2,562
Views
560
CrossRef citations to date
0
Altmetric
Review

Clinical epidemiology of Alzheimer’s disease: assessing sex and gender differences

, &
Pages 37-48 | Published online: 08 Jan 2014

Abstract

With the aging of the population, the burden of Alzheimer’s disease (AD) is rapidly expanding. More than 5 million people in the US alone are affected with AD and this number is expected to triple by 2050. While men may have a higher risk of mild cognitive impairment (MCI), an intermediate stage between normal aging and dementia, women are disproportionally affected with AD. One explanation is that men may die of competing causes of death earlier in life, so that only the most resilient men may survive to older ages. However, many other factors should also be considered to explain the sex differences. In this review, we discuss the differences observed in men versus women in the incidence and prevalence of MCI and AD, in the structure and function of the brain, and in the sex-specific and gender-specific risk and protective factors for AD. In medical research, sex refers to biological differences such as chromosomal differences (eg, XX versus XY chromosomes), gonadal differences, or hormonal differences. In contrast, gender refers to psychosocial and cultural differences between men and women (eg, access to education and occupation). Both factors play an important role in the development and progression of diseases, including AD. Understanding both sex- and gender-specific risk and protective factors for AD is critical for developing individualized interventions for the prevention and treatment of AD.

Introduction

Alzheimer’s disease (AD) is the most prevalent type of dementia, comprising about 60%–70% of all dementia cases.Citation1 Beta-amyloid plaques, neurofibrillary tangles, and neurodegeneration are the hallmark pathologic characteristics of AD. Clinically, AD is a progressive disorder characterized by loss of memory and overall cognitive functioning and by behavioral symptoms such as apathy, depression, and anxiety; vocabulary and crystallized abilities are preserved. The burden of AD is high, with more than 5 million people currently affected in the US alone. Presently, one in nine people aged 65 and older has AD and more than one in three people aged 85 and older are affected.Citation2 With the increasing age of the population, it is estimated that 14–16 million Americans will be diagnosed with the disease by 2050 unless new treatments to prevent or delay the onset of AD are identified.Citation3,Citation4 Women are disproportionally affected by AD; they are more likely to become caregivers to AD patients, and are also more likely to develop AD.Citation2 In contrast, some studies suggest that men are at greater risk of developing mild cognitive impairment (MCI), a state between the normal cognitive changes associated with aging and early dementia.Citation5,Citation6 Sex-related differences in the rate of progression after a diagnosis of AD and in the response to treatments have also been reported. In the present review, we will first summarize sex differences in cognitive aging, in the prevalence and incidence of MCI and AD, and in the rate of progression after an AD diagnosis. We will then discuss sex-specific differences in the neuroimaging measures used to study AD. Lastly, we will review potential reasons for the described differences between men and women considering factors both related to biology (sex) and to society and culture (gender).Citation7Citation9

Sex versus gender

An Institute of Medicine report published in 2001 concluded that “being male or female is an important fundamental variable that should be considered when designing and analyzing basic and clinical research.”Citation7 In medical research, sex refers to biological differences such as chromosomal (eg, XX versus XY chromosomes), gonadal, or hormonal differences. In contrast, gender refers to psychosocial and cultural differences between men and women (eg, access to education and occupation).Citation10 Both factors play an important role in the development and progression of diseases, including AD.Citation8,Citation9 In this review, potential sex and gender differences that may influence the difference in prevalence and incidence rates of MCI and AD among men and women will be discussed.

Sex differences in cognitive aging, in the prevalence and incidence of MCI and AD, and in the rate of progression after a diagnosis of AD

Cognitive aging

The shrinking of the brain and expansion of the ventricles is part of the natural maturational process of the brain during normal aging. Longitudinal studies that have used “normal” cognitive status as an inclusion criterion have found that even “normal” aging may contribute to subtle declines in cognitive functioning.Citation11,Citation12 There are significant sex differences in the normal aging process.Citation13 The most consistent cross-sectional difference at all ages is that women perform better on verbal memory tasks and men perform better on visuospatial tasks.Citation14Citation16 However, longitudinal studies have shown inconsistent sex differences, either reporting steeper annual rates of cognitive decline in men,Citation17 women,Citation14 or no sex differences.Citation18 One of the major factors causing these inconsistent findings may be the cognitive reserve profiles (discussed later) of specific cohorts (eg, the Lothian Birth cohorts of 1921 and 1936).Citation19

The trajectory of cognitive decline due to ongoing pathological insults to the brain (“pathological aging”) has been shown to deviate from the normal aging process. There is significant accelerated decline in cognitive functioning and brain volume loss years before the onset of MCI or dementia.Citation20Citation23 However, there have been no systematic studies of sex differences in the cognitive decline prior to onset of clinical symptoms (ie, preclinical stages of the disease).

MCI

MCI is considered an intermediate state between the cognitive changes associated with aging and mild dementia, particularly of the Alzheimer type.Citation24Citation26 Indeed, the risk of dementia is higher in persons with MCI compared to cognitively normal individuals.Citation27Citation30 The prevalence of MCI in persons older than 65 years of age ranges from 10% to 20%, depending on the population studied and on the diagnostic criteria utilized.Citation6,Citation27,Citation28,Citation31Citation34 Some studies suggest a higher prevalence of MCI in men,Citation6,Citation35,Citation36 while others suggest either a higher prevalence in womenCitation33,Citation37 or no sex difference.Citation38Citation40 The incidence rate of MCI has been estimated to be about 1%–4% per year in cognitively normal individuals aged 65 and older.Citation27,Citation40Citation42 Some studies examining the incidence of MCI also report that there may be sex differences, but reports vary based on the study design, diagnostic criteria, and age distribution of the sample. In general, women have a higher incidence of MCI at older ages. Men consistently have a higher incidence of the non-amnestic type of MCI.Citation42,Citation43 Amnestic MCI is defined as cognitive impairment that includes the memory domain whereas non-amnestic MCI refers to impairment in other domains (eg, executive functioning, visuospatial, language), but no impairment in memory. While amnestic MCI is considered prodromal for AD, non-amnestic MCI is considered prodromal for non-AD dementias, such as vascular dementia.Citation25

AD

The prevalence of AD is significantly higher in women compared to men. Recent estimates suggest that almost two-thirds of the individuals diagnosed with AD are women.Citation3 A reason for the higher prevalence among women may be that they live longer, on average, than men.Citation44,Citation45 By contrast, incidence studies examining sex differences in AD are equivocal. The majority of studies conducted in the US have not observed sex differences in the rates of developing AD.Citation46Citation52 In contrast to these studies, the Cache County Study (Cache County, UT, USA), did report a higher incidence of AD in men than women until age 78, after which women had a higher incidence than men.Citation53 Similarly, the Mayo Clinic Study of Aging recently reported that the rate of progression from MCI to AD was similar in men and women aged 70–79, but higher in women than men after age 80.Citation54 Consistent with these last two studies, most studies from EuropeanCitation55Citation59 and AsianCitation60,Citation61 populations have also observed a higher incidence in women after the age of 80–85 years.

The reasons for these disparities across studies and geographic regions are not clear. Discrepancies could be due to the use of different diagnostic criteria for AD versus other forms of dementia, such as vascular dementia or Lewy body dementia. The differences may also be due to small sample sizes at the upper range of the age distribution, resulting in unstable estimates. Finally, some differences across Europe, Asia, and North America may be due to social, cultural, and historical events. For example, the impact of World War II and the following Cold War era have been very different across continents. Some of these historical events may have affected men and women differently. Notably, a meta-analysis of 13 studies of populations in the US, Europe, and Asia did show that women were at a significantly greater risk of developing AD, but not other dementias.Citation62 Interestingly, women also have a faster rate of cognitive and functional decline after a diagnosis of AD.Citation63,Citation64

Sex differences in neuroimaging measures of brain reserve: structure and function

The concept of brain reserve posits that subjects with higher reserve have a greater capacity to cope with pathological insults than those with low reserve, and that these individual differences in reserve mechanisms help explain why cognitive decline may be initiated at different times in relation to the onset of pathology for each individual. Specifically, the concept of brain reserve stemmed from the observation by Katzman et al that subjects with larger brains have greater capacity to withstand more pathology at the same level of cognitive performance.Citation65 Cognitive reserve is discussed later in the review.

Structure

The most striking difference between the brain anatomy of men and women is the larger head size and cerebral brain volume in men (~10%).Citation66 Therefore, one would expect men to be able to withstand more pathology compared to women. This hypothesis was supported by an autopsy study that found that women had significantly higher odds of a clinical diagnosis of AD at the same level of pathology.Citation67 While overall larger head sizes may suggest larger brain reserve in men, studies have consistently shown faster age-associated brain volume decline in men compared to women in cognitively normal individuals.Citation68Citation71 However, in patients with MCI and AD, brain volumes have been found to decline faster in women than men, supporting the evidence of faster progression of women from MCI to AD.Citation72 Thus, even after considering difference in head size, sexual dimorphism in the brain anatomy exists.Citation66,Citation73Citation75 For example, women typically have a higher percentage of grey matter in several brain regions, whereas men have a higher percentage of white matter.Citation76 While many of these differences are likely due to sex chromosomes and sex hormones,Citation77Citation79 the exact mechanism through which sex hormones influence brain structures is still poorly understood. Notably, one major flaw of the studies examining changes in brain structure with age has been the modeling of brain volume loss over the life span without taking into account hormonal changes in men and women over time.

Function

Functional imaging measures such as 18F-fluorodeoxyglucose positron emission tomography (FDG-PET) for measuring metabolism and resting state functional magnetic resonance imaging for measuring brain connectivity have shown significant differences between men and women.Citation76,Citation80 Typically, cerebral blood flow and connectivity have been found to be higher in women in the parietal association cortices and higher for men in the visual and motor cortices,Citation81,Citation82 providing evidence for brain function and behavior differences between the sexes. Several imaging studies have shown that sex differences in the brain circuitry contribute to significant performance differences on specific cognitive tasks; for example, men perform better on visually oriented tasks. In the context of cerebral metabolic deficits associated with cognitive impairment in dementia, two studies have shown that men have more pronounced cerebral metabolic deficits compared to women at the same level of cognitive impairment, suggesting that the greater brain reserve in men may be helping them withstand more pathology than women at the same level of dementia severity.Citation83,Citation84 Given the current hypothesis that regions of high connectivity in the brain harbor amyloid deposition,Citation85 there is clearly a need to investigate sex differences in the pathological cascade of AD.

Biological explanations for the sex differences

Genetics

While many studies have examined and reported the relationship (or lack of) between numerous genes and single-nucleotide polymorphisms (SNPs) and risk of AD, few studies have specifically examined whether the relationships vary by sex. One reason for this is the high number of individuals needed for genetic analyses, and lack of power to examine sex differences, particularly for genome-wide association studies. As a result, studies adjust for sex rather than stratify by sex, or examine interactions with sex.Citation79 The identification of the different genetic processes that may affect the risk of MCI and AD in men and women is imperative for individualized preventive and treatment plans.

The ε4 allele of the apolipoprotein ε (APOE) gene is the strongest known genetic risk factor for late-onset AD.Citation86,Citation87 Compared to non-carriers, heterozygous carriers of one ε4 allele are 3–4 times more likely to develop AD, whereas the risk for homozygous carriers is even higher.Citation87,Citation88 The APOE ε4 allele is specifically associated with an earlier age of onset of AD.Citation89 The majority of studies, including a large meta-analysis of 8,607 controls and 5,930 AD cases,Citation90 have reported that the effects of the ε4 genotype are more pronounced in women than in men.Citation90 Three studies reported that women with one ε4 allele had about a four-fold risk of AD, whereas men with one ε4 allele showed little increased risk.Citation90Citation92 The APOE ε4 allele also has a greater deleterious effect on hippocampal pathology, functional connectivity changes in the default mode network, cortical thickness, and memory performance in women compared with men at different stages of AD.Citation93Citation95 Additionally, a large autopsy study found that amyloid plaque and neurofibrillary tangle pathology was greatest among women who were ε4 carriers.Citation96

Other genes and SNPs have also been shown to increase risk and progression of AD in one sex, but not the other. A large study consisting of 16 research centers worldwide (including 4,711 patients and 4,537 controls) reported that the Met66 allele of Brain Derived Neurotrophic Factor (BDNF) gene, which reduces the transport of BDNF, is associated with an increased risk of AD in women (odds ratio =1.14, 95% confidence interval 1.05–1.24, P=0.002), but not in men.Citation97 This finding is biologically plausible since estrogen plays an important role in the expression of BDNF.Citation98 Postmenopausal women with the MET66 allele would therefore have both reduced transport and expression of BDNF, thus causing an increased risk of AD.

SNPs found to pose a risk of AD among men, but not women, include a SNP (rs688) of the low-density lipoprotein receptor and functional apolipoprotein E receptor,Citation99 the rs17571 SNP of the lysosomal protease cathepsin D,Citation100 SOS2 (involved in signal transduction pathways, including insulin signaling), and PCK1 (catalyzes the first step in hepatic gluconeogenesis).Citation101 Interestingly, a few SNPs have also been found to have an opposite predictive value for women compared with men. A diabetes-related gene, the G allele of NSP65 of the peroxisome proliferators-activated receptors gamma was associated with a significantly increased odds of AD in men, but a reduced odds in women.Citation101 In contrast, the 219K allele of the ATP Binding Cassette Transporter 1 (ABCA1) gene had a 1.75-fold increased risk of developing AD in women, but was found to be protective in men.Citation102

The biological explanations for these sex differences are not fully understood, in part because the physiological effects of many of the genetic polymorphisms have not been completely determined. Most studies finding sex differences link the association to sex hormone levels. For example, some of the physiological benefits of estrogen have been linked to ABCA1-mediated pathways.Citation103 However, there could also be gene–gene interactions (epistasis) of genes on an autosomal chromosome with genes on chromosome X or Y. With the continued observation of sex differences in the risk of AD for identified SNPs, better understanding of the resulting physiological changes that contribute to the sex difference is needed.

Hormones

Gonadal hormones act as critical neurotrophic factors in the perinatal period and throughout the lifespan. Both hormones and genetic differences (ie, X and Y chromosomes)Citation79 contribute to the physiological mechanisms underlying sexual dimorphism of the brain, including neurogenesis, axon guidance, synaptogenesis, and neurovascular development.Citation104 Following menopause, women experience relatively rapid loss of the ovarian sex hormones 17 beta-estradiol and progesterone. A bilateral oophorectomy prior to menopause causes an abrupt deficiency of estrogen, progesterone, testosterone, and a disruption of the hypothalamic–pituitary–ovarian axis.Citation105,Citation106 Men also experience significant declines in testosterone levels with age, but these declines are more gradual. Bioavailable testosterone declines 2%–3% per year after the age of 30.Citation107 Because testosterone can be metabolized to estrogen, men do not have the severe estrogen loss, even in late-life, that is experienced by women after menopause or abruptly after a bilateral oophorectomy prior to menopause.

Animal and cellular models have consistently shown the neuroprotective effects of estrogen which include: improving synapse formation on hippocampal dendritic spines,Citation108,Citation109 maintaining hippocampal function during aging;Citation110 improving cerebral blood flow and glucose metabolism,Citation111 increasing choline acetyltransferase activity in the basal forebrain and hippocampus (choline acetyltransferase is involved in the synthesis of acetylcholine, a neurotransmitter reduced in AD and implicated in memory function),Citation112,Citation113 reducing the aggregation of amyloid-beta and associated neurotoxicity,Citation114,Citation115 and preventing mitochondrial damage.Citation116 Despite the apparent benefits in animal and cellular models, the impact of estrogen loss (due to natural menopause or surgically induced) and of hormone replacement therapy (HRT), on the risk of AD in women remains controversial. To date, observational studies generally report reduced risks of AD in women who initiate HRT within a short period (typically <3 years) after natural menopause and after oophorectomy performed prior to menopause.Citation105,Citation117Citation122 For example, the Mayo Clinic Cohort Study of Oophorectomy and Aging showed an almost doubled risk of dementia in women who underwent bilateral oophorectomy before menopause.Citation123 However, women who initiated HRT after the bilateral oophorectomy, and continued utilizing HRT at least until the age of natural menopause (approximately 51 years), did not experience an increased risk of AD.Citation123 In contrast to these studies showing a beneficial effect of estrogen use, the Women’s Health Initiative Memory Study (WHIMS),Citation124 a large randomized clinical trial of HRT, reported a two-fold increased risk of dementia in women randomized to HRT after age 65 years. One explanation for the differences between observational studies and clinical trials is that observational findings could be the result of confounding. Women who use HRT typically have a higher socioeconomic status, higher education, and/or better health and therefore may be at lower risk of AD. However, another possibility is the timing of the estrogen therapy.Citation106,Citation125

Observational studies show that the use of HRT, when initiated around the time of menopause but not years after, reduces the risk of AD.Citation105,Citation117Citation122 In the Cache County Study, women who initiated HRT within 5 years of menopause had a 30% lower risk of AD compared to women who reported no use of HRT. However, women who began hormone therapy more than 5 years after menopause did not have a lowered risk. In fact, those who started hormone use when they were 65 years or older had almost a two-fold increased risk.Citation122 Similarly, in both the Multi-Institutional Research on Alzheimer Genetic Epidemiology (MIRAGE) study and in the Northern California Kaiser Permanente study, initiation of HRT in mid-life was associated with reduced risk of AD, whereas initiation of HRT several years after menopause was associated with an increased risk.Citation120,Citation121

In light of the observational results suggesting that the initiation of estrogen in the immediate years after menopause is protective, whereas later administration increases AD risk, the WHIMSCitation124 trial results are not surprising. WHIMS subjects were aged 65–79 years old at baseline. Thus, HRT was initiated 10–20 years after the onset of natural menopause.

There are two ongoing hypotheses for the lack of benefit, or even detrimental effects, when estrogen is initiated years after menopause or bilateral oophorectomy. The first, “window of opportunity,” hypothesis is based on the mechanistic findings that long-term estrogen depletion (LTED) can cause decreased levels of estrogen receptor (ER)-alpha, in the CA1 region of the hippocampus, a highly responsive region to estrogen therapy, resulting in cognitive enhancement and neuroprotection.Citation126 Therefore, the initiation of estrogen after LTED, when ER-alpha receptors are already downregulated, does not result in the neuroprotective benefits of estrogen. The second, “healthy cell bias of estrogen benefit,” hypothesis suggests that estrogen only yields neuroprotective benefits when applied to healthy neurons.Citation127 Neurons with damaged mitochondria, a feature of aging, will not benefit, and estrogen may even be detrimental under these conditions. It is likely that both hypotheses contribute to differential benefits of estrogen when initiated peri-menopausal compared to after LTED.

Social explanation for the gender differences

In addition to several biological explanations for the observed sex differences in the prevalence and incidence of MCI and dementia, the effects of sociocultural aspects, ie, gender differences, should also be studied. Gender refers to the cultural and psychosocial factors that impact our identity and modify our risk of disease via health perception, risk behavior, social and work-related stressors, personal and societal perceptions of men’s and women’s role, patient–doctor relationships, and adherence to therapy.Citation10,Citation128 Specific factors related to gender identity that may contribute to the risk of AD include education, occupation, diet and exercise, and smoking and drinking behaviors. Gender is also strongly linked with the concept of cognitive reserve such that a higher education/occupation and greater engagement in cognitive activities provides higher reserve against disease and results in varying cognitive aging trajectories among individuals.Citation128,Citation129 In this section, we discuss gender-related risk and protective factors for AD. All of these factors can be tied to the concept of cognitive reserve as proposed by Stern.Citation130 This theory posits that subjects with higher cognitive reserve (eg, higher education, better diet, or less stress) may have a greater capacity to cope with pathological insults to the brain, or that it may take longer for them to reach the threshold of dementia detection. Thus, individuals with high cognitive reserve would be less likely to display the cognitive symptoms associated with dementia compared to individuals with the same pathology and low cognitive reserve.Citation131,Citation132

Intellectual lifestyle: education, occupation, and cognitive activity

Low education and low occupational history (eg, unskilled versus skilled worker) have repeatedly been associated with either a higher prevalenceCitation133Citation137 or incidence of AD.Citation138Citation141 Cognitive activities have been shown to reduce the risk of dementia in the elderly.Citation142,Citation143 Intellectual lifestyle (education, occupation, and current cognitive activity) explains more than 10% of the variance in an individual’s cognitive performance.Citation131 Innate cognitive ability is also important, and can lead to higher education and better occupation. Indeed, low childhood mental ability and IQ is associated with lower cognitive ability in late-life,Citation144 with an increased risk of dementia,Citation145 and with increased mortality.Citation146

Recent Pittsburgh compound B positron emission tomography (PiB-PET) and FDG-PET imaging studies have also shown that subjects with higher education or occupational engagement have more pathological changes when compared to subjects with lower education at the same level of cognitive performance (ie, they have greater brain reserve).Citation147Citation150 The mechanism by which low education and occupation are thought to increase risk of AD is by lowering cognitive reserve. A longitudinal study that followed 9,000 people semi-annually for 15 years found that the main effect of education was to increase the baseline cognitive performance of individuals.Citation151 Thus, subjects with higher education take longer to reach the dementia threshold. Sex differences in cognitive reserve and risk factors will further bias studies by causing different thresholds for detection of disease.

In the past century men have had more opportunities for higher education and higher occupational attainment than women. This is particularly true for individuals aged 70 and older who are now at greatest risk of developing AD, suggesting a higher education/occupation related reserve in men. In contrast, women generally engage in more cognitive activities such as reading books, arts and crafts, group, and social activities. While these cognitive activities impact reserve, the effect is much less than the impact of education and occupation.Citation131

Indeed, differential age, period, and cohort effects in educational and occupational attainment may play particularly important roles for late-life cognitive trajectories and risk of AD. The Seattle Longitudinal Study showed that individuals born in later (1914–1948) versus early (1886–1913) cohorts have better cognitive performance at the age of 70 years, and also slower rates of cognitive decline.Citation152,Citation153 Notably, the differences between the younger and older cohorts in cognitive gains were much greater for women than men. This research highlights the importance of gender-specific societal changes in intellectual lifestyle over time by cohort and specific historical periods (eg, during versus after World War II), and its subsequent impact on cognitive aging trajectories and risk of AD.

At the most recent census, the educational attainment in the US was higher in women than men,Citation154 and there also has been a dramatic shift in occupational engagement due to changing gender roles. For example, men and women have experienced different access to education and occupation in North America compared to Europe and Asia in the early part of this century. These gender-related differences may explain the observed geographic differences in the prevalence and incidence of AD that are described above.Citation46Citation61 Indeed, it is possible that with greater educational and occupational attainment in women, the sex differences will diminish. The changing trends of intellectual lifestyles in men and women may contribute to changing epidemiologic patterns for AD and dementia across countries and over time.

Exercise

Gender roles can affect exercise participation as parenthood and marital status have been shown to be significantly related to whether women exercise.Citation155 Several studies suggest that exercise and cardiorespiratory fitness are associated with a reduced risk of MCI and ADCitation156Citation160 and with a slower rate of decline after a diagnosis of AD.Citation161 While women are thought to be more “health-seeking” than men, it has been estimated that American women tend to get less exercise than men over the lifespan.Citation162 Studies on sex differences in exercise patterns and risk of AD are ambiguous. Some studies suggest that exercise lowers the risk of cognitive decline and AD more in women than men.Citation163,Citation164 In contrast, another study suggested that women who exercise tend to receive less of a protective effect than men.Citation165 These conflicting results may be due to the stage of life in which exercise is measured because most studies have measured self-reported physical activity in late-life. One study of over 9,000 women collected self-reported information on physical activity when the women were in their teens, age 30, age 50, and in later-life.Citation158 Physical activity at all time points was associated with a reduced risk of cognitive impairment in late-life. However, physical activity in the teenage years was associated with the greatest reduction in risk. Women who were not active as teenagers, but who were physically active at age 30 and 50, also had a reduced risk, but not as much as those who were active as teenagers. Further, among women who were physically active as teenagers, late-life physical activity did not appear to further reduce the risk of cognitive impairment, suggesting that early activity, when the brain is developing, may be most important.

The benefits of teen activity on late-life cognition are likely multifactorial. Teens who are active have better cognitive performance.Citation166 Youth physical activity may contribute to a cognitive reserve, similar to the effects of education, which would therefore have long-term effects on cognition.Citation167 Physical inactivity in the teenage years is also associated with obesity and diabetes;Citation168 both are risk factors for AD.Citation169 Physical activity and exercise are much more strongly encouraged for girls and teenage women now compared to the early- and mid-20th century; time will determine the impact that this trend may have on the sex difference in the prevalence of AD. Notably, while there is increasing focus on exercise, overall lifestyle is becoming more sedentary. Low activity throughout the day may be more beneficial than 30 minutes of moderate physical activity combined with 10 hours of sedentary behavior. Little research to date has focused on sex differences in sedentary behavior and how these differences may relate to risk of AD.

Smoking

Acetylcholine is a neurotransmitter that is decreased in Alzheimer’s patients. Indeed, the current US Food and Drug Administration-approved medications for AD primarily focus on inhibiting the degradation of acetylcholine. Nicotinic acetylcholine receptors are especially reduced in AD. Therefore, it was hypothesized that nicotine could be used to prevent or delay the progression of AD, and that smoking may be associated with a reduced risk of AD. Indeed, nicotine has been shown to increase cognitive performance in both animals and human smokers.Citation170 However, the results of clinical trials examining the use of a nicotine patch in AD patients have been mixed with some studies showing reduced cognitive decline,Citation171 and others showing no beneficial effect.Citation172 Larger studies are ongoing. Although it is possible that nicotine could be beneficial for AD, cigarette smoking contains several other toxins, has carcinogenic effects, is a known risk factor for cardiovascular and pulmonary disease, and therefore may increase the risk of AD. Additionally, many smokers also drink, and the interaction between cigarette smoking and heavy alcohol use may be especially detrimental for cognition.Citation173 A recent study suggests a strong interaction between smoking and alcohol use in predicting rate of cognitive decline, such that cigarette smokers who were heavy alcohol users had significantly faster rates of decline than smokers who were moderate alcohol users.Citation173

Cigarette smoking exacerbates Alzheimer’s pathology in transgenic mice and rats, including amyloidogenesis, tau phosphorylation, neuroinflammation, and neurodegeneration.Citation174,Citation175 Among humans, initial case–control studies reported that cigarette smoking was associated with a reduced risk of AD.Citation176Citation178 However, these studies may have been biased because smoking is strongly associated with cardiovascular disease and premature death.Citation179 Thus, smokers who survive to old age, when they are at greater risk of AD, may be more resilient to the negative effects of smoking and aging-related diseases. Subsequent cohort studies, especially those examining smoking in mid-life, have found that smoking is a risk factor for AD.Citation180Citation182 Cigarette smoking has also been associated with greater regional brain atrophy in cognitively normal individuals.Citation183

Some studies suggest that men who smoke are at greater risk of developing AD compared to women who smoke,Citation180,Citation184 whereas other studies did not show a sex difference.Citation181 Traditionally, men have had a higher prevalence of smoking because it was more socially acceptable for men to smoke. It wasn’t until the 1920s and 1930s that more women began to smoke. However, smoking among women was still less than in men. In 1965, 51.9% of men versus 33.9% of women smoked.Citation185 In recent years, the gender gap has been narrowing such that in 2009, 23.5% of men and 17.9% of women were current smokers. The differential changes in smoking by gender may impact the subsequent incidence rate of AD among women and men.

Conclusion

With the aging of the baby boomer generation, the prevalence of AD is reaching an epidemic size. By 2050, 14–16 million Americans will be diagnosed with the disease. Many more individuals will provide either formal or informal care for AD patients. There is currently no cure for this devastating disease. Current approved medications are symptomatic, and do not modify the underlying disease pathology. Although randomized clinical trials of medications to reduce amyloid and other targets are ongoing, a push towards understanding the factors associated with the risk and progression of AD is critical to identify possible preventive measures and potential new treatment targets. Future clinical trials of new therapies for AD should consider a deliberate stratification by sex, and should have adequate sample size to test for a therapeutic effect in men and women separately. A drug may have efficacy in only one sex, or the effect may be stronger in one sex.

A sex-specific or gender-specific focus in AD research is still not mainstream. However, as described in this review, the prevalence and incidence of AD, and brain structure and function, vary by sex and gender. There are also clear sex- and gender-specific risk factors for AD. Ignoring these differences will impede research and treatments. Further, this information is critical for predicting the future disease burden. For example, at the beginning of this century, men had higher education and occupational attainment. However, currently women, on average, have higher educational attainment than men. It is important to study these historical, social, and cultural trends to determine their impact on the future prevalence and incidence of AD. Understanding these sex differences and gender differences will help to define individualized treatment and preventive interventions for AD.

Acknowledgment

This work was supported in part by P50 AG44170 and U01 AG006786. Mayo Clinic is a National Institutes of Health-designed Specialized Center of Research on Sex Differences.

Disclosure

The authors report no conflicts of interest in this work.

References

  • ReitzCBrayneCMayeuxREpidemiology of Alzheimer diseaseNat Rev Neurol20117313715221304480
  • ThiesWBleilerL2013 Alzheimer’s disease facts and figuresAlzheimer’s Dement20139220824523507120
  • HebertLEWeuveJScherrPAEvansDAAlzheimer disease in the United States (2010–2050) estimated using the 2010 censusNeurology201380191778178323390181
  • HebertLEScherrPABieniasJLBennettDAEvansDAAlzheimer disease in the US population: prevalence estimates using the 2000 censusArch Neurol20036081119112212925369
  • PetersenRCClinical practice. Mild cognitive impairmentN Engl J Med2011364232227223421651394
  • PetersenRCRobertsROKnopmanDSPrevalence of mild cognitive impairment is higher in men: the Mayo Clinic Study of AgingNeurology2010751088989720820000
  • Committee on Understanding the Biology of Sex and Gender Differences, Board on Health Sciences Policy, Institute of MedicineExploring the Biological Contributions to Human Health: Does Sex Matter?WizemannTMPardueMLWashington, DCNational Academy Press2001
  • CarterCLResnickEMMallampalliMKalbarczykASex and gender differences in Alzheimer’s disease: recommendations for future researchJ Womens Health (Larchmt)201221101018102322917473
  • VestRSPikeCJGender, sex steroid hormones, and Alzheimer’s diseaseHorm Behav201363230130722554955
  • Regitz-ZagrosekVSex and Gender Aspects in Clinical MedicineOertelt-PrigioneSRegitz-ZagrosekVLondonSpringer-Verlag Limited2012
  • HickmanSEHowiesonDBDameASextonGKayeJLongitudinal analysis of the effects of the aging process on neuropsychological test performance in the healthy young-old and oldest-oldDev Neuropsychol200017332333711056847
  • NicholsMEMeadorKJLoringDWPoonLWClaytonGMMartinPAge-related changes in the neurologic examination of healthy sexagenarians, octogenarians, and centenariansJ Geriatr Psychiatry Neurol199471178192823
  • BeeriMSSchmeidlerJSanoMAge, gender, and education norms on the CERAD neuropsychological battery in the oldest oldNeurology20066761006101017000969
  • Proust-LimaCAmievaHLetenneurLOrgogozoJMJacqmin-GaddaHDartiguesJFGender and education impact on brain aging: a general cognitive factor approachPsychol Aging200823360862018808250
  • van ExelEGusseklooJde CraenAJCognitive function in the oldest old: women perform better than menJ Neurol Neurosurg Psychiatry2001711293211413258
  • van HoorenSAValentijnAMBosmaHPondsRWvan BoxtelMPJollesJCognitive functioning in healthy older adults aged 64–81: a cohort study into the effects of age, sex, and educationNeuropsychol Dev Cogn B Aging Neuropsychol Cogn2007141405417164189
  • WiederholtWCCahnDButtersNMSalmonDPKritz-SilversteinDBarrett-ConnorEEffects of age, gender and education on selected neuropsychological tests in an elderly community cohortJ Am Geriatr Soc19934166396478505462
  • BarnesLLWilsonRSSchneiderJABieniasJLEvansDABennettDAGender, cognitive decline, and risk of AD in older personsNeurology200360111777178112796530
  • DearyIJGowAJPattieAStarrJMCohort profile: the Lothian Birth Cohorts of 1921 and 1936Int J Epidemiol20124161576158422253310
  • CarlsonNEMooreMMDameATrajectories of brain loss in aging and the development of cognitive impairmentNeurology2008701182883318046010
  • GroberEHallCBLiptonRBZondermanABResnickSMKawasCMemory impairment, executive dysfunction, and intellectual decline in preclinical Alzheimer’s diseaseJ Int Neuropsychol Soc200814226627818282324
  • HowiesonDBCarlsonNEMooreMMTrajectory of mild cognitive impairment onsetJ Int Neuropsychol Soc200814219219818282317
  • TierneyMCMoineddinRMcDowellIPrediction of all-cause dementia using neuropsychological tests within 10 and 5 years of diagnosis in a community-based sampleJ Alzheimers Dis20102241231124020930315
  • BennettDAWilsonRSSchneiderJANatural history of mild cognitive impairment in older personsNeurology200259219820512136057
  • PetersenRCMild cognitive impairment as a diagnostic entityJ Intern Med2004256318319415324362
  • WinbladBPalmerKKivipeltoMMild cognitive impairment – beyond controversies, towards a consensus: report of the International Working Group on Mild Cognitive ImpairmentJ Intern Med2004256324024615324367
  • ManlyJJTangMXSchupfNSternYVonsattelJPMayeuxRFrequency and course of mild cognitive impairment in a multiethnic communityAnn Neurol200863449450618300306
  • RavagliaGFortiPMontesiFMild cognitive impairment: epidemiology and dementia risk in an elderly Italian populationJ Am Geriatr Soc2008561515818028343
  • LuckTLuppaMWieseBAgeCoDe Study GroupPrediction of incident dementia: impact of impairment in instrumental activities of daily living and mild cognitive impairment-results from the German study on ageing, cognition, and dementia in primary care patientsAm J Geriatr Psychiatry2012201194395422706332
  • PetersenRCConceptual overviewPetersenRCMild Cognitive Impairment: Aging to Alzheimer’s DiseaseNew York, NYOxford University Press2003114
  • LopezOLJagustWJDeKoskySTPrevalence and classification of mild cognitive impairment in the Cardiovascular Health Study Cognition Study: part 1Arch Neurol200360101385138914568808
  • PlassmanBLLangaKMFisherGGPrevalence of cognitive impairment without dementia in the United StatesAnn Intern Med2008148642743418347351
  • Di CarloALamassaMBaldereschiMCIND and MCI in the Italian elderly: frequency, vascular risk factors, progression to dementiaNeurology200768221909191617536047
  • GanguliMChangCCSnitzBESaxtonJAVanderbiltJLeeCWPrevalence of mild cognitive impairment by multiple classifications: the Monongahela-Youghiogheny Healthy Aging Team (MYHAT) projectAm J Geriatr Psychiatry201018867468320220597
  • GanguliMDodgeHHShenCDeKoskySTMild cognitive impairment, amnestic type: an epidemiologic studyNeurology200463111512115249620
  • KoivistoKReinikainenKJHänninenTPrevalence of age-associated memory impairment in a randomly selected population from eastern FinlandNeurology19954547417477723964
  • LarrieuSLetenneurLOrgogozoJMIncidence and outcome of mild cognitive impairment in a population-based prospective cohortNeurology200259101594159912451203
  • KivipeltoMHelkalaELHänninenTMidlife vascular risk factors and late-life mild cognitive impairment: a population-based studyNeurology200156121683168911425934
  • HänninenTHallikainenMTuomainenSVanhanenMSoininenHPrevalence of mild cognitive impairment: a population-based study in elderly subjectsActa Neurol Scand2002106314815412174174
  • SolfrizziVPanzaFColaciccoAMItalian Longitudinal Study on Aging Working GroupVascular risk factors, incidence of MCI, and rates of progression to dementiaNeurology200463101882189115557506
  • LuckTLuppaMBrielSMild cognitive impairment: incidence and risk factors: results of the Leipzig Longitudinal Study of the AgedJ Am Geriatr Soc201058101903191020840461
  • RobertsROGedaYEKnopmanDSThe incidence of MCI differs by subtype and is higher in men: the Mayo Clinic Study of AgingNeurology201278534235122282647
  • CaraccioloBPalmerKMonasteroRWinbladBBäckmanLFratiglioniLOccurrence of cognitive impairment and dementia in the community: a 9-year-long prospective studyNeurology20087019 Pt 21778178518184916
  • PlassmanBLLangaKMFisherGGPrevalence of dementia in the United States: the aging, demographics, and memory studyNeuroepidemiology2007291–212513217975326
  • SeshadriSWolfPABeiserALifetime risk of dementia and Alzheimer’s disease. The impact of mortality on risk estimates in the Framingham StudyNeurology1997496149815049409336
  • RoccaWAChaRHWaringSCKokmenEIncidence of dementia and Alzheimer’s disease: a reanalysis of data from Rochester, Minnesota, 1975–1984Am J Epidemiol1998148151629663404
  • BachmanDLWolfPALinnRTIncidence of dementia and probable Alzheimer’s disease in a general population: the Framingham StudyNeurology1993433 Pt 15155198450993
  • GanguliMDodgeHHChenPBelleSDeKoskySTTen-year incidence of dementia in a rural elderly US community population: the MoVIES ProjectNeurology20005451109111610720283
  • KawasCGraySBrookmeyerRFozardJZondermanAAge-specific incidence rates of Alzheimer’s disease: the Baltimore Longitudinal Study of AgingNeurology200054112072207710851365
  • HebertLEScherrPAMcCannJJBeckettLAEvansDAIs the risk of developing Alzheimer’s disease greater for women than for men?Am J Epidemiol2001153213213611159157
  • KukullWAHigdonRBowenJDDementia and Alzheimer disease incidence: a prospective cohort studyArch Neurol200259111737174612433261
  • EdlandSDRoccaWAPetersenRCChaRHKokmenEDementia and Alzheimer disease incidence rates do not vary by sex in Rochester, MinnArch Neurol200259101589159312374497
  • MiechRABreitnerJCZandiPPKhachaturianASAnthonyJCMayerLIncidence of AD may decline in the early 90s for men, later for women: the Cache County studyNeurology200258220921811805246
  • RobertsROKnopmanDSMielkeMMHigher risk of progression to dementia in mild cognitive impairment cases who revert to normalNeurology2013
  • FratiglioniLViitanenMvon StraussETontodonatiVHerlitzAWinbladBVery old women at highest risk of dementia and Alzheimer’s disease: incidence data from the Kungsholmen Project, StockholmNeurology19974811321389008508
  • OttABretelerMMvan HarskampFStijnenTHofmanAIncidence and risk of dementia. The Rotterdam StudyAm J Epidemiol199814765745809521184
  • LetenneurLGilleronVCommengesDHelmerCOrgogozoJMDartiguesJFAre sex and educational level independent predictors of dementia and Alzheimer’s disease? Incidence data from the PAQUID projectJ Neurol Neurosurg Psychiatry199966217718310071096
  • AndersenKLaunerLJDeweyMEGender differences in the incidence of AD and vascular dementia: the EURODEM Studies. EURODEM Incidence Research GroupNeurology19995391992199710599770
  • RuitenbergAOttAvan SwietenJCHofmanABretelerMMIncidence of dementia: does gender make a difference?Neurobiol Aging200122457558011445258
  • YoshitakeTKiyoharaYKatoIIncidence and risk factors of vascular dementia and Alzheimer’s disease in a defined elderly Japanese population: the Hisayama StudyNeurology1995456116111687783883
  • LiuCKLaiCLTaiCTLinRTYenYYHowngSLIncidence and subtypes of dementia in southern Taiwan: impact of socio-demographic factorsNeurology1998506157215799633696
  • GaoSHendrieHCHallKSHuiSThe relationships between age, sex, and the incidence of dementia and Alzheimer disease: a meta-analysisArch Gen Psychiatry19985598098159736007
  • Agüero-TorresHFratiglioniLGuoZViitanenMWinbladBPrognostic factors in very old demented adults: a seven-year follow-up from a population-based survey in StockholmJ Am Geriatr Soc19984644444529560066
  • TschanzJTCorcoranCDSchwartzSProgression of cognitive, functional, and neuropsychiatric symptom domains in a population cohort with Alzheimer dementia: the Cache County Dementia Progression studyAm J Geriatr Psychiatry201119653254221606896
  • KatzmanRTerryRDeTeresaRClinical, pathological, and neurochemical changes in dementia: a subgroup with preserved mental status and numerous neocortical plaquesAnn Neurol19882321381442897823
  • GieddJNRaznahanAMillsKLLenrootRKReview: magnetic resonance imaging of male/female differences in human adolescent brain anatomyBiol Sex Differ2012311922908911
  • BarnesLLWilsonRSBieniasJLSchneiderJAEvansDABennettDASex differences in the clinical manifestations of Alzheimer disease pathologyArch Gen Psychiatry200562668569115939846
  • CoffeyCELuckeJFSaxtonJASex differences in brain aging: a quantitative magnetic resonance imaging studyArch Neurol19985521691799482358
  • OguroHOkadaKYamaguchiSKobayashiSSex differences in morphology of the brain stem and cerebellum with normal ageingNeuroradiology199840127887929877132
  • GurRCGunning-DixonFMTuretskyBIBilkerWBGurREBrain region and sex differences in age association with brain volume: a quantitative MRI study of healthy young adultsAm J Geriatr Psychiatry2002101728011790637
  • PfefferbaumARohlfingTRosenbloomMJChuWColrainIMSullivanEVVariation in longitudinal trajectories of regional brain volumes of healthy men and women (ages 10 to 85 years) measured with atlas-based parcellation of MRINeuroimage20136517619323063452
  • SkupMZhuHWangYAlzheimer’s Disease Neuroimaging InitiativeSex differences in grey matter atrophy patterns among AD and aMCI patients: results from ADNINeuroimage201156389090621356315
  • LudersEGaserCNarrKLTogaAWWhy sex matters: brain size independent differences in gray matter distributions between men and womenJ Neurosci20092945142651427019906974
  • GoodCDJohnsrudeISAshburnerJHensonRNFristonKJFrackowiakRSA voxel-based morphometric study of ageing in 465 normal adult human brainsNeuroimage2001141 Pt 1213611525331
  • GieddJNCastellanosFXRajapakseJCVaituzisACRapoportJLSexual dimorphism of the developing human brainProg Neuropsychopharmacol Biol Psychiatry1997218118512019460086
  • CosgroveKPMazureCMStaleyJKEvolving knowledge of sex differences in brain structure, function, and chemistryBiol Psychiatry200762884785517544382
  • WitteAVSavliMHolikAKasperSLanzenbergerRRegional sex differences in grey matter volume are associated with sex hormones in the young adult human brainNeuroimage20104921205121219796695
  • van AmelsvoortTComptonJMurphyDIn vivo assessment of the effects of estrogen on human brainTrends Endocrinol Metab200112627327611445445
  • LentiniEKasaharaMArverSSavicISex differences in the human brain and the impact of sex chromosomes and sex hormonesCereb Cortex201323102322233622891037
  • BiswalBBMennesMZuoXNToward discovery science of human brain functionProc Natl Acad Sci U S A2010107104734473920176931
  • HsiehTCLinWYDingHJSex- and age-related differences in brain FDG metabolism of healthy adults: an SPM analysisJ Neuroimaging2012221212721332873
  • GurRCMozleyLHMozleyPDSex differences in regional cerebral glucose metabolism during a resting stateScience199526751975285317824953
  • PerneczkyRDrzezgaADiehl-SchmidJLiYKurzAGender differences in brain reserve: an (18)F-FDG PET study in Alzheimer’s diseaseJ Neurol2007254101395140017934882
  • PerneczkyRDiehl-SchmidJFörstlHDrzezgaAKurzAMale gender is associated with greater cerebral hypometabolism in frontotemporal dementia: evidence for sex-related cognitive reserveInt J Geriatr Psychiatry200722111135114017479980
  • BucknerRLSnyderAZShannonBJMolecular, structural, and functional characterization of Alzheimer’s disease: evidence for a relationship between default activity, amyloid, and memoryJ Neurosci200525347709771716120771
  • StrittmatterWJSaundersAMSchmechelDApolipoprotein E: high-avidity binding to beta-amyloid and increased frequency of type 4 allele in late-onset familial Alzheimer diseaseProc Natl Acad Sci U S A1993905197719818446617
  • CorderEHSaundersAMStrittmatterWJGene dose of apolipoprotein E type 4 allele and the risk of Alzheimer’s disease in late onset familiesScience199326151239219238346443
  • BertramLTanziREThirty years of Alzheimer’s disease genetics: the implications of systematic meta-analysesNat Rev Neurosci200891076877818802446
  • MeyerMRTschanzJTNortonMCAPOE genotype predicts when – not whether – one is predisposed to develop Alzheimer diseaseNat Genet19981943213229697689
  • FarrerLACupplesLAHainesJLEffects of age, sex, and ethnicity on the association between apolipoprotein E genotype and Alzheimer disease. A meta-analysis. APOE and Alzheimer Disease Meta Analysis ConsortiumJAMA199727816134913569343467
  • PayamiHZareparsiSMonteeKRGender difference in apolipoprotein E-associated risk for familial Alzheimer disease: a possible clue to the higher incidence of Alzheimer disease in womenAm J Hum Genet19965848038118644745
  • BretskyPMBuckwalterJGSeemanTEEvidence for an interaction between apolipoprotein E genotype, gender, and Alzheimer diseaseAlzheimer Dis Assoc Disord199913421622110609670
  • FleisherAGrundmanMJackCRJrAlzheimer’s Disease Cooperative StudySex, apolipoprotein E epsilon 4 status, and hippocampal volume in mild cognitive impairmentArch Neurol200562695395715956166
  • LiuYPaajanenTWestmanEAddNeuroMed ConsortiumEffect of APOE ε4 allele on cortical thicknesses and volumes: the AddNeuroMed studyJ Alzheimers Dis201021394796620693633
  • DamoiseauxJSSeeleyWWZhouJAlzheimer’s Disease Neuroimaging InitiativeGender modulates the APOE ε4 effect in healthy older adults: convergent evidence from functional brain connectivity and spinal fluid tau levelsJ Neurosci201232248254826222699906
  • CorderEHGhebremedhinETaylorMGThalDROhmTGBraakHThe biphasic relationship between regional brain senile plaque and neurofibrillary tangle distributions: modification by age, sex, and APOE polymorphismAnn N Y Acad Sci20041019242815246987
  • FukumotoNFujiiTCombarrosOSexually dimorphic effect of the Val66Met polymorphism of BDNF on susceptibility to Alzheimer’s disease: new data and meta-analysisAm J Med Genet B Neuropsychiatr Genet2010153B123524219504537
  • SohrabjiFMirandaRCToran-AllerandCDIdentification of a putative estrogen response element in the gene encoding brain-derived neurotrophic factorProc Natl Acad Sci U S A1995922411110111147479947
  • ZouFGopalrajRKLokJSex-dependent association of a common low-density lipoprotein receptor polymorphism with RNA splicing efficiency in the brain and Alzheimer’s diseaseHum Mol Genet200817792993518065781
  • AlbayrakOTirniceriuARiemenschneiderMKurzAScheragAEgenspergerRThe cathepsin D (224C/T) polymorphism confers an increased risk to develop Alzheimer’s disease in menJ Gerontol A Biol Sci Med Sci201065321922420083556
  • HamiltonGProitsiPJehuLCandidate gene association study of insulin signaling genes and Alzheimer’s disease: evidence for SOS2, PCK1, and PPARgamma as susceptibility lociAm J Med Genet B Neuropsychiatr Genet2007144B450851617440948
  • SundarPDFeingoldEMinsterRLDeKoskySTKambohMIGender-specific association of ATP-binding cassette transporter 1 (ABCA1) polymorphisms with the risk of late-onset Alzheimer’s diseaseNeurobiol Aging200728685686216725228
  • SrivastavaRAEstrogen-induced regulation of the ATP-binding cassette transporter A1 (ABCA1) in mice: a possible mechanism of atheroprotection by estrogenMol Cell Biochem20022401–2677312487373
  • NugentBMTobetSALaraHEHormonal programming across the lifespanHorm Metab Res201244857758622700441
  • MorrisonJHBrintonRDSchmidtPJGoreACEstrogen, menopause, and the aging brain: how basic neuroscience can inform hormone therapy in womenJ Neurosci20062641103321034817035515
  • RoccaWAGrossardtBRShusterLTOophorectomy, menopause, estrogen treatment, and cognitive aging: clinical evidence for a window of opportunityBrain Res2011137918819820965156
  • FeldmanHALongcopeCDerbyCAAge trends in the level of serum testosterone and other hormones in middle-aged men: longitudinal results from the Massachusetts male aging studyJ Clin Endocrinol Metab200287258959811836290
  • MurphyDDSegalMRegulation of dendritic spine density in cultured rat hippocampal neurons by steroid hormonesJ Neurosci19961613405940688753868
  • AenlleKKKumarACuiLJacksonTCFosterTCEstrogen effects on cognition and hippocampal transcription in middle-aged miceNeurobiol Aging200930693294517950954
  • HanXAenlleKKBeanLARole of estrogen receptor α and β in preserving hippocampal function during agingJ Neurosci20133362671268323392694
  • WangQSantizoRBaughmanVLPelligrinoDAIadecolaCEstrogen provides neuroprotection in transient forebrain ischemia through perfusion-independent mechanisms in ratsStroke199930363063710066863
  • GibbsRBEstrogen and nerve growth factor-related systems in brain. Effects on basal forebrain cholinergic neurons and implications for learning and memory processes and agingAnn N Y Acad Sci1994743165196 discussion 197–1697802412
  • GibbsRBAggarwalPEstrogen and basal forebrain cholinergic neurons: implications for brain aging and Alzheimer’s disease-related cognitive declineHorm Behav1998342981119799621
  • NilsenJChenSIrwinRWIwamotoSBrintonRDEstrogen protects neuronal cells from amyloid beta-induced apoptosis via regulation of mitochondrial proteins and functionBMC Neurosci200677417083736
  • JaffeABToran-AllerandCDGreengardPGandySEEstrogen regulates metabolism of Alzheimer amyloid beta precursor proteinJ Biol Chem19942691813065130688175728
  • YaoJBrintonRDEstrogen regulation of mitochondrial bioenergetics: implications for prevention of Alzheimer’s diseaseAdv Pharmacol20126432737122840752
  • WaringSCRoccaWAPetersenRCO’BrienPCTangalosEGKokmenEPostmenopausal estrogen replacement therapy and risk of AD: a population-based studyNeurology199952596597010102413
  • LeBlancESJanowskyJChanBKNelsonHDHormone replacement therapy and cognition: systematic review and meta-analysisJAMA2001285111489149911255426
  • ZandiPPCarlsonMCPlassmanBLCache County Memory Study InvestigatorsHormone replacement therapy and incidence of Alzheimer disease in older women: the Cache County StudyJAMA2002288172123212912413371
  • HendersonVWBenkeKSGreenRCCupplesLAFarrerLAMIRAGE Study GroupPostmenopausal hormone therapy and Alzheimer’s disease risk: interaction with ageJ Neurol Neurosurg Psychiatry200576110310515608005
  • WhitmerRAQuesenberryCPZhouJYaffeKTiming of hormone therapy and dementia: the critical window theory revisitedAnn Neurol201169116316921280086
  • ShaoHBreitnerJCWhitmerRACache County InvestigatorsHormone therapy and Alzheimer disease dementia: new findings from the Cache County StudyNeurology201279181846185223100399
  • RoccaWABowerJHMaraganoreDMIncreased risk of cognitive impairment or dementia in women who underwent oophorectomy before menopauseNeurology200769111074108317761551
  • ShumakerSALegaultCKullerLWomen’s Health Initiative Memory StudyConjugated equine estrogens and incidence of probable dementia and mild cognitive impairment in postmenopausal women: Women’s Health Initiative Memory StudyJAMA2004291242947295815213206
  • RoccaWAGrossardtBRShusterLTOophorectomy, menopause, estrogen, and cognitive aging: the timing hypothesisNeurodegener Dis201071–316316620197698
  • ZhangQGHanDWangRMC terminus of Hsc70-interacting protein (CHIP)-mediated degradation of hippocampal estrogen receptor-alpha and the critical period hypothesis of estrogen neuroprotectionProc Natl Acad Sci U S A201110835E617E62421808025
  • BrintonRDThe healthy cell bias of estrogen action: mitochondrial bioenergetics and neurological implicationsTrends Neurosci2008311052953718774188
  • KreigerNEpidemiology and the People’s Health: Theory and ContextNew YorkOxford University Press2011
  • WhalleyLJDearyIJAppletonCLStarrJMCognitive reserve and the neurobiology of cognitive agingAgeing Res Rev20043436938215541707
  • SternYCognitive reserveNeuropsychologia200947102015202819467352
  • VemuriPLesnickTGPrzybelskiSAEffect of lifestyle activities on Alzheimer disease biomarkers and cognitionAnn Neurol201272573073823280791
  • SternYCognitive reserve in ageing and Alzheimer’s diseaseLancet Neurol201211111006101223079557
  • KatzmanREducation and the prevalence of dementia and Alzheimer’s diseaseNeurology199343113208423876
  • FratiglioniLGrutMForsellYPrevalence of Alzheimer’s disease and other dementias in an elderly urban population: relationship with age, sex, and educationNeurology19914112188618921745343
  • ZhangMYKatzmanRSalmonDThe prevalence of dementia and Alzheimer’s disease in Shanghai, China: impact of age, gender, and educationAnn Neurol19902744284372353798
  • BonaiutoSRoccaWALippiAEducation and occupation as risk factors for dementia: a population-based case-control studyNeuroepidemiology19951431011097777124
  • MortimerJAGravesABEducation and other socioeconomic determinants of dementia and Alzheimer’s diseaseNeurology1993438, Suppl 4S39S448389010
  • KarpAKåreholtIQiuCBellanderTWinbladBFratiglioniLRelation of education and occupation-based socioeconomic status to incident Alzheimer’s diseaseAm J Epidemiol2004159217518314718220
  • BickelHCooperBIncidence and relative risk of dementia in an urban elderly population: findings of a prospective field studyPsychol Med19942411791928208883
  • SternYGurlandBTatemichiTKTangMXWilderDMayeuxRInfluence of education and occupation on the incidence of Alzheimer’s diseaseJAMA199427113100410108139057
  • RoeCMXiongCMillerJPMorrisJCEducation and Alzheimer disease without dementia: support for the cognitive reserve hypothesisNeurology200768322322817224578
  • CroweMAndelRPedersenNLJohanssonBGatzMDoes participation in leisure activities lead to reduced risk of Alzheimer’s disease? A prospective study of Swedish twinsJ Gerontol B Psychol Sci Soc Sci2003585P249P25514507930
  • FabrigouleCDo leisure activities protect against Alzheimer’s disease?Lancet Neurol2002111112849540
  • GaleCRCooperRCraigLCognitive function in childhood and lifetime cognitive change in relation to mental wellbeing in four cohorts of older peoplePLoS ONE201279e4486022970320
  • WhalleyLJStarrJMAthawesRHunterDPattieADearyIJChildhood mental ability and dementiaNeurology200055101455145911094097
  • WhalleyLJDearyIJLongitudinal cohort study of childhood IQ and survival up to age 76BMJ2001322729081911290633
  • ScarmeasNZarahnEAndersonKEAssociation of life activities with cerebral blood flow in Alzheimer disease: implications for the cognitive reserve hypothesisArch Neurol200360335936512633147
  • GaribottoVBorroniBKalbeEEducation and occupation as proxies for reserve in aMCI converters and AD: FDG-PET evidenceNeurology200871171342134918936426
  • KemppainenNMAaltoSKarraschMCognitive reserve hypothesis: Pittsburgh Compound B and fluorodeoxyglucose positron emission tomography in relation to education in mild Alzheimer’s diseaseAnn Neurol200863111211818023012
  • RoeCMMintunMAD’AngeloGXiongCGrantEAMorrisJCAlzheimer disease and cognitive reserve: variation of education effect with carbon 11-labeled Pittsburgh Compound B uptakeArch Neurol200865111467147119001165
  • SchneiderALSharrettARPatelMDEducation and cognitive change over 15 years: the atherosclerosis risk in communities studyJ Am Geriatr Soc201260101847185323013064
  • GerstorfDRamNHoppmannCWillisSLSchaieKWCohort differences in cognitive aging and terminal decline in the Seattle Longitudinal StudyDev Psychol20114741026104121517155
  • SchaieKWWillisSLThe Seattle Longitudinal Study of Adult Cognitive DevelopmentISSBD Bull2010571242923536918
  • RyanCLSiebensJEducational Attainment in the United States: 2009. Current Population ReportsWashington, DCUS Department of Commerce, US Census Bureau2012 Available from: http://www.census.gov/prod/2012pubs/p20-566.pdfAccessed August 17, 2013
  • VerhoefMJLoveEJRoseMSWomen’s social roles and their exercise participationWomen Health199219415291295266
  • GedaYERobertsROKnopmanDSPhysical exercise, aging, and mild cognitive impairment: a population-based studyArch Neurol2010671808620065133
  • HamerMChidaYPhysical activity and risk of neurodegenerative disease: a systematic review of prospective evidencePsychol Med200939131118570697
  • MiddletonLEBarnesDELuiLYYaffeKPhysical activity over the life course and its association with cognitive performance and impairment in old ageJ Am Geriatr Soc20105871322132620609030
  • LiuRSuiXLaditkaJNCardiorespiratory fitness as a predictor of dementia mortality in men and womenMed Sci Sports Exerc201244225325921796048
  • BuchmanASBoylePAYuLShahRCWilsonRSBennettDATotal daily physical activity and the risk of AD and cognitive decline in older adultsNeurology201278171323132922517108
  • PitkäläKHPöystiMMLaakkonenMLEffects of the Finnish Alzheimer disease exercise trial (FINALEX): a randomized controlled trialJAMA Intern Med20131731089490123589097
  • NomaguchiKMBianchiSMExercise time: gender differences in the effects of marriage, parenthood, and employmentJ Marriage Fam2004662413430
  • HogervorstECliffordAStockJXinXBandelowSExercise to prevent cognitive decline and Alzheimer’s disease: for whom, when, what, and (most importantly) how much?J Alzheimers Dis Park201223e117
  • CliffordABandelowSHogervorstEThe effects of physical exercise on cognitive function in the elderly: A reviewGariépyQMénardRHandbook of Cognitive Aging: Causes, Processes and EffectsNew YorkNova Science Publishers2010109150
  • FallahNMitnitskiAMiddletonLRockwoodKModeling the impact of sex on how exercise is associated with cognitive changes and death in older CanadiansNeuroepidemiology2009331475419365142
  • SibleyBAEtnierJLThe relationship between physical activity and cognition in children: a meta-analysisPediatr Exerc Sci2003153243256
  • ScarmeasNSternYCognitive reserve and lifestyleJ Clin Exp Neuropsychol200325562563312815500
  • McGavockJSellersEDeanHPhysical activity for the prevention and management of youth-onset type 2 diabetes mellitus: focus on cardiovascular complicationsDiab Vasc Dis Res20074430531018158700
  • LuchsingerJAAdiposity, hyperinsulinemia, diabetes and Alzheimer’s disease: an epidemiological perspectiveEur J Pharmacol2008585111912918384771
  • RezvaniAHLevinEDCognitive effects of nicotineBiol Psychiatry200149325826711230877
  • WilsonALLangleyLKMonleyJNicotine patches in Alzheimer’s disease: pilot study on learning, memory, and safetyPharmacol Biochem Behav1995512–35095147667377
  • WhiteHKLevinEDFour-week nicotine skin patch treatment effects on cognitive performance in Alzheimer’s diseasePsychopharmacology (Berl)1999143215816510326778
  • Hagger-JohnsonGSabiaSBrunnerEJCombined impact of smoking and heavy alcohol use on cognitive decline in early old age: Whitehall II prospective cohort studyBr J Psychiatry2013203212012523846998
  • Moreno-GonzalezIEstradaLDSanchez-MejiasESotoCSmoking exacerbates amyloid pathology in a mouse model of Alzheimer’s diseaseNat Commun20134149523422663
  • HoYSYangXYeungSCCigarette smoking accelerated brain aging and induced pre-Alzheimer-like neuropathology in ratsPLoS ONE201275e3675222606286
  • KatzmanRAronsonMFuldPDevelopment of dementing illnesses in an 80-year-old volunteer cohortAnn Neurol19892543173242712531
  • GravesABvan DuijnCMChandraVAlcohol and tobacco consumption as risk factors for Alzheimer’s disease: a collaborative re-analysis of case-control studies. EURODEM Risk Factors Research GroupInt J Epidemiol199120Suppl 2S48S571833354
  • LeePNSmoking and Alzheimer’s disease: a review of the epidemiological evidenceNeuroepidemiology19941341311448090255
  • DollRPetoRWheatleyKGrayRSutherlandIMortality in relation to smoking: 40 years’ observations on male British doctorsBMJ199430969599019117755693
  • OttASlooterAJHofmanASmoking and risk of dementia and Alzheimer’s disease in a population-based cohort study: the Rotterdam StudyLancet19983519119184018439652667
  • RusanenMKivipeltoMQuesenberryCPJrZhouJWhitmerRAHeavy smoking in midlife and long-term risk of Alzheimer disease and vascular dementiaArch Intern Med2011171433333920975015
  • TyasSLWhiteLRPetrovitchHMid-life smoking and late-life dementia: the Honolulu-Asia Aging StudyNeurobiol Aging200324458959612714116
  • DurazzoTCInselPSWeinerMWAlzheimer Disease Neuroimaging InitiativeGreater regional brain atrophy rate in healthy elderly subjects with a history of cigarette smokingAlzheimers Dement20128651351923102121
  • LaunerLJAndersenKDeweyMERates and risk factors for dementia and Alzheimer’s disease: results from EURODEM pooled analyses. EURODEM Incidence Research Group and Work Groups. European Studies of DementiaNeurology199952178849921852
  • American Lung AssociationResearch and Program Services, Epidemiology and Statistics Unit. Trends in tobacco use2011 PDF available from http://www.lung.org/finding-cures/our-research/trend-reports/Tobacco-Trend-Report.pdfAccessed December 17, 2013