Next Article in Journal
Oncolytic Viruses in the Era of Omics, Computational Technologies, and Modeling: Thesis, Antithesis, and Synthesis
Next Article in Special Issue
Microglial Transforming Growth Factor-β Signaling in Alzheimer’s Disease
Previous Article in Journal
Identification and Functional Analysis of an Epsilon Class Glutathione S-Transferase Gene Associated with α-Pinene Adaptation in Monochamus alternatus
Previous Article in Special Issue
The Role of Insulin-like Growth Factor I in Mechanisms of Resilience and Vulnerability to Sporadic Alzheimer’s Disease
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Minding the Gap: Exploring Neuroinflammatory and Microglial Sex Differences in Alzheimer’s Disease

by
Erin G. Reed
1,* and
Phaedra R. Keller-Norrell
2
1
Department of Pharmaceutical Sciences, Northeast Ohio Medical University, Rootstown, OH 44242, USA
2
School of Biomedical Sciences, Kent State University, Kent, OH 44140, USA
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2023, 24(24), 17377; https://doi.org/10.3390/ijms242417377
Submission received: 20 November 2023 / Revised: 4 December 2023 / Accepted: 9 December 2023 / Published: 12 December 2023
(This article belongs to the Special Issue Recent Advances in Alzheimer’s Disease)

Abstract

:
Research into Alzheimer’s Disease (AD) describes a link between AD and the resident immune cells of the brain, the microglia. Further, this suspected link is thought to have underlying sex effects, although the mechanisms of these effects are only just beginning to be understood. Many of these insights are the result of policies put in place by funding agencies such as the National Institutes of Health (NIH) to consider sex as a biological variable (SABV) and the move towards precision medicine due to continued lackluster therapeutic options. The purpose of this review is to provide an updated assessment of the current research that summarizes sex differences and the research pertaining to microglia and their varied responses in AD.

1. Introduction

Alzheimer’s disease (AD) affects approximately 6.7 million Americans aged 65 or older, of whom approximately two-thirds are women [1]. The higher number of female AD cases (the prevalence of AD) is comprised of both the number of newly diagnosed cases (the incidence of AD) and the number of individuals living with a diagnosis. Whether the incidence of AD in the US is sexually dimorphic is unclear, but women do live longer following an AD diagnosis [2,3]. This suggests not only the existence of factors that increase the overall risk in females and/or decrease the risk in males but also an increased resilience to disease processes among women diagnosed with AD. Furthermore, sexual dimorphism is seen in clinical manifestations of the disease, disease progression, neuroimaging, and pathology [4,5,6,7,8,9,10,11,12,13]. The mechanisms underlying these disparities are unknown, but research suggests a variety of candidate mechanisms, including risks linked to sex, age, gender, genetics, and environment.
Several excellent reviews have recently been written describing microglial sex differences in brain development, maintenance, disease, and in response to lifestyle influences [14,15,16,17,18,19,20,21]. Although microglia recruit and respond to the adaptive immune system, and this interaction is becoming progressively more appreciated, the adaptive immune system in AD has been recently reviewed elsewhere [22]. We will therefore focus this review on the most recent findings, situating and contextualizing them in a rapidly expanding field to provide a comprehensive view of sex differences in AD, highlighting the contribution of microglia to these differences (summarized in Figure 1).
The authors would like to note the term sex used within this article will follow the Merriam-Webster usage guidelines (available at https://www.merriam-webster.com/dictionary/sex, accessed 29 June 2023). The authors recognize sex and gender have often been used interchangeably in western cultures, and that many languages and cultures make no distinction between the two terms [23]. However, within the context of this review, sex refers to physical traits. Gender, referring to cultural and/or societal constructs, is beyond the scope of this review. As the field continues to expand at a prodigious rate, it must coalesce around nomenclature and approaches to assess these differences to appropriately interpret and extend findings [24].

2. Sex Differences in AD

2.1. The First Patient Was a Woman

AD was first characterized by its namesake German psychiatrist, Dr. Alois Alzheimer, in 1901 when Alzheimer was introduced to Auguste Deter, a 51-year-old woman struggling with cognitive and behavioral abnormalities [25]. Alzheimer’s observations of Auguste and the subsequent evaluation of her post-mortem cerebral tissue samples by Alzheimer and two physicians, Gaetano Perusini and Francesco Bonfiglio, became the first clinical record of AD [25]. The gross histopathological observations made by Alzheimer and his team, namely the presence of neuritic β-amyloid (Aβ) plaques and neurofibrillary tangles, remain the hallmark diagnostic criteria for the final stage of AD progression [25,26].
Dr. Alzheimer’s seminal studies laid the foundation for the characterization of AD pathology in human subjects, and, in the century following his initial investigations, the origins of AD’s characteristic plaques and tangles are now understood. Additionally, new mechanisms of AD pathophysiology implicate peripheral body systems in the disorder, indicating that plaques and tangles are only one piece of the much larger puzzle that is AD.

2.2. Consistency across AD Subtypes

AD is categorized based on age at symptom onset and heritability. The arbitrary age of 65 years is used to distinguish early- and late-onset AD. Two or more family members having AD results in it being considered familial [27]. These categories give rise to four subtypes: familial early-onset AD (FEOAD), sporadic early-onset AD (SEOAD), familial late-onset AD (FLOAD), and sporadic late-onset AD (SLOAD). Clinical features vary between subgroups (reviewed in [28]) but overlap so substantially that subtypes cannot be clinically distinguished from each other [27]. Furthermore, common biological pathways, shared clinical and pathological features, and consistent sex effects suggest findings in one subtype (summarized in Table 1) will be relevant to the others.
In EOAD (early-onset AD), symptoms manifest prior to the age of 65 but in rare instances may begin as early as a patient’s 30s. EOAD accounts for approximately 10% of all AD patients [40]. The heritability of EOAD is 92–100% [41], and 10–15% of these familial cases are due to autosomal dominant transmission [42,43]. Numerous pathogenic loci within three genes—Amyloid Precursor Protein (APP), Presenilin 1 (PSEN1), and Presenilin 2 (PSEN2)—have been identified, each contributing to disease pathogenesis in a unique manner. It is important to note that mutations in these three genes only explain 5–10% of EOAD diagnoses [41,44], leaving 90–95% of EOAD cases without a definitive cause.
Among EOAD patients, females have elevated tau burden compared to their male counterparts [29]. They also present with broader atrophy associated with worse cognition at diagnosis [30]. Intriguingly, female EOAD carriers may have greater cognitive resilience to AD pathology and neurodegeneration [33] but exhibit a greater rate of neurodegeneration and memory impairment during disease progression [31,32]. The mechanisms underlying accelerated neurodegeneration in females is unknown but could be due to the sex-selective vulnerability of specific brain regions that have reciprocal connections to other regions [45,46], the interactions between tau pathology and sex-specific genes, chromosomes, and/or hormones [47], possible genetic modifying factors [28], and the sex-specific amyloid and/or tau dynamics.
Autosomal dominant mutations in APP, PSEN1, and PSEN2 have been used to generate a variety of mouse models to recapitulate the amyloid pathology with subsequent neuroinflammation and neuron loss (reviewed in [48,49]). As in human EOAD patients, mouse models exhibit sex differences in plaque burden and rates of deposition, neuroinflammation, and cognition. While these models have extensive benefits to understanding amyloidogenic processes, they do have drawbacks including: (1) the neuronal loss in these mice is not as great as in human patients; (2) the sex effects are at times the opposite as those in human patients; (3) the absence of tau pathology, leading investigators to introduce mutations found in other tauopathies such as frontotemporal dementia to replicate that pathology. It had been proposed that the accelerated amyloid deposition in female mice was an artifact of the model, arising from the estrogen response element within the promoter used to express these mutations [50,51]. However, recent studies, some of which will be discussed in detail below, have indicated non-estrogen sensitive (i.e., sex chromosome) mechanisms may be behind some of these differences.
In contrast to EOAD, LOAD exhibits symptom onset after the age of 65 and accounts for almost all AD cases. LOAD is a complex disorder with a heterogeneous etiology and 70–80% heritability [41,52], where 40% of total LOAD cases are familial [53]. LOAD is driven by several factors including genetics, lifestyle, environment, and neurodevelopmental disorders. More than 50 genetic risk loci have been identified [54], with Apolipoprotein E (APOE) having the strongest correlation [55] followed by Triggering receptor expressed on myeloid cells 2 (TREM2; [56,57]). These risk genes are associated with various biological processes, including the immune response/inflammation, lipid metabolism, and neuronal/synaptic function. Sex differences are observed in LOAD patients, with females showing faster hippocampal volume loss [12] and greater brain glucose hypometabolism [34]. Significantly, as in EOAD, females with LOAD may have greater cognitive resilience to early pathology and neurodegeneration but exhibit faster decline and progression to dementia as the disease progresses [4,6,7,12,35,36,37,38,39,58]. Several factors have been proposed as contributions to the sex differences in LOAD, including specific genes [59,60,61], inflammation [62], cardiovascular disease [63], and hormonal changes [64], although the exact mechanisms remain to be determined.
Numerous mouse models of LOAD exist. Since none of the LOAD risk alleles are independently necessary or sufficient to drive AD, investigators are generating mouse models of combinatorial risk variants [54]. As these models are developed and further characterized, it will be important to determine whether they recapitulate the sex-specific effects seen in human patients. Additional approaches involve metabolic dysregulation, traumatic brain injury, adeno-associated virus 1 (AAV1) gene transduction, toxin exposure, perturbed metal ion homeostasis, and aging (reviewed in [65]). Like in the EOAD models, many of these LOAD models show sex differences, but not all recapitulate those seen in human patients. The senescence-accelerated mouse prone 8 (SAMP8) model is of particular interest and relevance. Age is the single greatest risk factor for LOAD, and although the exact cause of accelerated senescence has yet to be determined, the SAMP8 model may shed light on the relationship between aging and AD [66], particularly in sex-specific gene regulation, as discussed below.
Sex differences occur across multiple domains in both human AD patients and in mouse models of the disease, regardless of the pathogenic mechanism. A disadvantage is that the mouse models are often only replicating a portion of the disease process, such as dominant mutations driving amyloid production. Despite these drawbacks, the consistent commonalities suggest that findings in one context may translate to others. Where there are differences between the sexes in the mechanisms underlying each type of AD, this finding provides critical avenues to develop additional therapeutics to target specific patient populations. In short, while each mouse model is an imperfect representation of the human AD condition, in a disease with such varied pathogenesis there is still much work to be done with the tools available to further our understanding and the development of effective therapeutics.

2.3. Accompanying Gene Expression

Not surprisingly, in both humans and mice the sex differences in disease manifestation and progression described above are accompanied by sex-specific gene expression patterns [67,68] and epigenetic profiles [69,70]. Integrative network analysis of human samples revealed sex-specific functional modules, pathways, and genes that were associated with clinical characteristics in males only and molecular networks that were more conserved temporally and spatially in females [71]. Brain-region-specific, sex-biased patterns may arise from differences in vulnerabilities and/or resilience in different brain regions at distinct stages of AD development [72]. Updates on the possible mechanisms underlying these vulnerabilities and/or resilience will be addressed in more detail in the following sections.
A gene commonly implicated across all AD subtypes is APOE. The predominant function of APOE is that of trafficking lipids in the central nervous system; however, it functions in several brain processes, including neuron development and function, formation of cytosolic lipid droplets, endolysosomal trafficking, mitochondrial metabolism, and innate immunity [73,74]. These varied roles contribute to the widespread effects of APOE on AD risk. There are three common isoforms—APOE2, APOE3, and APOE4—that are generated by single nucleotide polymorphisms (SNPs), resulting in differences of two amino acid residues [73]. Notably, APOE2 seems to have a protective effect, while APOE4 confers a significant risk for developing AD, although recent studies have suggested that the risks associated with APOE4 may be overestimated (reviewed in [75]). Furthermore, the biological pathways underlying the APOE4-associated risk are distinct from the protective effects of APOE2 and intersect with age-related changes in sex biology [76].
Work in mice demonstrated that ApoE is abundantly expressed in both the brain and in the periphery as distinct pools that are kept separate by the blood brain barrier (BBB). Within the brain, ApoE is predominantly produced by astrocytes, but can come from stimulated microglia [77,78,79]. In fact, in mice, microglial-derived ApoE4 signals through the ITGB8-TGFβ (integrin subunit beta 8-transforming growth factor-β) pathway to negatively regulate the microglial response to AD pathology [80]. ApoE4 produced by the liver, while not entering the brain, can exacerbate amyloid pathology [81], providing a potential peripheral therapeutic target to treat AD.
In human AD patients and in mouse models, females are more strongly affected by APOE status. Female-specific effects due to APOE4 are seen in cognitive impairment [82], tau accumulation [83], gene expression [84,85], brain metabolism [86], cerebral microbleed frequency [87], TREM2-dependent microglial activation [88], and astrogliosis [89]. This could be due in part to the APOE4 modulating expression of the estrogen receptor ERα. It has been proposed that males and females share genetic regulators of amyloidosis, but, as amyloid pathology progresses, it has been demonstrated that APOE exerts sex-specific effects on gene expression, resulting in sex bias in disease manifestation [60,61].
A top key regulator of female AD gene networks was determined to be lipoprotein receptor-related protein 10 (LRP10), potentially driving sex differences based on its high regulatory strength and network connectivity, sex-specific differential expression, and dependence on APOE4 gene dosage [72]. It is hypothesized that reduced LRP10 in APOE4-carrier female AD brains increases the amyloid burden through enhanced amyloid production and reduced clearance due to impaired protein trafficking [72].
Epigenetic modifications including methylation and acetylation regulate chromatin structure and gene expression without modifying the DNA sequence. They are important contributors to sex differences in brain function and AD vulnerability [90,91,92,93,94,95,96,97,98,99,100]. Significantly, they are modifiable, transmissible, and strongly influenced by environmental factors including hyperphosphorylated tau [101,102,103,104,105]. Epigenetic changes, including chromatin-modifying enzymes, epigenetic marks, and microRNAs (miRNAs), are observed in humans during normal aging and AD, and are found in the SAMP8 mouse model, potentially accounting for the accelerated senescence (reviewed in [106]). Sex-specific epigenetic changes in the dorsal hippocampus, a region important for learning and memory, suggest epigenetic mechanisms in this region are regulated in sexually dimorphic ways [70]. Zhang et al. recently conducted a comprehensive meta-analysis revealing several novel DNA methylations associated with the AD Braak stage in a sex-specific manner [69]. In fact, for many genes previously implicated in AD neuropathology, the effects are predominantly driven by only one sex, with enrichment of integrin activation in females and complement activation in males [69].
These studies provide important insights into the molecular contributions underlying sex differences in AD onset and disease progression. However, while many studies did evaluate regional differences, cell-type-specific changes were largely absent. This is significant as each brain cell type contributes to brain function in disease in a unique manner. Furthermore, various cell types may work in concert or in opposition to other cell types in response to pathology.

3. Sex-Specific Neuroinflammation

Inflammation and neurodegeneration are strongly correlated, which is evidenced by the presence of inflammatory cytokines in the brains and CSF (cerebrospinal fluid) of AD patients. These are produced in response to several AD-related processes, including amyloid plaques, neurofibrillary tangles, and signals coming from sick, dying, and dead neurons [107]. As these stimuli are never completely removed, the inflammatory response becomes chronic, exacerbating the pathology and furthering disease processes. The significance of this neuroinflammatory response is highlighted by the substantial number of immune-related genes found to be correlated with AD risk in several genetic studies.

3.1. Sexually Dimorphic Microglia

The cells primarily responsible for this inflammatory response in the AD brain are microglia. They derive from peripheral macrophages that infiltrate the brain early in embryonic development prior to the formation of the blood–brain barrier [108,109]. These microglial progenitors proliferate and colonize the brain, tiling themselves such that no two microglia survey the same brain area. This colony of cells continues to proliferate until the cells reach optimum density [108,110]. Throughout development and adulthood, microglia serve a variety of homeostatic needs, ranging from synaptic pruning to acting as macrophages when cells undergo apoptosis [111,112,113]. They secrete both pro- and anti-inflammatory cytokines in a context-dependent manner, as well as neurotrophic factors [112].
Microglia actively survey the brain parenchyma, constantly extending and retracting processes such that the entire brain parenchyma is assessed within a few hours [114]. Following disruption by injury, infection, or disease, microglia shift from this active surveillance state to one that is responsive to the disruption. This shift is reflected by a general change in their morphology, and the acquisition of distinct transcriptional profiles. With the advent of advanced techniques, a more comprehensive understanding of these phenotypes has led to a recent re-evaluation of microglial nomenclature and classification [115].
Whether microglia exhibit sex differences under homeostatic conditions has been under active investigation and has yielded sex-specific patterns that depend on brain region and age (summarized in Table 2). There are no differences in microglial number or density in the amygdala, hippocampus, or prefrontal cortex during the embryonic or early-neonatal periods [16,116,117]. During the first postnatal week, males have more phagocytic amoeboid microglia in the amygdala, while females have more in the hippocampus [16,116,118]. Microglia of the dentate gyrus do not exhibit sex differences in their number, density, or morphology at post-natal day 10 (P10) [119]. However, by adolescence, morphological sex differences are accompanied by sex differences in microglial density. Male microglia in the prefrontal cortex have more complex branching, particularly in the branches more proximal to the soma [117], and a higher density in the cortex and hippocampus, compared to a higher density in the amygdala in females [120]. Microglia in the hippocampus of females are larger and more phagocytic than those in males [121]. Between adolescence and adulthood, another shift occurs, where the branching of female microglia in the prefrontal cortex becomes more complex [117]. Males continue to have a higher density of microglia in the hippocampus and cortex [120]. However, using hierarchical clustering on principal components, a recent study of the microglia of the adult mouse central nervous system found no sex differences in microglial morphology [122]. These discrepancies in microglial density and morphology could arise from a variety of sources, including pathogen exposure in housing, tissue processing and immunostaining techniques, and quantitative approaches. Moreover, sex and strain differences have been noted in other immune cell types such as B and T lymphocytes, NK cells, immature myeloid cells, and macrophages [123], suggesting that evaluation of various mouse strains may further complicate both comparison and interpretations.
While the general features of microglia (i.e., their number and shape) provide conflicting evidence of sex differences, expression studies are much more definitive, indicative of latent sex differences where male and female microglia use different mechanisms (such as gene expression and signaling cascades) to achieve similar outcomes (including the number, shape, and response to insult). At embryonic day 14.5 (E14.5), microglia from male and female mice are not transcriptionally different from each other [124]; however, distinct transcriptomes are seen at E18.5, with enrichment of apoptotic and inflammatory genes in female microglia [125]. Homeostatic microglia from male and female neonatal and adult mice continue to express different genes [120,126,127,128,129] and miRNAs [130], such that the inflammatory milieu of the brain is different [131]. Notably, marginal sex differences were observed in the gene expression profiles of hippocampal microglia from young mice, but sex chromosomally and autosomally encoded differences emerged with aging, with a female bias towards senescence and inflammation [132]. These studies have led to the hypothesis that male and female microglia mature at different rates, with implications for their response to various stimuli. In fact, after immune challenge, there are sex differences in the cytokines found in the hippocampus [133] and in discrimination memory impairments [134]. Microglia migrate at sex-specific rates [120], and, after injury, microglial mobility is regulated by interferon (IFN) γ in males but not females [135]. Single-cell and bulk RNA sequencing have also provided evidence for sex-specific gene expression in human microglia [136], extending the relevance of the findings in mice to humans.
Table 2. Sex differences observed in microglia across the rodent lifespan.
Table 2. Sex differences observed in microglia across the rodent lifespan.
Age *SpeciesMicroglial Sex DifferenceReferences
E14.5Mouse No difference in transcriptome [124]
E17RatNo difference in number, morphology in amygdala, hippocampus[116]
E18.5MouseFemales: express more apoptotic, inflammatory genes[125]
Birth/P0RatNo difference in morphology in prefrontal cortex[117]
P0–P4RatMales: higher density in amygdala[16]
P3RatFemales: more phagocytic in hippocampus[118]
MouseFemales: express more inflammatory cytokines[126]
P4RatMales: more amoeboid in cortex, hippocampus, amygdala[116]
P8MouseFemales: larger, more phagocytic in hippocampus[121]
P10MouseNo differences in number, density, morphology in dentate gyrus[119]
3 weeksMouseMales: higher density in hippocampusFemales: higher density in amygdalaNo difference in density in striatum, cerebellum[120]
P28MouseMales: larger, more phagocytic in hippocampus[121]
P30RatFemales: more activated in cortex, hippocampus, amygdala[116]
Males: more complex branching in prefrontal cortex[117]
P60RatFemales: more activated in cortex, hippocampus, amygdala[116]
MouseFemales: more transcriptionally mature[127]
MouseFemales: increased inflammatory gene expression[127]
2–6 monthsMouseMales: IFN-dependent migration after injury[135]
12 weeksMouseMales: express more inflammatory genesFemales: more neuroprotective[129]
P90RatFemales: more complex branching in prefrontal cortex[117]
3 monthsMouseNo difference in morphology in any brain, spinal cord region[122]
13 weeksMouseMales: higher density in hippocampus, cortex, amygdalaNo difference in density in striatum, cerebellumMales: greater antigen presentation capability[120]
18 monthsMouseFemales: more phagocytic, reduced ability to respond to insult[137]
22–25 monthsMouseFemales: express more disease, senescence genes[132]
24 monthsMouseFemales: express more inflammatory genes[128]
* E, embryonic day; and P, postnatal day.

3.2. Sources of Microglial Sex Differences

The mechanisms giving rise to these sex differences in microglial gene expression and responses are currently being investigated and include both genetic and hormonal influences. Significantly, sex chromosomes and sex hormones can act in a synergistic or antagonistic manner on a given process [138].
From a genetic standpoint, every cell in the body, microglia included, contains a set of sex chromosomes. Placental mammalian females have two X chromosomes, and placental mammalian males have an X and a Y chromosome. As the X chromosome contains a plethora of genes, many with immune-related functions, and a high concentration of microRNAs that can regulate expression of autosomal genes, the expression of XX chromosome gene dosage must be normalized to the XY levels in males. This is accomplished through the action of the long non-coding (lnc) RNA Xist in a process referred to as X chromosome inactivation (XCI). Detailed reviews of this complex process have been recently published [139,140,141,142], so we will only summarize it here.
Xist is randomly transcribed from one X chromosome; one to two RNA molecules recruit chromatin-modifying proteins, transcriptional silencers, and other RNA-binding proteins to approximately 50 distinct foci along one X chromosome; subsequently, local protein gradients are generated to coat and render that X chromosome inactive (Xi; [139,140,141,142,143]). Interestingly, San Roman et al. found that the active X (Xa) and inactive X (Xi) transcriptomes to be modular, with Xi modulating Xa transcript levels in cis and in trans [144]. Furthermore, only ten X chromosome genes were identified as most likely to contribute to male–female differences in common diseases [144], providing critical insight into the expression of genes on the X chromosomes. The escape or disruption of XCI leads to the loss of gene dosage compensation, driving pathogenic immune responses [145]. X chromosome epigenetics may relate to an elevated AD risk [146], where there is a robust neuroinflammatory response contributing to disease onset and progression. Furthermore, the extent of XCI, particularly in the brain, decreases with age [147], further increasing X chromosome-related gene expression.
Another important event during aging that can alter expression from the sex chromosomes is loss of the Y chromosome (LOY). An increasing frequency of mitotic missegregation errors along with declining genomic instability and impaired DNA repair capabilities may lead to LOY [148,149,150]. This common post-zygotic structural mutation shows a robust association with AD [151,152], contributing to disease through immune system dysfunction [150,153,154]. It has been hypothesized that as microglia proliferate, they could be more prone to LOY accumulation [155]. A recent study showed LOY is enriched in microglia from AD patients, resulting in dysregulation of many genes associated with aging and inflammation [156]. This study demonstrates how perturbations in processes associated with age-related inflammation could lead to neurodegeneration.
Hormones, including gonadotropins, androgens, and estrogens, also contribute to brain sexualization (reviewed in [18]). During critical windows in the neonatal period, sequential surges of these hormones result in the life-long patterning of brain circuitry that can then be re-activated by circulating hormones in adulthood. Microglia are affected by these neonatal surges, as administration of estradiol or testosterone to female rodents during these critical windows phenotypically and molecularly masculinizes the microglia, as well as masculinizes rodent behavior in adolescence and adulthood ([117,129,157]). Furthermore, administration of indomethacin to males prevents the masculinizing effects of prostaglandin E2 [158,159,160]. Epigenetic mechanisms prevent masculinization of the brain; however, the details of how this occurs remain elusive [21]. Interestingly, a recent study suggested estrogens may restrain microglial immune responses, thereby reducing vulnerability to adverse behavioral changes [161].

3.3. Microglial Responses in AD

Upon recognition of injury or insult, microglia transition to a responsive state with sex and age playing a role in this response [128,129,162]. Single-cell and single-nuclei sequencing studies have rapidly expanded the understanding of diverse microglial responses to AD-related pathology in human patients and in rodent disease models. Notably, female mice indicate sex-dependent microglial activation in response to amyloid but not tau pathology [163]. Although some differentially expressed genes in microglia are common between amyloid and tau pathologies, microglia upregulate phagocytic, inflammatory, and proteostatic pathways in areas of higher amyloid and interleukin (IL)-1 in association with tau [164]. The gene signature of disease-associated microglia (DAM), also referred to as neurodegenerative microglia (MGnD), is strongly correlated with apoptosis and myelin debris, consequences of neurodegeneration and inflammation, and is associated with amyloid pathogenesis [165,166,167,168]. Initially, DAMs/MGnDs associate with amyloid plaques, aiding in amyloid clearance though inefficiently [165,169]. Their prolonged presence contributes to the pathogenesis of neurodegenerative diseases through the impairment of microglial homeostatic mechanisms such as phagocytosis, antigen presentation, cell motility, dysregulation of reactive oxygen species (ROS) generation, and increased cytokine expression, resulting in neuron loss [165,166]. Notably, sex differences exist within these gene signatures (reviewed in [170]), but how they arise and whether they induce or are secondary to disease processes is not entirely clear and is being assessed.
It is becoming increasingly appreciated that early-life adverse events or conditions may affect microglia such that they later behave in ways that promote AD pathogenesis. A common approach to studying these effects is through neonatal maternal separation (MS) of rodent pups from their mother. This early-life adversity drives sex-specific changes in microglial morphology and immune challenge responses at various ages [171]. MS also alters cortical microglial activation, hippocampal gene expression, synaptic markers, and immune cell populations in sex-specific ways at various ages, and exacerbates amyloid deposition, particularly in females [172,173]. Additionally, neonatal immune challenges result in female-specific changes in social behavior and microglial cell number [174]. A recent study reported that even prenatal environmental stressors activated the immune system such that postnatal microglial function and adult behavior was impaired in males [175]. Together, these findings indicate that developmental and/or early-life events can program microglia to respond in distinct ways, with implications for subsequent responses during adulthood.

4. Sex-Specific Impacts on Brain Cytoarchitecture

As mentioned above and recently reviewed extensively elsewhere, the brain is highly sexualized across multiple levels, from genes to cells, circuits, and behaviors [176,177,178]. Both sex chromosomes and signaling through gonadal hormone receptors affect brain structures and gene regulation in humans and mice [170,179,180]. Recent studies have reported sex-specific microglial regulation of cell genesis in the neonatal hippocampus [181], sexual dimorphism in stereotyped cell-type-based cortical architecture [182], hippocampal astrocytes [183], and oligodendrocyte precursor cells [184]. Sex differences exist in brain protein expression [185] and gene expression and behavior [186]. Furthermore sex-specific reliance on certain proteins impacts synaptic connectivity, microglial activity, and behavior [187].

4.1. Sex-Specific Neuronal Effects in AD

Female AD patients have greater brain atrophy and neurodegeneration than male AD patients, contributing to greater declines in memory, reasoning, language, and spatial orientation [2,7,10,35,188,189,190,191]. As discussed extensively elsewhere [192,193,194,195], olfactory dysfunction has been reported in human AD patients and in mouse models of AD. It is one of the earliest clinical symptoms of AD, thereby acting as a biomarker of disease, and exhibits a strong sex bias. Changes in brain region volumes and connectivity as well as neurotransmitters have been implicated in olfactory deficits (reviewed in [196]). In fact, women exhibit accelerated age-related loss of olfactory cortical neurons [197]. Gene expression (sex chromosomal and autosomal) and hormones have recently been identified as potential mechanisms that could contribute to sex differences in neuronal survival during AD pathogenesis.
As discussed above, in females one X chromosome must be silenced or inactivated to achieve proper gene dosage. However, some genes on the Xi escape this silencing and may contribute to or modulate neurodegenerative processes. One such gene is KDM6A, also known as Utx, which encodes a lysine-specific histone demethylase important in cognition [198,199,200,201,202,203,204,205,206]. XCI escape by KDM6A increases its expression in the brains of females, conferring resilience to AD-related neuronal vulnerability [3]. This effect is independent of its demethylase function and could provide novel therapeutic targets for treating cognitive deficits in both sexes [207].
Another gene important in neuronal survival is MGMT, which encodes a DNA methyltransferase that is important in protecting cells from apoptosis following DNA damage [208]. Women, particularly those who do not carry the APOE4 allele, express less MGMT and are more likely to have AD [209]. The lower expression of MGMT stems from sex-specific methylation patterns due to SNPs in the MGMT locus that interact with distal enhancers via chromatin loops [210,211]. The mechanisms underlying sex-specific methylation patterns and SNPs remain to be determined. The interaction of MGMT with APOE may occur due to convergence on signaling pathways associated with inflammation.
The neuroprotective effects of estrogen are widely appreciated, but recently the pituitary gonadotropin follicle-stimulating hormone (FSH) was implicated in female susceptibility to AD [212]. FSH levels rapidly increase in the perimenopausal phase, and elevated levels are strongly associated with the onset of AD [213,214,215]. FSH-induced neuronal apoptosis in a mouse model of AD and blocking FSH activity using an anti-FSH antibody prevented neuron loss [212]. These effects on neuronal survival were accompanied by changes in synapse number, cognition, and amyloid and tau pathology, revealing a new potential mechanism underlying the accelerated AD pathogenesis in women during menopause, providing another potential therapeutic target.
In addition to neuronal survival, neuronal activity also exhibits sex specificity. Glutamate is the main excitatory brain neurotransmitter and plays a key role in learning in memory [216,217]. In males but not females, the G-coupled metabotropic glutamate receptor mGluR5 on neurons tightly binds oligomeric amyloid in a cellular prion protein-dependent manner, suppressing autophagic signaling, resulting in cognitive deficits [218]. These sex-specific effects in mGluR5 binding and scaffolding are estrogen-independent, but the source of this difference remains to be determined. This study indicated the possibility of repurposing mGluR5-selective modulators to treat male AD patients, highlighting the need to stratify clinical trials assessing AD therapeutics by sex.

4.2. Sex-Specific Glial Effects in AD

Glia modulate neuronal activity. Astrocytes are the most abundant glial cells, playing critical roles in synaptic transmission and plasticity by providing trophic and metabolic support to neurons. Female hippocampal astrocytes in a mouse model of AD exhibited low inflammatory activity and calcium flow associated with low cannabinoid signaling compared to their male counterparts [219]. These sex differences were evident at birth, suggesting intrinsic sex differences in astrocyte activity that may eventually impact their response during disease progression.
Microglia also modulate the brain’s activity through removal of synapses as well as entire neurons. A subset of microglia, ARG1+ microglia, located primarily in the basal forebrain and ventral striatum during early-postnatal development in the mouse, were recently reported [220]. These microglia are enriched in phagocytic inclusions, exhibit a distinct molecular signature, and play a critical role in shaping neuronal circuits involved in cognition through their actions on cholinergic innervation and spine maturation. Microglia also communicate with astrocytes providing neuronal support and synapse pruning, as discussed above. How microglial functions in homeostatic and AD processes interact with recently identified chromosomal and hormonal factors contributing to neuronal resilience and protection remains to be determined.
Estrogens and androgens have long been understood to have neuroprotective and anti-inflammatory effects. However, together, these recent findings illustrated in Figure 2 provide novel mechanisms contributing to neuronal resilience and vulnerability, whether it be intrinsic to the neurons themselves, or secondary to glial-mediated effects. Autosomal or sex chromosomal genes are now appreciated to have effects on neurons and glia, and the identification of another hormone provides another potential therapeutic target.

5. Sex Differences in Amyloid Deposition

Aβ was first implicated in the pathogenesis of neurocognitive disorders in 1984, when meningovascular plaques from the brains of Down’s Syndrome patients were genetically sequenced and found to be comprised primarily of Aβ [221,222]. Subsequent studies sequenced the APP gene and confirmed that Aβ originated from the biochemical processing of APP. By 1991, a large body of evidence suggested that the plaques observed in AD were like those initially observed in Down’s Syndrome patients [222,223,224]. Females exhibit a greater amyloid burden compared to males at comparable disease stages in both humans and mice, which could arise from differential rates of amyloid production and/or clearance.

5.1. Aβ Production

Aβ is formed through sequential proteolytic processing of the transmembrane Amyloid Precursor Protein (APP), which is ubiquitous throughout the central nervous system (CNS; [224]). In healthy brains, cleavage of APP by α-secretase does not produce pathogenic Aβ but rather soluble byproducts [225]. In AD, APP is cleaved by β-secretase (BACE), resulting in two soluble ectodomains and two carboxy-terminal fragments [224]. These fragments are further cleaved by γ-secretase, leading to the production of pathogenic Aβ, p3, and the intracellular APP domain [224,226]. γ-secretase is a multi-subunit protease containing Nicastrin, Presenilin 1 and/or 2 (PSEN1/PSEN2), Presenilin 2 Enhancer (PEN2), and Anterior Pharynx-Defective 1 (APH-1). The Presenilins form the catalytic subunit of γ-secretase; thus, mutations in PSEN1/PSEN2 increase the activity of the γ-secretase proteolytic pathway, increasing the deposition of Aβ [227,228,229].
Aβ has two major isoforms found in AD, Aβ40 and Aβ42, with the latter linked to the deposition of neuritic plaques [230,231,232]. AD patients exhibit an increased Aβ42:Aβ40 ratio, which is thought to drive protein self-aggregation, contributing to Aβ plaque formation [233,234]. The accumulation of Aβ in AD follows a characteristic pattern, beginning with the neocortex [226,235]. As the disease progresses, Aβ accumulates in progressively deeper structures, ending with the brainstem [226,235]. Aβ deposition is thought to be the earliest pathology to develop in AD, preceding the onset of clinically observable symptoms by decades, though this hypothesis remains debated [236,237,238]. The Amyloid Hypothesis suggests Aβ deposition contributes to the development of later AD symptoms. However, the precise mechanisms underlying this catalysis remain to be determined, and the hypothesis has been serially revised with new findings.
Though plaque load does not correlate well with cognitive deficits, the amyloid cascade hypothesis suggests greater amyloid load eventually results in more significant disease. In humans, there is an association between antecedent attention-deficit/hyperactivity disorder and increased risk of dementia [239,240]. Within the striatum, female 5xFAD mice exhibit increased amyloid plaque associated with changes in dopamine signaling in the dorsal striatum, thereby resulting in hyperactivity of female but not male mice [241].
Some AD patients also exhibit cerebral amyloid angiopathy (CAA), which involves accumulation of amyloid protein in the leptomeningeal and cortical blood vessels. Interestingly, men have more severe CAA than women [242]. Perez et al. have recently shown that the transcription-regulating protein inhibitor of the DNA-binding protein 3 (ID3) is associated with CAA severity in women, while the nuclear respiratory factor 1 (NRF1) is associated with CAA severity in men [243], potentially providing new targets for personalized medicine and/or prevention strategies against CAA.
Secretion of Aβ via exosomes, which are small vesicles derived from the inward budding of intraluminal vesicles (ILVs) inside multiple vesicle bodies (MVBs; [244]), following its production through the series of enzymatic steps described above, promotes plaque formation in a variety of model systems (reviewed in [245]). Ceramides are integral components of cell membranes, and the sphingolipid ceramide neutral sphingomyelinase (nSMase) promotes the formation of exosomes [246,247]. In an AD mouse model, female but not male mice exhibit elevated ceramide and exosome levels and are uniquely sensitive to nSMase inhibition, which blocked exosome spreading and subsequent amyloid pathology and rescued cognition [248]. The reason that ceramide and exosome biogeneses have a more significant role in the amyloid pathology of females is unclear and remains to be determined.
Furthermore, stress modulates amyloid production and does so in sex-specific ways. Adult female APP/PS1 mice show significantly increased hippocampal Aβ, while males do not, due to differences in β-arrestin involvement in corticotropin-releasing factor receptor signaling pathways [249].
The relationship between hormones and amyloid levels is complex and seemingly contradictory. The primary estrogen, 17β-estradiol (E2), decreases Aβ production [250,251,252] and stimulates its degradation [253]. This would suggest that females, having higher concentrations of circulating E2, would have decreased amyloid burden compared to males, when in fact the opposite is true. Future work must tease apart the relative contributions of and the balance between estrogenic effects on amyloid production and clearance and its influence on other cellular processes that could in turn modulate those effects, such as neuronal survival and neuroinflammation.

5.2. Aβ Clearance

Early-onset forms of AD are thought to arise from the increased production of Aβ, while late-onset forms stem from reduced Aβ clearance [254,255]. This clearance occurs through both enzymatic and non-enzymatic processes (reviewed in [256]).
In the brain, Aβ is primarily degraded and cleared through the proteolytic machinery [257,258], and more than 20 different Aβ-degrading enzymes (ADEs) have been identified, including metallo-serine, aspartyl, cysteine, and threonine proteases [259]. To date, metalloproteases are the best studied ADEs, with matrix metalloprotease 2 (MMP2), MMP7, and MMP9 demonstrating Aβ-degrading activity and association with AD [256]. Tissue inhibitors of metalloproteases (TIMPs) regulate MMP activity [260] and have been linked to AD as well [137,261,262,263]. Notably, MMPs may have sex-specific mechanisms contributing to AD [264], potentially due to their interactions with estrogen [265]. Aksnes and colleagues found that amyloid pathology is associated with MMP3 in males but with TIMP4 in females [266]. Furthermore, there was a female-specific effect of MMP10 on cognitive and functional decline in AD patients, suggesting that MMPs and TIMPs could be useful biomarkers for sex differences and progression in AD [266].
Non-enzymatic clearance of Aβ occurs through a variety of pathways, including microglial phagocytosis (reviewed in [267]). As microglia age, they become senescent/dystrophic, exhibiting age-related changes in the expression of phagocytosis-related receptors and processes, limiting their capability to respond to pathogens or neurodegenerative processes [268]. Microglial aging is accompanied by increased phagocytosis of neuronal debris, with female microglia exhibiting a greater increase but a loss in ability to adapt its phagocytosis to inflammatory conditions [269]. A recent report found sST2, the secreted isoform of the interleukin-3 (IL-33) receptor, is a decoy receptor that inhibits microglial activation, increases in the blood and brain of females with AD, and is positively associated with disease progression [270]. Furthermore, women with APOE4 that have lower amounts of sST2 have a lower risk of AD [270]. The authors hypothesize that less sST2 facilitates microglial clearance of Aβ from the brain, thereby reducing AD.
Recently, two companion studies demonstrated that microglial-derived ApoE4 was detrimental to the microglial response to amyloid, and this was more pronounced in females [80,271]. ApoE is essential for seeding amyloid deposits [272,273,274] as well as being required for microglial conversion to a DAM/MgnD phenotype in the presence of amyloid [165,169]. Using different genetic and molecular approaches, these studies showed microglial ApoE3 induced microglial DAM/MGnD genes for plaque encapsulation and clearance, while microglial ApoE4 induced ITGB8-TGFβ signaling that impaired this DAM/MGnD response, thereby exacerbating amyloid pathology [80,271]. In both studies, female AD patients were more strongly affected by the presence of an APOE4 allele in these processes, though the mechanism behind this sex-specific effect was unclear.
Together, recent findings summarized in Figure 3 suggest sex differences in microglial inflammatory response mechanisms may contribute to distinct inflammatory profiles, thereby differentially stimulating amyloid production and/or modulating its clearance. ApoE is likely to function in many of these processes, participating not only in amyloid clearance and plaque seeding directly but also in the acquisition of microglial phenotypes that facilitate or inhibit amyloid production and clearance. Future work will need to elucidate mechanisms contributing to sex differences in microglial responses generally and ApoE4 sensitivity specifically, as they could provide avenues for more targeted interventions and therapies.

6. Sex Differences in Tau Pathology

The third pathological hallmark of AD is intraneuronal tangles composed of the hyperphosphorylated microtubule-associated protein, tau. Unlike amyloid plaques, tau pathology is well correlated with brain atrophy and cognitive decline [275,276,277].

6.1. Intracellular Tau Tangle Production

Microtubule Associated Protein Tau (MAPT) encodes the cytoskeletal phosphoprotein tau, which encourages the formation of microtubules from tubulin and is normally found in mature neurons [278]. The transcription of MAPT can result in six different tau isoforms, with varying numbers of microtubule-binding repeats and amino-binding terminal inserts [278]. Tau shows highly variable structural confirmations, only exhibiting conformational consistency in β pleating between microtubule-binding repeats [278]. Under normal physiological conditions, the ratio of tubulin to tau in mature neurons is approximately 10:1. With tau’s high binding affinity to tubulin, little tau remains unbound in healthy adult cells [278]. As aforementioned, tau is a phosphoprotein, with over 40 different sites where phosphorylation may occur [279]. The normal concentration of bound phosphate relative to the tau protein is 2–3 mol phosphate/1 mol of protein; however, in AD patients, tau exhibits a 3–4-fold increase in bound phosphate concentration. This inhibits protein function and induces tau aggregation, leading to the formation of neurofibrillary tangles (NFTs; [278,280,281]). Although NFTs may develop in AD patients, most of the hyperphosphorylated tau (hTau) generated remains in the cytosol, where it sequesters functional tau proteins and disrupts the formation of microtubules [278]. Both NFTs and cytosolic hTau disrupt normal cell function and lead to cognitive decline; however, when hTau is dephosphorylated, it returns to a normal, functional state, supporting the creation of microtubules [278,282].
As in neuronal loss and amyloid burden, sexual dimorphism is evident in tau deposition and tau-related clinical progression, with women being more strongly affected [12,38,283,284,285,286,287,288]. Higher tau levels occur in women, particularly in the context of elevated Aβ and especially in the context of an earlier age of menopause and late initiation of hormone replacement therapy (HRT; [289]).

6.2. Microglial Responses to Tau

Despite tau being a structural neuronal protein and contributing to intraneuronal neurofibrillary tangles, it propagates from neuron to neuron, where it can activate microglia (reviewed in [290]). Tau activates a variety of signaling cascades in microglia, including the Toll-like Receptor 4 (TLR4)- NOD-, LRR- and pyrin domain-containing 3 (NLRP3)-caspase 1 cascade for phagocytosis of living neurons [291]. Additionally, the cyclic GMP-AMP synthase (cGAS)-IFN signaling pathway suppresses MEF2C neuronal transcriptional networks to attenuate cognitive resilience [292]. Microglial NFκB signaling also drives tau spreading and toxicity [293]. The kinase Tumor progression locus 2 (TPL2; [294]) and insulin-like growth factor-binding protein like protein 1 (IGFBPL1; [295]) function as master regulators of the microglial inflammatory responses to tauopathy. Sex-specific microglial signaling pathways and miRNAs contribute to differences in the microglial transcriptome, perturbations in lipid metabolism and lipid-droplet accumulation, and differential recruitment of T cells, all of which may contribute to sex-disparate tau pathology [130,296,297].

6.3. Reversal/Removal of Pathological Tau

Though tau pathology is reversible experimentally, clinical trials of therapies targeting tau pathology have not yet yielded positive results, though candidate compounds continue to make their way through the clinical trial pipeline [298,299]. Further, tau pathology is intimately linked with the presence of Aβ plaques in AD, with Aβ clearance often aiding in the clearance of hTau in the early stages of the disease [300,301]. The mechanism underlying this close pathological relationship remains poorly understood, although some hypotheses point toward the proteasome [300,301,302].
Tau clearance occurs through the ubiquitin–proteasome system and the autophagy-lysosome pathway, with the final modification being ubiquitination [303,304,305]. The ubiquitin-specific peptidase 11 (USP11) deubiquitinates tau, augmenting its aggregation [306]. USP11 is an X-linked gene that escapes complete XCI, resulting in elevated USP11 expression in females, correlating strongly with tau pathology in females but not males; therefore, it potentially underpins heightened disease susceptibility in women [306].
There has been a significant shift in focus away from amyloid to tau pathology. The focus on amyloid has been called into question due to failure of anti-amyloid therapeutics in the clinic, and the appreciation for high correlation between tau levels and cognitive decline. Recent work has illuminated the response of microglia to tau and has begun to demonstrate sex-specific microglial responses, potentially contributing to distinct disease manifestations. Many questions remain to be answered, most notably the role of APOE4. As in amyloid pathology, APOE4 from neurons and astrocytes play important roles in the tau-mediated gliosis and neurodegeneration [307,308]. What remains to be determined is whether these effects are more acute in females compared to males, as in amyloid pathology.

7. Conclusions and Future Directions

In the century following Dr Alzheimer’s encounter with Auguste Deter, there has been substantial progress in understanding the basic mechanisms of the three major pathological hallmarks of AD: neuron loss, extracellular amyloid plaque deposits, and intracellular tau tangles. Furthermore, inflammation mediated by the innate immune cells of the brain, the microglia, is understood to be a key influence on those hallmark pathologies. While the disparate impact of AD on women has long been widely appreciated, the reasons for sex-specific disease onset and progression remain elusive. Early work focused on the neuroprotective and anti-inflammatory aspects of hormones. More recently, genetic effects driven by sex chromosomes and epigenetics have been shown to play a role in neuronal resilience and activity, and microglial-mediated amyloid and tau clearance mechanisms. Despite these advances, much work remains to be done, both in understanding sex-specific mechanisms and in leveraging these findings for a personalized approach to preventing and treating AD.
The mechanisms behind the increased sensitivity of women to the APOE4 isoform remain a significant gap in our understanding. This has major implications on disease, as APOE has such varied functions, from lipid transportation to amyloid clearance and microglial responses to amyloid and tau. The complex interplay of APOE isoforms and sex were highlighted in a recent paper demonstrating in mice that the ApoE-isoform and microbiota-dependent progression of tau pathology occurred in a sex-specific manner [309]. Whether targeting gut microbiota may provide a potential preventative or therapeutic approach to AD in one or both sexes remains to be determined.
Metabolism is another avenue ripe for future investigation. The AD brain exhibits metabolic dysregulation, leading to the Metabolic Reprogramming Theory of AD [310]. In human patients and in mouse models of the disease, there are sex differences in cerebrovascular and brain metabolism [311,312,313]. In fact, elevated brain metabolism in women is hypothesized to confer cognitive resilience against early-onset AD [36]. Furthermore, dietary fat is known to influence microglia [314], and a high-fat, high-salt diet induces sex-specific responses in the gut microbiome and in hypothalamic astrocytes and microglia [315]. Daily administration of an insulin-sensitizing compound ameliorated female-specific metabolic imbalances in a mouse model of AD [316]. Whether using drugs targeting genes associated with both lipid metabolism and neuroinflammation [317] will be efficacious in one or both sexes, if at all, remains to be determined.
Numerous other avenues for additional work exist and understanding them is required. AD, particularly late-onset sporadic forms, is incredibly heterogeneous and will require multiple approaches to effectively treat the increasing patient population. Some therapeutic targets will be consistent across groups of patients, such as ApoE, but how they are targeted and how effective they may be is likely to depend on sex and a variety of other intersecting factors.

Author Contributions

Conceptualization, E.G.R.; writing—original draft preparation, E.G.R. and P.R.K.-N.; writing—review and editing, E.G.R. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by the National Institutes of Health/National Institute on Aging (award number R01AG075897) and The BrightFocus Foundation (ADR number A2021036S) to E.G.R.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Acknowledgments

The authors thank Brad Casali and Hannah Zuppe, members of the laboratory, for their discussion and review of the manuscript.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Alzheimer’s Association. 2020 Alzheimer’s Disease Facts and Figures. Alzheimer’s Dement. 2020, 16, 391. [Google Scholar] [CrossRef] [PubMed]
  2. Sinforiani, E.; Citterio, A.; Zucchella, C.; Bono, G.; Corbetta, S.; Merlo, P.; Mauri, M. Impact of Gender Differences on the Outcome of Alzheimer’s Disease. Dement. Geriatr. Cogn. Disord. 2010, 30, 147–154. [Google Scholar] [CrossRef] [PubMed]
  3. Davis, E.J.; Broestl, L.; Abdulai-saiku, S.; Worden, K.; Bonham, L.W.; Miñones-moyano, E.; Moreno, A.J.; Wang, D.; Chang, K.; Williams, G.; et al. A Second X Chromosome Contributes to Resilience in a Mouse Model of Alzheimer’s Disease. Sci. Transl. Med. 2020, 12, eaaz5677. [Google Scholar] [CrossRef] [PubMed]
  4. Barnes, L.L.; Wilson, R.S.; Bienias, J.L.; Schneider, J.A.; Evans, D.A.; Bennett, D.A. Sex Differences in the Clinical Manifestations of Alzheimer Disease Pathology. Arch. Gen. Psychiatry 2005, 62, 685–691. [Google Scholar] [CrossRef]
  5. Benke, T.; Delazer, M.; Sanin, G.; Schmidt, H.; Seiler, S.; Ransmayr, G.; Dal-Bianco, P.; Uranüs, M.; Marksteiner, J.; Leblhuber, F.; et al. Cognition, Gender, and Functional Abilities in Alzheimer’s Disease: How Are They Related? J. Alzheimer’s Dis. 2013, 35, 247–252. [Google Scholar] [CrossRef] [PubMed]
  6. Holland, D.; Desikan, R.S.; Dale, A.M.; McEvoy, L.K. Higher Rates of Decline for Women and Apolipoprotein e Ε4 Carriers. Am. J. Neuroradiol. 2013, 34, 2287–2293. [Google Scholar] [CrossRef] [PubMed]
  7. Lin, K.A.; Choudhury, K.R.; Rathakrishnan, B.G.; Marks, D.M.; Petrella, J.R.; Doraiswamy, P.M. Marked Gender Differences in Progression of Mild Cognitive Impairment over 8 Years. Alzheimer’s Dement. 2015, 1, 103–110. [Google Scholar] [CrossRef]
  8. Pusswald, G.; Tropper, E.; Kryspin-Exner, I.; Moser, D.; Klug, S.; Auff, E.; Dal-Bianco, P.; Lehrner, J. Health-Related Quality of Life in Patients with Subjective Cognitive Decline and Mild Cognitive Impairment and Its Relation to Activities of Daily Living. J. Alzheimer’s Dis. 2015, 47, 479–486. [Google Scholar] [CrossRef]
  9. Tifratene, K.; Robert, P.; Metelkina, A.; Pradier, C.; Dartigues, J.F. Progression of Mild Cognitive Impairment to Dementia Due to AD in Clinical Settings. Neurology 2015, 85, 331–338. [Google Scholar] [CrossRef]
  10. Laws, K.R.; Irvine, K.; Gale, T.M. Sex Differences in Alzheimer’s Disease. Curr. Opin. Psychiatry 2018, 31, 133–139. [Google Scholar] [CrossRef]
  11. Gamberger, D.; Lavrač, N.; Srivatsa, S.; Tanzi, R.E.; Doraiswamy, P.M. Identification of Clusters of Rapid and Slow Decliners among Subjects at Risk for Alzheimer’s Disease. Sci. Rep. 2017, 7, 6763. [Google Scholar] [CrossRef] [PubMed]
  12. Koran, M.E.I.; Wagener, M.; Hohman, T.J. Sex Differences in the Association between AD Biomarkers and Cognitive Decline. Brain Imaging Behav. 2017, 11, 205–213. [Google Scholar] [CrossRef] [PubMed]
  13. Sohn, D.; Shpanskaya, K.; Lucas, J.E.; Petrella, J.R.; Saykin, A.J.; Tanzi, R.E.; Samatova, N.F.; Doraiswamy, P.M. Sex Differences in Cognitive Decline in Subjects with High Likelihood of Mild Cognitive Impairment Due to Alzheimer’s Disease. Sci. Rep. 2018, 8, 7490. [Google Scholar] [CrossRef] [PubMed]
  14. Bordt, E.A.; Ceasrine, A.M.; Bilbo, S.D. Microglia and Sexual Differentiation of the Developing Brain: A Focus on Ontogeny and Intrinsic Factors. Glia 2020, 68, 1085–1099. [Google Scholar] [CrossRef] [PubMed]
  15. Kodama, L.; Gan, L. Do Microglial Sex Differences Contribute to Sex Differences in Neurodegenerative Diseases? Trends Mol. Med. 2019, 25, 741–749. [Google Scholar] [CrossRef] [PubMed]
  16. VanRyzin, J.W.; Marquardt, A.E.; Pickett, L.A.; McCarthy, M.M. Microglia and Sexual Differentiation of the Developing Brain: A Focus on Extrinsic Factors. Glia 2020, 68, 1100–1113. [Google Scholar] [CrossRef] [PubMed]
  17. Han, J.; Fan, Y.; Zhou, K.; Blomgren, K.; Harris, R.A. Uncovering Sex Differences of Rodent Microglia. J. Neuroinflamm. 2021, 18, 74. [Google Scholar] [CrossRef]
  18. Bobotis, B.C.; Braniff, O.; Gargus, M.; Akinluyi, E.T.; Awogbindin, I.O.; Tremblay, M.È. Sex Differences of Microglia in the Healthy Brain from Embryonic Development to Adulthood and across Lifestyle Influences. Brain Res. Bull. 2023, 202, 110752. [Google Scholar] [CrossRef]
  19. Ocañas, S.R.; Ansere, V.A.; Kellogg, C.M.; Isola, J.V.V.; Chucair-Elliott, A.J.; Freeman, W.M. Chromosomal and Gonadal Factors Regulate Microglial Sex Effects in the Aging Brain. Brain Res. Bull. 2023, 195, 157–171. [Google Scholar] [CrossRef]
  20. Kadlecova, M.; Freude, K.; Haukedal, H. Complexity of Sex Differences and Their Impact on Alzheimer’s Disease. Biomedicines 2023, 11, 1261. [Google Scholar] [CrossRef]
  21. O’Connor, J.L.; Nissen, J.C. The Pathological Activation of Microglia Is Modulated by Sexually Dimorphic Pathways. Int. J. Mol. Sci. 2023, 24, 4739. [Google Scholar] [CrossRef] [PubMed]
  22. Lutshumba, J.; Wilcock, D.M.; Monson, N.L.; Stowe, A.M. Sex-Based Differences in Effector Cells of the Adaptive Immune System during Alzheimer’s Disease and Related Dementias. Neurobiol. Dis. 2023, 184, 106202. [Google Scholar] [CrossRef] [PubMed]
  23. Gygax, P.M.; Elmiger, D.; Zufferey, S.; Garnham, A.; Sczesny, S.; von Stockhausen, L.; Braun, F.; Oakhill, J. A Language Index of Grammatical Gender Dimensions to Study the Impact of Grammatical Gender on the Way We Perceive Women and Men. Front. Psychol. 2019, 10, 1604. [Google Scholar] [CrossRef] [PubMed]
  24. Eliot, L.; Beery, A.K.; Jacobs, E.G.; LeBlanc, H.F.; Maney, D.L.; McCarthy, M.M. Why and How to Account for Sex and Gender in Brain and Behavioral Research. J. Neurosci. 2023, 43, 6344–6356. [Google Scholar] [CrossRef] [PubMed]
  25. Dahm, R. Alzheimer’s Discovery. Curr. Biol. 2006, 16, R906–R910. [Google Scholar] [CrossRef]
  26. Jack, C.R.; Albert, M.S.; Knopman, D.S.; McKhann, G.M.; Sperling, R.A.; Carrillo, M.C.; Thies, B.; Phelps, C.H. Introduction to the Recommendations from the National Institute on Aging-Alzheimer’s Association Workgroups on Diagnostic Guidelines for Alzheimer’s Disease. Alzheimer’s Dement. 2011, 7, 257–262. [Google Scholar] [CrossRef]
  27. Bird, T.D. Genetic Aspects of Alzheimer Disease. Genet. Med. 2008, 10, 231–239. [Google Scholar] [CrossRef]
  28. Cacace, R.; Sleegers, K.; Van Broeckhoven, C. Molecular Genetics of Early-Onset Alzheimer’s Disease Revisited. Alzheimer’s Dement. 2016, 12, 733–748. [Google Scholar] [CrossRef]
  29. Cho, H.; Mundada, N.S.; Apostolova, L.G.; Carrillo, M.C.; Shankar, R.; Amuiri, A.N.; Zeltzer, E.; Windon, C.C.; Soleimani-Meigooni, D.N.; Tanner, J.A.; et al. Amyloid and Tau-PET in Early-Onset AD: Baseline Data from the Longitudinal Early-Onset Alzheimer’s Disease Study (LEADS). Alzheimer’s Dement. 2023, 19, S98–S114. [Google Scholar] [CrossRef]
  30. Contador, J.; Pérez-Millan, A.; Guillén, N.; Sarto, J.; Tort-Merino, A.; Balasa, M.; Falgàs, N.; Castellví, M.; Borrego-Écija, S.; Juncà-Parella, J.; et al. Sex Differences in Early-Onset Alzheimer’s Disease. Eur. J. Neurol. 2022, 29, 3623–3632. [Google Scholar] [CrossRef]
  31. Vila-Castelar, C.; Chen, Y.; Langella, S.; Lopera, F.; Zetterberg, H.; Hansson, O.; Dage, J.L.; Janelidzde, S.; Su, Y.; Chen, K.; et al. Sex Differences in Blood Biomarkers and Cognitive Performance in Individuals with Autosomal Dominant Alzheimer’s Disease. Alzheimer’s Dement. 2023, 19, 4127–4138. [Google Scholar] [CrossRef] [PubMed]
  32. Wagemann, O.; Li, Y.; Hassenstab, J.; Aschenbrenner, A.J.; McKay, N.S.; Gordon, B.A.; Benzinger, T.L.S.; Xiong, C.; Cruchaga, C.; Renton, A.E.; et al. Investigation of Sex Differences in Mutation Carriers of the Dominantly Inherited Alzheimer Network. Alzheimer’s Dement. 2023. [Google Scholar] [CrossRef] [PubMed]
  33. Vila-Castelar, C.; Tariot, P.N.; Sink, K.M.; Clayton, D.; Langbaum, J.B.; Thomas, R.G.; Chen, Y.; Su, Y.; Chen, K.; Hu, N.; et al. Sex Differences in Cognitive Resilience in Preclinical Autosomal-Dominant Alzheimer’s Disease Carriers and Non-Carriers: Baseline Findings from the API ADAD Colombia Trial. Alzheimer’s Dement. 2022, 18, 2272–2282. [Google Scholar] [CrossRef] [PubMed]
  34. Ramanan, V.K.; Castillo, A.M.; Knopman, D.S.; Graff-Radford, J.; Lowe, V.J.; Petersen, R.C.; Jack, C.R.; Mielke, M.M.; Vemuri, P. Association of Apolipoprotein E ε4, Educational Level, and Sex with Tau Deposition and Tau-Mediated Metabolic Dysfunction in Older Adults. JAMA Netw. Open 2019, 2, e1913909. [Google Scholar] [CrossRef] [PubMed]
  35. Sundermann, E.E.; Maki, P.M.; Rubin, L.H.; Lipton, R.B.; Landau, S.; Biegon, A. Female Advantage in Verbal Memory: Evidence of Sex-Specific Cognitive Reserve. Neurology 2016, 87, 1916–1924. [Google Scholar] [CrossRef] [PubMed]
  36. Sundermann, E.E.; Maki, P.M.; Reddy, S.; Bondi, M.W.; Biegon, A. Women’s Higher Brain Metabolic Rate Compensates for Early Alzheimer’s Pathology. Alzheimer’s Dement. 2020, 12, e12121. [Google Scholar] [CrossRef] [PubMed]
  37. Buckley, R.F.; Mormino, E.C.; Amariglio, R.E.; Properzi, M.J.; Rabin, J.S.; Lim, Y.Y.; Papp, K.V.; Jacobs, H.I.L.; Burnham, S.; Hanseeuw, B.J.; et al. Sex, Amyloid, and APOE Ε4 and Risk of Cognitive Decline in Preclinical Alzheimer’s Disease: Findings from Three Well-Characterized Cohorts. Alzheimer’s Dement. 2018, 14, 1193–1203. [Google Scholar] [CrossRef]
  38. Buckley, R.F.; Scott, M.R.; Jacobs, H.I.L.; Schultz, A.P.; Properzi, M.J.; Amariglio, R.E.; Hohman, T.J.; Mayblyum, D.V.; Rubinstein, Z.B.; Manning, L.; et al. Sex Mediates Relationships Between Regional Tau Pathology and Cognitive Decline. Ann. Neurol. 2020, 88, 921–932. [Google Scholar] [CrossRef]
  39. Digma, L.A.; Madsen, J.R.; Rissman, R.A.; Jacobs, D.M.; Brewer, J.B.; Banks, S.J. Women Can Bear a Bigger Burden: Ante- and Post-Mortem Evidence for Reserve in the Face of Tau. Brain Commun. 2020, 2, fcaa025. [Google Scholar] [CrossRef]
  40. Alzheimer’s Disease International. World Alzheimer Report 2009: The Global Prevalence of Dementia; Alzheimer’s Disease International: London, UK, 2009. [Google Scholar]
  41. Wingo, T.S.; Lah, J.J.; Levey, A.I.; Cutler, D.J. Autosomal Recessive Causes Likely in Early-Onset Alzheimer Disease. Arch. Neurol. 2012, 69, 59–64. [Google Scholar] [CrossRef]
  42. Campion, D.; Dumanchin, C.; Hannequin, D.; Dubois, B.; Belliard, S.; Puel, M.; Thomas-Anterion, C.; Michon, A.; Martin, C.; Charbonnier, F.; et al. Early-Onset Autosomal Dominant Alzheimer Disease: Prevalence, Genetic Heterogeneity, and Mutation Spectrum. Am. J. Hum. Genet. 1999, 65, 664–670. [Google Scholar] [CrossRef] [PubMed]
  43. Jarmolowicz, A.I.; Chen, H.Y.; Panegyres, P.K. The Patterns of Inheritance in Early-Onset Dementia: Alzheimer’s Disease and Frontotemporal Dementia. Am. J. Alzheimer’s Dis. Other Dement. 2015, 30, 299–306. [Google Scholar] [CrossRef] [PubMed]
  44. Brouwers, N.; Sleegers, K.; Van Broeckhoven, C. Molecular Genetics of Alzheimer’s Disease: An Update. Ann. Med. 2008, 40, 562–583. [Google Scholar] [CrossRef] [PubMed]
  45. Ding, S.L. Comparative Anatomy of the Prosubiculum, Subiculum, Presubiculum, Postsubiculum, and Parasubiculum in Human, Monkey, and Rodent. J. Comp. Neurol. 2013, 521, 4145–4162. [Google Scholar] [CrossRef] [PubMed]
  46. Insausti, R.; Muñoz-López, M.; Insausti, A.M.; Artacho-Pérula, E. The Human Periallocortex: Layer Pattern in Presubiculum, Parasubiculum and Entorhinal Cortex. A Review. Front. Neuroanat. 2017, 11, 84. [Google Scholar] [CrossRef] [PubMed]
  47. Davis, E.J.; Solsberg, C.W.; White, C.C.; Miñones-Moyano, E.; Sirota, M.; Chibnik, L.; Bennett, D.A.; De Jager, P.L.; Yokoyama, J.S.; Dubal, D.B. Sex-Specific Association of the X Chromosome with Cognitive Change and Tau Pathology in Aging and Alzheimer Disease. JAMA Neurol. 2021, 78, 1249–1254. [Google Scholar] [CrossRef] [PubMed]
  48. Onos, K.D.; Sukoff Rizzo, S.J.; Howell, G.R.; Sasner, M. Toward More Predictive Genetic Mouse Models of Alzheimer’s Disease. Brain Res. Bull. 2016, 122, 1–11. [Google Scholar] [CrossRef] [PubMed]
  49. Esquerda-Canals, G.; Montoliu-Gaya, L.; Güell-Bosch, J.; Villegas, S. Mouse Models of Alzheimer’s Disease. J. Alzheimer’s Dis. 2017, 57, 1171–1183. [Google Scholar] [CrossRef]
  50. Sadleir, K.R.; Eimer, W.A.; Cole, S.L.; Vassar, R. Aβ Reduction in BACE1 Heterozygous Null 5XFAD Mice Is Associated with Transgenic APP Level. Mol. Neurodegener. 2015, 10, 1. [Google Scholar] [CrossRef]
  51. Bundy, J.L.; Vied, C.; Badger, C.; Nowakowski, R.S. Sex-Biased Hippocampal Pathology in the 5XFAD Mouse Model of Alzheimer’s Disease: A Multi-Omic Analysis. J. Comp. Neurol. 2019, 527, 462–475. [Google Scholar] [CrossRef]
  52. Gatz, M.; Reynolds, C.A.; Fratiglioni, L.; Johansson, B.; Mortimer, J.A.; Berg, S.; Fiske, A.; Pedersen, N.L. Role of Genes and Environments for Explaining Alzheimer Disease. Arch. Gen. Psychiatry 2006, 63, 168–174. [Google Scholar] [CrossRef] [PubMed]
  53. Jarvik, G.P.; Larson, E.B.; Goddard, K.; Kukull, W.A.; Schellenberg, G.D.; Wijsman, E.M. Influence of Apolipoprotein E Genotype on the Transmission of Alzheimer Disease in a Community-Based Sample. Am. J. Hum. Genet. 1996, 58, 191. [Google Scholar] [PubMed]
  54. Sims, R.; Hill, M.; Williams, J. The Multiplex Model of the Genetics of Alzheimer’s Disease. Nat. Neurosci. 2020, 23, 311–322. [Google Scholar] [CrossRef] [PubMed]
  55. Karch, C.M.; Goate, A.M. Alzheimer’s Disease Risk Genes and Mechanisms of Disease Pathogenesis. Biol. Psychiatry 2015, 77, 43–51. [Google Scholar] [CrossRef] [PubMed]
  56. Guerreiro, R.; Wojtas, A.; Bras, J.; Carrasquillo, M.; Rogaeva, E.; Majounie, E.; Cruchaga, C.; Sassi, C.; Kauwe, J.S.K.; Younkin, S.; et al. TREM2 Variants in Alzheimer’s Disease. N. Engl. J. Med. 2013, 368, 117–127. [Google Scholar] [CrossRef] [PubMed]
  57. Jonsson, T.; Stefansson, H.; Steinberg, S.; Jonsdottir, I.; Jonsson, P.V.; Snaedal, J.; Bjornsson, S.; Huttenlocher, J.; Levey, A.I.; Lah, J.J.; et al. Variant of TREM2 Associated with the Risk of Alzheimer’s Disease. N. Engl. J. Med. 2013, 368, 107–116. [Google Scholar] [CrossRef] [PubMed]
  58. Irvine, K.; Laws, K.R.; Gale, T.M.; Kondel, T.K. Greater Cognitive Deterioration in Women than Men with Alzheimer’s Disease: A Meta Analysis. J. Clin. Exp. Neuropsychol. 2012, 34, 989–998. [Google Scholar] [CrossRef]
  59. Neu, S.C.; Pa, J.; Kukull, W.; Beekly, D.; Kuzma, A.; Gangadharan, P.; Wang, L.S.; Romero, K.; Arneric, S.P.; Redolfi, A.; et al. Apolipoprotein E Genotype and Sex Risk Factors for Alzheimer’s Disease. JAMA Neurol. 2017, 74, 1178–1189. [Google Scholar] [CrossRef]
  60. Dumitrescu, L.; Barnes, L.L.; Thambisetty, M.; Beecham, G.; Kunkle, B.; Bush, W.S.; Gifford, K.A.; Chibnik, L.B.; Mukherjee, S.; de Jager, P.L.; et al. Sex Differences in the Genetic Predictors of Alzheimer’s Pathology. Brain 2019, 142, 2581–2589. [Google Scholar] [CrossRef]
  61. Dumitrescu, L.; Mayeda, E.R.; Sharman, K.; Moore, A.M.; Hohman, T.J. Sex Differences in the Genetic Architecture of Alzheimer’s Disease. Curr. Genet. Med. Rep. 2019, 7, 13–21. [Google Scholar] [CrossRef]
  62. Fisher, D.W.; Bennett, D.A.; Dong, H. Sexual Dimorphism in Predisposition to Alzheimer’s Disease. Neurobiol. Aging 2018, 70, 308–324. [Google Scholar] [CrossRef] [PubMed]
  63. Kautzky-Willer, A.; Harreiter, J.; Pacini, G. Sex and Gender Differences in Risk, Pathophysiology and Complications of Type 2 Diabetes Mellitus. Endocr. Rev. 2016, 37, 278–316. [Google Scholar] [CrossRef] [PubMed]
  64. Pike, C.J.; Carroll, J.C.; Rosario, E.R.; Barron, A.M. Protective Actions of Sex Steroid Hormones in Alzheimer’s Disease. Front. Neuroendocrinol. 2009, 30, 239–258. [Google Scholar] [CrossRef] [PubMed]
  65. Zhang, L.; Chen, C.; Mak, M.S.H.; Lu, J.; Wu, Z.; Chen, Q.; Han, Y.; Li, Y.; Pi, R. Advance of Sporadic Alzheimer’s Disease Animal Models. Med. Res. Rev. 2020, 40, 431–458. [Google Scholar] [CrossRef]
  66. Nardini, E.; Hogan, R.; Flamier, A.; Bernier, G. Alzheimer’s Disease: A Tale of Two Diseases? Neural Regen. Res. 2021, 16, 1958–1964. [Google Scholar]
  67. Kim, K.H.; Moon, M.; Yu, S.B.; Mook-Jung, I.; Kim, J. Il RNA-Seq Analysis of Frontal Cortex and Cerebellum from 5XFAD Mice at Early Stage of Disease Pathology. J. Alzheimer’s Dis. 2012, 29, 793–808. [Google Scholar] [CrossRef]
  68. Guo, L.; Zhong, M.B.; Zhang, L.; Zhang, B.; Cai, D. Sex Differences in Alzheimer’s Disease: Insights from the Multiomics Landscape. Biol. Psychiatry 2022, 91, 61–71. [Google Scholar] [CrossRef]
  69. Zhang, L.; Young, J.I.; Gomez, L.; Silva, T.C.; Schmidt, M.A.; Cai, J.; Chen, X.; Martin, E.R.; Wang, L. Sex-Specific DNA Methylation Differences in Alzheimer’s Disease Pathology. Acta Neuropathol. Commun. 2021, 9, 77. [Google Scholar] [CrossRef]
  70. Ravanelli, F.; Musazzi, L.; Barbieri, S.S.; Rovati, G.; Popoli, M.; Barbon, A.; Ieraci, A. Differential Epigenetic Changes in the Dorsal Hippocampus of Male and Female SAMP8 Mice: A Preliminary Study. Int. J. Mol. Sci. 2023, 24, 13084. [Google Scholar] [CrossRef]
  71. Sun, L.L.; Yang, S.L.; Sun, H.; Li, W.D.; Duan, S.R. Molecular Differences in Alzheimer’s Disease between Male and Female Patients Determined by Integrative Network Analysis. J. Cell. Mol. Med. 2019, 23, 47–58. [Google Scholar] [CrossRef]
  72. Guo, L.; Cao, J.; Hou, J.; Li, Y.; Huang, M.; Zhu, L.; Zhang, L.; Lee, Y.; Duarte, M.L.; Zhou, X.; et al. Sex Specific Molecular Networks and Key Drivers of Alzheimer’s Disease. Mol. Neurodegener. 2023, 18, 39. [Google Scholar] [CrossRef] [PubMed]
  73. Sun, Y.Y.; Wang, Z.; Huang, H.C. Roles of ApoE4 on the Pathogenesis in Alzheimer’s Disease and the Potential Therapeutic Approaches. Cell. Mol. Neurobiol. 2023, 43, 3115–3136. [Google Scholar] [CrossRef] [PubMed]
  74. Windham, I.A.; Cohen, S. The Cell Biology of APOE in the Brain Cell Biology. Trends Cell. Biol. 2023. [Google Scholar] [CrossRef] [PubMed]
  75. Palmer, J.M.; Huentelman, M.; Ryan, L. More than Just Risk for Alzheimer’s Disease: APOE Ε4′s Impact on the Aging Brain. Trends Neurosci. 2023, 46, 750–763. [Google Scholar] [CrossRef]
  76. Walters, S.; Contreras, A.G.; Eissman, J.M.; Mukherjee, S.; Lee, M.L.; Choi, S.E.; Scollard, P.; Trittschuh, E.H.; Mez, J.B.; Bush, W.S.; et al. Associations of Sex, Race, and Apolipoprotein E Alleles with Multiple Domains of Cognition among Older Adults. JAMA Neurol. 2023, 80, 929–939. [Google Scholar] [CrossRef] [PubMed]
  77. Morikawa, M.; Fryer, J.D.; Sullivan, P.M.; Christopher, E.A.; Wahrle, S.E.; DeMattos, R.B.; O’Dell, M.A.; Fagan, A.M.; Lashuel, H.A.; Walz, T.; et al. Production and Characterization of Astrocyte-Derived Human Apolipoprotein E Isoforms from Immortalized Astrocytes and Their Interactions with Amyloid-β. Neurobiol. Dis. 2005, 19, 66–76. [Google Scholar] [CrossRef]
  78. Parhizkar, S.; Arzberger, T.; Brendel, M.; Kleinberger, G.; Deussing, M.; Focke, C.; Nuscher, B.; Xiong, M.; Ghasemigharagoz, A.; Katzmarski, N.; et al. Loss of TREM2 Function Increases Amyloid Seeding but Reduces Plaque-Associated ApoE. Nat. Neurosci. 2019, 22, 191–204. [Google Scholar] [CrossRef]
  79. Spangenberg, E.; Severson, P.L.; Hohsfield, L.A.; Crapser, J.; Zhang, J.; Burton, E.A.; Zhang, Y.; Spevak, W.; Lin, J.; Phan, N.Y.; et al. Sustained Microglial Depletion with CSF1R Inhibitor Impairs Parenchymal Plaque Development in an Alzheimer’s Disease Model. Nat. Commun. 2019, 10, 3758. [Google Scholar] [CrossRef]
  80. Yin, Z.; Rosenzweig, N.; Kleemann, K.L.; Zhang, X.; Brandão, W.; Margeta, M.A.; Schroeder, C.; Sivanathan, K.N.; Silveira, S.; Gauthier, C.; et al. APOE4 Impairs the Microglial Response in Alzheimer’s Disease by Inducing TGFβ-Mediated Checkpoints. Nat. Immunol. 2023, 24, 1839–1853. [Google Scholar] [CrossRef]
  81. Liu, C.C.; Zhao, J.; Fu, Y.; Inoue, Y.; Ren, Y.; Chen, Y.; Doss, S.V.; Shue, F.; Jeevaratnam, S.; Bastea, L.; et al. Peripheral ApoE4 Enhances Alzheimer’s Pathology and Impairs Cognition by Compromising Cerebrovascular Function. Nat. Neurosci. 2022, 25, 1020–1033. [Google Scholar] [CrossRef]
  82. Altmann, A.; Tian, L.; Henderson, V.W.; Greicius, M.D. Sex Modifies the APOE-Related Risk of Developing Alzheimer Disease. Ann. Neurol. 2014, 75, 563–573. [Google Scholar] [CrossRef]
  83. Hohman, T.J.; Dumitrescu, L.; Barnes, L.L.; Thambisetty, M.; Beecham, G.; Kunkle, B.; Gifford, K.A.; Bush, W.S.; Chibnik, L.B.; Mukherjee, S.; et al. Sex-Specific Association of Apolipoprotein e with Cerebrospinal Fluid Levels of Tau. JAMA Neurol. 2018, 75, 989–998. [Google Scholar] [CrossRef] [PubMed]
  84. Hsu, M.; Dedhia, M.; Crusio, W.E.; Delprato, A. Open Peer Review Sex Differences in Gene Expression Patterns Associated with the Allele APOE4 [Version 2; Peer Review: 2 Approved]. F1000Research 2019, 8, 387. [Google Scholar] [CrossRef] [PubMed]
  85. Shang, Y.; Mishra, A.; Wang, T.; Wang, Y.; Desai, M.; Chen, S.; Mao, Z.; Do, L.; Bernstein, A.S.; Trouard, T.P.; et al. Evidence in Support of Chromosomal Sex Influencing Plasma Based Metabolome vs APOE Genotype Influencing Brain Metabolome Profile in Humanized APOE Male and Female Mice. PLoS ONE 2020, 15, e0025302. [Google Scholar] [CrossRef] [PubMed]
  86. Rhea, E.M.; Hansen, K.; Pemberton, S.; Torres, E.R.S.; Holden, S.; Raber, J.; Banks, W.A. Effects of Apolipoprotein E Isoform, Sex, and Diet on Insulin BBB Pharmacokinetics in Mice. Sci. Rep. 2021, 11, 18636. [Google Scholar] [CrossRef] [PubMed]
  87. Cacciottolo, M.; Morgan, T.E.; Finch, C.E. Age, Sex, and Cerebral Microbleeds in EFAD Alzheimer Disease Mice. Neurobiol. Aging 2021, 103, 42–51. [Google Scholar] [CrossRef] [PubMed]
  88. Stephen, T.L.; Cacciottolo, M.; Balu, D.; Morgan, T.E.; Ladu, M.J.; Finch, C.E.; Pike, C.J. APOE Genotype and Sex Affect Microglial Interactions with Plaques in Alzheimer’s Disease Mice. Acta Neuropathol. Commun. 2019, 7, 82. [Google Scholar] [CrossRef]
  89. Taxier, L.R.; Philippi, S.M.; York, J.M.; Ladu, M.J.; Frick, K.M. The Detrimental Effects of APOE4 on Risk for Alzheimer’s Disease May Result from Altered Dendritic Spine Density, Synaptic Proteins, and Estrogen Receptor Alpha. Neurobiol. Aging 2022, 112, 74–86. [Google Scholar] [CrossRef]
  90. Menger, Y.; Bettscheider, M.; Murgatroyd, C.; Spengler, D. Sex Differences in Brain Epigenetics. Epigenomics 2010, 2, 807–821. [Google Scholar] [CrossRef]
  91. Dunn, G.A.; Morgan, C.P.; Bale, T.L. Sex-Specificity in Transgenerational Epigenetic Programming. Horm. Behav. 2011, 59, 290–295. [Google Scholar] [CrossRef]
  92. Akbarian, S.; Beeri, M.S.; Haroutunian, V. Epigenetic Determinants of Healthy and Diseased Brain Aging and Cognition. JAMA Neurol. 2013, 70, 711–718. [Google Scholar] [CrossRef] [PubMed]
  93. Xu, H.; Wang, F.; Liu, Y.; Yu, Y.; Gelernter, J.; Zhang, H. Sex-Biased Methylome and Transcriptome in Human Prefrontal Cortex. Hum. Mol. Genet. 2014, 23, 1260–1270. [Google Scholar] [CrossRef] [PubMed]
  94. De Jager, P.L.; Srivastava, G.; Lunnon, K.; Burgess, J.; Schalkwyk, L.C.; Yu, L.; Eaton, M.L.; Keenan, B.T.; Ernst, J.; McCabe, C.; et al. Alzheimer’s Disease: Early Alterations in Brain DNA Methylation at ANK1, BIN1, RHBDF2 and Other Loci. Nat. Neurosci. 2014, 17, 1156–1163. [Google Scholar] [CrossRef] [PubMed]
  95. Lunnon, K.; Smith, R.; Hannon, E.; De Jager, P.L.; Srivastava, G.; Volta, M.; Troakes, C.; Al-Sarraj, S.; Burrage, J.; Macdonald, R.; et al. Cross-Tissue Methylomic Profiling Strongly Implicates a Role for Cortex-Specific Deregulation of ANK1 in Alzheimer’s Disease Neuropathology. Nat. Neurosci. 2014, 17, 1164–1170. [Google Scholar] [CrossRef]
  96. McCarthy, M.M.; Nugent, B.M. At the Frontier of Epigenetics of Brain Sex Differences. Front. Behav. Neurosci. 2015, 9, 221. [Google Scholar] [CrossRef]
  97. Berson, A.; Nativio, R.; Berger, S.L.; Bonini, N.M. Epigenetic Regulation in Neurodegenerative Diseases. Trends Neurosci. 2018, 41, 587–598. [Google Scholar] [CrossRef]
  98. Gasparoni, G.; Bultmann, S.; Lutsik, P.; Kraus, T.F.J.; Sordon, S.; Vlcek, J.; Dietinger, V.; Steinmaurer, M.; Haider, M.; Mulholland, C.B.; et al. DNA Methylation Analysis on Purified Neurons and Glia Dissects Age and Alzheimer’s Disease-Specific Changes in the Human Cortex. Epigenet. Chromatin 2018, 11, 41. [Google Scholar] [CrossRef]
  99. Smith, R.G.; Hannon, E.; De Jager, P.L.; Chibnik, L.; Lott, S.J.; Condliffe, D.; Smith, A.R.; Haroutunian, V.; Troakes, C.; Al-Sarraj, S.; et al. Elevated DNA Methylation across a 48-Kb Region Spanning the HOXA Gene Cluster Is Associated with Alzheimer’s Disease Neuropathology. Alzheimer’s Dement. 2018, 14, 1580–1588. [Google Scholar] [CrossRef]
  100. Zhang, L.; Silva, T.C.; Young, J.I.; Gomez, L.; Schmidt, M.A.; Hamilton-Nelson, K.L.; Kunkle, B.W.; Chen, X.; Martin, E.R.; Wang, L. Epigenome-Wide Meta-Analysis of DNA Methylation Differences in Prefrontal Cortex Implicates the Immune Processes in Alzheimer’s Disease. Nat. Commun. 2020, 11, 6114. [Google Scholar] [CrossRef]
  101. Handy, D.E.; Castro, R.; Loscalzo, J. Epigenetic Modifications: Basic Mechanisms and Role in Cardiovascular Disease. Circulation 2011, 123, 2145–2156. [Google Scholar] [CrossRef]
  102. Frost, B.; Hemberg, M.; Lewis, J.; Feany, M.B. Tau Promotes Neurodegeneration through Global Chromatin Relaxation. Nat. Neurosci. 2014, 17, 357–366. [Google Scholar] [CrossRef] [PubMed]
  103. Mansuroglu, Z.; Benhelli-Mokrani, H.; Marcato, V.; Sultan, A.; Violet, M.; Chauderlier, A.; Delattre, L.; Loyens, A.; Talahari, S.; Bégard, S.; et al. Loss of Tau Protein Affects the Structure, Transcription and Repair of Neuronal Pericentromeric Heterochromatin. Sci. Rep. 2016, 6, 33047. [Google Scholar] [CrossRef] [PubMed]
  104. Lacal, I.; Ventura, R. Epigenetic Inheritance: Concepts, Mechanisms and Perspectives. Front. Mol. Neurosci. 2018, 11, 292. [Google Scholar] [CrossRef] [PubMed]
  105. Klein, H.U.; McCabe, C.; Gjoneska, E.; Sullivan, S.E.; Kaskow, B.J.; Tang, A.; Smith, R.V.; Xu, J.; Pfenning, A.R.; Bernstein, B.E.; et al. Epigenome-Wide Study Uncovers Large-Scale Changes in Histone Acetylation Driven by Tau Pathology in the Aging and Alzheimer Human Brain. Nat. Neurosci. 2019, 22, 37–46. [Google Scholar] [CrossRef]
  106. Griñán-Ferré, C.; Corpas, R.; Puigoriol-Illamola, D.; Palomera-Ávalos, V.; Sanfeliu, C.; Pallàs, M. Understanding Epigenetics in the Neurodegeneration of Alzheimer’s Disease: SAMP8 Mouse Model. J. Alzheimer’s Dis. 2018, 62, 943–963. [Google Scholar] [CrossRef]
  107. Scheiblich, H.; Trombly, M.; Ramirez, A.; Heneka, M.T. Neuroimmune Connections in Aging and Neurodegenerative Diseases. Trends Immunol. 2020, 41, 300–312. [Google Scholar] [CrossRef]
  108. Alliot, F.; Godin, I.; Pessac, B. Microglia Derive from Progenitors, Originating from the Yolk Sac, and Which Proliferate in the Brain. Dev. Brain Res. 1999, 117, 145–152. [Google Scholar] [CrossRef]
  109. Ginhoux, F.; Prinz, M. Origin of Microglia: Current Concepts and Past Controversies. Cold Spring Harb. Perspect. Biol. 2015, 7, a020537. [Google Scholar] [CrossRef]
  110. Verney, C.; Monier, A.; Fallet-Bianco, C.; Gressens, P. Early Microglial Colonization of the Human Forebrain and Possible Involvement in Periventricular White-Matter Injury of Preterm Infants. J. Anat. 2010, 217, 436–448. [Google Scholar] [CrossRef]
  111. Bennett, M.L.; Bennett, F.C.; Liddelow, S.A.; Ajami, B.; Zamanian, J.L.; Fernhoff, N.B.; Mulinyawe, S.B.; Bohlen, C.J.; Adil, A.; Tucker, A.; et al. New Tools for Studying Microglia in the Mouse and Human CNS. Proc. Natl. Acad. Sci. USA 2016, 113, E1738–E1746. [Google Scholar] [CrossRef]
  112. Wu, Y.; Dissing-Olesen, L.; MacVicar, B.A.; Stevens, B. Microglia: Dynamic Mediators of Synapse Development and Plasticity. Trends Immunol. 2015, 36, 605–613. [Google Scholar] [CrossRef]
  113. Perry, V.H.; Teeling, J. Microglia and Macrophages of the Central Nervous System: The Contribution of Microglia Priming and Systemic Inflammation to Chronic Neurodegeneration. Semin. Immunopathol. 2013, 35, 601–612. [Google Scholar] [CrossRef] [PubMed]
  114. Nimmerjahn, A.; Kirchhoff, F.; Helmchen, F. Neuroscience: Resting Microglial Cells Are Highly Dynamic Surveillants of Brain Parenchyma in Vivo. Science 2005, 308, 1314–1318. [Google Scholar] [CrossRef] [PubMed]
  115. Paolicelli, R.C.; Sierra, A.; Stevens, B.; Tremblay, M.E.; Aguzzi, A.; Ajami, B.; Amit, I.; Audinat, E.; Bechmann, I.; Bennett, M.; et al. Microglia States and Nomenclature: A Field at Its Crossroads. Neuron 2022, 110, 3458–3483. [Google Scholar] [CrossRef] [PubMed]
  116. Schwarz, J.M.; Sholar, P.W.; Bilbo, S.D. Sex Differences in Microglial Colonization of the Developing Rat Brain. J. Neurochem. 2012, 120, 948–963. [Google Scholar] [CrossRef] [PubMed]
  117. Simões-Henriques, C.F.; Rodrigues-Neves, A.C.; Sousa, F.J.; Gaspar, R.; Almeida, I.; Baptista, F.I.; Ambrósio, A.F.; Gomes, C.A. Neonatal Testosterone Voids Sexually Differentiated Microglia Morphology and Behavior. Front. Endocrinol. (Lausanne) 2023, 14, 1102068. [Google Scholar] [CrossRef]
  118. Nelson, L.H.; Warden, S.; Lenz, K.M. Sex Differences in Microglial Phagocytosis in the Neonatal Hippocampus. Brain Behav. Immun. 2017, 64, 11–22. [Google Scholar] [CrossRef]
  119. Guez-Barber, D.; Colon, L.M.; Raphael, D.; Wragan, M.A.; Yun, S.; Eisch, A.J. Female and Male Microglia Are Not Different in the Dentate Gyrus of Postnatal Day 10 Mice. Neurosci. Lett. 2023, 803, 137171. [Google Scholar] [CrossRef]
  120. Guneykaya, D.; Ivanov, A.; Hernandez, D.P.; Haage, V.; Wojtas, B.; Meyer, N.; Maricos, M.; Jordan, P.; Buonfiglioli, A.; Gielniewski, B.; et al. Transcriptional and Translational Differences of Microglia from Male and Female Brains. Cell Rep. 2018, 24, 2773–2783. [Google Scholar] [CrossRef]
  121. Weinhard, L.; Di Bartolomei, G.; Bolasco, G.; Machado, P.; Schieber, N.L.; Neniskyte, U.; Exiga, M.; Vadisiute, A.; Raggioli, A.; Schertel, A.; et al. Microglia Remodel Synapses by Presynaptic Trogocytosis and Spine Head Filopodia Induction. Nat. Commun. 2018, 9, 1228. [Google Scholar] [CrossRef]
  122. van Weering, H.R.J.; Nijboer, T.W.; Brummer, M.L.; Boddeke, E.W.G.M.; Eggen, B.J.L. Microglia Morphotyping in the Adult Mouse CNS Using Hierarchical Clustering on Principal Components Reveals Regional Heterogeneity but No Sexual Dimorphism. Glia 2023, 71, 2356–2371. [Google Scholar] [CrossRef] [PubMed]
  123. Hensel, J.A.; Khattar, V.; Ashton, R.; Ponnazhagan, S. Characterization of Immune Cell Subtypes in Three Commonly Used Mouse Strains Reveals Gender and Strain-Specific Variations. Lab. Investig. 2019, 99, 93–106. [Google Scholar] [CrossRef] [PubMed]
  124. Hammond, T.R.; Dufort, C.; Dissing-Olesen, L.; Giera, S.; Young, A.; Wysoker, A.; Walker, A.J.; Gergits, F.; Segel, M.; Nemesh, J.; et al. Single-Cell RNA Sequencing of Microglia throughout the Mouse Lifespan and in the Injured Brain Reveals Complex Cell-State Changes. Immunity 2019, 50, 253–271. [Google Scholar] [CrossRef] [PubMed]
  125. Thion, M.S.; Low, D.; Silvin, A.; Chen, J.; Grisel, P.; Schulte-Schrepping, J.; Blecher, R.; Ulas, T.; Squarzoni, P.; Hoeffel, G.; et al. Microbiome Influences Prenatal and Adult Microglia in a Sex-Specific Manner. Cell 2018, 172, 500–516. [Google Scholar] [CrossRef]
  126. Crain, J.M.; Nikodemova, M.; Watters, J.J. Microglia Express Distinct M1 and M2 Phenotypic Markers in the Postnatal and Adult Central Nervous System in Male and Female Mice. J. Neurosci. Res. 2013, 91, 1143–1151. [Google Scholar] [CrossRef]
  127. Hanamsagar, R.; Alter, M.D.; Block, C.S.; Sullivan, H.; Bolton, J.L.; Bilbo, S.D. Generation of a Microglial Developmental Index in Mice and in Humans Reveals a Sex Difference in Maturation and Immune Reactivity. Glia 2017, 65, 1504–1520. [Google Scholar] [CrossRef] [PubMed]
  128. Mangold, C.A.; Wronowski, B.; Du, M.; Masser, D.R.; Hadad, N.; Bixler, G.V.; Brucklacher, R.M.; Ford, M.M.; Sonntag, W.E.; Freeman, W.M. Sexually Divergent Induction of Microglial-Associated Neuroinflammation with Hippocampal Aging. J. Neuroinflamm. 2017, 14, 141. [Google Scholar] [CrossRef]
  129. Villa, A.; Gelosa, P.; Castiglioni, L.; Cimino, M.; Rizzi, N.; Pepe, G.; Lolli, F.; Marcello, E.; Sironi, L.; Vegeto, E.; et al. Sex-Specific Features of Microglia from Adult Mice. Cell Rep. 2018, 23, 3501–3511. [Google Scholar] [CrossRef]
  130. Kodama, L.; Guzman, E.; Etchegaray, J.I.; Li, Y.; Sayed, F.A.; Zhou, L.; Zhou, Y.; Zhan, L.; Le, D.; Udeochu, J.C.; et al. Microglial MicroRNAs Mediate Sex-Specific Responses to Tau Pathology. Nat. Neurosci. 2020, 23, 167–171. [Google Scholar] [CrossRef]
  131. Cyr, B.; Pablo, J.; Vaccari, D.R. Sex Differences in the Inflammatory Profile in the Brain of Young and Aged Mice. Cells 2023, 12, 1372. [Google Scholar] [CrossRef]
  132. Ocañas, S.R.; Pham, K.D.; Cox, J.E.J.; Keck, A.W.; Ko, S.; Ampadu, F.A.; Porter, H.L.; Ansere, V.A.; Kulpa, A.; Kellogg, C.M.; et al. Microglial Senescence Contributes to Female-Biased Neuroinflammation in the Aging Mouse Hippocampus: Implications for Alzheimer’s Disease. J. Neuroinflamm. 2023, 20, 188. [Google Scholar] [CrossRef] [PubMed]
  133. Loram, L.C.; Sholar, P.W.; Taylor, F.R.; Wiesler, J.L.; Babb, J.A.; Strand, K.A.; Berkelhammer, D.; Day, H.E.W.; Maier, S.F.; Watkins, L.R. Sex and Estradiol Influence Glial Pro-Inflammatory Responses to Lipopolysaccharide in Rats. Psychoneuroendocrinology 2012, 37, 1688–1699. [Google Scholar] [CrossRef] [PubMed]
  134. McNaughton, K.A.; Williamson, L.L. Effects of Sex and Pro-Inflammatory Cytokines on Context Discrimination Memory. Behav. Brain Res. 2023, 442, 114320. [Google Scholar] [CrossRef] [PubMed]
  135. Boghozian, R.; Sharma, S.; Narayana, K.; Cheema, M.; Brown, C.E. Sex and Interferon Gamma Signaling Regulate Microglia Migration in the Adult Mouse Cortex in Vivo. PNAS 2023, 120, e2302892120. [Google Scholar] [CrossRef] [PubMed]
  136. Young, A.M.H.; Kumasaka, N.; Calvert, F.; Hammond, T.R.; Knights, A.; Panousis, N.; Park, J.S.; Schwartzentruber, J.; Liu, J.; Kundu, K.; et al. A Map of Transcriptional Heterogeneity and Regulatory Variation in Human Microglia. Nat. Genet. 2021, 53, 861–868. [Google Scholar] [CrossRef] [PubMed]
  137. Py, N.A.; Bonnet, A.E.; Bernard, A.; Marchalant, Y.; Charrat, E.; Checler, F.; Khrestchatisky, M.; Baranger, K.; Rivera, S.; Ruano, D.; et al. Differential Spatio-Temporal Regulation of MMPs in the 5xFAD Mouse Model of Alzheimer’s Disease: Evidence for a pro-Amyloidogenic Role of MT1-MMP. Front. Aging Neurosci. 2014, 6, 247. [Google Scholar] [CrossRef] [PubMed]
  138. Palaszynski, K.M.; Smith, D.L.; Kamrava, S.; Burgoyne, P.S.; Arnold, A.P.; Voskuhl, R.R. A Yin-Yang Effect between Sex Chromosome Complement and Sex Hormones on the Immune Response. Endocrinology 2005, 146, 3280–3285. [Google Scholar] [CrossRef]
  139. Jégu, T.; Aeby, E.; Lee, J.T. The X Chromosome in Space. Nat. Rev. Genet. 2017, 18, 377–389. [Google Scholar] [CrossRef]
  140. Galupa, R.; Heard, E. X-Chromosome Inactivation: A Crossroads Between Chromosome Architecture and Gene Regulation. Annu. Rev. Genet. 2018, 52, 535–566. [Google Scholar] [CrossRef]
  141. Fang, H.; Disteche, C.M.; Berletch, J.B. X Inactivation and Escape: Epigenetic and Structural Features. Front. Cell Dev. Biol. 2019, 7, 219. [Google Scholar] [CrossRef]
  142. Brockdorff, N.; Bowness, J.S.; Wei, G. Progress toward Understanding Chromosome Silencing by Xist RNA. Genes Dev. 2020, 34, 733–744. [Google Scholar] [CrossRef] [PubMed]
  143. Markaki, Y.; Gan Chong, J.; Wang, Y.; Jacobson, E.C.; Luong, C.; Tan, S.Y.X.; Maestrini, D.; Banerjee, A.K.; Mistry, B.A.; Dror, I.; et al. Xist Nucleates Local Protein Gradients to Propagate Silencing across the X Chromosome. Cell 2021, 184, 6174–6192. [Google Scholar] [CrossRef]
  144. San Roman, A.K.; Godfrey, A.K.; Skaletsky, H.; Bellott, D.W.; Groff, A.F.; Harris, H.L.; Blanton, L.V.; Hughes, J.F.; Brown, L.; Phou, S.; et al. The Human Inactive X Chromosome Modulates Expression of the Active X Chromosome. Cell Genom. 2023, 3, 100259. [Google Scholar] [CrossRef] [PubMed]
  145. Brooks, W.H.; Renaudineau, Y. Epigenetics and Autoimmune Diseases: The X Chromosome-Nucleolus Nexus. Front. Genet. 2015, 6, 22. [Google Scholar] [CrossRef] [PubMed]
  146. Bajic, V.P.; Essack, M.; Zivkovic, L.; Stewart, A.; Zafirovic, S.; Bajic, V.B.; Gojobori, T.; Isenovic, E.; Spremo-Potparevic, B. The X Files: “The Mystery of X Chromosome Instability in Alzheimer’s Disease”. Front. Genet. 2020, 10, 1368. [Google Scholar] [CrossRef] [PubMed]
  147. Fang, H.; Deng, X.; Disteche, C.M. X-Factors in Human Disease: Impact of Gene Content and Dosage Regulation. Hum. Mol. Genet. 2021, 30, R285–R295. [Google Scholar] [CrossRef] [PubMed]
  148. Wright, D.J.; Day, F.R.; Kerrison, N.D.; Zink, F.; Cardona, A.; Sulem, P.; Thompson, D.J.; Sigurjonsdottir, S.; Gudbjartsson, D.F.; Helgason, A.; et al. Genetic Variants Associated with Mosaic Y Chromosome Loss Highlight Cell Cycle Genes and Overlap with Cancer Susceptibility. Nat. Genet. 2017, 49, 674–679. [Google Scholar] [CrossRef] [PubMed]
  149. Terao, C.; Momozawa, Y.; Ishigaki, K.; Kawakami, E.; Akiyama, M.; Loh, P.R.; Genovese, G.; Sugishita, H.; Ohta, T.; Hirata, M.; et al. GWAS of Mosaic Loss of Chromosome Y Highlights Genetic Effects on Blood Cell Differentiation. Nat. Commun. 2019, 10, 4719. [Google Scholar] [CrossRef]
  150. Thompson, D.J.; Genovese, G.; Halvardson, J.; Ulirsch, J.C.; Wright, D.J.; Terao, C.; Davidsson, O.B.; Day, F.R.; Sulem, P.; Jiang, Y.; et al. Genetic Predisposition to Mosaic Y Chromosome Loss in Blood. Nature 2019, 575, 652–657. [Google Scholar] [CrossRef]
  151. Dumanski, J.P.; Lambert, J.C.; Rasi, C.; Giedraitis, V.; Davies, H.; Grenier-Boley, B.; Lindgren, C.M.; Campion, D.; Dufouil, C.; Pasquier, F.; et al. Mosaic Loss of Chromosome Y in Blood Is Associated with Alzheimer Disease. Am. J. Hum. Genet. 2016, 98, 1208–1219. [Google Scholar] [CrossRef]
  152. Cáceres, A.; Jene, A.; Esko, T.; Pérez-Jurado, L.A.; González, J.R. Extreme Downregulation of Chromosome Y and Cancer Risk in Men. J. Natl. Cancer Inst. 2020, 112, 913–920. [Google Scholar] [CrossRef] [PubMed]
  153. Dumanski, J.P.; Halvardson, J.; Davies, H.; Rychlicka-Buniowska, E.; Mattisson, J.; Moghadam, B.T.; Nagy, N.; Węglarczyk, K.; Bukowska-Strakova, K.; Danielsson, M.; et al. Immune Cells Lacking Y Chromosome Show Dysregulation of Autosomal Gene Expression. Cell. Mol. Life Sci. 2021, 78, 4019–4033. [Google Scholar] [CrossRef]
  154. Mattisson, J.; Danielsson, M.; Hammond, M.; Davies, H.; Gallant, C.J.; Nordlund, J.; Raine, A.; Edén, M.; Kilander, L.; Ingelsson, M.; et al. Leukocytes with Chromosome Y Loss Have Reduced Abundance of the Cell Surface Immunoprotein CD99. Sci. Rep. 2021, 11, 15160. [Google Scholar] [CrossRef] [PubMed]
  155. Orta, A.H.; Bush, S.J.; Gutiérrez-Mariscal, M.; Castro-Obregón, S.; Jaimes-Hoy, L.; Grande, R.; Vázquez, G.; Gorostieta-Salas, E.; Martínez-Pacheco, M.; Díaz-Barba, K.; et al. Rats Exhibit Age-Related Mosaic Loss of Chromosome Y. Commun. Biol. 2021, 4, 1418. [Google Scholar] [CrossRef] [PubMed]
  156. Vermeulen, M.C.; Pearse, R.; Young-Pearse, T.; Mostafavi, S. Mosaic Loss of Chromosome Y in Aged Human Microglia. Genome Res. 2022, 32, 1795–1807. [Google Scholar] [CrossRef] [PubMed]
  157. Nelson, L.H.; Lenz, K.M. The Immune System as a Novel Regulator of Sex Differences in Brain and Behavioral Development. J. Neurosci. Res. 2017, 95, 447–461. [Google Scholar] [CrossRef] [PubMed]
  158. Todd, B.J.; Schwarz, J.M.; McCarthy, M.M. Prostaglandin-E2: A Point of Divergence in Estradiol-Mediated Sexual Differentiation. Horm. Behav. 2005, 48, 512–521. [Google Scholar] [CrossRef] [PubMed]
  159. Wright, C.L.; McCarthy, M.M. Prostaglandin E2-Induced Masculinization of Brain and Behavior Requires Protein Kinase A, AMPA/Kainate, and Metabotropic Glutamate Receptor Signaling. J. Neurosci. 2009, 29, 13274–13282. [Google Scholar] [CrossRef]
  160. Lenz, K.M.; Nugent, B.M.; Haliyur, R.; McCarthy, M.M. Microglia Are Essential to Masculinization of Brain and Behavior. J. Neurosci. 2013, 33, 2761–2772. [Google Scholar] [CrossRef]
  161. Sanchez, K.; Wu, S.L.; Kakkar, R.; Darling, J.S.; Harper, C.S.; Fonken, L.K. Ovariectomy in Mice Primes Hippocampal Microglia to Exacerbate Behavioral Sickness Responses. Brain Behav. Immun. Health 2023, 30, 100638. [Google Scholar] [CrossRef]
  162. Colton, C.A.; Wilcock, D.M. Assessing Activation States in Microglia. CNS Neurol Disord. Drug Targets 2010, 9, 174–191. [Google Scholar] [CrossRef] [PubMed]
  163. Biechele, G.; Franzmeier, N.; Blume, T.; Ewers, M.; Luque, J.M.; Eckenweber, F.; Sacher, C.; Beyer, L.; Ruch-Rubinstein, F.; Lindner, S.; et al. Glial Activation Is Moderated by Sex in Response to Amyloidosis but Not to Tau Pathology in Mouse Models of Neurodegenerative Diseases. J. Neuroinflamm. 2020, 17, 374. [Google Scholar] [CrossRef] [PubMed]
  164. Smith, A.M.; Davey, K.; Tsartsalis, S.; Khozoie, C.; Fancy, N.; Tang, S.S.; Liaptsi, E.; Weinert, M.; McGarry, A.; Muirhead, R.C.J.; et al. Diverse Human Astrocyte and Microglial Transcriptional Responses to Alzheimer’s Pathology. Acta Neuropathol. 2022, 143, 75–91. [Google Scholar] [CrossRef] [PubMed]
  165. Keren-Shaul, H.; Spinrad, A.; Weiner, A.; Matcovitch-Natan, O.; Dvir-Szternfeld, R.; Ulland, T.K.; David, E.; Baruch, K.; Lara-Astaiso, D.; Toth, B.; et al. A Unique Microglia Type Associated with Restricting Development of Alzheimer’s Disease. Cell 2017, 169, 1276–1290. [Google Scholar] [CrossRef] [PubMed]
  166. Deczkowska, A.; Keren-Shaul, H.; Weiner, A.; Colonna, M.; Schwartz, M.; Amit, I. Disease-Associated Microglia: A Universal Immune Sensor of Neurodegeneration. Cell 2018, 173, 1073–1081. [Google Scholar] [CrossRef]
  167. Rangaraju, S.; Dammer, E.B.; Syed, R.A.; Rathakrishnan, P.; Xiao, H.; Gao, T.; Duong, D.M.; Pennington, M.W.; Lah, J.J.; Seyfried, N.T.; et al. Identification and Therapeutic Modulation of a Pro-Inflammatory Subset of Disease-Associated-Microglia in Alzheimer’s Disease. Mol. Neuodegener. 2018, 13, 24. [Google Scholar] [CrossRef]
  168. Mathys, H.; Davila-Velderrain, J.; Peng, Z.; Gao, F.; Mohammadi, S.; Young, J.Z.; Menon, M.; He, L.; Abdurrob, F.; Jiang, X.; et al. Single-Cell Transcriptomic Analysis of Alzheimer’s Disease. Nature 2019, 570, 332–337. [Google Scholar] [CrossRef]
  169. Krasemann, S.; Madore, C.; Cialic, R.; Baufeld, C.; Calcagno, N.; El Fatimy, R.; Beckers, L.; O’Loughlin, E.; Xu, Y.; Fanek, Z.; et al. The TREM2-APOE Pathway Drives the Transcriptional Phenotype of Dysfunctional Microglia in Neurodegenerative Diseases. Immunity 2017, 47, 566–581. [Google Scholar] [CrossRef]
  170. Ocañas, S.R.; Ansere, V.A.; Tooley, K.B.; Hadad, N.; Chucair-Elliott, A.J.; Stanford, D.R.; Rice, S.; Wronowski, B.; Pham, K.D.; Hoffman, J.M.; et al. Differential Regulation of Mouse Hippocampal Gene Expression Sex Differences by Chromosomal Content and Gonadal Sex. Mol. Neurobiol. 2022, 59, 4669–4702. [Google Scholar] [CrossRef]
  171. Gildawie, K.R.; Orso, R.; Peterzell, S.; Thompson, V.; Brenhouse, H.C. Sex Differences in Prefrontal Cortex Microglia Morphology: Impact of a Two-Hit Model of Adversity throughout Development. Neurosci. Lett. 2020, 738, 135381. [Google Scholar] [CrossRef]
  172. Bachiller, S.; Hidalgo, I.; Garcia, M.G.; Boza-Serrano, A.; Paulus, A.; Denis, Q.; Haikal, C.; Manouchehrian, O.; Klementieva, O.; Li, J.Y.; et al. Early-Life Stress Elicits Peripheral and Brain Immune Activation Differently in Wild Type and 5xFAD Mice in a Sex-Specific Manner. J. Neuroinflamm. 2022, 19, 151. [Google Scholar] [CrossRef] [PubMed]
  173. Garcia, M.G.; Paulus, A.; Vázquez-Reyes, S.; Klementieva, O.; Gouras, G.K.; Bachiller, S.; Deierborg, T. Maternal Separation Differentially Modulates Early Pathology by Sex in 5xFAD Alzheimer’s Disease-Transgenic Mice. Brain Behav. Immun. Health 2023, 32, 100663. [Google Scholar] [CrossRef] [PubMed]
  174. Smith, C.J.; Kingsbury, M.A.; Dziabis, J.E.; Hanamsagar, R.; Malacon, K.E.; Tran, J.N.; Norris, H.A.; Gulino, M.; Bordt, E.A.; Bilbo, S.D. Neonatal Immune Challenge Induces Female-Specific Changes in Social Behavior and Somatostatin Cell Number. Brain Behav. Immun. 2020, 90, 332–345. [Google Scholar] [CrossRef] [PubMed]
  175. Block, C.L.; Eroglu, O.; Mague, S.D.; Dzirasa, K.; Eroglu, C.; Bilbo, S.D. Prenatal Environmental Stressors Impair Postnatal Microglia Function and Adult Behavior in Males. Cell Rep. 2022, 40, 111161. [Google Scholar] [CrossRef] [PubMed]
  176. Arambula, S.E.; McCarthy, M.M. Neuroendocrine-Immune Crosstalk Shapes Sex-Specific Brain Development. Endocrinology 2020, 161, bqaa055. [Google Scholar] [CrossRef] [PubMed]
  177. Cabrera Zapata, L.E.; Garcia-Segura, L.M.; Cambiasso, M.J.; Arevalo, M.A. Genetics and Epigenetics of the X and Y Chromosomes in the Sexual Differentiation of the Brain. Int. J. Mol. Sci. 2022, 23, 12288. [Google Scholar] [CrossRef] [PubMed]
  178. Terrin, F.; Tesoriere, A.; Plotegher, N.; Valle, L.D. Sex and Brain: The Role of Sex Chromosomes and Hormones in Brain Development and Parkinson’s Disease. Cells 2023, 12, 1486. [Google Scholar] [CrossRef]
  179. Gegenhuber, B.; Wu, M.V.; Bronstein, R.; Tollkuhn, J. Gene Regulation by Gonadal Hormone Receptors Underlies Brain Sex Differences Genomic Targets of ERα in the Brain. Nature 2022, 606, 153. [Google Scholar] [CrossRef]
  180. Guma, E.; Beauchamp, A.; Liu, S.; Levitis, E.; Clasen, L.S.; Torres, E.; Blumenthal, J.; Lalonde, F.; Qiu, L.R.; Hrncir, H.; et al. A Cross-Species Neuroimaging Study of Sex Chromosome Dosage Effects on Human and Mouse Brain Anatomy. J. Neurosci. 2023, 43, 1321–1333. [Google Scholar] [CrossRef]
  181. Nelson, L.H.; Peketi, P.; Lenz, K.M. Microglia Regulate Cell Genesis in a Sex-Dependent Manner in the Neonatal Hippocampus. Neuroscience 2021, 453, 237–255. [Google Scholar] [CrossRef]
  182. Kim, Y.; Yang, G.R.; Pradhan, K.; Venkataraju, K.U.; Bota, M.; del Molino, L.C.G.; Fitzgerald, G.; Ram, K.; He, M.; Levine, J.M.; et al. Brain-Wide Maps Reveal Stereotyped Cell-Type-Based Cortical Architecture and Subcortical Sexual Dimorphism in Brief Kim et Al. Cell 2017, 171, 456–469. [Google Scholar] [CrossRef] [PubMed]
  183. Guebel, D.V. Human Hippocampal Astrocytes: Computational Dissection of Their Transcriptome, Sexual Differences and Exosomes across Ageing and Mild-Cognitive Impairment. Eur. J. Neurosci. 2023, 58, 2677–2707. [Google Scholar] [CrossRef] [PubMed]
  184. Yasuda, K.; Maki, T.; Kinoshita, H.; Kaji, S.; Toyokawa, M.; Nishigori, R.; Kinoshita, Y.; Ono, Y.; Kinoshita, A.; Takahashi, R. Sex-Specific Differences in Transcriptomic Profiles and Cellular Characteristics of Oligodendrocyte Precursor Cells. Stem Cell Res. 2020, 46, 101866. [Google Scholar] [CrossRef] [PubMed]
  185. Wingo, A.P.; Liu, Y.; Gerasimov, E.S.; Vattathil, S.M.; Liu, J.; Cutler, D.J.; Epstein, M.P.; Blokland, G.A.M.; Thambisetty, M.; Troncoso, J.C.; et al. Sex Differences in Brain Protein Expression and Disease. Nat. Med. 2023, 29, 2224–2232. [Google Scholar] [CrossRef] [PubMed]
  186. Baumgartner, N.E.; Biraud, M.C.; Lucas, E.K. Sex Differences in Socioemotional Behavior and Changes in Ventral Hippocampal Transcription across Aging in C57Bl/6J Mice. Neurobiol. Aging 2023, 130, 141–153. [Google Scholar] [CrossRef] [PubMed]
  187. Dawson, M.S.; Gordon-Fleet, K.; Yan, L.; Tardos, V.; He, H.; Mui, K.; Nawani, S.; Asgarian, Z.; Catani, M.; Fernandes, C.; et al. Sexual Dimorphism in the Social Behaviour of Cntnap2-Null Mice Correlates with Disrupted Synaptic Connectivity and Increased Microglial Activity in the Anterior Cingulate Cortex. Commun. Biol. 2023, 6, 846. [Google Scholar] [CrossRef]
  188. Tschanz, J.T.; Corcoran, C.D.; Schwartz, S.; Treiber, K.; Green, R.C.; Norton, M.C.; Mielke, M.M.; Piercy, K.; Steinberg, M.; Rabins, P.V.; et al. Progression of Cognitive, Functional and Neuropsychiatric Symptom Domains in a Population Cohort with Alzheimer’s Dementia the Cache County Dementia Progression Study. Am. J. Geriatr. Psychiatry 2011, 19, 532–542. [Google Scholar] [CrossRef]
  189. Filon, J.R.; Intorcia, A.J.; Sue, L.I.; Vazquez Arreola, E.; Wilson, J.; Davis, K.J.; Sabbagh, M.N.; Belden, C.M.; Caselli, R.J.; Adler, C.H.; et al. Gender Differences in Alzheimer Disease: Brain Atrophy, Histopathology Burden, and Cognition. J. Neuropathol. Exp. Neurol. 2016, 75, 748–754. [Google Scholar] [CrossRef]
  190. Sundermann, E.E.; Biegon, A.; Rubin, L.H.; Lipton, R.B.; Mowrey, W.; Landau, S.; Maki, P.M. Better Verbal Memory in Women than Men in MCI despite Similar Levels of Hippocampal Atrophy. Neurology 2016, 86, 1368–1376. [Google Scholar] [CrossRef]
  191. Malpetti, M.; Ballarini, T.; Presotto, L.; Garibotto, V.; Tettamanti, M.; Perani, D.; Alzheimer’s Disease Neuroimaging Initiative (ADNI) Database; Network for Efficiency and Standardization of Dementia Diagnosis (NEST-DD) Database. Gender Differences in Healthy Aging and Alzheimer’s Dementia: A 18F-FDG-PET Study of Brain and Cognitive Reserve. Hum. Brain Mapp. 2017, 38, 4212–4227. [Google Scholar] [CrossRef]
  192. Wang, S.M.; Kang, D.W.; Um, Y.H.; Kim, S.; Lee, C.U.; Lim, H.K. Olfactory Dysfunction Is Associated with Cerebral Amyloid Deposition and Cognitive Function in the Trajectory of Alzheimer’s Disease. Biomolecules 2023, 13, 1336. [Google Scholar] [CrossRef] [PubMed]
  193. Fatuzzo, I.; Niccolini, G.F.; Zoccali, F.; Cavalcanti, L.; Bellizzi, M.G.; Riccardi, G.; de Vincentiis, M.; Fiore, M.; Petrella, C.; Minni, A.; et al. Neurons, Nose, and Neurodegenerative Diseases: Olfactory Function and Cognitive Impairment. Int. J. Mol. Sci. 2023, 24, 2117. [Google Scholar] [CrossRef] [PubMed]
  194. Audronyte, E.; Pakulaite-Kazliene, G.; Sutnikiene, V.; Kaubrys, G. Odor Discrimination as a Marker of Early Alzheimer’s Disease. J. Alzheimer’s Dis. 2023, 94, 1169–1178. [Google Scholar] [CrossRef] [PubMed]
  195. Zhang, J.; Zhao, Z.; Sun, S.; Li, J.; Wang, Y.; Dong, J.; Yang, S.; Lou, Y.; Yang, J.; Li, W.; et al. Olfactory Evaluation in Alzheimer’s Disease Model Mice. Brain Sci. 2022, 12, 607. [Google Scholar] [CrossRef] [PubMed]
  196. Zou, Y.M.; Lu, D.; Liu, L.P.; Zhang, H.H.; Zhou, Y.Y. Olfactory Dysfunction in Alzheimer’s Disease. Neuropsychiatr. Dis. Treat. 2016, 12, 869–875. [Google Scholar] [CrossRef] [PubMed]
  197. Alotaibi, M.M.; De Marco, M.; Venneri, A. Sex Differences in Olfactory Cortex Neuronal Loss in Aging. Front. Hum. Neurosci. 2023, 17, 1130200. [Google Scholar] [CrossRef]
  198. Greenfield, A.; Carrel, L.; Pennisi, D.; Philippe, C.; Quaderi, N.; Siggers, P.; Steiner, K.; Tam, P.P.L.; Monaco, A.P.; Willard, H.F.; et al. The UTX Gene Escapes X Inactivation in Mice and Humans. Hum. Mol. Genet. 1998, 7, 737–742. [Google Scholar] [CrossRef]
  199. Lederer, D.; Grisart, B.; Digilio, M.C.; Benoit, V.; Crespin, M.; Ghariani, S.C.; Maystadt, I.; Dallapiccola, B.; Verellen-Dumoulin, C. Deletion of KDM6A, a Histone Demethylase Interacting with MLL2, in Three Patients with Kabuki Syndrome. Am. J. Hum. Genet. 2012, 90, 119–124. [Google Scholar] [CrossRef]
  200. Miyake, N.; Mizuno, S.; Okamoto, N.; Ohashi, H.; Shiina, M.; Ogata, K.; Tsurusaki, Y.; Nakashima, M.; Saitsu, H.; Niikawa, N.; et al. KDM6A Point Mutations Cause Kabuki Syndrome. Hum. Mutat. 2013, 34, 108–110. [Google Scholar] [CrossRef]
  201. Miyake, N.; Koshimizu, E.; Okamoto, N.; Mizuno, S.; Ogata, T.; Nagai, T.; Kosho, T.; Ohashi, H.; Kato, M.; Sasaki, G.; et al. MLL2 and KDM6A Mutations in Patients with Kabuki Syndrome. Am. J. Med. Genet. A 2013, 161, 2234–2243. [Google Scholar] [CrossRef]
  202. Berletch, J.B.; Ma, W.; Yang, F.; Shendure, J.; Noble, W.S.; Disteche, C.M.; Deng, X. Escape from X Inactivation Varies in Mouse Tissues. PLoS Genet. 2015, 11, e1005079. [Google Scholar] [CrossRef] [PubMed]
  203. Van Laarhoven, P.M.; Neitzel, L.R.; Quintana, A.M.; Geiger, E.A.; Zackai, E.H.; Clouthier, D.E.; Artinger, K.B.; Ming, J.E.; Shaikh, T.H. Kabuki Syndrome Genes KMT2D and KDM6A: Functional Analyses Demonstrate Critical Roles in Craniofacial, Heart and Brain Development. Hum. Mol. Genet. 2015, 24, 4443–4453. [Google Scholar] [CrossRef]
  204. Bögershausen, N.; Gatinois, V.; Riehmer, V.; Kayserili, H.; Becker, J.; Thoenes, M.; Simsek-Kiper, P.Ö.; Barat-Houari, M.; Elcioglu, N.H.; Wieczorek, D.; et al. Mutation Update for Kabuki Syndrome Genes KMT2D and KDM6A and Further Delineation of X-Linked Kabuki Syndrome Subtype 2. Hum. Mutat. 2016, 37, 847–864. [Google Scholar] [CrossRef] [PubMed]
  205. Yang, P.; Tan, H.; Xia, Y.; Yu, Q.; Wei, X.; Guo, R.; Peng, Y.; Chen, C.; Li, H.; Mei, L.; et al. De Novo Exonic Deletion of KDM6A in a Chinese Girl with Kabuki Syndrome: A Case Report and Brief Literature Review. Am. J. Med. Genet. A 2016, 170, 1613–1621. [Google Scholar] [CrossRef] [PubMed]
  206. Tang, G.B.; Zeng, Y.Q.; Liu, P.P.; Mi, T.W.; Zhang, S.F.; Dai, S.K.; Tang, Q.Y.; Yang, L.; Xu, Y.J.; Yan, H.L.; et al. The Histone H3K27 Demethylase UTX Regulates Synaptic Plasticity and Cognitive Behaviors in Mice. Front. Mol. Neurosci. 2017, 10, 267. [Google Scholar] [CrossRef] [PubMed]
  207. Shaw, C.K.; Abdulai-Saiku, S.; Marino, F.; Wang, D.; Davis, E.J.; Panning, B.; Dubal, D.B. X Chromosome Factor Kdm6a Enhances Cognition Independent of Its Demethylase Function in the Aging XY Male Brain. J. Gerontol. A Biol. Sci. Med. Sci. 2023, 78, 938–943. [Google Scholar] [CrossRef]
  208. Warren, C.L.; Kratochvil, N.C.S.; Hauschild, K.E.; Foister, S.; Brezinski, M.L.; Dervan, P.B.; Phillips, G.M.; Ansari, A.Z. Defining the Sequence-Recognition Profile of DNA-Binding Molecules. Proc. Natl. Acad. Sci. USA 2006, 103, 867–872. [Google Scholar] [CrossRef]
  209. Oláh, Z.; Kálmán, J.; Tóth, M.E.; Zvara, Á.; Sántha, M.; Ivitz, E.; Janka, Z.; Pákáski, M. Proteomic Analysis of Cerebrospinal Fluid in Alzheimer’s Disease: Wanted Dead or Alive. J. Alzheimer’s Dis. 2015, 44, 1303–1312. [Google Scholar] [CrossRef]
  210. Sobreira, D.R.; Joslin, A.C.; Zhang, Q.; Williamson, I.; Hansen, G.T.; Farris, K.M.; Sakabe, N.J.; Sinnott-Armstrong, N.; Bozek, G.; Jensen-Cody, S.O.; et al. Extensive Pleiotropism and Allelic Heterogeneity Mediate Metabolic Effects of IRX3 and IRX5. Science 2021, 372, 1085–1091. [Google Scholar] [CrossRef]
  211. Chung, J.; Das, A.; Sun, X.; Sobreira, D.R.; Leung, Y.Y.; Igartua, C.; Mozaffari, S.; Chou, Y.F.; Thiagalingam, S.; Mez, J.; et al. Genome-Wide Association and Multi-Omics Studies Identify MGMT as a Novel Risk Gene for Alzheimer’s Disease among Women. Alzheimer’s Dement. 2022, 19, 896–908. [Google Scholar] [CrossRef]
  212. Xiong, J.; Kang, S.S.; Wang, Z.; Liu, X.; Kuo, T.-C.; Korkmaz, F.; Padilla, A.; Miyashita, S.; Chan, P.; Zhang, Z.; et al. FSH Blockade Improves Cognition in Mice with Alzheimer’s Disease. Nature 2022, 603, 470–476. [Google Scholar] [CrossRef] [PubMed]
  213. Randolph, J.F.; Zheng, H.; Sowers, M.F.R.; Crandall, C.; Crawford, S.; Gold, E.B.; Vuga, M. Change in Follicle-Stimulating Hormone and Estradiol across the Menopausal Transition: Effect of Age at the Final Menstrual Period. J. Clin. Endocrinol. Metab. 2011, 96, 746–754. [Google Scholar] [CrossRef] [PubMed]
  214. Short, R.A.; O’brien, P.C.; Graff-Radford, N.R. Elevated Gonadotropin Levels in Patients with Alzheimer Disease. Mayo Clin. Proc. 2001, 76, 906–909. [Google Scholar] [CrossRef] [PubMed]
  215. Bowen, R.L.; Isley, J.P.; Atkinson, R.L. An Association of Elevated Serum Gonadotropin Concentrations and Alzheimer Disease? J. Neuroendocrinol. 2000, 12, 351–354. [Google Scholar] [CrossRef] [PubMed]
  216. Niswender, C.M.; Conn, P.J. Metabotropic Glutamate Receptors: Physiology, Pharmacology, and Disease. Annu. Rev. Pharmacol. Toxicol. 2010, 50, 295–322. [Google Scholar] [CrossRef] [PubMed]
  217. Hamilton, A.; Zamponi, G.W.; Ferguson, S.S.G. Glutamate Receptors Function as Scaffolds for the Regulation of β-Amyloid and Cellular Prion Protein Signaling Complexes. Mol. Brain 2015, 8, 18. [Google Scholar] [CrossRef] [PubMed]
  218. Abd-Elrahman, K.S.; Albaker, A.; de Souza, J.M.; Ribeiro, F.M.; Schlossmacher, M.G.; Tiberi, M.; Hamilton, A.; Ferguson, S.S. Aβ Oligomers Induce Pathophysiological MGluR5 Signaling in Alzheimer’s Disease Model Mice in a Sex-Selective Manner. Sci. Signal 2020, 13, eabd2494. [Google Scholar] [CrossRef]
  219. Pacheco-Sánchez, B.; Tovar, R.; Ben Rabaa, M.; Sánchez-Salido, L.; Vargas, A.; Suárez, J.; Rodríguez de Fonseca, F.; Rivera, P. Sex-Dependent Altered Expression of Cannabinoid Signaling in Hippocampal Astrocytes of the Triple Transgenic Mouse Model of Alzheimer’s Disease: Implications for Controlling Astroglial Activity. Int. J. Mol. Sci. 2023, 24, 12598. [Google Scholar] [CrossRef]
  220. Stratoulias, V.; Ruiz, R.; Kanatani, S.; Osman, A.M.; Keane, L.; Armengol, J.A.; Rodríguez-Moreno, A.; Murgoci, A.-N.; García-Domínguez, I.; Alonso-Bellido, I.; et al. ARG1-Expressing Microglia Show a Distinct Molecular Signature and Modulate Postnatal Development and Function of the Mouse Brain. Nat. Neurosci. 2023, 26, 1008–1020. [Google Scholar] [CrossRef]
  221. Glenner, G.G.; Wong, C.W. Alzheimer’s Disease: Initial Report of the Purification and Characterization of a Novel Cerebrovascular Amyloid Protein. Biochem. Biophys. Res. Commun. 1984, 120, 885–890. [Google Scholar] [CrossRef]
  222. Masters, C.L.; Simms, G.; Weinman, N.A.; Multhaup, G.; McDonald, B.L.; Beyreuther, K. Amyloid Plaque Core Protein in Alzheimer Disease and Down Syndrome. Proc. Natl. Acad. Sci. USA 1985, 82, 4245–4249. [Google Scholar] [CrossRef] [PubMed]
  223. Cras, P.; Kawai, M.; Lowery, D.; Gonzalez-DeWhitt, P.; Greenberg, B.; Perry, G. Senile Plaque Neurites in Alzheimer Disease Accumulate Amyloid Precursor Protein. Proc. Natl. Acad. Sci. USA 1991, 88, 7552–7556. [Google Scholar] [CrossRef] [PubMed]
  224. Müller, U.C.; Zheng, H. Physiological Functions of APP Family Proteins. Cold Spring Harb. Perspect. Med. 2012, 2, a006288. [Google Scholar] [CrossRef] [PubMed]
  225. De Strooper, B.; Saftig, P.; Craessaerts, K.; Vanderstichele, H.; Guhde, G.; Annaert, W.; Von Figura, K.; Van Leuven, F. Deficiency of Presenilin-1 Inhibits the Normal Cleavage of Amyloid Precursor Protein. Nature 1998, 391, 387–390. [Google Scholar] [CrossRef]
  226. Hampel, H.; Hardy, J.; Blennow, K.; Chen, C.; Perry, G.; Kim, S.H.; Villemagne, V.L.; Aisen, P.; Vendruscolo, M.; Iwatsubo, T.; et al. The Amyloid-β Pathway in Alzheimer’s Disease. Mol. Psychiatry 2021, 26, 5481–5503. [Google Scholar] [CrossRef]
  227. Bai, X.C.; Yan, C.; Yang, G.; Lu, P.; Ma, D.; Sun, L.; Zhou, R.; Scheres, S.H.W.; Shi, Y. An Atomic Structure of Human γ-Secretase. Nature 2015, 525, 212–217. [Google Scholar] [CrossRef]
  228. Zhang, J.; Ke, K.-F.; Liu, Z.; Qiu, Y.-H.; Peng, Y.-P. Th17 Cell-Mediated Neuroinflammation Is Involved in Neurodegeneration of Aβ1-42-Induced Alzheimer’s Disease Model Rats. PLoS ONE 2013, 8, e75786. [Google Scholar] [CrossRef]
  229. Alzheimer’s Foundation. AlzGene Mutations Database. Available online: https://www.alzforum.org/mutations/app (accessed on 10 July 2023).
  230. Gu, L.; Guo, Z. Alzheimer’s Aβ42 and Aβ40 Peptides Form Interlaced Amyloid Fibrils. J. Neurochem. 2013, 126, 305–311. [Google Scholar] [CrossRef]
  231. Chen, G.F.; Xu, T.H.; Yan, Y.; Zhou, Y.R.; Jiang, Y.; Melcher, K.; Xu, H.E. Amyloid Beta: Structure, Biology and Structure-Based Therapeutic Development. Acta Pharmacol. Sin. 2017, 38, 1205–1235. [Google Scholar] [CrossRef]
  232. Yang, Y.H.; Huang, L.C.; Hsieh, S.W.; Huang, L.J. Dynamic Blood Concentrations of Aβ1–40 and Aβ1–42 in Alzheimer’s Disease. Front. Cell Dev. Biol. 2020, 8, 768. [Google Scholar] [CrossRef]
  233. Hartley, D.; Blumenthal, T.; Carrillo, M.; DiPaolo, G.; Esralew, L.; Gardiner, K.; Granholm, A.C.; Iqbal, K.; Krams, M.; Lemere, C.; et al. Down Syndrome and Alzheimer’s Disease: Common Pathways, Common Goals. Alzheimer’s Dement. 2015, 11, 700–709. [Google Scholar] [CrossRef]
  234. Cai, X.D.; Golde, T.E.; Younkin, S.G. Release of Excess Amyloid Beta Protein from a Mutant Amyloid Beta Protein Precursor. Science 1993, 259, 514–516. [Google Scholar] [CrossRef] [PubMed]
  235. Thal, D.R.; Rüb, U.; Orantes, M.; Braak, H. Phases of A Beta-Deposition in the Human Brain and Its Relevance for the Development of AD. Neurology 2002, 58, 1791–1800. [Google Scholar] [CrossRef] [PubMed]
  236. Hardy, J.; Selkoe, D.J. The Amyloid Hypothesis of Alzheimer’s Disease: Progress and Problems on the Road to Therapeutics. Science 2002, 297, 353–356. [Google Scholar] [CrossRef] [PubMed]
  237. Hardy, J.A.; Higgins, G.A. Alzheimer’s Disease: The Amyloid Cascade Hypothesis. Science 1992, 256, 184–185. [Google Scholar] [CrossRef] [PubMed]
  238. Jansen, W.J.; Ossenkoppele, R.; Knol, D.L.; Tijms, B.M.; Scheltens, P.; Verhey, F.R.J.; Visser, P.J.; Aalten, P.; Aarsland, D.; Alcolea, D.; et al. Prevalence of Cerebral Amyloid Pathology in Persons without Dementia: A Meta-Analysis. JAMA 2015, 313, 1924–1938. [Google Scholar] [CrossRef] [PubMed]
  239. Golimstok, A.; Rojas, J.I.; Romano, M.; Zurru, M.C.; Doctorovich, D.; Cristiano, E. Previous Adult Attention-Deficit and Hyperactivity Disorder Symptoms and Risk of Dementia with Lewy Bodies: A Case-Control Study. Eur. J. Neurol. 2011, 18, 78–84. [Google Scholar] [CrossRef]
  240. Tzeng, N.S.; Chung, C.H.; Lin, F.H.; Yeh, C.B.; Huang, S.Y.; Lu, R.B.; Chang, H.A.; Kao, Y.C.; Yeh, H.W.; Chiang, W.S.; et al. Risk of Dementia in Adults With ADHD: A Nationwide, Population-Based Cohort Study in Taiwan. J. Atten. Disord. 2019, 23, 995–1006. [Google Scholar] [CrossRef]
  241. Lansdell, T.A.; Xu, H.; Galligan, J.J.; Dorrance, A.M.; Pérez, E.A. Effects of Striatal Amyloidosis on the Dopaminergic System and Behavior: A Comparative Study in Male and Female 5XFAD Mice. J. Alzheimer’s Dis. 2023, 94, 1361–1375. [Google Scholar] [CrossRef]
  242. Shinohara, M.; Murray, M.E.; Frank, R.D.; Shinohara, M.; DeTure, M.; Yamazaki, Y.; Tachibana, M.; Atagi, Y.; Davis, M.D.; Liu, C.C.; et al. Impact of Sex and APOE4 on Cerebral Amyloid Angiopathy in Alzheimer’s Disease. Acta Neuropathol. 2016, 132, 225–234. [Google Scholar] [CrossRef]
  243. Perez, C.M.; Gong, Z.; Yoo, C.; Roy, D.; Deoraj, A.; Felty, Q. Inhibitor of DNA Binding Protein 3 (ID3) and Nuclear Respiratory Factor 1 (NRF1) Mediated Transcriptional Gene Signatures Are Associated with the Severity of Cerebral Amyloid Angiopathy. Mol. Neurobiol. 2023. [Google Scholar] [CrossRef]
  244. Colombo, M.; Raposo, G.; Théry, C. Biogenesis, Secretion, and Intercellular Interactions of Exosomes and Other Extracellular Vesicles. Annu. Rev. Cell Dev. Biol. 2014, 30, 255–289. [Google Scholar] [CrossRef] [PubMed]
  245. Malm, T.; Loppi, S.; Kanninen, K.M. Exosomes in Alzheimer’s Disease. Neurochem. Int. 2016, 97, 193–199. [Google Scholar] [CrossRef] [PubMed]
  246. Trajkovic, K.; Hsu, C.; Chiantia, S.; Rajendran, L.; Wenzel, D.; Wieland, F.; Schwille, P.; Brügger, B.; Simons, M. Ceramide Triggers Budding of Exosome Vesicles into Multivesicular Endosomes. Science 2008, 319, 1244–1247. [Google Scholar] [CrossRef] [PubMed]
  247. Jazvinšćak Jembrek, M.; Hof, P.R.; Šimić, G. Ceramides in Alzheimer’s Disease: Key Mediators of Neuronal Apoptosis Induced by Oxidative Stress and Aβ Accumulation. Oxid. Med. Cell. Longev. 2015, 2015, 346783. [Google Scholar] [CrossRef]
  248. Mowry, F.E.; Espejo-Porras, F.; Jin, S.; Quadri, Z.; Wu, L.; Bertolio, M.; Jarvis, R.; Reynolds, C.; Alananzeh, R.; Bieberich, E.; et al. Chronic NSMase Inhibition Suppresses Neuronal Exosome Spreading and Sex-Specifically Attenuates Amyloid Pathology in APP Knock-in Alzheimer’s Disease Mice. Neurobiol. Dis. 2023, 184, 106213. [Google Scholar] [CrossRef]
  249. Edwards, H.M.; Wallace, C.E.; Gardiner, W.D.; Doherty, B.M.; Harrigan, R.T.; Yuede, K.M.; Yuede, C.M.; Cirrito, J.R. Sex-Dependent Effects of Acute Stress on Amyloid-β in Male and Female Mice. Brain 2023, 146, 2268–2274. [Google Scholar] [CrossRef]
  250. Chang, D.; Kwan, J.; Timiras, P.S. Estrogens Influence Growth, Maturation, and Amyloid Beta-Peptide Production in Neuroblastoma Cells and in a Beta-APP Transfected Kidney 293 Cell Line. Adv. Exp. Med. Biol. 1997, 429, 261–271. [Google Scholar] [CrossRef]
  251. Xu, H.; Gouras, G.K.; Greenfield, J.P.; Vincent, B.; Naslund, J.; Mazzarelli, L.; Fried, G.; Jovanovic, J.N.; Seeger, M.; Relkin, N.R.; et al. Estrogen Reduces Neuronal Generation of Alzheimer Beta-Amyloid Peptides. Nat. Med. 1998, 4, 447–451. [Google Scholar] [CrossRef]
  252. Hu, Y.T.; Chen, X.L.; Zhang, Y.N.; McGurran, H.; Stormmesand, J.; Breeuwsma, N.; Sluiter, A.; Zhao, J.; Swaab, D.; Bao, A.M. Sex Differences in Hippocampal β-Amyloid Accumulation in the Triple-Transgenic Mouse Model of Alzheimer’s Disease and the Potential Role of Local Estrogens. Front. Neurosci. 2023, 17, 1117584. [Google Scholar] [CrossRef]
  253. Liang, K.; Yang, L.; Yin, C.; Xiao, Z.; Zhang, J.; Liu, Y.; Huang, J. Estrogen Stimulates Degradation of β-Amyloid Peptide by Up-Regulating Neprilysin. J. Biol. Chem. 2010, 285, 935–942. [Google Scholar] [CrossRef] [PubMed]
  254. Mawuenyega, K.G.; Sigurdson, W.; Ovod, V.; Munsell, L.; Kasten, T.; Morris, J.C.; Yarasheski, K.E.; Bateman, R.J. Decreased Clearance of CNS Amyloid-β in Alzheimer’s Disease. Science 2010, 330, 1774. [Google Scholar] [CrossRef] [PubMed]
  255. Hatami, A.; Monjazeb, S.; Milton, S.; Glabe, C.G. Familial Alzheimer’s Disease Mutations within the Amyloid Precursor Protein Alter the Aggregation and Conformation of the Amyloid-β Peptide. J. Biol. Chem. 2017, 292, 3172–3185. [Google Scholar] [CrossRef] [PubMed]
  256. Ullah, R.; Lee, E.J. Advances in Amyloid-β Clearance in the Brain and Periphery: Implications for Neurodegenerative Diseases. Exp. Neurobiol. 2023, 32, 216–246. [Google Scholar] [CrossRef] [PubMed]
  257. Turner, A.J.; Nalivaeva, N.N. New Insights into the Roles of Metalloproteinases in Neurodegeneration and Neuroprotection. Int. Rev. Neurobiol. 2007, 82, 113–135. [Google Scholar] [CrossRef] [PubMed]
  258. Yoon, S.S.; Jo, S.A. Mechanisms of Amyloid-β Peptide Clearance: Potential Therapeutic Targets for Alzheimer’s Disease. Biomol Ther. 2012, 20, 245–255. [Google Scholar] [CrossRef] [PubMed]
  259. Żukowska, J.; Moss, S.J.; Subramanian, V.; Acharya, K.R. Molecular Basis of Selective Amyloid-β Degrading Enzymes in Alzheimer’s Disease. FEBS J. 2023. [Google Scholar] [CrossRef]
  260. Yong, V.W. Metalloproteinases: Mediators of Pathology and Regeneration in the CNS. Nat. Rev. Neurosci. 2005, 6, 931–944. [Google Scholar] [CrossRef]
  261. Peress, N.; Perillo, E.; Zucker, S. Localization of Tissue Inhibitor of Matrix Metalloproteinases in Alzheimer’s Disease and Normal Brain. J. Neuropathol. Exp. Neurol. 1995, 54, 16–22. [Google Scholar] [CrossRef]
  262. Dunckley, T.; Beach, T.G.; Ramsey, K.E.; Grover, A.; Mastroeni, D.; Walker, D.G.; LaFleur, B.J.; Coon, K.D.; Brown, K.M.; Caselli, R.; et al. Gene Expression Correlates of Neurofibrillary Tangles in Alzheimer’s Disease. Neurobiol. Aging 2006, 27, 1359–1371. [Google Scholar] [CrossRef]
  263. Hoe, H.S.; Cooper, M.J.; Burns, M.P.; Lewis, P.A.; Van Der Brug, M.; Chakraborty, G.; Cartagena, C.M.; Pak, D.T.S.; Cookson, M.R.; Rebeck, G.W. The Metalloprotease Inhibitor TIMP-3 Regulates Amyloid Precursor Protein and Apolipoprotein E Receptor Proteolysis. J. Neurosci. 2007, 27, 10895–10905. [Google Scholar] [CrossRef] [PubMed]
  264. Trentini, A.; Manfrinato, M.C.; Castellazzi, M.; Bellini, T. Sex-Related Differences of Matrix Metalloproteinases (MMPs): New Perspectives for These Biomarkers in Cardiovascular and Neurological Diseases. J. Pers. Med. 2022, 12, 1196. [Google Scholar] [CrossRef] [PubMed]
  265. Rosenberg, G.A. Matrix Metalloproteinases and Their Multiple Roles in Neurodegenerative Diseases. Lancet Neurol. 2009, 8, 205–216. [Google Scholar] [CrossRef] [PubMed]
  266. Aksnes, M.; Edwin, T.H.; Saltvedt, I.; Eldholm, R.S.; Chaudhry, F.A.; Halaas, N.B.; Myrstad, M.; Watne, L.O.; Knapskog, A.B. Sex-Specific Associations of Matrix Metalloproteinases in Alzheimer’s Disease. Biol. Sex Differ. 2023, 14, 35. [Google Scholar] [CrossRef] [PubMed]
  267. Ni, J.; Xie, Z.; Quan, Z.; Meng, J.; Qing, H. How Brain “cleaners” Fail: Mechanisms and Therapeutic Value of Microglial Phagocytosis in Alzheimer’s Disease. Glia 2023. [Google Scholar] [CrossRef] [PubMed]
  268. Liang, S.; Domon, H.; Hosur, K.B.; Wang, M.; Hajishengallis, G. Age-Related Alterations in Innate Immune Receptor Expression and Ability of Macrophages to Respond to Pathogen Challenge In Vitro. Mech. Ageing Dev. 2009, 130, 538–546. [Google Scholar] [CrossRef]
  269. Yanguas-Casás, N.; Crespo-Castrillo, A.; Arevalo, M.; Garcia-Segura, L.M. Aging and Sex: Impact on Microglia Phagocytosis. Aging Cell 2020, 19, e13182. [Google Scholar] [CrossRef]
  270. Jiang, Y.; Zhou, X.; Wong, H.Y.; Ouyang, L.; Ip, F.C.F.; Chau, V.M.N.; Lau, S.-F.; Wu, W.; Wong, D.Y.K.; Seo, H.; et al. An IL1RL1 Genetic Variant Lowers Soluble ST2 Levels and the Risk Effects of APOE-Ε4 in Female Patients with Alzheimer’s Disease. Nat. Aging 2022, 2, 616–634. [Google Scholar] [CrossRef]
  271. Liu, C.C.; Wang, N.; Chen, Y.; Inoue, Y.; Shue, F.; Ren, Y.; Wang, M.; Qiao, W.; Ikezu, T.C.; Li, Z.; et al. Cell-Autonomous Effects of APOE4 in Restricting Microglial Response in Brain Homeostasis and Alzheimer’s Disease. Nat. Immunol. 2023, 24, 1854–1866. [Google Scholar] [CrossRef]
  272. Ulrich, J.D.; Ulland, T.K.; Mahan, T.E.; Nyström, S.; Peter Nilsson, K.; Song, W.M.; Zhou, Y.; Reinartz, M.; Choi, S.; Jiang, H.; et al. ApoE Facilitates the Microglial Response to Amyloid Plaque Pathology. J. Exp. Med. 2018, 215, 1047–1058. [Google Scholar] [CrossRef]
  273. Liu, C.C.; Zhao, N.; Fu, Y.; Wang, N.; Linares, C.; Tsai, C.W.; Bu, G. ApoE4 Accelerates Early Seeding of Amyloid Pathology. Neuron 2017, 96, 1024–1032. [Google Scholar] [CrossRef] [PubMed]
  274. Holtzman, D.M.; Bales, K.R.; Tenkova, T.; Fagan, A.M.; Parsadanian, M.; Sartorius, L.J.; Mackey, B.; Olney, J.; McKeel, D.; Wozniak, D.; et al. Apolipoprotein E Isoform-Dependent Amyloid Deposition and Neuritic Degeneration in a Mouse Model of Alzheimer’s Disease. Proc. Natl. Acad. Sci. USA 2000, 97, 2892–2897. [Google Scholar] [CrossRef] [PubMed]
  275. Giannakopoulos, P.; Herrmann, F.R.; Bussière, T.; Bouras, C.; Kövari, E.; Perl, D.P.; Morrison, J.H.; Gold, G.; Hof, P.R. Tangle and Neuron Numbers, but Not Amyloid Load, Predict Cognitive Status in Alzheimer’s Disease. Neurology 2003, 60, 1495–1500. [Google Scholar] [CrossRef] [PubMed]
  276. Jack, C.R.; Holtzman, D.M. Biomarker Modeling of Alzheimer’s Disease. Neuron 2013, 80, 1347–1358. [Google Scholar] [CrossRef] [PubMed]
  277. Musiek, E.S.; Holtzman, D.M. Three Dimensions of the Amyloid Hypothesis: Time, Space, and “Wingmen”. Nat. Neurosci. 2015, 18, 800–806. [Google Scholar] [CrossRef] [PubMed]
  278. Iqbal, K.; Liu, F.; Gong, C.-X.; Grundke-Iqbal, I. Tau in Alzheimer Disease and Related Tauopathies. Curr. Alzheimer Res. 2010, 7, 656. [Google Scholar] [CrossRef] [PubMed]
  279. Gong, C.-X.; Iqbal, K. Hyperphosphorylation of Microtubule-Associated Protein Tau: A Promising Therapeutic Target for Alzheimer Disease. Curr. Med. Chem. 2008, 15, 2321–2328. [Google Scholar] [CrossRef] [PubMed]
  280. Kopke, E.; Tung, Y.-C.; Shaikh, S.; Alonso, A.D.C.; Iqbal, K.; Grundke-Iqbals, I. Microtubule-Associated Protein Tau: Abnormal Phosphorylation of a Non-Paired Helical Filament Pool in Alzheimer’s Disease. J. Biol. Chem. 1993, 268, 24374–24384. [Google Scholar] [CrossRef]
  281. Metaxas, A.; Kempf, S.J. Neurofibrillary Tangles in Alzheimer’s Disease: Elucidation of the Molecular Mechanism by Immunohistochemistry and Tau Protein Phospho-Proteomics. Neural Regen. Res. 2016, 11, 1579–1581. [Google Scholar] [CrossRef]
  282. Wang, J.Z.; Gong, C.X.; Zaidi, T.; Grundke-Iqbal, I.; Iqbal, K. Dephosphorylation of Alzheimer Paired Helical Filaments by Protein Phosphatase-2A and -2B. J. Biol. Chem. 1995, 270, 4854–4860. [Google Scholar] [CrossRef]
  283. Buckley, R.F.; Mormino, E.C.; Rabin, J.S.; Hohman, T.J.; Landau, S.; Hanseeuw, B.J.; Jacobs, H.I.L.; Papp, K.V.; Amariglio, R.E.; Properzi, M.J.; et al. Sex Differences in the Association of Global Amyloid and Regional Tau Deposition Measured by Positron Emission Tomography in Clinically Normal Older Adults. JAMA Neurol. 2019, 76, 542–551. [Google Scholar] [CrossRef] [PubMed]
  284. Lindbergh, C.A.; Casaletto, K.B.; Staffaroni, A.M.; La Joie, R.; Iaccarino, L.; Edwards, L.; Tsoy, E.; Elahi, F.; Walters, S.M.; Cotter, D.; et al. Sex-Related Differences in the Relationship Between β-Amyloid and Cognitive Trajectories in Older Adults. Neuropsychology 2020, 34, 835–850. [Google Scholar] [CrossRef] [PubMed]
  285. Luchsinger, J.A.; Palta, P.; Rippon, B.; Soto, L.; Ceballos, F.; Pardo, M.; Laing, K.; Igwe, K.; Johnson, A.; Tomljanovic, Z.; et al. Sex Differences in in Vivo Alzheimer’s Disease Neuropathology in Late Middle-Aged Hispanics. J. Alzheimer’s Dis. 2020, 74, 1243–1252. [Google Scholar] [CrossRef] [PubMed]
  286. Wisch, J.K.; Meeker, K.L.; Gordon, B.A.; Flores, S.; Dincer, A.; Grant, E.A.; Benzinger, T.L.; Morris, J.C.; Ances, B.M. Sex-Related Differences in Tau Positron Emission Tomography (PET) and the Effects of Hormone Therapy (HT). Alzheimer Dis. Assoc. Disord. 2021, 35, 164–168. [Google Scholar] [CrossRef]
  287. Palta, P.; Rippon, B.; Tahmi, M.; Pardo, M.; Johnson, A.; Tomljanovic, Z.; He, H.; Laing, K.K.; Razlighi, Q.R.; Teresi, J.A.; et al. Sex Differences in In-Vivo Tau Neuropathology in a Multi-Ethnic Sample of Late Middle-Aged Adults. Neurobiol. Aging 2021, 103, 109–116. [Google Scholar] [CrossRef]
  288. Buckley, R.F.; O’Donnell, A.; McGrath, E.R.; Jacobs, H.I.L.; Lois, C.; Satizabal, C.L.; Ghosh, S.; Rubinstein, Z.B.; Murabito, J.M.; Sperling, R.A.; et al. Menopause Status Moderates Sex Differences in Tau Burden: A Framingham PET Study. Ann. Neurol. 2022, 92, 11–22. [Google Scholar] [CrossRef]
  289. Coughlan, G.T.; Betthauser, T.J.; Boyle, R.; Koscik, R.L.; Klinger, H.M.; Chibnik, L.B.; Jonaitis, E.M.; Yau, W.-Y.W.; Wenzel, A.; Christian, B.T.; et al. Association of Age at Menopause and Hormone Therapy Use with Tau and β-Amyloid Positron Emission Tomography. JAMA Neurol. 2023, 80, 462. [Google Scholar] [CrossRef]
  290. Tamburini, B.; Badami, G.D.; La Manna, M.P.; Shekarkar Azgomi, M.; Caccamo, N.; Dieli, F. Emerging Roles of Cells and Molecules of Innate Immunity in Alzheimer’s Disease. Int. J. Mol. Sci. 2023, 24, 11922. [Google Scholar] [CrossRef]
  291. Pampuscenko, K.; Morkuniene, R.; Krasauskas, L.; Smirnovas, V.; Brown, G.C.; Borutaite, V. Extracellular Tau Stimulates Phagocytosis of Living Neurons by Activated Microglia via Toll-like 4 Receptor–NLRP3 Inflammasome–Caspase-1 Signalling Axis. Sci. Rep. 2023, 13, 10813. [Google Scholar] [CrossRef]
  292. Udeochu, J.C.; Amin, S.; Huang, Y.; Fan, L.; Torres, E.R.S.; Carling, G.K.; Liu, B.; McGurran, H.; Coronas-Samano, G.; Kauwe, G.; et al. Tau Activation of Microglial CGAS–IFN Reduces MEF2C-Mediated Cognitive Resilience. Nat. Neurosci. 2023, 26, 737–750. [Google Scholar] [CrossRef]
  293. Wang, C.; Fan, L.; Khawaja, R.R.; Liu, B.; Zhan, L.; Kodama, L.; Chin, M.; Li, Y.; Le, D.; Zhou, Y.; et al. Microglial NF-ΚB Drives Tau Spreading and Toxicity in a Mouse Model of Tauopathy. Nat. Commun. 2022, 13, 1969. [Google Scholar] [CrossRef] [PubMed]
  294. Wang, Y.; Wu, T.; Tsai, M.C.; Rezzonico, M.G.; Abdel-Haleem, A.M.; Xie, L.; Gandham, V.D.; Ngu, H.; Stark, K.; Glock, C.; et al. TPL2 Kinase Activity Regulates Microglial Inflammatory Responses and Promotes Neurodegeneration in Tauopathy Mice. Elife 2023, 12, e83451. [Google Scholar] [CrossRef] [PubMed]
  295. Pan, L.; Cho, K.S.; Wei, X.; Xu, F.; Lennikov, A.; Hu, G.; Tang, J.; Guo, S.; Chen, J.; Kriukov, E.; et al. IGFBPL1 Is a Master Driver of Microglia Homeostasis and Resolution of Neuroinflammation in Glaucoma and Brain Tauopathy. Cell Rep. 2023, 42, 112889. [Google Scholar] [CrossRef] [PubMed]
  296. Chen, X.; Firulyova, M.; Manis, M.; Herz, J.; Smirnov, I.; Aladyeva, E.; Wang, C.; Bao, X.; Finn, M.B.; Hu, H.; et al. Microglia-Mediated T Cell Infiltration Drives Neurodegeneration in Tauopathy. Nature 2023, 615, 668–677. [Google Scholar] [CrossRef] [PubMed]
  297. Lee, J.; Dimitry, J.M.; Song, J.H.; Son, M.; Sheehan, P.W.; King, M.W.; Travis Tabor, G.; Goo, Y.A.; Lazar, M.A.; Petrucelli, L.; et al. Microglial REV-ERBα Regulates Inflammation and Lipid Droplet Formation to Drive Tauopathy in Male Mice. Nat. Commun. 2023, 14, 5197. [Google Scholar] [CrossRef] [PubMed]
  298. Van Dyck, C.H.; Nygaard, H.B.; Chen, K.; Donohue, M.C.; Raman, R.; Rissman, R.A.; Brewer, J.B.; Koeppe, R.A.; Chow, T.W.; Rafii, M.S.; et al. Effect of AZD0530 on Cerebral Metabolic Decline in Alzheimer Disease: A Randomized Clinical Trial. JAMA Neurol. 2019, 76, 1219–1229. [Google Scholar] [CrossRef]
  299. Ji, C.; Sigurdsson, E.M. Current Status of Clinical Trials on Tau Immunotherapies. Drugs 2021, 81, 1135–1152. [Google Scholar] [CrossRef]
  300. Jin, M.; Shepardson, N.; Yang, T.; Chen, G.; Walsh, D.; Selkoe, D.J. Soluble Amyloid Beta-Protein Dimers Isolated from Alzheimer Cortex Directly Induce Tau Hyperphosphorylation and Neuritic Degeneration. Proc. Natl. Acad. Sci. USA 2011, 108, 5819–5824. [Google Scholar] [CrossRef]
  301. Oddo, S.; Billings, L.; Kesslak, J.P.; Cribbs, D.H.; LaFerla, F.M. Aβ Immunotherapy Leads to Clearance of Early, but Not Late, Hyperphosphorylated Tau Aggregates via the Proteasome. Neuron 2004, 43, 321–332. [Google Scholar] [CrossRef]
  302. Tarasoff-Conway, J.M.; Carare, R.O.; Osorio, R.S.; Glodzik, L.; Butler, T.; Fieremans, E.; Axel, L.; Rusinek, H.; Nicholson, C.; Zlokovic, B.V.; et al. Clearance Systems in the Brain—Implications for Alzheimer Disease. Nat. Rev. Neurol. 2015, 11, 457–470. [Google Scholar] [CrossRef]
  303. Ihara, Y.; Morishima-Kawashima, M.; Nixon, R. The Ubiquitin-Proteasome System and the Autophagic-Lysosomal System in Alzheimer Disease. Cold Spring Harb. Perspect. Med. 2012, 2, a006361. [Google Scholar] [CrossRef] [PubMed]
  304. Lee, M.J.; Lee, J.H.; Rubinsztein, D.C. Tau Degradation: The Ubiquitin–Proteasome System versus the Autophagy-Lysosome System. Prog. Neurobiol. 2013, 105, 49–59. [Google Scholar] [CrossRef] [PubMed]
  305. Ciechanover, A.; Kwon, Y.T. Degradation of Misfolded Proteins in Neurodegenerative Diseases: Therapeutic Targets and Strategies. Exp. Mol. Med. 2015, 47, e147. [Google Scholar] [CrossRef] [PubMed]
  306. Yan, Y.; Wang, X.; Chaput, D.; Shin, M.-K.; Koh, Y.; Gan, L.; Pieper, A.A.; Woo, J.-A.A.; Kang, D.E. X-Linked Ubiquitin-Specific Peptidase 11 Increases Tauopathy Vulnerability in Women. Cell 2022, 185, 3913–3930. [Google Scholar] [CrossRef] [PubMed]
  307. Koutsodendris, N.; Blumenfeld, J.; Agrawal, A.; Traglia, M.; Grone, B.; Zilberter, M.; Yip, O.; Rao, A.; Nelson, M.R.; Hao, Y.; et al. Neuronal APOE4 Removal Protects against Tau-Mediated Gliosis, Neurodegeneration and Myelin Deficits. Nat. Aging 2023, 3, 275–296. [Google Scholar] [CrossRef]
  308. Wang, C.; Xiong, M.; Gratuze, M.; Artyomov, M.; Ulrich, J.D.; Holtzman, D.M. Selective Removal of Astrocytic APOE4 Strongly Protects against Tau-Mediated Neurodegeneration and Decreases Synaptic Phagocytosis by Microglia. Neuron 2021, 109, 1657–1674. [Google Scholar] [CrossRef] [PubMed]
  309. Seo, D.; O’Donnell, D.; Jain, N.; Ulrich, J.D.; Herz, J.; Li, Y.; Lemieux, M.; Cheng, J.; Hu, H.; Serrano, J.R.; et al. ApoE Isoform- and Microbiota-Dependent Progression of Neurodegeneration in a Mouse Model of Tauopathy. Science 2023, 379, eadd1236. [Google Scholar] [CrossRef] [PubMed]
  310. Demetrius, L.A.; Eckert, A.; Grimm, A. Sex Differences in Alzheimer’s Disease: Metabolic Reprogramming and Therapeutic Intervention. Trends Endocrinol. Metab. 2021, 32, 963–979. [Google Scholar] [CrossRef]
  311. Bouter, C.; Irwin, C.; Franke, T.N.; Beindorff, N.; Bouter, Y. Quantitative Brain Positron Emission Tomography in Female 5XFAD Alzheimer Mice: Pathological Features and Sex-Specific Alterations. Front. Med. (Lausanne) 2021, 8, 745064. [Google Scholar] [CrossRef]
  312. Jullienne, A.; Szu, J.I.; Quan, R.; Trinh, M.V.; Norouzi, T.; Noarbe, B.P.; Bedwell, A.A.; Eldridge, K.; Persohn, S.C.; Territo, P.R.; et al. Cortical Cerebrovascular and Metabolic Perturbations in the 5xFAD Mouse Model of Alzheimer’s Disease. Front. Aging Neurosci. 2023, 15, 1220036. [Google Scholar] [CrossRef]
  313. Robison, L.S.; Gannon, O.J.; Salinero, A.E.; Abi-Ghanem, C.; Kelly, R.D.; Riccio, D.A.; Mansour, F.M.; Zuloaga, K.L. Sex Differences in Metabolic Phenotype and Hypothalamic Inflammation in the 3xTg-AD Mouse Model of Alzheimer’s Disease. Biol. Sex Differ. 2023, 4, 51. [Google Scholar] [CrossRef] [PubMed]
  314. Ceasrine, A.M.; Bilbo, S.D. Dietary Fat: A Potent Microglial Influencer. Trends Endocrinol. Metab. 2022, 33, 196–205. [Google Scholar] [CrossRef] [PubMed]
  315. Daly, C.M.; Saxena, J.; Singh, J.; Bullard, M.R.; Bondy, E.O.; Saxena, A.; Buffalino, R.E.; Melville, M.F.; Freeman, L.R. Sex Differences in Response to a High Fat, High Sucrose Diet in Both the Gut Microbiome and Hypothalamic Astrocytes and Microglia. Nutr. Neurosci. 2022, 25, 321. [Google Scholar] [CrossRef] [PubMed]
  316. López-Gambero, A.J.; Pacheco-Sánchez, B.; Rosell-Valle, C.; Medina-Vera, D.; Navarro, J.A.; Del Mar Fernández-Arjona, M.; De Ceglia, M.; Sanjuan, C.; Simon, V.; Cota, D.; et al. Dietary Administration of D-Chiro-Inositol Attenuates Sex-Specific Metabolic Imbalances in the 5xFAD Mouse Model of Alzheimer’s Disease. Biomed. Pharmacother. 2022, 150, 112994. [Google Scholar] [CrossRef]
  317. Shippy, D.C.; Ulland, T.K. Lipid Metabolism Transcriptomics of Murine Microglia in Alzheimer’s Disease and Neuroinflammation. Sci. Rep. 2023, 13, 14800. [Google Scholar] [CrossRef]
Figure 1. Summary figure of microglial sex differences contributing to the pathological features of Alzheimer’s disease discussed in this review. The composition of sex chromosomes (XX/XY) and/or hormones contribute to sex-specific features of microglia, including their number (#), morphology, gene and protein expression, and function. These differences may promote or inhibit sex-specific processes and pathways in neurons, amyloid plaque load, and tau tangle pathology to bias disease onset and progression in a sex-specific manner. Created with BioRender.com (accessed on 4 December 2023).
Figure 1. Summary figure of microglial sex differences contributing to the pathological features of Alzheimer’s disease discussed in this review. The composition of sex chromosomes (XX/XY) and/or hormones contribute to sex-specific features of microglia, including their number (#), morphology, gene and protein expression, and function. These differences may promote or inhibit sex-specific processes and pathways in neurons, amyloid plaque load, and tau tangle pathology to bias disease onset and progression in a sex-specific manner. Created with BioRender.com (accessed on 4 December 2023).
Ijms 24 17377 g001
Figure 2. Sex-specific expression patterns of transcription factors, follicle-stimulating hormone (FSH), and mGluR5 contribute to differential patterns in neuronal survival and overall brain atrophy. Pink boxes represent female-specific effects, while blue boxes represent male-specific effects. Arrows represent altered expression levels contributing to AD risk/presentation. Yellow boxes represent processes that exist within both sexes. Created with BioRender.com (accessed on 4 December 2023).
Figure 2. Sex-specific expression patterns of transcription factors, follicle-stimulating hormone (FSH), and mGluR5 contribute to differential patterns in neuronal survival and overall brain atrophy. Pink boxes represent female-specific effects, while blue boxes represent male-specific effects. Arrows represent altered expression levels contributing to AD risk/presentation. Yellow boxes represent processes that exist within both sexes. Created with BioRender.com (accessed on 4 December 2023).
Ijms 24 17377 g002
Figure 3. Sex-specific processes contributing to differential amyloid plaque load through either Aβ production or Aβ clearance mechanisms. Burgundy boxes represent female-specific processes, while blue boxes represent male-specific processes. Created with BioRender.com (accessed on 4 December 2023).
Figure 3. Sex-specific processes contributing to differential amyloid plaque load through either Aβ production or Aβ clearance mechanisms. Burgundy boxes represent female-specific processes, while blue boxes represent male-specific processes. Created with BioRender.com (accessed on 4 December 2023).
Ijms 24 17377 g003
Table 1. Sex differences in clinical phenotypes observed in AD subtypes.
Table 1. Sex differences in clinical phenotypes observed in AD subtypes.
AD Subtype *Clinical MetricSex Difference *References
EOADTau burdenF > M[29]
Brain atrophy, cognition at diagnosisF > M[30]
Rate of neurodegeneration and impairmentF > M[31,32]
Cognitive resilienceF > M[33]
LOADRate of hippocampal volume lossF > M[12]
Brain glucose hypometabolismF > M[34]
Rate of cognitive decline, progression to dementiaF > M[4,6,7,12,35,36,37,38,39]
* EOAD, early-onset AD; LOAD, late-onset AD; F, females; and M, males.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Reed, E.G.; Keller-Norrell, P.R. Minding the Gap: Exploring Neuroinflammatory and Microglial Sex Differences in Alzheimer’s Disease. Int. J. Mol. Sci. 2023, 24, 17377. https://doi.org/10.3390/ijms242417377

AMA Style

Reed EG, Keller-Norrell PR. Minding the Gap: Exploring Neuroinflammatory and Microglial Sex Differences in Alzheimer’s Disease. International Journal of Molecular Sciences. 2023; 24(24):17377. https://doi.org/10.3390/ijms242417377

Chicago/Turabian Style

Reed, Erin G., and Phaedra R. Keller-Norrell. 2023. "Minding the Gap: Exploring Neuroinflammatory and Microglial Sex Differences in Alzheimer’s Disease" International Journal of Molecular Sciences 24, no. 24: 17377. https://doi.org/10.3390/ijms242417377

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop