JA, Smith JD, 2002. Ovariectomy of young mutant amyloid precursor protein transgenic mice leads to increased mortality. J. Mol. Neurosci 19, 163e166. [DOI] [PubMed] [Google Scholar]
- Li G, Shofer JB, Petrie EC, Yu C-E, Wilkinson CW, Figlewicz DP, Shutes-David A, Zhang J, Montine TJ, Raskind MA, Quinn JF, Galasko DR, Peskind ER, 2017a. Cerebrospinal fluid biomarkers for Alzheimer’s and vascular disease vary by age, gender, and APOE genotype in cognitively normal adults. Alzheimers Res. Ther 9, 48. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Li G-D, Bi R, Zhang D-F, Xu M, Luo R, Wang D, Fang Y, Li T, Zhang C, Yao Y-G, 2017b. Female-specific effect of the BDNF gene on Alzheimer’s disease. Neurobiol. Aging 53, 192.e11e192.e19. [DOI] [PubMed] [Google Scholar]
- Lim YY, Villemagne VL, Laws SM, Pietrzak RH, Snyder PJ, Ames D, Ellis KA, Harrington K, Rembach A, Martins RN, Rowe CC, Masters CL, Maruff P, 2015. APOE and BDNF polymorphisms moderate amyloid b-related cognitive decline in preclinical Alzheimer’s disease. Mol. Psychiatry 20, 1322e1328. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lin KA, Choudhury KR, Rathakrishnan BG, Marks DM, Petrella JR, Doraiswamy PM, 2015. Marked gender differences in progression of mild cognitive impairment over 8 years. Alzheimers Dement (N Y) 1, 103e110. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lin KA, Doraiswamy PM, 2014. When mars versus venus is not a cliche: gender differences in the neurobiology of Alzheimer’s disease. Front. Neurol 5, 288. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Liu CC, Kanekiyo T, Xu H, Bu G, 2013. Apolipoprotein E and Alzheimer disease: risk, mechanisms and therapy. Nat. Rev. Neurol 9, 106e118. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lue LF, Brachova L, Civin WH, Rogers J, 1996. Inflammation, A beta deposition, and neurofibrillary tangle formation as correlates of Alzheimer’s disease neurodegeneration. J. Neuropathol. Exp. Neurol 55, 1083e1088. [PubMed] [Google Scholar]
- Masters CL, Bateman R, Blennow K, Rowe CC, Sperling RA, Cummings JL, 2015. Alzheimer’s disease. Nat. Rev. Dis. Primers 1, 15056. [DOI] [PubMed] [Google Scholar]
- Matyi J, Tschanz JT, Rattinger GB, Sanders C, Vernon EK, Corcoran C, Kauwe JSK, Buhusi M, 2017. Sex differences in risk for Alzheimer’s disease related to neurotrophin gene polymorphisms: the Cache county memory study. J. Gerontol. A. Biol. Sci. Med. Sci 72, 1607e1613. [DOI] [PMC free article] [PubMed] [Google Scholar]
- McClelland EE, Smith JM, 2011. Gender specific differences in the immune response to infection. Arch. Immunol. Ther. Exp 59, 203e213. [DOI] [PubMed] [Google Scholar]
- Miech RA, Breitner JC, Zandi PP, Khachaturian AS, Anthony JC, Mayer L, 2002. Incidence of AD may decline in the early 90s for men, later for women: the Cache County study. Neurology 58 (2), 209e218. [DOI] [PubMed] [Google Scholar]
- Mielke MM, Vemuri P, Rocca WA, 2014. Clinical epidemiology of Alzheimer’s disease: assessing sex and gender differences. Clin. Epidemiol 6, 37e48. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Morrison JH, Brinton RD, Schmidt PJ, Gore AC, 2006. Estrogen, menopause, and the aging brain: how basic neuroscience can inform hormone therapy in women. J. Neurosci 26, 10332e10348. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Mortensen EL, Hogh P, 2001. A gender difference in the association between APOE genotype and age-related cognitive decline. Neurology 57, 89e95. [DOI] [PubMed] [Google Scholar]
- Mucke L, Selkoe DJ, 2012. Neurotoxicity of amyloid b-protein: synaptic and network dysfunction. Cold Spring Harbor Perspect. Med 2, a006338. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Mulnard RA, Cotman CW, Kawas C, van Dyck CH, Sano M, Doody R, Koss E, Pfeiffer E, Jin S, Gamst A, Grundman M, Thomas R, Thal LJ, 2000. Estrogen replacement therapy for treatment of mild to moderate Alzheimer disease: a randomized controlled trial. Alzheimer’s Disease Cooperative Study. JAMA 283, 1007e1015. [DOI] [PubMed] [Google Scholar]
- Neu SC, Pa J, Kukull W, Beekly D, Kuzma A, Gangadharan P, Wang L-S, Romero K, Arneric SP, Redolfi A, Orlandi D, Frisoni GB, Au R, Devine S, Auerbach S, Espinosa A, Boada M, Ruiz A, Johnson SC, Koscik R, Wang J-J, Hsu W-C, Chen Y-L, Toga AW, 2017. Apolipoprotein E genotype and sex risk factors for Alzheimer disease: a meta-analysis. JAMA Neurol. 74, 1178e1189. [DOI] [PMC free article] [PubMed] [Google Scholar]
- O’Brien JT, Ames D, Schweitzer I, Colman P, Desmond P, Tress B, 1996. Clinical and magnetic resonance imaging correlates of hypothalamic-pituitary-adrenal axis function in depression and Alzheimer’s disease. Br. J. Psychiatry 168, 679e687. [DOI] [PubMed] [Google Scholar]
- Overk CR, Perez SE, Ma C, Taves MD, Soma KK, Mufson EJ, 2013. Sex steroid levels and AD-like pathology in 3xTgAD mice. J. Neuroendocrinol 25, 131e144. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Paganini-Hill A, Henderson VW, 1994. Estrogen deficiency and risk of Alzheimer’s disease in women. Am. J. Epidemiol 140, 256e261. [DOI] [PubMed] [Google Scholar]
- Park HJ, Ran Y, Jung JI, Holmes O, Price AR, Smithson L, Ceballos-Diaz C, Han C, Wolfe MS, Daaka Y, Ryabinin AE, Kim SH, Hauger RL, Golde TE, Felsenstein KM, 2015. The stress response neuropeptide CRF increases amyloid-beta production by regulating gamma-secretase activity. EMBO J. 34, 1674e1686. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Patel NS, Paris D, Mathura V, Quadros AN, Crawford FC, Mullan MJ, 2005. Inflammatory cytokine levels correlate with amyloid load in transgenic mouse models of Alzheimer’s disease. J. Neuroinflammation 2, 9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Payami H, Montee KR, Kaye JA, Bird TD, Yu CE, Wijsman EM, Schellenberg GD, 1994. Alzheimer’s disease, apolipoprotein E4, and gender. JAMA 271, 1316e1317. [PubMed] [Google Scholar]
- Pedersen WA, Wan R, Mattson MP, 2001. Impact of aging on stress-responsive neuroendocrine systems. Mech. Ageing Dev 122, 963e983. [DOI] [PubMed] [Google Scholar]
- Perry VH, Holmes C, 2014. Microglial priming in neurodegenerative disease. Nat. Rev. Neurol 10, 217e224. [DOI] [PubMed] [Google Scholar]
- Pike CJ, Carroll JC, Rosario ER, Barron AM, 2009. Protective actions of sex steroid hormones in Alzheimer’s disease. Front. Neuroendocrinol 30, 239e258. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Plassman BL, Langa KM, Fisher GG, Heeringa SG, Weir DR, Ofstedal MB, Burke JR, Hurd MD, Potter GG, Rodgers WL, Steffens DC, Willis RJ, Wallace RB, 2007. Prevalence of dementia in the United States: the aging, demographics, and memory study. Neuroepidemiology 29, 125e132. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Poirier J, Davignon J, Bouthillier D, Kogan S, Bertrand P, Gauthier S, 1993. Apolipoprotein E polymorphism and Alzheimer’s disease. Lancet 342, 697e699. [DOI] [PubMed] [Google Scholar]
- Qin XY, Cao C, Cawley NX, Liu TT, Yuan J, Loh YP, Cheng Y, 2017. Decreased peripheral brain-derived neurotrophic factor levels in Alzheimer’s disease: a meta-analysis study (N¼7277). Mol. Psychiatry 22, 312e320. [DOI] [PubMed] [Google Scholar]
- Raber J, Wong D, Buttini M, Orth M, Bellosta S, Pitas RE, Mahley RW, Mucke L, 1998. Isoform-specific effects of human apolipoprotein E on brain function revealed in ApoE knockout mice: increased susceptibility of females. Proc. Natl. Acad. Sci. U. S. A 95, 10914e10919. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Rasmuson S, Nasman B, Olsson T, 2011. Increased serum levels of dehydroepiandrosterone (DHEA) and interleukin-6 (IL-6) in women with mild to moderate Alzheimer’s disease. Int. Psychogeriatr 23, 1386e1392. [DOI] [PubMed] [Google Scholar]
- Rehman HU, 2002. Role of CRH in the pathogenesis of dementia of Alzheimer’s type and other dementias. Curr. Opin. Investig. Drugs 3, 1637e1642. [PubMed] [Google Scholar]
- Reisberg B, Borenstein J, Salob SP, Ferris SH, Franssen E, Georgotas A, 1987. Behavioral symptoms in Alzheimer’s disease: phenomenology and treatment. J. Clin. Psychiatry 48 (Suppl), 9e15. [PubMed] [Google Scholar]
- Riedel BC, Thompson PM, Brinton RD, 2016. Age, APOE and sex: triad of risk of Alzheimer’s disease. J. Steroid Biochem. Mol. Biol 160, 134e147. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Rissman RA, Staup MA, Lee AR, Justice NJ, Rice KC, Vale W, Sawchenko PE, 2012. Corticotropin-releasing factor receptor-dependent effects of repeated stress on tau phosphorylation, solubility, and aggregation. Proc. Natl. Acad. Sci. U. S. A 109, 6277e6282. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Robert SJ, Zugaza JL, Fischmeister R, Gardier AM, Lezoualc’h F, 2001. The human serotonin 5-HT4 receptor regulates secretion of non-amyloidogenic precursor protein. J. Biol. Chem 276, 44881e44888. [DOI] [PubMed] [Google Scholar]
- Roberts RO, Geda YE, Knopman DS, Cha RH, Pankratz VS, Boeve BF, Tangalos EG, Ivnik RJ, Rocca WA, Petersen RC, 2012. The incidence of MCI differs by subtype and is higher in men: the Mayo Clinic Study of Aging. Neurology 78, 342e351. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Rocca WA, Cha RH, Waring SC, Kokmen E, 1998. Incidence of dementia and Alzheimer’s disease: a reanalysis of data from Rochester, Minnesota, 1975–1984. Am. J. Epidemiol 148, 51e62. [DOI] [PubMed] [Google Scholar]
- Rocca WA, Grossardt BR, Shuster LT, 2011. Oophorectomy, menopause, estrogen treatment, and cognitive aging: clinical evidence for a window of opportunity. Brain Res. 1379, 188e198. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Rohe M, Synowitz M, Glass R, Paul SM, Nykjaer A, Willnow TE, 2009. Brain-Derived neurotrophic factor reduces amyloidogenic processing through control of SORLA gene expression. J. Neurosci 29, 15472e15478. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Rosario ER, Carroll J, Pike CJ, 2010. Testosterone regulation of Alzheimer-like neuropathology in male 3xTg-AD mice involves both estrogen and androgen pathways. Brain Res 1359, 281e290. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Rosario ER, Chang L, Head EH, Stanczyk FZ, Pike CJ, 2011. Brain levels of sex steroid hormones in men and women during normal aging and in Alzheimer’s disease. Neurobiol. Aging 32, 604e613. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sampedro F, Vilaplana E, de Leon MJ, Alcolea D, Pegueroles J, Montal V, Carmona-Iragui M, Sala I, Sanchez-Saudinos MB, Anton-Aguirre S, Morenas-Rodriguez E, Camacho V, Falcon C, Pavia J, Ros D, Clarimon J, Blesa R, Lleo A, Fortea J, 2015. APOE-by-sex interactions on brain structure and metabolism in healthy elderly controls. Oncotarget 6, 26663e26674. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sanchez-Mut JV, Aso E, Heyn H, Matsuda T, Bock C, Ferrer I, Esteller M, 2014. Promoter hypermethylation of the phosphatase DUSP22 mediates PKA-dependent TAU phosphorylation and CREB activation in Alzheimer’s disease. Hippocampus 24, 363e368. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sarkar S, Jun S, Simpkins JW, 2015. Estrogen amelioration of Abeta-induced defects in mitochondria is mediated by mitochondrial signaling pathway involving ERbeta, AKAP and Drp1. Brain Res. 1616, 101e111. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Scharfman HE, MacLusky NJ, 2006. Estrogen and brain-derived neurotrophic factor (BDNF) in hippocampus: complexity of steroid hormone-growth factor interactions in the adult CNS. Front. Neuroendocrinol 27, 415e435. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Schneider JA, Arvanitakis Z, Leurgans SE, Bennett DA, 2009. The neuropathology of probable Alzheimer disease and mild cognitive impairment. Ann. Neurol 66, 200e208. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Schwarz JM, Sholar PW, Bilbo SD, 2012. Sex differences in microglial colonization of the developing rat brain. J. Neurochem 120, 948. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Selmi C, 2008. The X in sex: how autoimmune diseases revolve around sex chromosomes. Best Pract. Res. Clin. Rheumatol 22, 913e922. [DOI] [PubMed] [Google Scholar]
- Sen A, Nelson TJ, Alkon DL, 2015. ApoE4 and Aβ oligomers reduce BDNF expression via HDAC nuclear translocation. J. Neurosci 35, 7538e7551. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sen A, Nelson TJ, Alkon DL, 2017. ApoE isoforms differentially regulates cleavage and secretion of BDNF. Mol. Brain 10, 19. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Seshadri S, Wolf PA, Beiser A, Au R, McNulty K, White R, D’Agostino RB, 1997. Lifetime risk of dementia and Alzheimer’s disease. The impact of mortality on risk estimates in the Framingham Study. Neurology 49, 1498e1504. [DOI] [PubMed] [Google Scholar]
- Shumaker SA, Legault C, Rapp SR, Thal L, Wallace RB, Ockene JK, Hendrix SL, Jones BN 3rd, Assaf AR, Jackson RD, Kotchen JM, Wassertheil-Smoller S, Wactawski-Wende J, 2003. Estrogen plus progestin and the incidence of dementia and mild cognitive impairment in postmenopausal women: the Women’s Health Initiative Memory Study: a randomized controlled trial. JAMA 289, 2651e2662. [DOI] [PubMed] [Google Scholar]
- Sierksma AS, Vanmierlo T, De Vry J, Raijmakers ME, Steinbusch HW, van den Hove DL, Prickaerts J, 2012. Effects of prenatal stress exposure on soluble Abeta and brain-derived neurotrophic factor signaling in male and female APPswe/PS1dE9 mice. Neurochem. Int 61, 697e701. [DOI] [PubMed] [Google Scholar]
- Sotiropoulos I, Silva J, Kimura T, Rodrigues AJ, Costa P, Almeida OF, Sousa N, Takashima A, 2015. Female hippocampus vulnerability to environmental stress, a precipitating factor in Tau aggregation pathology. J. Alzheimers Dis 43, 763e774. [DOI] [PubMed] [Google Scholar]
- Sundermann EE, Biegon A, Rubin LH, Lipton RB, Landau S, Maki PM, 2017. Does the female advantage in verbal memory contribute to underestimating Alzheimer’s disease pathology in women versus men? J. Alzheimers Dis 56, 947e957. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sundermann EE, Biegon A, Rubin LH, Lipton RB, Mowrey W, Landau S, Maki PM Alzheimer’s Disease Neuroimaging Initiative, 2016. Better verbal memory in women than men in MCI despite similar levels of hippocampal atrophy. Neurology 86, 1368e1376. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Swanwick GR, Kirby M, Bruce I, Buggy F, Coen RF, Coakley D, Lawlor BA, 1998. Hypothalamic-pituitary-adrenal axis dysfunction in Alzheimer’s disease: lack of association between longitudinal and cross-sectional findings. Am. J. Psychiatry 155, 286e289. [DOI] [PubMed] [Google Scholar]
- Tai LM, Thomas R, Marottoli FM, Koster KP, Kanekiyo T, Morris AWJ, Bu G, 2016. The role of APOE in cerebrovascular dysfunction. Acta. Neuropathol 131, 709e723. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Tamagno E, Guglielmotto M, Giliberto L, Vitali A, Borghi R, Autelli R, Danni O, Tabaton M, 2009. JNK and ERK1/2 pathways have a dual opposite effect on the expression of BACE1. Neurobiol. Aging 30, 1563e1573. [DOI] [PubMed] [Google Scholar]
- Tang MX, Jacobs D, Stern Y, Marder K, Schofield P, Gurland B, Andrews H, Mayeux R, 1996. Effect of oestrogen during menopause on risk and age at onset of Alzheimer’s disease. Lancet 348, 429e432. [DOI] [PubMed] [Google Scholar]
- Terry RD, Masliah E, Salmon DP, Butters N, DeTeresa R, Hill R, Hansen LA, Katzman R, 1991. Physical basis of cognitive alterations in Alzheimer’s disease: synapse loss is the major correlate of cognitive impairment. Ann. Neurol 30, 572e580. [DOI] [PubMed] [Google Scholar]
- Thathiah A, De Strooper B, 2011. The role of G protein-coupled receptors in the pathology of Alzheimer’s disease. Nat. Rev. Neurosci 12, 73e87. [DOI] [PubMed] [Google Scholar]
- Thathiah A, Horre K, Snellinx A, Vandewyer E, Huang Y, Ciesielska M, De Kloe G, Munck S, De Strooper B, 2013. Beta-arrestin 2 regulates Abeta generation and gamma-secretase activity in Alzheimer’s disease. Nat. Med 19, 43e49. [DOI] [PubMed] [Google Scholar]
- Trojanowski JQ, Revesz T Neuropathology Working Group on, M.S.A, 2007. Proposed neuropathological criteria for the post mortem diagnosis of multiple system atrophy. Neuropathol. Appl. Neurobiol 33, 615e620. [DOI] [PubMed] [Google Scholar]
- Umegaki H, Ikari H, Nakahata H, Endo H, Suzuki Y, Ogawa O, Nakamura A, Yamamoto T, Iguchi A, 2000. Plasma cortisol levels in elderly female subjects with Alzheimer’s disease: a cross-sectional and longitudinal study. Brain Res. 881, 241e243. [DOI] [PubMed] [Google Scholar]
- Ungar L, Altmann A, Greicius MD, 2014. Apolipoprotein E, gender, and Alzheimer’s disease: an overlooked, but potent and promising interaction. Brain Imaging Behav. 8, 262e273. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Verdile G, Laws SM, Henley D, Ames D, Bush AI, Ellis KA, Faux NG, Gupta VB, Li QX, Masters CL, Pike KE, Rowe CC, Szoeke C, Taddei K, Villemagne VL, Martins RN, 2014. Associations between gonadotropins, testosterone and beta amyloid in men at risk of Alzheimer’s disease. Mol. Psychiatry 19, 69e75. [DOI] [PubMed] [Google Scholar]
- Wahrendorf M, Reinhardt JD, Siegrist J, 2013. Relationships of disability with age among adults aged 50 to 85: evidence from the United States, England and Continental Europe. PLoS One 8, e71893. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wang J, Tanila H, Puolivali J, Kadish I, van Groen T, 2003. Gender differences in the amount and deposition of amyloidbeta in APPswe and PS1 double transgenic mice. Neurobiol. Dis 14, 318e327. [DOI] [PubMed] [Google Scholar]
- Wang PN, Liao SQ, Liu RS, Liu CY, Chao HT, Lu SR, Yu HY, Wang SJ, Liu HC, 2000. Effects of estrogen on cognition, mood, and cerebral blood flow in AD: a controlled study. Neurology 54, 2061e2066. [DOI] [PubMed] [Google Scholar]
- Wang Y, Cella M, Mallinson K, Ulrich JD, Young KL, Robinette ML, Gifillian S, Krishnan GM, Sudhakar S, Zinselmeyer BH, Holtzman DM, Cirrito JR, Colonna M, 2015. TREM2 lipid sensing sustains microglia response in an Alzheimer’s disease model. Cell 160, 1061. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wang YQ, Qu DH, Wang K, 2016. Therapeutic approaches to Alzheimer’s disease through stimulating of non-amyloidogenic processing of amyloid precursor protein. Eur. Rev. Med. Pharmacol. Sci 20, 2389e2403. [PubMed] [Google Scholar]
- Ward DD, Summers MJ, Saunders NL, Janssen P, Stuart KE, Vickers JC, 2014. APOE and BDNF Val66Met polymorphisms combine to influence episodic memory function in older adults. Behav. Brain Res 271, 309e315. [DOI] [PubMed] [Google Scholar]
- Whiteford HA, Ferrari AJ, Degenhardt L, Feigin V, Vos T, 2015. The global burden of mental, neurological and substance use disorders: an analysis from the global burden of disease study 2010. PLoS One 10, e0116820. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Whitmer RA, Sidney S, Selby J, Johnston SC, Yaffe K, 2005. Midlife cardiovascular risk factors and risk of dementia in late life. Neurology 64, 277e281. [DOI] [PubMed] [Google Scholar]
- Willem M, Tahirovic S, Busche MA, Ovsepian SV, Chafai M, Kootar S, Hornburg D, Evans LD, Moore S, Daria A, Hampel H, Muller V, Giudici C, Nuscher B, Wenninger-Weinzierl A, Kremmer E, Heneka MT, Thal DR, Giedraitis V, Lannfelt L, Muller U, Livesey FJ, Meissner F, Herms J, Konnerth A, Marie H, Haass C, 2015. Eta-Secretase processing of APP inhibits neuronal activity in the hippocampus. Nature 526, 443e447. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wolfe MS, 2013. Alzheimer’s gamma-secretase under arrestin. Nat. Med 19, 22e24. [DOI] [PubMed] [Google Scholar]
- Xu H, Sweeney D, Greengard P, Gandy S, 1996. Metabolism of Alzheimer beta-amyloid precursor protein: regulation by protein kinase A in intact cells and in a cell-free system. Proc. Natl. Acad. Sci. U. S. A 93, 4081e4084. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Yeh F-C, Kao C-F, Kuo P-H, 2015. Explore the features of brain-derived neurotrophic factor in mood disorders. PLoS One 10, e0128605. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zandi PP, Carlson MC, Plassman BL, Welsh-Bohmer KA, Mayer LS, Steffens DC, Breitner JCS Cache County Memory Study Investigators, 2002. Hormone replacement therapy and incidence of Alzheimer disease in older women: the Cache county study. JAMA 288, 2123e2129. [DOI] [PubMed] [Google Scholar]
- Zhang B, Gaiteri C, Bodea L-G, Wang Z, McElwee J, Podtelezhnikov AA, Zhang C, Xie T, Tran L, Dobrin R, Fluder E, Clurman B, Melquist S, Narayanan M, Suver C, Shah H, Mahajan M, Gillis T, Mysore J, MacDonald ME, Lamb JR, Bennett DA, Molony C, Stone DJ, Gudnason V, Myers AJ, Schadt EE, Neumann H, Zhu J, Emilsson V, 2013. Integrated systems approach identifies genetic nodes and networks in late-onset Alzheimer’s disease. Cell 153, 707e720. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zheng H, Xu H, Uljon SN, Gross R, Hardy K, Gaynor J, Lafrancois J, Simpkins J, Refolo LM, Petanceska S, Wang R, Duff K, 2002. Modulation of A(beta) peptides by estrogen in mouse models. J. Neurochem 80, 191e196. [DOI] [PubMed] [Google Scholar]