Abstract
Alzheimer’s disease (AD) is a common neurodegenerative disorder that presents with cognitive impairment and behavioral disturbance. Approximately 5.5 million people in the United States live with AD, most of whom are over the age of 65 with two-thirds being woman. There have been major advancements over the last decade or so in the understanding of AD neuropathological changes and genetic involvement. However, studies of sex impact in AD have not been adequately integrated into the investigation of disease development and progression. It becomes indispensable to acknowledge in both basic science and clinical research studies the importance of understanding sex-specific differences in AD pathophysiology and pathogenesis, which could guide future effort in the discovery of novel targets for AD. Here, we review the latest and most relevant literature on this topic, highlighting the importance of understanding sex dimorphism from a molecular perspective and its association to clinical trial design and development in AD research field.
Keywords: Alzheimer’s Disease, Risk Factors, Pathogenesis, Sex Differences, Molecular Impact
1. Introduction
Neurodegeneration involves a progressive functional and structural loss of both neurons and glia that ultimately contribute to massive cell death within the central nervous system. Advances in precision medicine approaches have been instrumental in studying mechanisms underlying neurodegenerative processes. Different mechanisms have been implicated including aggregation of misfolded proteins, impairment in degradation pathways, defective axonal transportation, DNA and membrane damage, mitochondrial dysfunction, and programmed cell death (Gitler and Tsuiji, 2016). On the other hand, sex and gender differences have been increasingly recognized for their impacts on etiology, incidence, severity, progression and treatment outcomes of several neurodegenerative disorders. Sex refers to the physiological and biological disparities between men and women with chromosomal variations and gonadal hormones as the primary contributors of these differences at cellular and systemic levels. Gender alludes to a combination of influences (i.e. social, cultural, and environmental) that affect biological factors within men and women. In other words, gender is implanted by biology but molded by experience and environment (Roselli, 2018).
While major progresses have made in understanding Alzheimer’s disease (AD) pathogenesis (Podcasy and Epperson, 2016), there has been lack of attention until recently on sex differences in AD. A growing amount of evidence suggests that sex and gender differences may play important roles in the heterogeneities of AD prevalence, clinical manifestations such as behavior and cognitive performance, disease course and prognosis, as well as pathology (Heun, 2002; Jack et al., 2013; Pini et al., 2016). For example, women have increased risks of developing AD than men (Nebel, 2018). Sex differences have also been observed in treatment responses of patients in clinical trials (Kim et al., 2015). It has been increasingly recognized that sex- and gender-related differences cannot be completely explained by longer lifespans of women (Nebel et al., 2018). Furthermore, sex differences in AD are stratified by many factors. Therefore, a greater understanding of sex and gender differences from a molecular to a clinical point of view could greatly improve symptom awareness, risk modification and prevention, as well as clinical management of AD. In the present review, we provide updates about current understanding of sex and gender differences in brain aging and AD-related pathological processes, emphasizing on how developmental, genetic, comorbid and environmental factors impact on sex dimorphism in AD.
2. Sex Differences during Nervous System Development and Aging
2.1. Development of Nervous System and Neural Network
Sex dimorphism starts at early stage of brain development and continues its dynamic molding throughout the entire human lifespan. There are many structural and functional differences between men and women during the development of nervous system and neural network. For example, males have higher brain volumes with relatively higher white matter content whereas females have lower volumes of brain and cerebrospinal fluid (CSF) with relatively higher gray matter content (Allen et al., 2002; Cosgrove et al., 2007). In consistent with these observations, neuroimaging analysis demonstrated that males have more myelinated fibers than females at the start of puberty (Herting et al., 2012). Functional magnetic resonance imaging studies indicated that cognitively healthy adult males present with higher within-hemispheric connectivity, whereas females showed higher degrees of network efficiency and cortical connectivity (Ingalhalikar et al., 2014; Kanaan et al., 2012). Moreover, brain regional differences were noted in blood flow of males and females, with higher blood flow in parietal cortex of women and in motor and visual cortex of men (Gur et al., 1995; Hsieh et al., 2012). It has been suggested that regional effects of sex hormones with specific expression profiles of sex hormone receptors could at least partially explain sex dimorphism in brain development, as well as sex differences in structural and functional network connectivity (Giedd et al., 1999; Li and Singh, 2014; Tomasi and Volkow, 2012).
2.2. Aging processes
It should be noted that sex dimorphism may extend beyond the development and adolescent stages. Some neuroimaging studies in normal aging subjects have shown that men present greater age-related cortical volume loss than women (Jack et al., 2015; Jack et al., 2017). There are significantly more cortical atrophy and glucose metabolic deficits in males than in females during aging (Murphy et al., 1996). Another study reported that men present a greater degree of white matter microstructural damage than female healthy older participants, suggesting a greater degree of brain reserve in men to maintain cognitive performances at similar levels of women despite more severe structural damage (O’Dwyer et al., 2012).
Many mechanisms have been proposed for sex differences in longevity such as sex asymmetries in the X chromosome and mitochondrial genome inheritance (Frank and Hurst, 1996; Tower, 2006) and sex-specific selection (Maklakov et al., 2009; Maklakov and Lummaa, 2013). Studies also suggest that the changes in biological sex and sex hormone mediated memory circuitry have become evidence in midlife (Pudas et al., 2018). This time-period in women transitioning through menopause may play an important role in cognitive decline during aging (Jacobs and Goldstein, 2018). For example, low estradiol levels correlated with poor performance on memory retrieval task and changes in hippocampal connectivity (Jacobs et al., 2016). In addition, postmenopausal women demonstrated different hippocampal responses during verbal memory tasks when compared to premenopausal women (Jacobs et al., 2016; Jacobs et al., 2017). It has been suggested that reduction in estrogen levels during aging and menopausal periods are associated with heightened oxidative stress and mitochondrial dysfunction (Beckman and Ames, 1998), exacerbated pro-inflammatory responses (Berchtold et al., 2008), and reduced synaptic plasticity (Baudry et al., 2013). For example, female cells were found less vulnerable to oxidative insults than males (Ide et al., 2002; Matarrese et al., 2011). In addition, female animals had lower levels of reactive oxygen species (Vina et al., 2003) and reduced mitochondrial DNA damage than male counterparts (Borras et al., 2003). One interesting study that assessed cognitive resilience in healthy older adults with AD risk genes including apolipoprotein E4 (ApoE4) in a 9-year follow-up period indicated sex-specific predictors of memory resilience, e.g. pet ownership and history of depression in men, whereas marital status, subjective health, mobility and current alcohol use in women (McDermott et al., 2017), indicating sex-specific modifiable factors for healthy aging. A recent study using system biology approaches to analyze large sets of microarray data from hippocampal brain regions of normal aging individuals, identified the effects mediated by sex-aging interaction that lead to changes in functions of mitochondria, autophagy, and miRNA expression. This information may elucidate important pathways and mechanisms underlying sex dimorphism in aging brain (Guebel and Torres, 2016).
3. Sex Differences in Alzheimer’s disease
Unlike healthy aging population, several lines of data strongly indicate that female AD subjects are more affected by disease processes than male patients are (Filon et al., 2016; Malpetti et al., 2017). AD women showed a greater decline in essentially all cognitive domains than AD men (Laws et al., 2018). Irvine et al. reported significant male advantages over female in five cognitive domains (Irvine et al., 2012). Consistently, two independent studies concluded statistically significant worsening of verbal and visuospatial performance in female mild cognitive impairment (MCI) and AD subjects than male counterparts, regardless of apolipoprotein E4 (ApoE4) genotype status (Gale et al., 2016; Tensil et al., 2018). Data from the Alzheimer’s Disease Neuroimaging Initiative (ADNI) study suggest the rate of cognitive decline in women is twice as fast as that of men even after corrected for the ApoE genotypes (Lin et al., 2015). Furthermore, there have been sex differences in behavioral symptoms observed of AD subjects despite the relatively small sample sizes in these studies, such as an increased likelihood of agitation (Mega et al., 1996) and socially inappropriate behaviors (Kitamura et al., 2012; Ott et al., 1996; Ott et al., 2000) in AD men, and an increased likelihood of depressive symptoms (Teri et al., 1989) and delusions (Karttunen et al., 2011) in AD female subjects.
The biomarker studies indicate no clear sex differences in brain amyloid or neurofibrillary tangle burden of AD subjects as determined by postmortem neuropathological analysis (Barnes et al., 2005; Shinohara et al., 2016), or by measurement of cerebrospinal fluid amyloid-β (Aβ42) and tau levels (Mattsson et al., 2017). Consistently, no sex differences are present in amyloid positivity or tau accumulation between male and female MCI subjects as determine by PET imaging studies (Jack et al., 2017; Jansen et al., 2015; Johnson et al., 2016). On the other hand, sex differences in brain atrophy rates have been observed in many brain regions of normal aging, MCI, and AD subjects (Hua et al., 2010; Skup et al., 2011). In MCI and AD women, the atrophy rates are 1–1.5% faster than those in men (Ardekani et al., 2016; Hua et al., 2010). Surprisingly, female MCI subjects with positive biomarker studies such as increased amyloid plaque and neurofibrillary tangle burden (Barnes et al., 2005) or CSF biomarker changes (Koran et al., 2017) had significantly higher chances of developing dementia or neurodegeneration than male counterparts, suggesting a potential interaction between sex and AD biomarkers in the disease prognosis. Several animal models have been used to determine the molecular mechanisms underlying sex-specific differences in AD. A clear sex-specific effect in amyloid load and cognitive deficits have been characterized in AD transgenic mouse models such as 3xTg, APP/PS1 and Tg2576 lines (Dubal et al., 2012), as well as human ApoE4 mouse models such as ApoE4 KI (Bour et al., 2008; Reverte et al., 2012) and EFAD lines (Tai et al., 2017). The sex effects on tau hyper-phosphorylation are rather controversial (Yue et al., 2011).
One possible mechanism is related to differences in γ–secretase activities between females and males as suggested in mouse model studies (Placanica et al., 2009). More importantly, sex hormones contribute to sex dimorphism in AD-related pathological processes. Sex steroid hormones are important risk factors in AD development because they are fundamental determinants of sex differences in brain function and cognition. The mechanisms of action of sex hormones are mediated by binding of specific steroid hormones with their specific receptors which are also nuclear transcription factors, followed by modulation of transcriptional activities after binding (DeMayo et al., 2002). The incidence of AD is higher in postmenopausal women, which has been linked to abrupt reduction of estrogens and progesterone (Geerlings et al., 2001). Studies have demonstrated significant decrease in verbal memory related to alterations in the hippocampal function during menopausal transition due to declines in estradiol levels (Jacobs et al., 2016; Rentz et al., 2017). The peri-menopausal cognitive decline is believed to be temporary with evidence indicated recovery during postmenopausal stages by some researchers (Greendale et al., 2009), whereas others showed persistent cognitive decline (Jacobs et al., 2016; Rentz et al., 2017).
The decrease of circulating estrogen and progesterone with age has served as the foundation to test whether estrogen intervention may help protect against AD progression. Several early clinical studies in women with AD undergoing hormone therapy suggested that estrogen preserves cognitive function (Doraiswamy et al., 1997a; Doraiswamy et al., 1997b; Henderson et al., 1994). Another recent study reported a significant lower risk of developing AD in women with estrogen therapy in comparison with individuals who had placebo (Manson et al., 2017). However, many studies have found that hormone treatment in older aged women (≥65) led to a two-fold increase in risks of developing dementia (Shumaker et al., 2003; Shumaker et al., 2004). These studies suggested that starting hormone therapy at older ages may have adverse consequences. Overall, the current thinking is that women who initiated hormone therapy early during menopausal transition may lower the risks of developing AD compared to women received treatment at later stages (Henderson et al., 2005; Whitmer et al., 2011).
A possible mechanism by which estrogens exert anti-neurodegenerative effects in the brain is through the modulation of Aβ generation (Xu et al., 1998). Estrogen is shown to reduce Aβ production in neurons, revealing a neuroprotective mechanism against AD progression. Other possible protective mechanisms include the regulation of basal forebrain activities (Luine, 1985), synaptic plasticity (Woolley et al., 1997), neurotrophin signaling (Toran-Allerand, 1996), and reduction of free radical induced oxidative stress (Behl et al., 1995). Moreover, estrogen has been shown to regulate vesicle formation from late secretory pathway (Scammell et al., 1986), and vesicle trafficking from the trans-Golgi-network (TGN) to the plasma membrane (Platt et al., 1991). Specifically, estrogen stimulates biogenesis of APP-containing vesicles from the TGN, promotes APP delivery to cell surfaces, and thereby significantly decreases Aβ production (Greenfield et al., 2002).
Decreased testosterone during aging constitutes a risk factor for AD in men. Studies on AD-like transgenic mouse model have shown that depletion of androgen exacerbates AD-related neuropathological changes, which is rescued by treatment with androgen (Pike et al., 2009). Testosterone mediates most of its effects, including the regulation of Aβ neuropathology, through both androgen and estrogen pathways, since it is metabolized in the brain into androgen dihydrotestosterone (DHT) and estrogen 17β-estradiol (E2). In contrast, the regulation of tau hyper-phosphorylation by testosterone may be mainly regulated by estrogen pathways, as neuronal tangle burden is reduced by testosterone and E2 but not DHT (Rosario, 2010).
It should be noted that the effects of hormones on brain aging and neurodegenerative processes are important contributors of sex differences. However, they are not he only sex-based influences. Many factors contribute to sex differences in cognition, behavior and neural processes. In the following section, we will discuss the impact of genetic risk factors, comorbid and environmental factors contributing to sex differences in AD pathogenesis.
4.1. The Impact of Genetic Risk Factors on Sex Differences in AD
4.1.1. ApoE
ApoE serves as an important lipid binding protein that facilitates the transport of phospholipids and cholesterol in a myriad of different tissue types through receptor mediated interactions at cell surfaces (Bu, 2009; Verghese et al., 2013). There are 3 genetic variants of ApoE: APOE2, APOE3 and APOE4, where the APOE4 allele confers the strongest genetic risk factor for AD development (Saunders et al., 1993; Strittmatter et al., 1993). ApoE4 has direct effects on Aβ aggregation and clearance (Kim et al., 2009; Ossenkoppele et al., 2015; Verghese et al., 2013). Moreover, ApoE4 isoform is susceptible to proteolytic cleavage, resulting in the release of neurotoxic fragments (Mahley and Huang, 2012). Evidence further suggests that ApoE affects tau pathogenesis independent of Aβ pathology, and ApoE4 exhibits a gain of toxic effect on tau pathologies (Shi et al., 2017). A recent study has also shown that ApoE4 contributes to elevation of hyper-phosphorylated tau levels after traumatic brain injury (TBI) exposure that could subsequently induce neurodegeneration and neuro-inflammation (Cao et al., 2017).
While the AD risk posed by ApoE4 is observed in both males and females, females with at least one copy ApoE4 often exhibit faster cognitive decline and deterioration than males with at least one copy of ApoE4 (Farrer et al., 1997). A recent study showed that carriers of ApoE3/4 genotype between age of 65 and 75 had risks of developing AD when compared to ApoE4 non-carriers (odd ratios 4.37 ApoE4 women and 3.14 in ApoE4 men). Moreover, ApoE3/4 female carriers of age 55 to 70 had increased risks of cognitive impairment than male carriers (Neu et al., 2017). Consistently, other studies also demonstrated that female ApoE4 carriers have a higher risk of converting into AD (Corder et al., 2004) and a faster decline of cognitive function when compared to non-ApoE4 carrier women or men with any APOE genotype (Beydoun et al., 2012). It is interesting to note that the gender effect is most pronounced among ApoE3 and ApoE4 heterozygotes (Farrer et al., 1997), and that these disparities are most apparent in women with menopause (Burger et al., 2002). It is suggested that alterations in estrogen levels may account for increased risks of developing AD among menopausal women compared to men.
Since estrogen level decline stemming from menopause is implicated in AD related cognitive vulnerabilities, estrogen therapy was first implemented to ameliorate AD related cognitive deficits. Interestingly, there was a notably trend in which women with ApoE4 receiving estrogen therapy exhibited more cognitive decline than ApoE4 women without estrogen treatment. This contrasts with ApoE2 and ApoE3 women carriers who demonstrated cognitive improvement from estrogen therapy (Burger et al., 2002). One possible explanation is the interplay between estrogen receptor alpha (ERα) and estrogen receptor beta (ERβ). Both ERα and ERβ can regulate the expression of ApoE lipoprotein as demonstrated in vitro in HT-22 cells and hippocampal neurons, and in vivo in mice (Wang et al., 2006). Upon stimulating ERα with either 17β-estradiol (E2) or propylpyrazole triol, an ERα-selective ligand, ApoE protein expression was up-regulated. In contrast, subjecting ERβ to an ERβ-selective ligand such as diarylpropionitrile down-regulated ApoE gene expression and protein levels (Wang et al., 2006). It is possible that ApoE4 carriers receiving estrogen therapy suffered an exacerbated ApoE4 overexpression stemming from ERα stimulation while the ApoE2 and ApoE3 carriers on estrogen therapy benefited from the protective effects of ApoE2 and ApoE3 over-expressions, which could account for the complex impact that estrogen therapy has on postmenopausal AD individuals that are ApoE3 and ApoE2 carriers versus ApoE4 carriers (Yaffe et al., 2000). From a therapeutic standpoint, this highlights not only ERα and ERβ as potential drug targets for AD treatment but also the need for a more personalized therapeutic approach; what works for ApoE2 and ApoE3 carriers might not benefit and may in fact be detrimental to ApoE4 carriers.
In addition to gene expression, estrogen also serves to regulate various pertinent metabolic pathways in the brain such as glycolysis and the Krebs cycle within the mitochondria (12). Studies with ovariectomy mice models that mimic postmenopausal hormonal conditions demonstrated a shift in bioenergetics that diminished glucogenic pathways via shunting of glucose transport in the brain and upregulated ketogenic compensatory pathways (Yao et al., 2012). This clearly suggests that estrogen promotes glucogenic metabolic pathways and lessens the need for ketogenic pathways for adenosine triphosphate (ATP) generation in female brains. Previous studies pertaining to AD have also implicated metabolic shift from one that is glucogenic to a more ketogenic system that relies on utilizing white matter for fuel in ApoE4 carriers (Reiman et al., 2005). This seems to suggest that ApoE4 brains heavily rely on a dual metabolic pathway consisting of both glucose and keto bodies. For ApoE4 carriers, estrogen therapy that suppresses ketogenic pathways may compromise ATP production because the brain of ApoE4 carriers may be less acclimated to an exclusive glucogenic pathway. Conversely, promoting glucogenic metabolic pathways while simultaneously mitigating ketogenic pathway in brains that are more tune to using glucose as the primary metabolic pathway, as is the case with ApoE2 and ApoE3 carriers, may confer a more positive and conducive bioenergetic profile for generating ATP. This may offer another contributing factor and explanation for why estrogen therapy disproportionally benefits ApoE2, and ApoE3 carriers but harms ApoE4 individuals. Ultimately, the effects observed from estrogen therapy may be a culmination of mechanisms that amalgamates that what is observed in the clinical setting.
Lastly, in addition to influencing energy profiles, ApoE4 may also influence expression of β-site APP cleavage enzyme (BACE1), a key enzyme in the amyloidogenic pathways responsible for generating amyloid-beta (Vassar, 2004). In AD mice models expressing APPSwe and tauP301L (3xTg) with various ApoE genetic profiles, ApoE4 female carriers were disproportionally overexpressing BACE1 compared to male ApoE4 and other ApoE genetic counterparts (Hou et al., 2015). These results confirm what is currently established in AD research; females with ApoE4 carry the highest risk for developing AD (Payami et al., 1994). While the exact mechanism for this phenomenon is uncertain, there may be two possible explanations. The first possibility is the relationship between ApoE and cholesterol. ApoE4 has been shown to increase cholesterol concentration in both CSF and blood (Notkola et al., 1998; Papassotiropoulos et al., 2002). Elevated cholesterol levels have also been implicated to stimulated BACE1 activity (Runz et al., 2002). Therefore, ApoE4 females accelerate BACE1 by indirectly modulating cholesterol quantities. A second possibility is increased APP recycling by ApoE4, which in turn increases for APP exposure to BACE1 for cleavage (Yang et al., 2003).
Nevertheless, several AD clinical trials studies have demonstrated the role of ApoE as an important determinant for treatment responses, indicating major differences between ApoE4 carriers and non-carriers and between males and females (Hanson et al., 2015; Salloway et al., 2014). For example, hormone therapy may exhibit beneficial effects on cognition, but only seen in non-ApoE4 carriers (Kunzler et al, 2014). On the other hand, hormone therapy reduced AD risks in ApoE4 carriers more so than in non-ApoE4 carriers (Tang et al., 1996). Further research is needed to address molecular mechanisms underlying sex-specific impact on AD pathogenesis in ApoE4 carriers and non-carriers over the life span. In addition, the effects of ApoE4 genotype and its correlation with sex differences should be taken into considerations when evaluating the efficacy of AD clinical trials.
4.1.2. Other AD genes:
Presenilin 1 (PS1) gene is an important AD gene. PS1 protein, along with its genetic counterpart, presenilin 2 (PS2) protein encompasses a key component of the γ-secretase complex and its catalytic core, which cleave amyloid precursor protein (APP) to generate Aβ (Wolfe, 2006). Mutations in PS1 and PS2 could result in faulty cleavage of APP, leading to elevated Aβ generation and altered Aβ42/Aβ40 ratio (Hutton and Hardy, 1997). There are some experimental evidence suggesting that the effects of PS1 on AD pathogenesis might differ based on genders. It was shown that Aβ levels were increased with aging in APP/PS1 transgenic mouse models. Additionally, female cohorts consistently demonstrated greater Aβ burden compared to males at the same age (Wang et al., 2003). A separate study conducted by Piscopo et al found that neocortical and hippocampal PS1 expression was much higher in female wildtype mice than in males. However, these differences in PS1 expression were attenuated after brain maturation (Thakur and Ghosh, 2007). Nevertheless, these results support that sex dimorphic differences in PS1 expression might play a role in differential brain development, and that male and female brain might have different mechanisms for regulating PS1 expression (Ghosh and Thakur, 2008a; Ghosh and Thakur, 2008b; Thakur and Ghosh, 2007).
Interestingly, PS1 expression may be modulated by gonadal sex hormones. It has been shown that PS1 expression in cerebral cortex was down-regulated by 17β-estradiol and testosterone in both male and female mouse models (Ghosh and Thakur, 2008a). Therefore, the reduction in estrogen levels after menopause may contribute to increased PS1 expression leading to amyloid plaque buildup. Males on the other hand experience a less dramatic hormone loss compared to females. Additionally, males may also convert testosterone into estradiol (Wu et al., 2009), which confers more protection against PS1-mediated AD pathology. Clearly, sex hormones regulate PS1 expression, and more research is need in this area to fully elucidate AD gender differences pertaining to PS1.
Besides PS1/PS2, brain-derived neurotrophic factor (BDNF) plays an important role in memory formation (Kemppainen et al., 2012; Rabbitts et al., 1985). It also protects neurons against Aβ-induced toxicities (Doi et al., 2013; Rohe et al., 2009). The association of BDNF polymorphism with AD was controversial (Li et al., 2017b). Several studies identified female-specific association between BNDF polymorphism and AD. A meta-analysis identified the BDNF Val66Met polymorphism increased AD risks in females (Chen et al., 2014a). These studies were supported by others (Fukumoto et al., 2010; Li et al., 2017a; Li et al., 2017b). Matyi et al also found two additional single nucleotide polymorphisms (SNPs) of BDNF highly associated with AD women (Matyi et al., 2017). On the other hand, the expression of BDNF was reduced in AD patients (Connor et al., 1997; Hock et al., 2000; Li et al., 2017a; Peng et al., 2005; Qin et al., 2017), and in AD mouse models (Francis et al., 2012; Naert and Rivest, 2012). In APP/PS1 mouse models, female mice presented with a dramatic drop of BDNF levels upon stress, whereas males had an increase in BDNF levels after exposure to chronic stress (Autry et al., 2009). In addition, ApoE4 reduces BDNF levels through epigenetic regulation of histone deacetylase (HDAC) activities (Sen et al., 2015; Sen et al., 2017). Future studies are necessary to characterize molecular mechanisms underlying sex dimorphism of AD susceptibility in the presence of risk genes, and to investigate how they interact with each other to affect AD pathogenesis.
4.2. The Interaction between Sex and Comorbid Risk Factors in AD Pathogenesis
A number of studies have identified cardio-metabolic risk factors such as obesity, type 2 diabetes (T2DM), and metabolic syndrome associated with AD (Barnes and Yaffe, 2011). However, only a few studies analyzed how sex differences influence these factors during the progression of AD (Kautzky-Willer et al., 2016).
4.2.1. Vascular risk factors
It is known that vasculature system in men is significantly different from that of women. While microvascular disease is a major contributor to cardiovascular disease in women, obstructive coronary artery disease is a main contributor in men (Bairey Merz et al., 2006). Women also present with an increased risk of diabetic complications including myocardial infarction and depression, both of which are AD risk factors (Kautzky-Willer et al., 2016). Recent studies examined the association between the risks of developing dementia and various cardiovascular risk factors like coronary artery disease (CAD), atrial fibrillation, myocardial infarct, hypertension and heart failure, as well as sex-specific effects (Kim et al., 2018). For example, men have a much higher incidence of CAD than women in all ages (Kivipelto et al., 2001a; Kivipelto et al., 2001b), which is believed to be due to protective effects of estrogen against atherosclerosis, oxidative stress and inflammation in women (Mendelsohn and Karas, 2005). CAD is associated with cognitive decline (Roberts et al., 2010) with brain microvascular lesions as proposed mechanisms (Rosano et al., 2005). Studies have shown that microvascular lesions reduced intracranial blood flow, impaired blood-brain barrier (BBB) integrity and reduced Aβ clearance through BBB leading to brain damage (Bell and Zlokovic, 2009; Kovacic et al., 2011a; Kovacic et al., 2011b).
In fact, BBB failure is one of the central mechanisms underlying development of cerebral small vessel diseases and dementia. It contributes to neurodegeneration and rapid progression from vascular defects to major neurological diseases (Sweeney, 2019; Sweeney, 2019b). The genetic mutations of AD genes PS1 and PS2 have been associated with the BBB malfunction and higher risks for AD (Sweeney, 2019b) (Basun, 2008; Cohen, 2009). Genome-wide association studies have identified AD risk loci on genes like PICALM, CLU and SORL1 that are well-known to regulate BBB transport and clearance function (Carrasquillo, 2010; Corneveaux, 2010; Harold, 2009; Lambert, 2009; Rogaeva, 2007). A connection between PICALM polymorphisms and AD has been identified, and lower levels of PICALM may increase AD risks (Harold, 2009; Lambert, 2009). The amino terminal tail of PICALM contains a PIP2-binding domain which allows PICALM to sense membrane curvature and modulates the size of clathrin-coated vesicles (CCV). PICALM also has a key role in the internalization and intracellular traffic via the SNARE-mediated fusion of CCV to endosomes (Miller, 2011). These functions play important roles in tau clearance from autophagy pathway (Moreau, 2014) and Aβ clearance across the BBB (Zhao, 2015). CLU is another important AD risk gene (Carrasquillo, 2010; Corneveaux, 2010), which encodes for APOJ and regulates lipid transport and membrane recycling (Calero, 1999; Nuutien, 2009). In the brain, astrocytes secrete APOJ that binds to soluble Aβ (Nuutien, 2009), and APOJ regulates Aβ clearance by promoting Aβ42 efflux though the BBB (Tanzi, 2012). Currently there is no reported literature regarding the sex-specific effects on the function of these genes in AD.
Similarly, the incidence of atrial fibrillation, MI and heart failure is much higher in men than in women with limited studies examining their association with dementia (Kim et al., 2018). Hypertension is known to increase risk for cerebrovascular disorders but whether it directly affects the risk of developing dementia remains controversial (Kim et al., 2018). A recent study demonstrated an association between diastolic blood pressure and Aβ levels (Shah et al., 2012). Interestingly, middle age hypertension is highly associated with increased risks of dementia in females but not in males (Gilsanz et al., 2017). Future studies are needed to understand the sex-specific effects of cardiovascular risk factors on AD progression, specifically on how they vary by different types of disorders and by age.
Some studies have found that stroke prevalence in men is significantly higher than in women and that men develop strokes at much earlier ages (mean age 68.6 year old versus 72.9 year old in women) (Appelros et al., 2009). In contrast, others have shown that women possess a higher risk of developing stroke given their longer life expectancy (Seshadri et al., 2006) and the increased likelihood of developing comorbid conditions such as thrombosis and diabetes (Cheng and Kong, 2016). Several lines of studies support the important role of insulin-like growth factor-1 (IGF-1) in acute ischemic stroke. For example, lower serum IGF-1 levels are correlated with an increased risk of stroke, whereas higher levels associated with better outcomes post stroke (De Smedt et al., 2011; Dong et al., 2014; Tang et al., 2014). Similar findings have been related to AD, where low IGF-1 levels are associated with an increased risk of AD progression, and higher levels with larger brain volumes, even in normal individuals without history of stroke or dementia (Westwood et al., 2014). Furthermore, data suggested that levels of IGF-1 were reduced during aging (Rosario, 2010), and much more so in males than in females (Waters et al., 2003).
The proposed mechanisms for IGF-1’s protective effects against neurodegeneration include anti-inflammation, anti-thrombotic and survival-promoting effects (Jin et al., 2013; Li et al., 2010; Patel et al., 2014). Studies in animal models have identified neuroprotective effects of IGF-1 against ischemic strokes in both males and females, at young and old ages (Sohrabji, 2017). The IGF-1 replacement treatment post-stroke was found to rescue neurotoxicity, ameliorate stroke-associated brain damage and motor impairment, reduce neuro-inflammation and BBB disruption in middle-aged ovariectomized or estrogen deficient female rats (Bake et al., 2014; Selvamani and Sohrabji; Selvamani and Sohrabji). Moreover, modulating IGF-1 levels by miRNA manipulations demonstrated significant sex differences in treatment effectiveness, with effects only seen in females but not in males or estrogen deficient females (ovariectomized animals), suggesting that the IGF-1 treatment may be strongly influenced by hormonal environment (Selvamani et al.).
New evidence suggests a sexual dimorphism in the cellular death pathways during ischemic stroke (Gibson; Liu et al., 2011). Poly-ADP ribose (PAR) is important molecule inducing programmed cell death (Strosznajder et al., 2012). Poly (ADP-ribose) polymerase-1 (PARP-1) is a key enzyme regulating PAR levels with important roles in transcriptional regulation and long-term potentiation (Hernandez et al., 2009). In AD patients, pathological activation of PARP-1 led to PAR accumulation in the brains (Cozzi et al., 2006). In addition, PARP-1 polymorphisms are identified in AD (Liu et al., 2010). PARP-1 was found to be activated in AD with an increased Aβ generation (Martire et al., 2013). Moreover, Aβ activates PARP-1 resulting in PAR-modulated mitochondrial release of apoptosis-inducing factor (AIF), ultimately causing cell death (Yu et al., 2006). PARP-1 activation plays a major role in cardiovascular diseases (Song et al., 2013; Sun et al., 2015). The downstream effectors of PARP-1 including AIF and PAR have been shown to induce cell death after ischemic insults only in male mice (Yuan et al., 2009). Recent animal studies have further demonstrated sexually dimorphic neuroprotective effects through the inhibition of PARP-1 that is seen only in male mice, but not in females (Lang and McCullough; Li and McCullough; Liu et al., 2011).
Another type of vascular risk factors unique to females is known as hypertensive pregnancy disorders, which affects more than 10% of all pregnancies. These disorders include preeclampsia, eclampsia, and chronic and gestational hypertension (Mielke et al., 2016; Postma et al., 2014). There has been direct association among these conditions with impaired cognition as well as multiple brain lesions (Aukes et al., 2007; Mielke et al., 2016; Postma et al., 2014). However, most of published studies were limited by small sample sizes and short time periods of cognitive function assessment (less than a decade after development of hypertensive pregnancy disorders). A recent large cohort study of multiethnic women around 60 years of age found that females with prior history of hypertensive pregnancy disorders had greater degrees of brain atrophies decades after their pregnancies when compared to normotensive pregnancies (Mielke et al., 2016). It remains unclear whether there is a shared mechanism that increases the risk of developing hypertensive pregnancy disorders and cognitive impairment after pregnancy, or if hypertensive pregnancy disorders directly increase the risk of developing cognitive impairment. The current research suggests that alterations in serum levels of angiogenic factors contribute to the pathogenesis of hypertensive pregnancy disorders (Hod et al., 2015). Angiogenesis is closely regulated by angiogenic factors to maintain optimal microenvironment for blood vessels. Reduced levels of angiogenic factors such as vascular endothelial growth factor (VEGF) and transforming growth factor beta led to endothelial dysfunction and clinical manifestations of pregnancy hypertensive disorders (Romero and Chaiworapongsa, 2013; Venkatesha et al., 2006). Therefore, it becomes critical to better predict or prevent hypertensive pregnancies, and more importantly to understand the mechanisms by which angiogenic factors contribute to hypertensive pregnancy disorders and their associated cognitive deficits.
4.2.2. Sleep
Sleep plays a critical role in the maintenance of health and modulates behavioral and physiological functions. Sex-related differences in risks of developing sleep disorders are well established. While studies show that young healthy adult women tend to have better sleep quality than men (Goel et al., 2005), most studies in patients of all ages indicate that women present more sleep problems including insomnia and inadequate sleep compared to men (Bixler et al., 2002; Zhang and Wing, 2006). Women are more prone to present clinical symptoms from inadequate sleep, and they may also be more inclined to report symptoms. In general, there is a correlation between a decline in slow-wave sleep and aging processes (Carrier et al., 2001). Sleep disorders increase with age, reaching a peak for women during menopause (Bixler et al., 2001). Impaired circadian rhythm and sleep disorders are frequently seen among individuals with AD, which influence the development and progression of AD pathology (Ju et al., 2014). For example, the production and clearance of Aβ are correlated with the wake/sleep cycles. While wakefulness is associated with Aβ production, clearance of Aβ occurs mainly during the sleep stage (Cedernaes et al., 2017). Shorter sleep duration and poorer sleep quality are associated with increased Aβ accumulation and risks of developing AD in older adults (Lim et al., 2012; Spira et al., 2013).
It has been reported that brain has its own lymphatic-like clearance system named the glymphatic system which operates via a network of perivascular clearing mechanisms (Kress et al., 2014). The glymphatic system is suggested to function exclusively during sleep (Xie et al., 2013). Moreover, animal studies have characterized a clearance system in the brain that involves bulk flow of interstitial fluid and water-channel protein, aquoporin-4 (AQP4) localized throughout astrocytic processes (Lundgaard et al., 2017). AQP4 function has been directly linked to Aβ clearance, most likely through the glymphatic system as suggested by the AQP4 knockdown mice (Yang et al., 2012). It is hypothesized that poor-quality sleep is associated with reduced Aβ clearance and increased Aβ accumulation in the brain due to impaired AQP4 function (Rainey-Smith et al., 2018). A recent study proposed a synaptic homeostasis hypothesis, where sleep is suggested to selectively attenuate synaptic strength between neurons (Tononi and Cirelli, 2014). With electrophysiological, molecular, and behavioral studies, this report suggested that during wakefulness, demand of synaptic strengthening is high and requires more energy. Conversely, sleep promotes synaptic weakening of neurons, decreases cellular stress, and allows the reestablishment of energy reserves in the brain. This and other studies suggest that sleep has an important role in re-sculpting synaptic landscape and restoring brain energy reserves. Future research is needed to understand in depth the association between sleep and the risk of developing neurodegenerative diseases, especially AD, and whether the risk changes within different sex.
4.2.3. Psychiatric comorbidity
Depression is another well-known risk factor for AD in older adults (Ownby et al., 2006). It has been reported that women have two-fold increase of risk developing depression than men, which peaks during menopause (Bromberger et al., 2011). Clinically significant depressive symptoms in women were associated with an increased risk of developing cognitive impairment (Goveas et al., 2011). In contrast, another study indicated that men with depressive symptoms have a significantly higher risk of developing AD compared to women counterparts (Dal Forno et al., 2005). A possible explanation for the discrepancies between these two studies is that male subjects were more likely to present with severe depression upon evaluation since men were less willing to admit depression symptoms, which could post a higher risk of developing AD when compared to their female counterparts.
Hormonal changes may be key factors underlying sex-specific differences in depression. The biggest indicator is that the increased prevalence of depression seems to correlate well with hormonal changes in women most notably during puberty and menopause (Soares and Zitek, 2008). This is bolstered by the protective effects of estrogen against depression in post-menopausal women (Gordon and Girdler, 2014). However, men have a lower rate of depression even without estrogen. One possible explanation is the ability of male brains to convert circulating testosterone into 17β-estradiol, and to maintain a more consistent 17β-estradiol reservoir for protection against depression without having drastic androgen spikes (Wu et al., 2009).
The sexual dimorphic brain activities in depression may also be observed at the transcriptional level. Labonté B et al analyzed the extent of transcriptional expression differences across six regions of postmortem human brains of male and female depression and control subjects. They demonstrated an upregulation in transcription signatures across all six brain regions of male and female depression subjects when compared to controls, but with few overlaps in transcriptional signatures between the two genders, suggesting a gender based dimorphic transcriptional signatures in patients who had depression (Labonte et al., 2017). These transcriptional expression differences were also found in C57BL/6 mice subjected to chronic variable stress (Labonte et al., 2017). One specific genetic expression of interest is DUSP6, which can modulate synaptic signaling activity via ERK1/2 phosphorylation (Mazzucchelli et al., 2002; Muda et al., 1996). Specifically, DUSP6 down-regulation in pyramidal neurons of ventromedial prefrontal cortex in female C57BL/6 mice induced an increase in frequency of spontaneous excitatory postsynaptic currents compared to control. Conversely, male showed no such distinction in DUSP6 down-regulation (Labonte et al., 2017). This is one of the examples how different transcriptional expression profiles exist between males and females, which could be responsible for brain dimorphism between the two genders.
Stress has also been linked to AD and increased cortisol levels during stress have been associated with cognitive impairment (Csernansky et al., 2006; Pedersen et al., 2001). Changes in stress hormone signaling have been found in AD patients (Reisberg et al., 1987). For example, the levels of corticotrophin releasing factor 1 (CRF1) were found much higher in hippocampal brain regions of AD patients (De Souza, 1995). The signaling changes in hypothalamus-pituitary-adrenal axis are associated with hippocampal atrophy (O’Brien et al., 1996). Sex dimorphic responses to stress have been reported with women being more vulnerable to stress-related disorders. One study reported that levels of cortisol in female mild-to-moderate AD patients were much higher compared to males (Rasmuson et al., 2011). Several studies using AD mouse models further supported sex-specific differences in response to stress challenges (Devi et al., 2010; Sierksma et al., 2012; Sierksma et al., 2013; Sotiropoulos et al., 2015). It is hypothesized that heightened CRF1 signaling is associated with increased AD pathologies including elevated Aβ and pTau generation, as well as worsened Aβ toxicity, and that the sex dimorphic activation of CRF1 and GRCR-PKA signaling may contribute to sex dimorphism in AD pathogenesis (Fisher et al., 2018).
4.2.4. Diabetes Mellitus
The relationship between type 2 diabetic mellitus (T2DM) and AD has been well studied with high prevalence of both diseases among the aging population. Growing epidemiological research suggests that there is a strong comorbid association between both diseases (Cha et al., 2014), possibly because T2DM and AD have similar underlying pathophysiological processes (Moreira, 2012). Previous research spanning over the last decade implicates insulin insensitivity to AD pathology from in vitro experiments, clinical observations, and animal models (Chen et al., 2014b; Craft, 2006; Schioth et al., 2012). It has been found that chronic hyperglycemia in the brain is associated with chronic hyperinsulinemia, leading to brain insulin resistance manifested as impaired insulin signaling and disrupted BBB integrity leading to cognitive impairment and neurodegeneration (Neth and Craft, 2017; Yoo et al., 2016). Abnormal insulin signaling also increased Aβ accumulation and tau hyper-phosphorylation (Cao et al., 2007; Currais et al., 2012; Devi et al., 2012; Ho et al., 2004; Mehla et al., 2014; Vandal et al., 2014), as well as inhibited APP degradation leading to increased Aβ production (Yang et al., 2013). Interestingly, the association between AD prevalence and insulin insensitivity is stronger among female patients than in males (Ekblad et al., 2015). Additionally, clinical trials using intranasal insulin administration showed memory enhancement particularly among women and non-ApoE4 carriers, which further supports the role of insulin in AD pathology progression (Benedict et al., 2008; Reger et al., 2006).
A possible explanation of increased insulin insensitivity among women that leads to AD progression is the loss of estrogen after menopause (Asthana, 2003). It is well established that estrogen confers protection against weight gain and insulin insensitivity via estradiol-induced up-regulation of heat shock protein HSP72 and diminished inflammatory response (Chung et al., 2008). This result is also corroborated in two separate studies using animal models treated with high fat diet (HFD). In the first study, a greater increased in insulin insensitivity and glucose intolerance was found in male mice treated with HFD than in female mice (Estrany et al., 2013), suggesting sex hormones at play. While testosterone does confer some protection against weight gain and insulin insensitivity (Zitzmann, 2009), it seems that estrogen is more protective than testosterone as seen in the HFD mouse model studies (Estrany et al., 2013). In the second study, estrogen receptor knockout mice treated with HFD presented with increased insulin resistance and glucose intolerance (Riant et al., 2009). These results are consistent with the fact that males accentuate a consistent but slow decrease in testosterone levels through aging while females experience a sudden drop in estrogen during menopause and hence why females are more afflicted by comorbidities later in life that can be modulated by sex hormones as exemplified by insulin insensitivity in this case. This is bolstered by a separate study where protection against obesity is lost in post-menopausal women (Meyer et al., 2011).
4.2.5. Thyroid dysfunction
Hypo- and hyperthyroidism are implicated as AD risk factors in the elderly population (de Jong et al.; Kalmijn et al., 2000). A large cohort study reported an association between changed levels of thyroid-stimulating hormone (TSH) and dementia exclusively among women (Tan et al., 2008). Suggested mechanisms include alterations on Aβ and tau deposition, neuronal survival, and indirect mediation through vascular risk factors (de Jong et al., 2009; Tan et al., 2008).
Reduced levels of TSH before any memory decline may predict the conversion of MCI to AD (Annerbo et al., 2006). Several studies indicated that thyroid hormones regulate APP expression, and modulate APP processing and secretion (Belandia et al., 1998; Latasa et al., 1998). For example, low levels of thyroid hormones increased APP expression and Aβ levels leading to AD. On the other hand, studies implicate that lower TSH levels could be a consequence of AD rather than the cause. The AD-associated neurodegenerative processes may reduce secretion of TSH-releasing hormone (TRH) and blunt pituitary responses to TRH, manifested as reduced TSH and thyroxine (T4) levels (Shi et al., 1993). TRH depletion itself can also induce AD progression by promoting tau hyper-phosphorylation (Luo et al., 2002). In rodents, treatments with TRH analogues increased acetylcholine synthesis and release (Ogasawara et al., 1996). Hyperthyroidism with increased T4 levels has been found in sub-clinical dementia patients. It is believed that hippocampal atrophy occurred during early stages of AD generates less feedback regulation on the hypothalamus-pituitary-thyroid axis, resulting in higher levels of free T4 (den Heijer et al., 2006). On the other hand, hyperthyroidism patients are found to have increased oxidative stress and lower antioxidant metabolite levels (Bianchi et al., 1999), which could potentially increase AD risks through inflammation.
Overall, comorbidity factors are important contributors to cognitive and functional decline during AD progression. They bring considerable implications for both patients and healthcare providers since more expertise, service, and requirements are needed compared to when dealing with only one condition. Clinical patients would benefit greatly from improving multidisciplinary in-depth studies to understand the underlying mechanisms, emphasizing on the pathological time-course and sex difference implications. A major challenge in comorbidity studies is that most research relies upon statistical analysis of clinical readouts. To fully understand the mechanistic link between comorbid conditions and AD pathogenesis, and how they differ between sexes, an integration of molecular information available among diseases and how they altered by sex are needed.
4.3. The Interaction between Environmental Risk Factors and Sex in AD Pathogenesis
Many environmental factors such as lifestyle (e.g. diet and exercise), inflammation and immune responses, prior exposure to traumatic brain injury and stress may interact with sex to impact the development and/or progression of AD.
4.3.1. Lifestyle risk factors
Numerous studies have suggested obesity as an AD risk factor (Feart et al., 2009; Grant, 1999). Fat consumption and caloric intake are among the most significant dietary risk factors for AD, both contributing to oxidative stress either directly or indirectly. Studies have shown that high fat diet accelerated cognitive decline and AD-related neuropathology in human and animal models (Barron et al., 2013; Elias et al., 2003). The sex-specific differences have been implicated in obesity, adiposity, insulin resistance and metabolic syndromes in both experimental animal models and human studies (Franconi et al., 2008; Regitz-Zagrosek et al., 2006). It is speculated that cognitive decline and AD-related neuropathology after exposure to high fat diet might be different between males and females. Recently, studies in animal models and human subjects have demonstrated that long-term exposure to high fat diet exerted more profound impairments in cognitive function or synaptic plasticity in males than in females (Elias et al., 2003; Hwang et al., 2010). Higher insulin resistance was observed in obese male cohorts suggesting male vulnerability in this regard. Conversely, the Mediterranean diet, rich in fruits, vegetables, fish and olive oil, has been studied for AD prevention trials and is considered highly beneficial (Feart et al., 2009; Morris et al., 2015a; Morris et al., 2015b). It is important to determine any possible sex-specific differences in response to AD preventive trials like healthy diet treatment. The sex dimorphic effects of obesity are manifested in the degrees of BBB disruption, oxidative stress, and inflammation. Overweight and obese women have increased BBB disruption while obese men have significantly more gray matter loss (Gustafson et al., 2007; Taki et al., 2008). Obesity and inflammation are linked through involvement of oxidative stress and free radical production. It has been found that obesity induces a much greater inflammatory response with an increased likelihood of developing metabolic disturbances in women (Ahonen et al., 2012; Khera et al., 2009; Thorand et al., 2007).
Sex differences have been suggested in response to physical exercise as well. Studies have shown that physical exercise improved cognitive performance in AD transgenic mice and ameliorated AD-related white matter changes in early stage of diseases (Nichol et al., 2009; Yuede et al., 2009; Zhou et al., 2018). It was found that female AD mice have lower volume of white matter and less myelinated fibers. However, after running exercise, a greater improvement of axonal diameter of myelinated fibers was observed in the white matter of the female AD mice, suggesting that exercise may improve cognitive function more effectively in female AD transgenic mice (Zhou et al., 2018). Studies have also indicated greater benefits in cognition from aerobic training (AT) in females than in males (Baker et al., 2010; Colcombe and Kramer, 2003). Higher levels of physical activity lower dementia risks more so in women than in men (Laurin et al., 2001; Middleton et al., 2008). Studies led by Varma et indicated older females but not males who underwent greater amounts of walking activities over one week measured objectively presented with larger hippocampal volumes (Varma et al., 2015; Varma et al., 2016). At the molecular level, Cotman et al. (2007) proposed induction of neurotrophic factors such as BDNF, IGF-1 and VEGF as a possible mechanism underlying that beneficial effects of AT (Cotman et al., 2007). Animal studies further strengthen the concept that brain BDNF mediates beneficial effects of AT by showing that inhibition of hippocampal BDNF signaling blocks the effects of AT on cognitive function (Maass et al., 2016; Vaynman et al., 2004a; Vaynman et al., 2004b). Interestingly, the AT-induced increase in brain BDNF levels may be sex-dependent (Chan and Ye, 2017; Szuhany et al., 2015; Venezia et al., 2016), with greater effects observed in females than in males both from animal studies (Barha et al., 2017b) and human studies (Barha et al., 2017a).
4.3.2. Inflammation and neuro-immune responses
Several lines of evidence support the importance of inflammation in AD. Recent genetic studies identified polymorphisms of genes involved in immune responses such as CD33 (Bertram et al., 2008), TREM2 (Kleinberger et al., 2014) and CR1 (Lambert et al., 2009). Environmental factors that increase inflammation such as obesity and TBI are associated with increased risks of AD (Breunig et al., 2013; Moser and Pike, 2016). The inflammatory response is also a common effect following viral infection (Brothers et al., 2018; Kumar et al., 2016; Van Lenten et al., 2001). In AD, activated microglia surrounding amyloid plaques produce higher levels of pro-inflammatory cytokines and accelerate Aβ accumulation (Glass et al., 2010; Sastre et al., 2008; Sheng et al., 2003). A study performed on a group of individuals over the age of 55 found that individuals taking non-steroidal anti-inflammatory drugs (NSAIDs) for two years presented with a lower risk of AD progression than individuals not taking NSAIDs (in t’ Veld et al., 2001).
Sex-specific differences have been observed in immune system including neuro-immune modulation of memory and cognitive function (Tronson and Collette, 2017). For example, greater immune activities are seen in females than in males with different pathways and cells triggered after stimulation (Klein et al., 2015; Moxley et al., 2002). These differences correlate with higher susceptibility to infections in men, and higher incidence of autoimmune disorders in women (Fairweather et al., 2008). It is suggested that sex hormones and sex chromosomes contribute to sex-specific differences in immune system (Tronson and Collette, 2017). Similarly, sex differences are also seen in the neuro-immune system with unique immune-neuron interaction, cytokine responses and signaling in males and females (Sorge et al., 2011). The interactions between neurons and microglia via chemokine CX3CL1 are critical for memory formation, and immune challenges modify memory through activation of microglia (Sheridan and Murphy, 2013). Following immune stimuli, males and females demonstrated different cytokine activation. For example, after systemic LPS injection, males show a stronger and sustained hippocampal activation of cytokines than females (Hudson et al., 2014; Pyter et al., 2013). Dysregulation of microglia and astrocyte-mediated neuro-immune responses has been strongly implicated in AD and TBI (Dzamba et al., 2016; Sajja et al., 2016). However, how the function of these cells affected in sex-specific manners remains unaddressed. It is also important to identify sex-specific cellular and signaling mechanisms in neuro-immune activation which could provide critical information for developing sex-specific AD therapies.
4.3.3. TBI
TBI is also one of the most studied environmental risk factors for AD, increasing the chance of developing the pathology nearly four-fold (DeKosky et al., 2010; Schofield et al., 1997). Epidemiological studies following TBI and its association to neurodegeneration have suggested that genetic risk factors such as ApoE4 can modulate the effect of the impact (Cao et al., 2017; Hayes et al., 2017). The risk magnitude for AD may increase with TBI frequency, age of when the injury took place, and sex differences among the population. While males account for the majority TBI patients and have been overly represented in preclinical and clinical studies, TBI in females still a major contributor to morbidity and mortality among children and elderly subjects (Colantonio et al., 2010; Scott et al., 2015). One review by Berry et al. demonstrated that moderate-to-severe TBI female subjects at peri-menopausal and post-menopausal stages had lower risks of mortality than males, but this advantage was not seen in pre-menopausal female patients (Berry et al., 2009). In addition, female patients after moderate-to-severe TBI exposure had few complications and shorter hospital stay than males (Berry et al., 2009).
TBI is often associated with higher risks of developing mental health comorbid conditions. Data suggest that female TBI patients are more prone to develop or report symptoms of depression, anxiety, headache and impaired memory when compared to men (Colantonio et al., 2010; Farace and Alves, 2000; Scott et al., 2015). Similarly, studies of veterans with TBI found that female veterans with a history of TBI are more likely to develop depression and anxiety whereas male TBI veterans are more likely to have alcohol use disorder and post-traumatic stress disorder (PTSD) (Epstein et al., 2019). In animal model studies, female mice performed worse in spatial working memory tasks than males post-TBI exposure (Tucker et al., 2016).
Mechanisms such as sex hormone regulation, sex differences in cerebral blood flow autoregulation as well sex-specific microglial responses in TBI have been proposed (Caplan et al., 2017). For example, male mice after cortical compact injury (CCI) presented with more profound pro-inflammatory microglial phenotypes and astrogliosis when compared to females. Subsequently, cytokine profiles associated with increased neuronal cell death and brain lesion were identified in male mice post CCI (Villapol et al., 2017). A greater density of microglia around lesion site was also reported in male mice post cortical stab lesions when compared to females (Acaz-Fonseca et al., 2015). A recent report shows that expression patterns of brain cannabinoid receptors (CB1 and CB2) are differentially modified in female and male rats after repeated stress challenges, suggesting a potential mechanism underlying sex-related differences after TBI exposure and TBI/PTSD development (Xing et al., 2014). Together, the sex dimorphism in TBI have been reported in animal models but more studies are needed in human clinical studies. It becomes essential for future investigations to determine the mechanisms of developing post-TBI neurodegeneration and cognitive decline based on sex, age, and other factors that may modify the risk or protection.
It should be noted that comorbid diseases share some common mechanisms. Hence, one therapy used to treat one condition could also be a risk for initiating or accelerating the progression of another disorder (Bohlega and Al-Foghom, 2013; Shin and Chung, 2012). Interestingly, most studies have been focusing on specific biological pathways without considering how dysregulated effectors can modulate the comorbid association among diseases (i.e. SNPs, epigenetic modifications, miRNA, genes and proteins). Therefore, there is an urgent need to create a comprehensive model that takes into considerations of molecular mechanisms involved in comorbidities of AD in time-dependent and sex-specific manners.
5. Sex-specific treatment responses in AD
The stratification of sex when evaluating drug efficacies and safety profiles has made significant impact in clinical practice such as aspirin and warfarin in stroke (Berger et al., 2006; Cordonnier et al., 2017). On the other hand, the studies of sex differences in AD treatment are rather limited. Systematic reviews of currently available results from randomized clinical trials regarding efficacy and safety of cholinesterase inhibitors and memantine in total 20,688 patients found very limited data studying sex differences in these regards (Canevelli et al., 2017; Mehta et al., 2017). Similarly, limited data are available on sex stratified effects of drugs tested in recent AD clinical trials such as phase III studies of γ–secretase inhibitor semagacestat (Henley et al., 2009), anti-Aβ antibody solanezumab (Doody et al., 2014) and bapineuzumab (Salloway et al., 2014), or BACE inhibitor verubecestat (Egan et al., 2018). Several preventive trials such as FINGER and MAPT are yet to release any data for assessing sex specific effects on lifestyle stratification (Ferretti et al., 2018). One interesting study of a phase II clinical trial of intranasal insulin in amnestic MCI subjects reported greater effects on delayed story recall in males than in females, whereas greater benefits on activities of daily living in women than in men (Claxton et al., 2013). The effects were further stratified by ApoE genotypes, with worsened cognitive performance seen in ApoE4 females but improved performance in ApoE4 males. The interaction of sex and ApoE was also seen in some clinical studies of hormone replacement therapy (HRT) with ApoE4 women benefitting the least (Burkhardt et al., 2004; Ryan et al., 2009; Yaffe et al., 2000). The clinical benefits of HRT remain controversial and require more future long-term follow up studies in early HRT treatment (Ferretti et al., 2018). Together, the importance of tailoring sex-specific diagnosis, prognosis, prevention and treatment strategies of AD patients is increasingly recognized.
6. Future perspectives for understanding sex-specific differences in AD
High-throughput and bioinformatics technologies have helped us tremendously in understanding molecular and genetic basis of sex dimorphism in aging and AD. A new study analyzed genome wide transcriptome datasets from various brain regions of AD men and women using system biology network approaches (Sun et al., 2019). They have identified pathways and genes associated with AD in males and revealed that female molecular networks were more conserved than male ones across different brain regions at various disease stages. An ongoing research effort utilizing systems biology approaches including multiscale network analysis has identified several key molecular pathways and regulators that are specific in AD females and males, further classified by ApoE genotypes (Lei et al., manuscript in preparation). Together, these findings will contribute to a deeper understanding of sex differences in individuals with AD and could help greatly in the development of personalized and effective strategies for both AD diagnosis and treatment.
Prior studies have unveiled several important aspects of sex differences in AD as we discussed in this paper. However, in the era of precision medicine, a more comprehensive understanding of molecular mechanisms underlying sex dimorphism in AD is urgently needed for better development of diagnostic markers and target-driven therapies for AD (Mielke et al., 2014; Miller et al., 2013). New research should not only focus on the biological sex, but also focus on factors that contribute to male and female disparity. It is important to understand the interaction between sex and other risk factors of AD such as genetic and environmental factors. As the field is moving toward early detection and preventive strategies, future AD research should always evaluate sex differences, particularly when designing clinical trial studies.
Highlights.
Sex differences in Alzheimer’s disease (AD) play a pivotal role in the prevalence, clinical course, responses to treatment during clinical trials, prognosis, and severity of pathological changes.
A greater understanding of sex differences in AD from a molecular to a clinical point of view may improve symptom awareness, risk factor prevention, and clinical outcomes.
Our review on recent and relevant literature about sex differences in AD highlights its impact on how development, aging, genetic, comorbidity, and environmental factors contribute to disease pathogenesis.
Acknowledgments
DC is supported by NIH R01 (1R01AG048923) and RF1 (1RF1AG054014), by Department of Veteran Affairs BLR&D (1I01BX003380) and RR&D (1I01RX002290), as well as by New York State SCI Foundation. CAT is supported by New York State SCI Foundation. LZ is supported by NIH R01 (1R01AG048923 for DC) and RF1 (1RF1AG054014 for DC). JC is supported by NIH R01 (1R01AG048923 for DC) and RF1 (1RF1AG054014 for DC), and NNSFC (81771162 for JC).
Abbreviations
- Aβ
Amyloid-β
- AD
Alzheimer’s disease
- ApoE
Apolipoprotein E
- ApoE2
Apolipoprotein E2
- ApoE
Apolipoprotein E3
- ApoE
Apolipoprotein E4
- AIF
Apoptosis-Inducing Factor
- APP
Amyloid precursor protein
- AQP4
Aquoporin-4
- AT
Aerobic Training
- ATP
Adenosine Triphosphate
- BBB
Blood Brain Barrier
- CAD
Coronary Artery Disease
- BACE1
β-site APP cleavage enzyme
- BDNF
Brain-Derived Neurotrophic Factor
- CCI
Cortical Compact Injury
- CCV
Clathrin-Coated Vesicles
- CRF1
Corticotrophin Releasing Factor 1
- CSF
Cerebral Spinal Fluid
- DHT
Dihydrotestosterone
- E2
Estrogen 17β-Estradiol
- ERα
Estrogen Receptor Alpha
- ERβ
Estrogen Receptor Beta
- HFD
High Fat Diet
- HRT
Hormone Replacement Therapy
- HT
Hormone therapy
- IGF-1
Insulin-Like Growth Factor-1
- LDLR
Low-density lipoprotein receptor
- MCI
Mild Cognitive Impairments
- NDs
Neurodegenerative diseases
- NFTs
Neurofibrillary tangles
- NSAIDs
Non-Steroidal Anti-Inflammatory Drugs
- PAR
Poly-ADP ribose
- PARP-1
Poly (ADP-ribose) polymerase-1
- PS1
Presenilin 1
- PS2
Presenilin 2
- SNPs
Single Nucleotide Polymorphisms
- T2DM
Type 2 diabetes mellitus
- T3
Triiodothyronine
- T4
Thyroxine
- TBI
Traumatic brain injury
- TRH
TSH-releasing hormone
- TSH
Thyroid-stimulating hormone
- VEGF
Vascular Endothelial Growth Factor
Footnotes
Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.
References
- Acaz-Fonseca E, et al. , 2015. Sex differences in glia reactivity after cortical brain injury. Glia 63, 1966–1981. [DOI] [PubMed] [Google Scholar]
- Ahonen T, et al. , 2012. Sex differences in the association of adiponectin and low-grade inflammation with changes in the body mass index from youth to middle age. Gend Med 9, 1–8. [DOI] [PubMed] [Google Scholar]
- Allen JS, Damasio H, Grabowski TJ, 2002. Normal neuroanatomical variation in the human brain: an MRI-volumetric study. Am J Phys Anthropol 118, 341–58. [DOI] [PubMed] [Google Scholar]
- Annerbo S, Wahlund LO, Lokk J, 2006. The significance of thyroid-stimulating hormone and homocysteine in the development of Alzheimer’s disease in mild cognitive impairment: a 6-year follow-up study. Am J Alzheimers Dis Other Demen 21, 182–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Appelros P, Stegmayr B, Terent A, 2009. Sex differences in stroke epidemiology: a systematic review. Stroke 40, 1082–90. [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, 847–57. [DOI] [PubMed] [Google Scholar]
- Asthana S, 2003. Estrogen and cognition: the story so far. J Gerontol A Biol Sci Med Sci 58, 322–3. [DOI] [PubMed] [Google Scholar]
- Aukes AM, et al. , 2007. Pregnancy prevents hypertensive remodeling and decreases myogenic reactivity in posterior cerebral arteries from Dahl salt-sensitive rats: a role in eclampsia? Am J Physiol Heart Circ Physiol 292, H1071–6. [DOI] [PubMed] [Google Scholar]
- Autry AE, et al. , 2009. Gender-specific impact of brain-derived neurotrophic factor signaling on stress-induced depression-like behavior. Biol Psychiatry 66, 84–90. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bairey Merz CN, et al. , 2006. Insights from the NHLBI-Sponsored Women’s Ischemia Syndrome Evaluation (WISE) Study: Part II: gender differences in presentation, diagnosis, and outcome with regard to gender-based pathophysiology of atherosclerosis and macrovascular and microvascular coronary disease. J Am Coll Cardiol 47, S21–9. [DOI] [PubMed] [Google Scholar]
- Bake S, et al. , 2014. Blood brain barrier and neuroinflammation are critical targets of IGF-1-mediated neuroprotection in stroke for middle-aged female rats. PLoS One 9, e91427. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Baker LD, et al. , 2010. Effects of aerobic exercise on mild cognitive impairment: a controlled trial. Arch Neurol 67, 71–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Barha CK, et al. , 2017a. Sex differences in exercise efficacy to improve cognition: A systematic review and meta-analysis of randomized controlled trials in older humans. Front Neuroendocrinol 46, 71–85. [DOI] [PubMed] [Google Scholar]
- Barha CK, et al. , 2017b. Sex differences in aerobic exercise efficacy to improve cognition: A systematic review and meta-analysis of studies in older rodents. Front Neuroendocrinol 46, 86–105. [DOI] [PubMed] [Google Scholar]
- Barnes DE, Yaffe K, 2011. The projected effect of risk factor reduction on Alzheimer’s disease prevalence. Lancet Neurol 10, 819–28. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Barnes LL, et al. , 2005. Sex differences in the clinical manifestations of Alzheimer disease pathology. Arch Gen Psychiatry 62, 685–91. [DOI] [PubMed] [Google Scholar]
- Barron AM, et al. , 2013. Sex-specific effects of high fat diet on indices of metabolic syndrome in 3xTg-AD mice: implications for Alzheimer’s disease. PLoS One 8, e78554. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Baudry M, Bi X, Aguirre C, 2013. Progesterone-estrogen interactions in synaptic plasticity and neuroprotection. Neuroscience 239, 280–94. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Beckman KB, Ames BN, 1998. The free radical theory of aging matures. Physiol Rev 78, 547–81. [DOI] [PubMed] [Google Scholar]
- Behl C, et al. , 1995. 17-beta estradiol protects neurons from oxidative stress-induced cell death in vitro. Biochem Biophys Res Commun 216, 473–82. [DOI] [PubMed] [Google Scholar]
- Belandia B, et al. , 1998. Thyroid hormone negatively regulates the transcriptional activity of the beta-amyloid precursor protein gene. J Biol Chem 273, 30366–71. [DOI] [PubMed] [Google Scholar]
- Bell RD, Zlokovic BV, 2009. Neurovascular mechanisms and blood-brain barrier disorder in Alzheimer’s disease. Acta Neuropathol 118, 103–13. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Benedict C, et al. , 2008. Differential sensitivity of men and women to anorexigenic and memory-improving effects of intranasal insulin. J Clin Endocrinol Metab 93, 1339–44. [DOI] [PubMed] [Google Scholar]
- Berchtold NC, et al. , 2008. Gene expression changes in the course of normal brain aging are sexually dimorphic. Proc Natl Acad Sci U S A 105, 15605–10. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Berger JS, et al. , 2006. Aspirin for the primary prevention of cardiovascular events in women and men: a sex-specific meta-analysis of randomized controlled trials. JAMA 295, 306–13. [DOI] [PubMed] [Google Scholar]
- Berry C, et al. , 2009. The effect of gender on patients with moderate to severe head injuries. J Trauma 67, 950–3. [DOI] [PubMed] [Google Scholar]
- Bertram L, et al. , 2008. Genome-wide association analysis reveals putative Alzheimer’s disease susceptibility loci in addition to APOE. Am J Hum Genet 83, 623–32. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Beydoun MA, et al. , 2012. Sex differences in the association of the apolipoprotein E epsilon 4 allele with incidence of dementia, cognitive impairment, and decline. Neurobiol Aging 33, 720–731 e4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bianchi G, et al. , 1999. Oxidative stress and anti-oxidant metabolites in patients with hyperthyroidism: effect of treatment. Horm Metab Res 31, 620–4. [DOI] [PubMed] [Google Scholar]
- Bixler EO, et al. , 2001. Prevalence of sleep-disordered breathing in women: effects of gender. Am J Respir Crit Care Med 163, 608–13. [DOI] [PubMed] [Google Scholar]
- Bixler EO, et al. , 2002. Insomnia in central Pennsylvania. J Psychosom Res 53, 589–92. [DOI] [PubMed] [Google Scholar]
- Bohlega SA, Al-Foghom NB, 2013. Drug-induced Parkinson`s disease. A clinical review. Neurosciences (Riyadh) 18, 215–21. [PubMed] [Google Scholar]
- Borras C, et al. , 2003. Mitochondria from females exhibit higher antioxidant gene expression and lower oxidative damage than males. Free Radic Biol Med 34, 546–52. [DOI] [PubMed] [Google Scholar]
- Bour A, et al. , 2008. Middle-aged human apoE4 targeted-replacement mice show retention deficits on a wide range of spatial memory tasks. Behav Brain Res 193, 174–82. [DOI] [PubMed] [Google Scholar]
- Breunig JJ, Guillot-Sestier MV, Town T, 2013. Brain injury, neuroinflammation and Alzheimer’s disease. Front Aging Neurosci 5, 26. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bromberger JT, et al. , 2011. Major depression during and after the menopausal transition: Study of Women’s Health Across the Nation (SWAN). Psychol Med 41, 1879–88. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Brothers HM, Gosztyla ML, Robinson SR, 2018. The Physiological Roles of Amyloid-beta Peptide Hint at New Ways to Treat Alzheimer’s Disease. Front Aging Neurosci 10, 118. [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, 333–44. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Burger HG, et al. , 2002. Hormonal changes in the menopause transition. Recent Prog Horm Res 57, 257–75. [DOI] [PubMed] [Google Scholar]
- Burkhardt MS, et al. , 2004. Oestrogen replacement therapy may improve memory functioning in the absence of APOE epsilon4. J Alzheimers Dis 6, 221–8. [DOI] [PubMed] [Google Scholar]
- Canevelli M, et al. , 2017. Sex and gender differences in the treatment of Alzheimer’s disease: A systematic review of randomized controlled trials. Pharmacol Res 115, 218–223. [DOI] [PubMed] [Google Scholar]
- Cao D, et al. , 2007. Intake of sucrose-sweetened water induces insulin resistance and exacerbates memory deficits and amyloidosis in a transgenic mouse model of Alzheimer disease. J Biol Chem 282, 36275–82. [DOI] [PubMed] [Google Scholar]
- Cao J, et al. , 2017. ApoE4-associated phospholipid dysregulation contributes to development of Tau hyper-phosphorylation after traumatic brain injury. Sci Rep 7, 11372. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Caplan HW, Cox CS, Bedi SS, 2017. Do microglia play a role in sex differences in TBI? J Neurosci Res 95, 509–517. [DOI] [PubMed] [Google Scholar]
- Carrier J, et al. , 2001. The effects of age and gender on sleep EEG power spectral density in the middle years of life (ages 20–60 years old). Psychophysiology 38, 232–42. [PubMed] [Google Scholar]
- Cedernaes J, et al. , 2017. Candidate mechanisms underlying the association between sleep-wake disruptions and Alzheimer’s disease. Sleep Med Rev 31, 102–111. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Cha DS, et al. , 2014. Major depressive disorder and type II diabetes mellitus: mechanisms underlying risk for Alzheimer’s disease. CNS Neurol Disord Drug Targets 13, 1740–9. [DOI] [PubMed] [Google Scholar]
- Chan CB, Ye K, 2017. Sex differences in brain-derived neurotrophic factor signaling and functions. J Neurosci Res 95, 328–335. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Chen J, et al. , 2014a. 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, 724–33. [DOI] [PubMed] [Google Scholar]
- Chen Y, et al. , 2014b. Deregulation of brain insulin signaling in Alzheimer’s disease. Neurosci Bull 30, 282–94. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Cheng EY, Kong MH, 2016. Gender Differences of Thromboembolic Events in Atrial Fibrillation. Am J Cardiol 117, 1021–7. [DOI] [PubMed] [Google Scholar]
- Chung J, et al. , 2008. HSP72 protects against obesity-induced insulin resistance. Proc Natl Acad Sci U S A 105, 1739–44. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Claxton A, et al. , 2013. Sex and ApoE genotype differences in treatment response to two doses of intranasal insulin in adults with mild cognitive impairment or Alzheimer’s disease. J Alzheimers Dis 35, 789–97. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Colantonio A, et al. , 2010. Gender differences in self reported long term outcomes following moderate to severe traumatic brain injury. BMC Neurol 10, 102. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Colcombe S, Kramer AF, 2003. Fitness effects on the cognitive function of older adults: a meta-analytic study. Psychol Sci 14, 125–30. [DOI] [PubMed] [Google Scholar]
- Connor B, et al. , 1997. Brain-derived neurotrophic factor is reduced in Alzheimer’s disease. Brain Res Mol Brain Res 49, 71–81. [DOI] [PubMed] [Google Scholar]
- Corder EH, et al. , 2004. The biphasic relationship between regional brain senile plaque and neurofibrillary tangle distributions: modification by age, sex, and APOE polymorphism. Ann N Y Acad Sci 1019, 24–8. [DOI] [PubMed] [Google Scholar]
- Cordonnier C, et al. , 2017. Stroke in women - from evidence to inequalities. Nat Rev Neurol 13, 521–532. [DOI] [PubMed] [Google Scholar]
- Cosgrove KP, Mazure CM, Staley JK, 2007. Evolving knowledge of sex differences in brain structure, function, and chemistry. Biol Psychiatry 62, 847–55. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Cotman CW, Berchtold NC, Christie LA, 2007. Exercise builds brain health: key roles of growth factor cascades and inflammation. Trends Neurosci 30, 464–72. [DOI] [PubMed] [Google Scholar]
- Cozzi A, et al. , 2006. Poly(ADP-ribose) accumulation and enhancement of postischemic brain damage in 110-kDa poly(ADP-ribose) glycohydrolase null mice. J Cereb Blood Flow Metab 26, 684–95. [DOI] [PubMed] [Google Scholar]
- Craft S, 2006. Insulin resistance syndrome and Alzheimer disease: pathophysiologic mechanisms and therapeutic implications. Alzheimer Dis Assoc Disord 20, 298–301. [DOI] [PubMed] [Google Scholar]
- Csernansky JG, et al. , 2006. Plasma cortisol and progression of dementia in subjects with Alzheimer-type dementia. Am J Psychiatry 163, 2164–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Currais A, et al. , 2012. Diabetes exacerbates amyloid and neurovascular pathology in aging-accelerated mice. Aging Cell 11, 1017–26. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Dal Forno G, et al. , 2005. Depressive symptoms, sex, and risk for Alzheimer’s disease. Ann Neurol 57, 381–7. [DOI] [PubMed] [Google Scholar]
- de Jong FJ, et al. , 2009. Thyroid function, the risk of dementia and neuropathologic changes: the Honolulu-Asia aging study. Neurobiol Aging 30, 600–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- De Smedt A, et al. , 2011. Insulin-like growth factor I serum levels influence ischemic stroke outcome. Stroke 42, 2180–5. [DOI] [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, 789–819. [DOI] [PubMed] [Google Scholar]
- DeKosky ST, Ikonomovic MD, Gandy S, 2010. Traumatic brain injury--football, warfare, and longterm effects. N Engl J Med 363, 1293–6. [DOI] [PubMed] [Google Scholar]
- DeMayo FJ, et al. , 2002. Mechanisms of action of estrogen and progesterone. Ann N Y Acad Sci 955, 48–59; discussion 86–8, 396–406. [DOI] [PubMed] [Google Scholar]
- den Heijer T, et al. , 2006. Use of hippocampal and amygdalar volumes on magnetic resonance imaging to predict dementia in cognitively intact elderly people. Arch Gen Psychiatry 63, 57–62. [DOI] [PubMed] [Google Scholar]
- Devi L, et al. , 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]
- Devi L, et al. , 2012. Mechanisms underlying insulin deficiency-induced acceleration of beta-amyloidosis in a mouse model of Alzheimer’s disease. PLoS One 7, e32792. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Doi Y, et al. , 2013. Fingolimod phosphate attenuates oligomeric amyloid beta-induced neurotoxicity via increased brain-derived neurotrophic factor expression in neurons. PLoS One 8, e61988. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Dong X, et al. , 2014. The relationship between serum insulin-like growth factor I levels and ischemic stroke risk. PLoS One 9, e94845. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
- Doody RS, et al. , 2014. Phase 3 trials of solanezumab for mild-to-moderate Alzheimer’s disease. N Engl J Med 370, 311–21. [DOI] [PubMed] [Google Scholar]
- Doraiswamy PM, et al. , 1997a. Memory, language, and praxis in Alzheimer’s disease: norms for outpatient clinical trial populations. Psychopharmacol Bull 33, 123–8. [PubMed] [Google Scholar]
- Doraiswamy PM, et al. , 1997b. The Alzheimer’s Disease Assessment Scale: patterns and predictors of baseline cognitive performance in multicenter Alzheimer’s disease trials. Neurology 48, 1511–7. [DOI] [PubMed] [Google Scholar]
- Dubal DB, Broestl L, Worden K, 2012. Sex and gonadal hormones in mouse models of Alzheimer’s disease: what is relevant to the human condition? Biol Sex Differ 3, 24. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Dzamba D, et al. , 2016. Glial Cells - The Key Elements of Alzheimer s Disease. Curr Alzheimer Res 13, 894–911. [DOI] [PubMed] [Google Scholar]
- Egan MF, et al. , 2018. Randomized Trial of Verubecestat for Mild-to-Moderate Alzheimer’s Disease. N Engl J Med 378, 1691–1703. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ekblad LL, et al. , 2015. Insulin resistance is associated with poorer verbal fluency performance in women. Diabetologia 58, 2545–53. [DOI] [PubMed] [Google Scholar]
- Elias MF, et al. , 2003. Lower cognitive function in the presence of obesity and hypertension: the Framingham heart study. Int J Obes Relat Metab Disord 27, 260–8. [DOI] [PubMed] [Google Scholar]
- Epstein EL, et al. , 2019. Posttraumatic stress disorder and traumatic brain Injury: Sex differences in veterans. Psychiatry Res 274, 105–111. [DOI] [PubMed] [Google Scholar]
- Estrany ME, et al. , 2013. High-fat diet feeding induces sex-dependent changes in inflammatory and insulin sensitivity profiles of rat adipose tissue. Cell Biochem Funct 31, 504–10. [DOI] [PubMed] [Google Scholar]
- Fairweather D, Frisancho-Kiss S, Rose NR, 2008. Sex differences in autoimmune disease from a pathological perspective. Am J Pathol 173, 600–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Farace E, Alves WM, 2000. Do women fare worse? A metaanalysis of gender differences in outcome after traumatic brain injury. Neurosurg Focus 8, e6. [DOI] [PubMed] [Google Scholar]
- Farrer LA, et al. , 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, 1349–56. [PubMed] [Google Scholar]
- Feart C, et al. , 2009. Adherence to a Mediterranean diet, cognitive decline, and risk of dementia. JAMA 302, 638–48. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ferretti MT, et al. , 2018. Sex differences in Alzheimer disease - the gateway to precision medicine. Nat Rev Neurol 14, 457–469. [DOI] [PubMed] [Google Scholar]
- Filon JR, et al. , 2016. Gender Differences in Alzheimer Disease: Brain Atrophy, Histopathology Burden, and Cognition. J Neuropathol Exp Neurol [DOI] [PMC free article] [PubMed]
- Fisher DW, Bennett DA, Dong H, 2018. Sexual dimorphism in predisposition to Alzheimer’s disease. Neurobiol Aging 70, 308–324. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Francis BM, et al. , 2012. Object recognition memory and BDNF expression are reduced in young TgCRND8 mice. Neurobiol Aging 33, 555–63. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Franconi F, et al. , 2008. Are the available experimental models of type 2 diabetes appropriate for a gender perspective? Pharmacol Res 57, 6–18. [DOI] [PubMed] [Google Scholar]
- Frank SA, Hurst LD, 1996. Mitochondria and male disease. Nature 383, 224. [DOI] [PubMed] [Google Scholar]
- Fukumoto N, et al. , 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, 235–42. [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 38, 527–33. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Geerlings MI, et al. , 2001. Reproductive period and risk of dementia in postmenopausal women. JAMA 285, 1475–81. [DOI] [PubMed] [Google Scholar]
- Ghosh S, Thakur MK, 2008a. PS1 expression is downregulated by gonadal steroids in adult mouse brain. Neurochem Res 33, 365–9. [DOI] [PubMed] [Google Scholar]
- Ghosh S, Thakur MK, 2008b. PS2 protein expression is upregulated by sex steroids in the cerebral cortex of aging mice. Neurochem Int 52, 363–7. [DOI] [PubMed] [Google Scholar]
- Gibson CL, 2013. Cerebral ischemic stroke: is gender important? J Cereb Blood Flow Metab 33, 1355–61. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Giedd JN, et al. , 1999. Brain development during childhood and adolescence: a longitudinal MRI study. Nat Neurosci 2, 861–3. [DOI] [PubMed] [Google Scholar]
- Gilsanz P, et al. , 2017. Female sex, early-onset hypertension, and risk of dementia. Neurology 89, 1886–1893. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gitler AD, Tsuiji H, 2016. There has been an awakening: Emerging mechanisms of C9orf72 mutations in FTD/ALS. Brain Res 1647, 19–29. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Glass CK, et al. , 2010. Mechanisms underlying inflammation in neurodegeneration. Cell 140, 918–34. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Goel N, Kim H, Lao RP, 2005. Gender differences in polysomnographic sleep in young healthy sleepers. Chronobiol Int 22, 905–15. [DOI] [PubMed] [Google Scholar]
- Gordon JL, Girdler SS, 2014. Hormone replacement therapy in the treatment of perimenopausal depression. Curr Psychiatry Rep 16, 517. [DOI] [PubMed] [Google Scholar]
- Goveas JS, et al. , 2011. Depressive symptoms and incidence of mild cognitive impairment and probable dementia in elderly women: the Women’s Health Initiative Memory Study. J Am Geriatr Soc 59, 57–66. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Grant WB, 1999. Dietary links to Alzheimer’s disease: 1999 update. J Alzheimers Dis 1, 197–201. [DOI] [PubMed] [Google Scholar]
- Greendale GA, et al. , 2009. Effects of the menopause transition and hormone use on cognitive performance in midlife women. Neurology 72, 1850–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Greenfield JP, et al. , 2002. Estrogen lowers Alzheimer beta-amyloid generation by stimulating trans-Golgi network vesicle biogenesis. J Biol Chem 277, 12128–36. [DOI] [PubMed] [Google Scholar]
- Guebel DV, Torres NV, 2016. Sexual Dimorphism and Aging in the Human Hyppocampus: Identification, Validation, and Impact of Differentially Expressed Genes by Factorial Microarray and Network Analysis. Front Aging Neurosci 8, 229. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gur RC, et al. , 1995. Sex differences in regional cerebral glucose metabolism during a resting state. Science 267, 528–31. [DOI] [PubMed] [Google Scholar]
- Gustafson DR, et al. , 2007. Mid-life adiposity factors relate to blood-brain barrier integrity in late life. J Intern Med 262, 643–50. [DOI] [PubMed] [Google Scholar]
- Hanson AJ, Craft S, Banks WA, 2015. The APOE genotype: modification of therapeutic responses in Alzheimer’s disease. Curr Pharm Des 21, 114–20. [DOI] [PubMed] [Google Scholar]
- Hayes JP, et al. , 2017. Mild traumatic brain injury is associated with reduced cortical thickness in those at risk for Alzheimer’s disease. Brain 140, 813–825. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Henderson VW, et al. , 1994. Estrogen replacement therapy in older women. Comparisons between Alzheimer’s disease cases and nondemented control subjects. Arch Neurol 51, 896–900. [DOI] [PubMed] [Google Scholar]
- Henderson VW, et al. , 2005. Postmenopausal hormone therapy and Alzheimer’s disease risk: interaction with age. J Neurol Neurosurg Psychiatry 76, 103–5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Henley DB, et al. , 2009. Development of semagacestat (LY450139), a functional gamma-secretase inhibitor, for the treatment of Alzheimer’s disease. Expert Opin Pharmacother 10, 1657–64. [DOI] [PubMed] [Google Scholar]
- Hernandez AI, et al. , 2009. Poly-(ADP-ribose) polymerase-1 is necessary for long-term facilitation in Aplysia. J Neurosci 29, 9553–62. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Herting MM, et al. , 2012. The impact of sex, puberty, and hormones on white matter microstructure in adolescents. Cereb Cortex 22, 1979–92. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Heun R, 2002. Does gender play a role in Alzheimer therapy? Expert Rev Neurother 2, 589–91. [DOI] [PubMed] [Google Scholar]
- Ho L, et al. , 2004. Diet-induced insulin resistance promotes amyloidosis in a transgenic mouse model of Alzheimer’s disease. FASEB J 18, 902–4. [DOI] [PubMed] [Google Scholar]
- Hock C, et al. , 2000. Region-specific neurotrophin imbalances in Alzheimer disease: decreased levels of brain-derived neurotrophic factor and increased levels of nerve growth factor in hippocampus and cortical areas. Arch Neurol 57, 846–51. [DOI] [PubMed] [Google Scholar]
- Hod T, Cerdeira AS, Karumanchi SA, 2015. Molecular Mechanisms of Preeclampsia. Cold Spring Harb Perspect Med 5. [DOI] [PMC free article] [PubMed]
- Hou X, et al. , 2015. Differential contributions of ApoE4 and female sex to BACE1 activity and expression mediate Abeta deposition and learning and memory in mouse models of Alzheimer’s disease. Front Aging Neurosci 7, 207. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hsieh TC, et al. , 2012. Sex- and age-related differences in brain FDG metabolism of healthy adults: an SPM analysis. J Neuroimaging 22, 21–7. [DOI] [PubMed] [Google Scholar]
- Hua X, et al. , 2010. Sex and age differences in atrophic rates: an ADNI study with n=1368 MRI scans. Neurobiol Aging 31, 1463–80. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hudson SP, Jacobson-Pick S, Anisman H, 2014. Sex differences in behavior and pro-inflammatory cytokine mRNA expression following stressor exposure and re-exposure. Neuroscience 277, 239–49. [DOI] [PubMed] [Google Scholar]
- Hutton M, Hardy J, 1997. The presenilins and Alzheimer’s disease. Hum Mol Genet 6, 1639–46. [DOI] [PubMed] [Google Scholar]
- Hwang LL, et al. , 2010. Sex differences in high-fat diet-induced obesity, metabolic alterations and learning, and synaptic plasticity deficits in mice. Obesity (Silver Spring) 18, 463–9. [DOI] [PubMed] [Google Scholar]
- Ide T, et al. , 2002. Greater oxidative stress in healthy young men compared with premenopausal women. Arterioscler Thromb Vasc Biol 22, 438–42. [DOI] [PubMed] [Google Scholar]
- in t’ Veld BA, et al. , 2001. Nonsteroidal antiinflammatory drugs and the risk of Alzheimer’s disease. N Engl J Med 345, 1515–21. [DOI] [PubMed] [Google Scholar]
- Ingalhalikar M, et al. , 2014. Sex differences in the structural connectome of the human brain. Proc Natl Acad Sci U S A 111, 823–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Irvine K, et al. , 2012. Greater cognitive deterioration in women than men with Alzheimer’s disease: a meta analysis. J Clin Exp Neuropsychol 34, 989–98. [DOI] [PubMed] [Google Scholar]
- Jack CR Jr., et al. , 2013. Tracking pathophysiological processes in Alzheimer’s disease: an updated hypothetical model of dynamic biomarkers. Lancet Neurol 12, 207–16. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Jack CR Jr., et al. , 2015. Age, Sex, and APOE epsilon4 Effects on Memory, Brain Structure, and beta-Amyloid Across the Adult Life Span. JAMA Neurol 72, 511–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Jack CR Jr., et al. , 2017. Age-specific and sex-specific prevalence of cerebral beta-amyloidosis, tauopathy, and neurodegeneration in cognitively unimpaired individuals aged 50–95 years: a cross-sectional study. Lancet Neurol 16, 435–444. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Jacobs EG, et al. , 2016. Impact of Sex and Menopausal Status on Episodic Memory Circuitry in Early Midlife. J Neurosci 36, 10163–73. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Jacobs EG, et al. , 2017. Reorganization of Functional Networks in Verbal Working Memory Circuitry in Early Midlife: The Impact of Sex and Menopausal Status. Cereb Cortex 27, 2857–2870. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Jacobs EG, Goldstein JM, 2018. The Middle-Aged Brain: Biological sex and sex hormones shape memory circuitry. Curr Opin Behav Sci 23, 84–91. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Jansen WJ, et al. , 2015. Prevalence of cerebral amyloid pathology in persons without dementia: a meta-analysis. JAMA 313, 1924–38. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Jin R, et al. , 2013. Role of inflammation and its mediators in acute ischemic stroke. J Cardiovasc Transl Res 6, 834–51. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Johnson KA, et al. , 2016. Tau positron emission tomographic imaging in aging and early Alzheimer disease. Ann Neurol 79, 110–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ju YE, Lucey BP, Holtzman DM, 2014. Sleep and Alzheimer disease pathology--a bidirectional relationship. Nat Rev Neurol 10, 115–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kalmijn S, et al. , 2000. Subclinical hyperthyroidism and the risk of dementia. The Rotterdam study. Clin Endocrinol (Oxf) 53, 733–7. [DOI] [PubMed] [Google Scholar]
- Kanaan RA, et al. , 2012. Gender differences in white matter microstructure. PLoS One 7, e38272. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Karttunen K, et al. , 2011. Neuropsychiatric symptoms and quality of life in patients with very mild and mild Alzheimer’s disease. Int J Geriatr Psychiatry 26, 473–82. [DOI] [PubMed] [Google Scholar]
- Kautzky-Willer A, Harreiter J, Pacini G, 2016. Sex and Gender Differences in Risk, Pathophysiology and Complications of Type 2 Diabetes Mellitus. Endocr Rev 37, 278–316. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kemppainen S, et al. , 2012. Impaired TrkB receptor signaling contributes to memory impairment in APP/PS1 mice. Neurobiol Aging 33, 1122 e23–39. [DOI] [PubMed] [Google Scholar]
- Khera A, et al. , 2009. Sex differences in the relationship between C-reactive protein and body fat. J Clin Endocrinol Metab 94, 3251–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kim J, et al. , 2009. Overexpression of low-density lipoprotein receptor in the brain markedly inhibits amyloid deposition and increases extracellular A beta clearance. Neuron 64, 632–44. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kim MY, et al. , 2018. Sex Differences in Cardiovascular Risk Factors for Dementia. Biomol Ther (Seoul) 26, 521–532. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kim S, et al. , 2015. Gender differences in risk factors for transition from mild cognitive impairment to Alzheimer’s disease: A CREDOS study. Compr Psychiatry 62, 114–22. [DOI] [PubMed] [Google Scholar]
- Kitamura T, et al. , 2012. Gender differences in clinical manifestations and outcomes among hospitalized patients with behavioral and psychological symptoms of dementia. J Clin Psychiatry 73, 1548–54. [DOI] [PubMed] [Google Scholar]
- Kivipelto M, et al. , 2001a. Midlife vascular risk factors and late-life mild cognitive impairment: A population-based study. Neurology 56, 1683–9. [DOI] [PubMed] [Google Scholar]
- Kivipelto M, et al. , 2001b. Midlife vascular risk factors and Alzheimer’s disease in later life: longitudinal, population based study. BMJ 322, 1447–51. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Klein SL, Marriott I, Fish EN, 2015. Sex-based differences in immune function and responses to vaccination. Trans R Soc Trop Med Hyg 109, 9–15. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kleinberger G, et al. , 2014. TREM2 mutations implicated in neurodegeneration impair cell surface transport and phagocytosis. Sci Transl Med 6, 243ra86. [DOI] [PubMed] [Google Scholar]
- Koran MEI, et al. , 2017. Sex differences in the association between AD biomarkers and cognitive decline. Brain Imaging Behav 11, 205–213. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kovacic JC, et al. , 2011a. Cellular senescence, vascular disease, and aging: Part 1 of a 2-part review. Circulation 123, 1650–60. [DOI] [PubMed] [Google Scholar]
- Kovacic JC, et al. , 2011b. Cellular senescence, vascular disease, and aging: part 2 of a 2-part review: clinical vascular disease in the elderly. Circulation 123, 1900–10. [DOI] [PubMed] [Google Scholar]
- Kress BT, et al. , 2014. Impairment of paravascular clearance pathways in the aging brain. Ann Neurol 76, 845–61. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kumar DK, et al. , 2016. Amyloid-beta peptide protects against microbial infection in mouse and worm models of Alzheimer’s disease. Sci Transl Med 8, 340ra72. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Labonte B, et al. , 2017. Sex-specific transcriptional signatures in human depression. Nat Med 23, 1102–1111. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lambert JC, et al. , 2009. Genome-wide association study identifies variants at CLU and CR1 associated with Alzheimer’s disease. Nat Genet 41, 1094–9. [DOI] [PubMed] [Google Scholar]
- Lang JT, McCullough LD, 2008. Pathways to ischemic neuronal cell death: are sex differences relevant? J Transl Med 6, 33. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Latasa MJ, Belandia B, Pascual A, 1998. Thyroid hormones regulate beta-amyloid gene splicing and protein secretion in neuroblastoma cells. Endocrinology 139, 2692–8. [DOI] [PubMed] [Google Scholar]
- Laurin D, et al. , 2001. Physical activity and risk of cognitive impairment and dementia in elderly persons. Arch Neurol 58, 498–504. [DOI] [PubMed] [Google Scholar]
- Laws KR, Irvine K, Gale TM, 2018. Sex differences in Alzheimer’s disease. Curr Opin Psychiatry 31, 133–139. [DOI] [PubMed] [Google Scholar]
- Lei G, et al. , Gender specific molecular networks and drivers of Alzheimer’s Disease. Manuscript in preparation
- Li G, et al. , 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 GD, et al. , 2017b. Female-specific effect of the BDNF gene on Alzheimer’s disease. Neurobiol Aging 53, 192 e11–192 e19. [DOI] [PubMed] [Google Scholar]
- Li J, McCullough LD, 2009. Sex differences in minocycline-induced neuroprotection after experimental stroke. J Cereb Blood Flow Metab 29, 670–4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Li R, Singh M, 2014. Sex differences in cognitive impairment and Alzheimer’s disease. Front Neuroendocrinol 35, 385–403. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Li S, et al. , 2010. An age-related sprouting transcriptome provides molecular control of axonal sprouting after stroke. Nat Neurosci 13, 1496–504. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lim SS, et al. , 2012. A comparative risk assessment of burden of disease and injury attributable to 67 risk factors and risk factor clusters in 21 regions, 1990–2010: a systematic analysis for the Global Burden of Disease Study 2010. Lancet 380, 2224–60. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lin KA, et al. , 2015. Marked gender differences in progression of mild cognitive impairment over 8 years. Alzheimers Dement (N Y) 1, 103–110. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Liu F, et al. , 2011. Sex differences in the response to poly(ADP-ribose) polymerase-1 deletion and caspase inhibition after stroke. Stroke 42, 1090–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Liu HP, et al. , 2010. Evaluation of the poly(ADP-ribose) polymerase-1 gene variants in Alzheimer’s disease. J Clin Lab Anal 24, 182–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Luine VN, 1985. Estradiol increases choline acetyltransferase activity in specific basal forebrain nuclei and projection areas of female rats. Exp Neurol 89, 484–90. [DOI] [PubMed] [Google Scholar]
- Lundgaard I, et al. , 2017. Glymphatic clearance controls state-dependent changes in brain lactate concentration. J Cereb Blood Flow Metab 37, 2112–2124. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Luo L, et al. , 2002. Thyrotropin releasing hormone (TRH) in the hippocampus of Alzheimer patients. J Alzheimers Dis 4, 97–103. [DOI] [PubMed] [Google Scholar]
- Maass A, et al. , 2016. Relationships of peripheral IGF-1, VEGF and BDNF levels to exercise-related changes in memory, hippocampal perfusion and volumes in older adults. Neuroimage 131, 142–54. [DOI] [PubMed] [Google Scholar]
- Mahley RW, Huang Y, 2012. Apolipoprotein e sets the stage: response to injury triggers neuropathology. Neuron 76, 871–85. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Maklakov AA, Bonduriansky R, Brooks RC, 2009. Sex differences, sexual selection, and ageing: an experimental evolution approach. Evolution 63, 2491–503. [DOI] [PubMed] [Google Scholar]
- Maklakov AA, Lummaa V, 2013. Evolution of sex differences in lifespan and aging: causes and constraints. Bioessays 35, 717–24. [DOI] [PubMed] [Google Scholar]
- Malpetti M, et al. , 2017. Gender differences in healthy aging and Alzheimer’s Dementia: A (18) F-FDG-PET study of brain and cognitive reserve. Hum Brain Mapp 38, 4212–4227. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Manson JE, et al. , 2017. Menopausal Hormone Therapy and Long-term All-Cause and Cause-Specific Mortality: The Women’s Health Initiative Randomized Trials. JAMA 318, 927–938. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Martire S, et al. , 2013. PARP-1 modulates amyloid beta peptide-induced neuronal damage. PLoS One 8, e72169. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Matarrese P, et al. , 2011. Gender disparity in susceptibility to oxidative stress and apoptosis induced by autoantibodies specific to RLIP76 in vascular cells. Antioxid Redox Signal 15, 2825–36. [DOI] [PubMed] [Google Scholar]
- Mattsson N, et al. , 2017. Clinical validity of cerebrospinal fluid Abeta42, tau, and phospho-tau as biomarkers for Alzheimer’s disease in the context of a structured 5-phase development framework. Neurobiol Aging 52, 196–213. [DOI] [PubMed] [Google Scholar]
- Matyi J, et al. , 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, 1607–1613. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Mazzucchelli C, et al. , 2002. Knockout of ERK1 MAP kinase enhances synaptic plasticity in the striatum and facilitates striatal-mediated learning and memory. Neuron 34, 807–20. [DOI] [PubMed] [Google Scholar]
- McDermott KL, et al. , 2017. Memory Resilience to Alzheimer’s Genetic Risk: Sex Effects in Predictor Profiles. J Gerontol B Psychol Sci Soc Sci 72, 937–946. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Mega MS, et al. , 1996. The spectrum of behavioral changes in Alzheimer’s disease. Neurology 46, 130–5. [DOI] [PubMed] [Google Scholar]
- Mehla J, Chauhan BC, Chauhan NB, 2014. Experimental induction of type 2 diabetes in aging-accelerated mice triggered Alzheimer-like pathology and memory deficits. J Alzheimers Dis 39, 145–62. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Mehta N, et al. , 2017. Systematic Review of Sex-Specific Reporting of Data: Cholinesterase Inhibitor Example. J Am Geriatr Soc 65, 2213–2219. [DOI] [PubMed] [Google Scholar]
- Mendelsohn ME, Karas RH, 2005. Molecular and cellular basis of cardiovascular gender differences. Science 308, 1583–7. [DOI] [PubMed] [Google Scholar]
- Meyer MR, et al. , 2011. Obesity, insulin resistance and diabetes: sex differences and role of oestrogen receptors. Acta Physiol (Oxf) 203, 259–69. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Middleton L, Kirkland S, Rockwood K, 2008. Prevention of CIND by physical activity: different impact on VCI-ND compared with MCI. J Neurol Sci 269, 80–4. [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, 37–48. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Mielke MM, et al. , 2016. Impaired Cognition and Brain Atrophy Decades After Hypertensive Pregnancy Disorders. Circ Cardiovasc Qual Outcomes 9, S70–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Miller JA, et al. , 2013. Genes and pathways underlying regional and cell type changes in Alzheimer’s disease. Genome Med 5, 48. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Moreira PI, 2012. Alzheimer’s disease and diabetes: an integrative view of the role of mitochondria, oxidative stress, and insulin. J Alzheimers Dis 30 Suppl 2, S199–215. [DOI] [PubMed] [Google Scholar]
- Morris MC, et al. , 2015a. MIND diet slows cognitive decline with aging. Alzheimers Dement 11, 1015–22. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Morris MC, et al. , 2015b. MIND diet associated with reduced incidence of Alzheimer’s disease. Alzheimers Dement 11, 1007–14. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Moser VA, Pike CJ, 2016. Obesity and sex interact in the regulation of Alzheimer’s disease. Neurosci Biobehav Rev 67, 102–18. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Moxley G, et al. , 2002. Sexual dimorphism in innate immunity. Arthritis Rheum 46, 250–8. [DOI] [PubMed] [Google Scholar]
- Muda M, et al. , 1996. The dual specificity phosphatases M3/6 and MKP-3 are highly selective for inactivation of distinct mitogen-activated protein kinases. J Biol Chem 271, 27205–8. [DOI] [PubMed] [Google Scholar]
- Murphy DG, et al. , 1996. Sex differences in human brain morphometry and metabolism: an in vivo quantitative magnetic resonance imaging and positron emission tomography study on the effect of aging. Arch Gen Psychiatry 53, 585–94. [DOI] [PubMed] [Google Scholar]
- Naert G, Rivest S, 2012. Age-related changes in synaptic markers and monocyte subsets link the cognitive decline of APP(Swe)/PS1 mice. Front Cell Neurosci 6, 51. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Nebel RA, et al. , 2018. Understanding the impact of sex and gender in Alzheimer’s disease: A call to action. Alzheimers Dement 14, 1171–1183. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Neth BJ, Craft S, 2017. Insulin Resistance and Alzheimer’s Disease: Bioenergetic Linkages. Front Aging Neurosci 9, 345. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Neu SC, et al. , 2017. Apolipoprotein E Genotype and Sex Risk Factors for Alzheimer Disease: A Meta-analysis. JAMA Neurol 74, 1178–1189. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Nichol K, et al. , 2009. Exercise improves cognition and hippocampal plasticity in APOE epsilon4 mice. Alzheimers Dement 5, 287–94. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Notkola IL, et al. , 1998. Serum total cholesterol, apolipoprotein E epsilon 4 allele, and Alzheimer’s disease. Neuroepidemiology 17, 14–20. [DOI] [PubMed] [Google Scholar]
- O’Brien JT, et al. , 1996. Clinical and magnetic resonance imaging correlates of hypothalamic-pituitary-adrenal axis function in depression and Alzheimer’s disease. Br J Psychiatry 168, 679–87. [DOI] [PubMed] [Google Scholar]
- O’Dwyer L, et al. , 2012. Sexual dimorphism in healthy aging and mild cognitive impairment: a DTI study. PLoS One 7, e37021. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ogasawara T, et al. , 1996. NS-3, a TRH-analog, reverses memory disruption by stimulating cholinergic and noradrenergic systems. Pharmacol Biochem Behav 53, 391–9. [DOI] [PubMed] [Google Scholar]
- Ossenkoppele R, et al. , 2015. Prevalence of amyloid PET positivity in dementia syndromes: a meta-analysis. JAMA 313, 1939–49. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ott BR, et al. , 1996. Gender differences in the behavioral manifestations of Alzheimer’s disease. J Am Geriatr Soc 44, 583–7. [DOI] [PubMed] [Google Scholar]
- Ott BR, Lapane KL, Gambassi G, 2000. Gender differences in the treatment of behavior problems in Alzheimer’s disease. SAGE Study Group. Systemic Assessment of Geriatric drug use via Epidemiology. Neurology 54, 427–32. [DOI] [PubMed] [Google Scholar]
- Ownby RL, et al. , 2006. Depression and risk for Alzheimer disease: systematic review, meta-analysis, and metaregression analysis. Arch Gen Psychiatry 63, 530–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Papassotiropoulos A, et al. , 2002. 24S-hydroxycholesterol in cerebrospinal fluid is elevated in early stages of dementia. J Psychiatr Res 36, 27–32. [DOI] [PubMed] [Google Scholar]
- Patel L, et al. , 2014. Circulating insulin-like growth factor-binding protein 3 levels, independent of insulin-like growth factor 1, associate with truncal fat and systolic blood pressure in South Asian and white European preschool children. Horm Res Paediatr 81, 109–17. [DOI] [PubMed] [Google Scholar]
- Payami H, et al. , 1994. Alzheimer’s disease, apolipoprotein E4, and gender. JAMA 271, 1316–7. [PubMed] [Google Scholar]
- Pedersen WA, Wan R, Mattson MP, 2001. Impact of aging on stress-responsive neuroendocrine systems. Mech Ageing Dev 122, 963–83. [DOI] [PubMed] [Google Scholar]
- Peng S, et al. , 2005. Precursor form of brain-derived neurotrophic factor and mature brain-derived neurotrophic factor are decreased in the pre-clinical stages of Alzheimer’s disease. J Neurochem 93, 1412–21. [DOI] [PubMed] [Google Scholar]
- Pike CJ, et al. , 2009. Protective actions of sex steroid hormones in Alzheimer’s disease. Front Neuroendocrinol 30, 239–58. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Pini L, et al. , 2016. Brain atrophy in Alzheimer’s Disease and aging. Ageing Res Rev 30, 25–48. [DOI] [PubMed] [Google Scholar]
- Placanica L, Zhu L, Li YM, 2009. Gender- and age-dependent gamma-secretase activity in mouse brain and its implication in sporadic Alzheimer disease. PLoS One 4, e5088. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Platt EJ, et al. , 1991. Altered effects of glucocorticoids on the trafficking and processing of mouse mammary tumor virus glycoproteins constitutively expressed in rat hepatoma cells in the absence of nonglycosylated viral components. Mol Endocrinol 5, 1696–706. [DOI] [PubMed] [Google Scholar]
- Podcasy JL, Epperson CN, 2016. Considering sex and gender in Alzheimer disease and other dementias. Dialogues Clin Neurosci 18, 437–446. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Postma IR, et al. , 2014. Neurocognitive functioning following preeclampsia and eclampsia: a long-term follow-up study. Am J Obstet Gynecol 211, 37 e1–9. [DOI] [PubMed] [Google Scholar]
- Pudas S, et al. , 2018. Longitudinal Evidence for Increased Functional Response in Frontal Cortex for Older Adults with Hippocampal Atrophy and Memory Decline. Cereb Cortex 28, 936–948. [DOI] [PubMed] [Google Scholar]
- Pyter LM, et al. , 2013. Sex differences in the effects of adolescent stress on adult brain inflammatory markers in rats. Brain Behav Immun 30, 88–94. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Qin XY, et al. , 2017. Decreased peripheral brain-derived neurotrophic factor levels in Alzheimer’s disease: a meta-analysis study (N=7277). Mol Psychiatry 22, 312–320. [DOI] [PubMed] [Google Scholar]
- Rabbitts PH, et al. , 1985. Metabolism of c-myc gene products: c-myc mRNA and protein expression in the cell cycle. EMBO J 4, 2009–15. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Rainey-Smith SR, et al. , 2018. Genetic variation in Aquaporin-4 moderates the relationship between sleep and brain Abeta-amyloid burden. Transl Psychiatry 8, 47. [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, 1386–92. [DOI] [PubMed] [Google Scholar]
- Reger MA, et al. , 2006. Effects of intranasal insulin on cognition in memory-impaired older adults: modulation by APOE genotype. Neurobiol Aging 27, 451–8. [DOI] [PubMed] [Google Scholar]
- Regitz-Zagrosek V, Lehmkuhl E, Weickert MO, 2006. Gender differences in the metabolic syndrome and their role for cardiovascular disease. Clin Res Cardiol 95, 136–47. [DOI] [PubMed] [Google Scholar]
- Reiman EM, et al. , 2005. Correlations between apolipoprotein E epsilon4 gene dose and brain-imaging measurements of regional hypometabolism. Proc Natl Acad Sci U S A 102, 8299–302. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Reisberg B, et al. , 1987. Behavioral symptoms in Alzheimer’s disease: phenomenology and treatment. J Clin Psychiatry 48 Suppl, 9–15. [PubMed] [Google Scholar]
- Rentz DM, et al. , 2017. Sex differences in episodic memory in early midlife: impact of reproductive aging. Menopause 24, 400–408. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Reverte I, et al. , 2012. Behavioral phenotype and BDNF differences related to apoE isoforms and sex in young transgenic mice. Exp Neurol 237, 116–25. [DOI] [PubMed] [Google Scholar]
- Riant E, et al. , 2009. Estrogens protect against high-fat diet-induced insulin resistance and glucose intolerance in mice. Endocrinology 150, 2109–17. [DOI] [PubMed] [Google Scholar]
- Roberts RO, et al. , 2010. Coronary heart disease is associated with non-amnestic mild cognitive impairment. Neurobiol Aging 31, 1894–902. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Rohe M, et al. , 2009. Brain-derived neurotrophic factor reduces amyloidogenic processing through control of SORLA gene expression. J Neurosci 29, 15472–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Romero R, Chaiworapongsa T, 2013. Preeclampsia: a link between trophoblast dysregulation and an antiangiogenic state. J Clin Invest 123, 2775–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Rosano C, et al. , 2005. Coronary artery calcium: associations with brain magnetic resonance imaging abnormalities and cognitive status. J Am Geriatr Soc 53, 609–15. [DOI] [PubMed] [Google Scholar]
- Rosario PW, 2010. Normal values of serum IGF-1 in adults: results from a Brazilian population. Arq Bras Endocrinol Metabol 54, 477–81. [DOI] [PubMed] [Google Scholar]
- Roselli CE, 2018. Neurobiology of gender identity and sexual orientation. J Neuroendocrinol 30, e12562. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Runz H, et al. , 2002. Inhibition of intracellular cholesterol transport alters presenilin localization and amyloid precursor protein processing in neuronal cells. J Neurosci 22, 1679–89. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ryan J, et al. , 2009. Characteristics of hormone therapy, cognitive function, and dementia: the prospective 3C Study. Neurology 73, 1729–37. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sajja VS, Hlavac N, VandeVord PJ, 2016. Role of Glia in Memory Deficits Following Traumatic Brain Injury: Biomarkers of Glia Dysfunction. Front Integr Neurosci 10, 7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Salloway S, et al. , 2014. Two phase 3 trials of bapineuzumab in mild-to-moderate Alzheimer’s disease. N Engl J Med 370, 322–33. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sastre M, Walter J, Gentleman SM, 2008. Interactions between APP secretases and inflammatory mediators. J Neuroinflammation 5, 25. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Saunders AM, et al. , 1993. Association of apolipoprotein E allele epsilon 4 with late-onset familial and sporadic Alzheimer’s disease. Neurology 43, 1467–72. [DOI] [PubMed] [Google Scholar]
- Scammell JG, Burrage TG, Dannies PS, 1986. Hormonal induction of secretory granules in a pituitary tumor cell line. Endocrinology 119, 1543–8. [DOI] [PubMed] [Google Scholar]
- Schioth HB, et al. , 2012. Brain insulin signaling and Alzheimer’s disease: current evidence and future directions. Mol Neurobiol 46, 4–10. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Schofield PW, et al. , 1997. Alzheimer’s disease after remote head injury: an incidence study. J Neurol Neurosurg Psychiatry 62, 119–24. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Scott C, et al. , 2015. A comparison of adult outcomes for males compared to females following pediatric traumatic brain injury. Neuropsychology 29, 501–8. [DOI] [PubMed] [Google Scholar]
- Selvamani A, Sohrabji F, 2010a. Reproductive age modulates the impact of focal ischemia on the forebrain as well as the effects of estrogen treatment in female rats. Neurobiol Aging 31, 1618–28. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Selvamani A, Sohrabji F, 2010b. The neurotoxic effects of estrogen on ischemic stroke in older female rats is associated with age-dependent loss of insulin-like growth factor-1. J Neurosci 30, 6852–61. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Selvamani A, et al. , 2012. An antagomir to microRNA Let7f promotes neuroprotection in an ischemic stroke model. PLoS One 7, e32662. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sen A, Nelson TJ, Alkon DL, 2015. ApoE4 and Abeta Oligomers Reduce BDNF Expression via HDAC Nuclear Translocation. J Neurosci 35, 7538–51. [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, et al. , 2006. The lifetime risk of stroke: estimates from the Framingham Study. Stroke 37, 345–50. [DOI] [PubMed] [Google Scholar]
- Shah NS, et al. , 2012. Midlife blood pressure, plasma beta-amyloid, and the risk for Alzheimer disease: the Honolulu Asia Aging Study. Hypertension 59, 780–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sheng JG, et al. , 2003. Lipopolysaccharide-induced-neuroinflammation increases intracellular accumulation of amyloid precursor protein and amyloid beta peptide in APPswe transgenic mice. Neurobiol Dis 14, 133–45. [DOI] [PubMed] [Google Scholar]
- Sheridan GK, Murphy KJ, 2013. Neuron-glia crosstalk in health and disease: fractalkine and CX3CR1 take centre stage. Open Biol 3, 130181. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Shi Y, et al. , 2017. ApoE4 markedly exacerbates tau-mediated neurodegeneration in a mouse model of tauopathy. Nature 549, 523–527. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Shi ZX, Levy A, Lightman SL, 1993. Hippocampal input to the hypothalamus inhibits thyrotrophin and thyrotrophin-releasing hormone gene expression. Neuroendocrinology 57, 576–80. [DOI] [PubMed] [Google Scholar]
- Shin HW, Chung SJ, 2012. Drug-induced parkinsonism. J Clin Neurol 8, 15–21. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Shinohara M, et al. , 2016. Impact of sex and APOE4 on cerebral amyloid angiopathy in Alzheimer’s disease. Acta Neuropathol 132, 225–34. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Shumaker SA, et al. , 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, 2651–62. [DOI] [PubMed] [Google Scholar]
- Shumaker SA, et al. , 2004. Conjugated equine estrogens and incidence of probable dementia and mild cognitive impairment in postmenopausal women: Women’s Health Initiative Memory Study. JAMA 291, 2947–58. [DOI] [PubMed] [Google Scholar]
- Sierksma AS, et al. , 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, 697–701. [DOI] [PubMed] [Google Scholar]
- Sierksma AS, et al. , 2013. Behavioral and neurobiological effects of prenatal stress exposure in male and female APPswe/PS1dE9 mice. Neurobiol Aging 34, 319–37. [DOI] [PubMed] [Google Scholar]
- Skup M, et al. , 2011. Sex differences in grey matter atrophy patterns among AD and aMCI patients: results from ADNI. Neuroimage 56, 890–906. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Soares CN, Zitek B, 2008. Reproductive hormone sensitivity and risk for depression across the female life cycle: a continuum of vulnerability? J Psychiatry Neurosci 33, 331–43. [PMC free article] [PubMed] [Google Scholar]
- Song ZF, et al. , 2013. Poly (ADP-ribose) polymerase inhibitor reduces heart ischaemia/reperfusion injury via inflammation and Akt signalling in rats. Chin Med J (Engl) 126, 1913–7. [PubMed] [Google Scholar]
- Sorge RE, et al. , 2011. Spinal cord Toll-like receptor 4 mediates inflammatory and neuropathic hypersensitivity in male but not female mice. J Neurosci 31, 15450–4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sotiropoulos I, et al. , 2015. Female hippocampus vulnerability to environmental stress, a precipitating factor in Tau aggregation pathology. J Alzheimers Dis 43, 763–74. [DOI] [PubMed] [Google Scholar]
- Spira AP, et al. , 2013. Self-reported sleep and beta-amyloid deposition in community-dwelling older adults. JAMA Neurol 70, 1537–43. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Strittmatter WJ, et al. , 1993. Apolipoprotein E: high-avidity binding to beta-amyloid and increased frequency of type 4 allele in late-onset familial Alzheimer disease. Proc Natl Acad Sci U S A 90, 1977–81. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Strosznajder JB, et al. , 2012. Poly(ADP-ribose) polymerase-1 in amyloid beta toxicity and Alzheimer’s disease. Mol Neurobiol 46, 78–84. [DOI] [PubMed] [Google Scholar]
- Sun LL, et al. , 2019. Molecular differences in Alzheimer’s disease between male and female patients determined by integrative network analysis. J Cell Mol Med 23, 47–58. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sun Y, et al. , 2015. Poly(ADP-ribose) polymerase 1 inhibition prevents interleukin-1beta-induced inflammation in human osteoarthritic chondrocytes. Acta Biochim Biophys Sin (Shanghai) 47, 422–30. [DOI] [PubMed] [Google Scholar]
- Szuhany KL, Bugatti M, Otto MW, 2015. A meta-analytic review of the effects of exercise on brain-derived neurotrophic factor. J Psychiatr Res 60, 56–64. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Tai LM, et al. , 2017. EFAD Transgenic Mice as a Human APOE Relevant Preclinical Model of Alzheimer’s Disease. J Lipid Res [DOI] [PMC free article] [PubMed]
- Taki Y, et al. , 2008. Relationship between body mass index and gray matter volume in 1,428 healthy individuals. Obesity (Silver Spring) 16, 119–24. [DOI] [PubMed] [Google Scholar]
- Tan ZS, et al. , 2008. Thyroid function and the risk of Alzheimer disease: the Framingham Study. Arch Intern Med 168, 1514–20. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Tang JH, et al. , 2014. Insulin-like growth factor-1 as a prognostic marker in patients with acute ischemic stroke. PLoS One 9, e99186. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
- Tang MX, et al. , 1996. Effect of oestrogen during menopause on risk and age at onset of Alzheimer’s disease. Lancet 348, 429–32. [DOI] [PubMed] [Google Scholar]
- Tensil M, et al. , 2018. Sex Differences in Neuropsychological Test Performance in Alzheimer’s Disease and the Influence of the ApoE Genotype. Alzheimer Dis Assoc Disord 32, 145–149. [DOI] [PubMed] [Google Scholar]
- Teri L, et al. , 1989. Behavioral disturbance, cognitive dysfunction, and functional skill. Prevalence and relationship in Alzheimer’s disease. J Am Geriatr Soc 37, 109–16. [DOI] [PubMed] [Google Scholar]
- Thakur MK, Ghosh S, 2007. Age and sex dependent alteration in presenilin expression in mouse cerebral cortex. Cell Mol Neurobiol 27, 1059–67. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Thorand B, et al. , 2007. Sex differences in the prediction of type 2 diabetes by inflammatory markers: results from the MONICA/KORA Augsburg case-cohort study, 1984–2002. Diabetes Care 30, 854–60. [DOI] [PubMed] [Google Scholar]
- Tomasi D, Volkow ND, 2012. Gender differences in brain functional connectivity density. Hum Brain Mapp 33, 849–60. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Tononi G, Cirelli C, 2014. Sleep and the price of plasticity: from synaptic and cellular homeostasis to memory consolidation and integration. Neuron 81, 12–34. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Toran-Allerand CD, 1996. The estrogen/neurotrophin connection during neural development: is co-localization of estrogen receptors with the neurotrophins and their receptors biologically relevant? Dev Neurosci 18, 36–48. [DOI] [PubMed] [Google Scholar]
- Tower J, 2006. Sex-specific regulation of aging and apoptosis. Mech Ageing Dev 127, 705–18. [DOI] [PubMed] [Google Scholar]
- Tronson NC, Collette KM, 2017. (Putative) sex differences in neuroimmune modulation of memory. J Neurosci Res 95, 472–486. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Tucker LB, Fu AH, McCabe JT, 2016. Performance of Male and Female C57BL/6J Mice on Motor and Cognitive Tasks Commonly Used in Pre-Clinical Traumatic Brain Injury Research. J Neurotrauma 33, 880–94. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Van Lenten BJ, et al. , 2001. High-density lipoprotein loses its anti-inflammatory properties during acute influenza a infection. Circulation 103, 2283–8. [DOI] [PubMed] [Google Scholar]
- Vandal M, et al. , 2014. Insulin reverses the high-fat diet-induced increase in brain Abeta and improves memory in an animal model of Alzheimer disease. Diabetes 63, 4291–301. [DOI] [PubMed] [Google Scholar]
- Varma VR, et al. , 2015. Low-intensity daily walking activity is associated with hippocampal volume in older adults. Hippocampus 25, 605–15. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Varma VR, Tang X, Carlson MC, 2016. Hippocampal sub-regional shape and physical activity in older adults. Hippocampus 26, 1051–60. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Vassar R, 2004. BACE1: the beta-secretase enzyme in Alzheimer’s disease. J Mol Neurosci 23, 105–14. [DOI] [PubMed] [Google Scholar]
- Vaynman S, Ying Z, Gomez-Pinilla F, 2004a. Exercise induces BDNF and synapsin I to specific hippocampal subfields. J Neurosci Res 76, 356–62. [DOI] [PubMed] [Google Scholar]
- Vaynman S, Ying Z, Gomez-Pinilla F, 2004b. Hippocampal BDNF mediates the efficacy of exercise on synaptic plasticity and cognition. Eur J Neurosci 20, 2580–90. [DOI] [PubMed] [Google Scholar]
- Venezia AC, et al. , 2016. Sex-dependent and independent effects of long-term voluntary wheel running on Bdnf mRNA and protein expression. Physiol Behav 156, 8–15. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Venkatesha S, et al. , 2006. Soluble endoglin contributes to the pathogenesis of preeclampsia. Nat Med 12, 642–9. [DOI] [PubMed] [Google Scholar]
- Verghese PB, et al. , 2013. ApoE influences amyloid-beta (Abeta) clearance despite minimal apoE/Abeta association in physiological conditions. Proc Natl Acad Sci U S A 110, E1807–16. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Villapol S, Loane DJ, Burns MP, 2017. Sexual dimorphism in the inflammatory response to traumatic brain injury. Glia 65, 1423–1438. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Vina J, et al. , 2003. Mitochondrial theory of aging: importance to explain why females live longer than males. Antioxid Redox Signal 5, 549–56. [DOI] [PubMed] [Google Scholar]
- Wang J, et al. , 2003. Gender differences in the amount and deposition of amyloidbeta in APPswe and PS1 double transgenic mice. Neurobiol Dis 14, 318–27. [DOI] [PubMed] [Google Scholar]
- Wang JM, Irwin RW, Brinton RD, 2006. Activation of estrogen receptor alpha increases and estrogen receptor beta decreases apolipoprotein E expression in hippocampus in vitro and in vivo. Proc Natl Acad Sci U S A 103, 16983–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Waters DL, et al. , 2003. Serum Sex Hormones, IGF-1, and IGFBP3 Exert a Sexually Dimorphic Effect on Lean Body Mass in Aging. J Gerontol A Biol Sci Med Sci 58, 648–52. [DOI] [PubMed] [Google Scholar]
- Westwood AJ, et al. , 2014. Insulin-like growth factor-1 and risk of Alzheimer dementia and brain atrophy. Neurology 82, 1613–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Whitmer RA, et al. , 2011. Timing of hormone therapy and dementia: the critical window theory revisited. Ann Neurol 69, 163–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wolfe MS, 2006. The gamma-secretase complex: membrane-embedded proteolytic ensemble. Biochemistry 45, 7931–9. [DOI] [PubMed] [Google Scholar]
- Woolley CS, et al. , 1997. Estradiol increases the sensitivity of hippocampal CA1 pyramidal cells to NMDA receptor-mediated synaptic input: correlation with dendritic spine density. J Neurosci 17, 1848–59. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wu MV, et al. , 2009. Estrogen masculinizes neural pathways and sex-specific behaviors. Cell 139, 61–72. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Xie L, et al. , 2013. Sleep drives metabolite clearance from the adult brain. Science 342, 373–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Xing G, et al. , 2014. Differential Expression of Brain Cannabinoid Receptors between Repeatedly Stressed Males and Females may Play a Role in Age and Gender-Related Difference in Traumatic Brain Injury: Implications from Animal Studies. Front Neurol 5, 161. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Xu H, et al. , 1998. Estrogen reduces neuronal generation of Alzheimer beta-amyloid peptides. Nat Med 4, 447–51. [DOI] [PubMed] [Google Scholar]
- Yaffe K, et al. , 2000. Estrogen use, APOE, and cognitive decline: evidence of gene-environment interaction. Neurology 54, 1949–54. [DOI] [PubMed] [Google Scholar]
- Yang LB, et al. , 2003. Elevated beta-secretase expression and enzymatic activity detected in sporadic Alzheimer disease. Nat Med 9, 3–4. [DOI] [PubMed] [Google Scholar]
- Yang W, et al. , 2012. Aquaporin-4 mediates astrocyte response to beta-amyloid. Mol Cell Neurosci 49, 406–14. [DOI] [PubMed] [Google Scholar]
- Yang Y, et al. , 2013. High glucose promotes Abeta production by inhibiting APP degradation. PLoS One 8, e69824. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Yao J, et al. , 2012. Ovarian hormone loss induces bioenergetic deficits and mitochondrial beta-amyloid. Neurobiol Aging 33, 1507–21. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Yoo DY, et al. , 2016. Chronic type 2 diabetes reduces the integrity of the blood-brain barrier by reducing tight junction proteins in the hippocampus. J Vet Med Sci 78, 957–62. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Yu SW, et al. , 2006. Apoptosis-inducing factor mediates poly(ADP-ribose) (PAR) polymer-induced cell death. Proc Natl Acad Sci U S A 103, 18314–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Yuan M, et al. , 2009. Sex differences in the response to activation of the poly (ADP-ribose) polymerase pathway after experimental stroke. Exp Neurol 217, 210–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Yue M, et al. , 2011. Sex difference in pathology and memory decline in rTg4510 mouse model of tauopathy. Neurobiol Aging 32, 590–603. [DOI] [PubMed] [Google Scholar]
- Yuede CM, et al. , 2009. Effects of voluntary and forced exercise on plaque deposition, hippocampal volume, and behavior in the Tg2576 mouse model of Alzheimer’s disease. Neurobiol Dis 35, 426–32. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zhang B, Wing YK, 2006. Sex differences in insomnia: a meta-analysis. Sleep 29, 85–93. [DOI] [PubMed] [Google Scholar]
- Zhou CN, et al. , 2018. Sex Differences in the White Matter and Myelinated Fibers of APP/PS1 Mice and the Effects of Running Exercise on the Sex Differences of AD Mice. Front Aging Neurosci 10, 243. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zitzmann M, 2009. Testosterone deficiency, insulin resistance and the metabolic syndrome. Nat Rev Endocrinol 5, 673–81. [DOI] [PubMed] [Google Scholar]
- Kunzler J, et al. , 2014. APOE modulates the effect of estrogen therapy on Aβ accumulation EFAD-Tg mice. Neurosci Lett 560: 131–136. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sohrabji F, et al. , 2017. Sex Differences in Stroke Therapies. J Neurosci Res 95(1–2): 681–691. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Basun H, et al. , 2008. Clinical and neuropathological features of the arctic APP gene mutation causing early-onset Alzheimer disease. Arch Neurol 65, 499–505. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Calero M, et al. , 1999. Functional and structural properties of lipid-associated apolipoprotein J (clusterin). Biochem J 344 Pt 2, 375𠄳83. [PMC free article] [PubMed] [Google Scholar]
- Carrasquillo MM, et al. , 2010. Replication of CLU, CR1, and PICALM associations with alzheimer disease. Arch Neurol 67, 961–4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Cohen DM, et al. , 2009. Blood-spinal cord barrier permeability in experimental spinal cord injury: dynamic contrast-enhanced MRI. NMR Biomed 22, 332–41. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Corneveaux JJ, et al. , 2010. Association of CR1, CLU and PICALM with Alzheimer's disease in a cohort of clinically characterized and neuropathologically verified individuals. Hum Mol Genet 19, 3295–301. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Harold D, et al. , 2009. Genome-wide association study identifies variants at CLU and PICALM associated with Alzheimer’s disease. Nat Genet 41, 1088–93. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Miller SE, et al. , 2011. The molecular basis for the endocytosis of small R-SNAREs by the clathrin adaptor CALM. Cell 147, 1118–31. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Nuutinen T, et al. , 2009. Clusterin: a forgotten player in Alzheimer’s disease. Brain Res Rev 61, 89–104. [DOI] [PubMed] [Google Scholar]
- Rogaeva E, et al. , 2007. The neuronal sortilin-related receptor SORL1 is genetically associated with Alzheimer disease. Nat Genet 39, 168–77. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sweeney MD, et al. , 2019a. Vascular dysfunction-The disregarded partner of Alzheimer’s disease. Alzheimers Dement. 15, 158–167. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sweeney MD, et al. , 2019b. Blood-Brain Barrier: From Physiology to Disease and Back. Physiol Rev 99, 21–78. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Tanzi RE, 2012. The genetics of Alzheimer disease. Cold Spring Harb Perspect Med 2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zhao Z, et al. , 2015. Central role for PICALM in amyloid-beta blood-brain barrier transcytosis and clearance. Nat Neurosci 18, 978–87. [DOI] [PMC free article] [PubMed] [Google Scholar]