Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2020 Jul 1.
Published in final edited form as: Brain Behav Immun. 2019 Mar 11;79:39–55. doi: 10.1016/j.bbi.2019.03.010

Neuroimmunology of the female brain across the lifespan: plasticity to psychopathology

RM Barrientos 1,2,3, PJ Brunton 4,5, KM Lenz 1,6, L Pyter 1,2, SJ Spencer 7,*
PMCID: PMC6591071  NIHMSID: NIHMS1523922  PMID: 30872093

Abstract

The female brain is highly dynamic and can fundamentally remodel throughout the normal ovarian cycle as well as in critical life stages including perinatal development, pregnancy and old-age. As such, females are particularly vulnerable to infections, psychological disorders, certain cancers, and cognitive impairments. We will present the latest evidence on the female brain; how it develops through the neonatal period; how it changes through the ovarian cycle in normal individuals; how it adapts to pregnancy and postpartum; how it responds to illness and disease, particularly cancer; and, finally, how it is shaped by old age. Throughout, we will highlight female vulnerability to and resilience against disease and dysfunction in the face of environmental challenges.

1. Introduction

The female brain is remarkably dynamic, responding uniquely to the early life environment, aging and to various life stages in between. In this review we will highlight the key themes the psychoneuroimmunology research community is contributing to the understanding of the female brain (Figure 1). In particular, we focus on specific life stages. First, we outline how the female brain develops and how this development can be perturbed by environmental influences, particularly by neuroimmune challenges. Next, we address how the healthy adult female brain is dynamically remodeled as circulating sex hormones change. Pregnancy is an important part of many females’ lives wherein the neuroimmune system changes fundamentally to protect the growing fetus and we discuss the latest work on how the brain adapts. As females mature and age, they face unique challenges, including susceptibility to sex hormone-responsive diseases (and treatments) around menopause that are associated with persistent behavioral comorbidities, like breast cancer and age-related cognitive decline. Here we outline some of the female-specific psychoneuroimmune changes that occur at these times (Figure 1). With this review, it is our deliberate intention to present what is known about female neuroimmunology without, where possible, invoking a comparison to males. Until very recently, there has been a conspicuous paucity of research focusing solely on females, despite meta-analysis demonstrating female mouse models are not more variable than males, even when cycle stage is not controlled for (Prendergast et al., 2014). For example, with the exception of reproductive topics, female-only studies accounted for 20% or fewer of the papers published in 2009 (Beery and Zucker, 2011). As our discussion shows, female brains and neuroimmune responses can be very different from those of males. However, simply pursuing a comparative approach in research design is restrictive as it tends to limit any investigation of females to how they do, or do not, differ from males and discourages exploration of female-unique responses. Thus, neglecting to specifically study females in biomedical research restricts our understanding of women’s health and disease progression to assumptions that they are indistinct from that of men, and limits the possibilities of creating female-specific therapies and pharmaceuticals. This oversight may be very costly, not just in terms of research and healthcare dollars, but also in terms of human suffering and understanding potential applied health advantages of females (Krukowski et al., 2018; Rummel, 2018). We envisage that as our understanding of female psychoneuroimmunology progresses, so will our capacity to tailor drug delivery systems to accommodate monthly and even circadian sex hormone fluctuations; to prevent pre-term birth by strategically enhancing a woman’s adaptive neuroimmune responses to pregnancy; and to identify and mimic how females are relatively protected against early life challenges. The findings discussed here suggest we are beginning to make headway into this important topic.

Figure 1.

Figure 1.

Summary diagram of the key changes in the psychoneuroimmunology of the female brain across the lifespan. Across the lifespan, from the perinatal stage to advanced age, females exhibit dramatic shifts in sex steroid hormones. These range from very low (during the perinatal period), to moderate, oscillating levels (during young adulthood), to very high (during late pregnancy), to precipitously declining levels (with cancer treatments) and finally, to gradually declining levels (in menopause and into advanced age). These hormonal shifts play an important role in the behavioral phenotype demonstrated at each developmental stage, altering behaviors such as mood and cognition. Preclinical studies have informed our understanding of the neuroimmune phenotype that is characteristic of each stage of life, and which may be modulated by fluctuations in sex hormones. Reproduced with the permission of The Ohio State University.

2. Perinatal development of the female brain

The basics of female brain development

In mammals, female-typical brain development begins with the process of sex determination, driven by sex chromosomes. With a sex chromosome complement of XX, female development proceeds along a ‘default process’, so called because it occurs in the absence of active hormonal signals, but not because it does not involve active regulation by sex chromosome genes (Blecher and Erickson, 2007). In the absence of intervening signals from the Y chromosome, primordial gonadal tissue differentiates into the ovaries (Blecher and Erickson, 2007). The ovaries are relatively quiescent during development in comparison to the testes, only beginning to secrete substantial amounts of the female typical hormones, estrogens and progestins, at puberty (Jost et al., 1973). Despite this quiescence, female mice that lack aromatase, the enzyme responsible for producing estrogens, display disrupted sociosexual behavior in adulthood; but these behavioral changes can be normalized by prepubertal treatment with estrogens (Bakker and Baum, 2008; Brock et al., 2011). These data indicate that female-typical hormones contribute to female brain development early on (Blecher and Erickson, 2007; McCarthy and Arnold, 2011). The source of the hormones in question could be extragonadal, as the brain synthesizes significant amounts of estrogens (Konkle and McCarthy, 2011; McCarthy, 2008).

One X chromosome in each female cell is silenced via the process of X-inactivation (Gartler and Riggs, 1983). Thus, every female tissue is mosaic, either expressing paternal or maternal X-chromosome genes. The pattern of X-inactivation across brain regions and neuron subtypes is not uniform (Gregg et al., 2010; Wu et al., 2014), and such mosaicism may be a major (and totally unexplored) source of variability across brain cells that is relevant for individual differences in the behavioral function of females. The process of X-inactivation is also not entirely complete, and both immune and epigenetic regulatory genes have been shown to escape inactivation in different female tissues (Carrel and Willard, 2005). There is a high concentration of immune-related genes on the X chromosome, and, if they escape inactivation, these could underlie the female-biased risk for autoimmunity (Bianchi et al., 2012). A fascinating study in mice shows the maternal X gene for interleukin (IL)-18 is more highly expressed in the female prefrontal cortex than in the male (Gregg et al., 2010), but the mechanistic role of this or other X-chromosome immune genes have not been explored in the context of female brain development.

Neuroimmune contributors to normal female brain and behavioral development

Neuroimmune cells are key mediators of normal brain development. Microglia, the brain’s resident immune cells, in particular, are crucial to regulating developmental cell genesis (Cunningham et al., 2013; Shigemoto-Mogami et al., 2014), apoptosis (Marin-Teva et al., 2004; Wakselman et al., 2008), myelination (Hagemeyer et al., 2017), synaptogenesis and synaptic pruning (Miyamoto et al., 2016; Paolicelli et al., 2011; Schafer et al., 2012; Weinhard et al., 2018a). The process of sexual differentiation, too, is dependent upon neuroimmune cells and their signaling. For example, the anteroventral periventricular area of the hypothalamus (AVPV), is larger in female rodents than in males, and is crucial for female hormonal cycling later in life (Semaan and Kauffman, 2010; Sumida et al., 1993). Higher rates of developmental cell survival in the AVPV underlie this sex difference (Waters and Simerly, 2009), and the neuroimmune mediators, tumor necrosis factor (TNF) receptor 2 as well as nuclear factor κ-B (NFκB) protect neurons from cell death to lead to this brain region developing in a female-typical manner (Krishnan et al., 2009). In the nearby medial preoptic area (POA) of the hypothalamus, females have fewer activated microglia (microglia that are hypo-ramified, phagocytic, and inflammatory (Davalos et al., 2005; Nimmerjahn et al., 2005; Sominsky et al., 2018)) and mast cells than males, and active signaling by neuroimmune cells contributes to brain masculinization (Lenz et al., 2013; Lenz et al., 2018). But it is entirely possible that these cells are also crucial for the feminization process, given that the POA regulates maternal behavior and female hormonal cycling.

Females are also protected from male-typical brain sexual differentiation via epigenetic means. The POA is responsible for both female estrous cycling and maternal behavior as well as male copulatory behavior (Lenz et al., 2012). DNA methylation levels are higher in the female POA than in the male and interfering with methylation in females leads to significant increases in male-typical gene expression and masculinization of sexual behavior (Nugent et al., 2015). Treating females with an inhibitor of DNA methylation enzymes leads to masculinization of sexual behavior (Nugent et al., 2015), but also the expression of immune-related genes increases four-fold in the female brain (McCarthy et al., 2017), meaning that many of the genes that are epigenetically silenced in the female brain are related to the immune system. It follows that early life immune activation could elevate expression for these genes that are typically silenced, and thereby perturb the normal process of brain feminization.

During development, female microglia across many brain regions, including the hippocampus, amygdala, parietal cortex and hypothalamus, show a more ameboid morphology than they do in later life (Schwarz et al., 2012). Female microglia also show a transient peak in phagocytic tone in the developing hippocampus relative to males, including greater numbers of phagocytic cups on microglia and increased expression of genes related to phagocytosis, including Cd68, Tyrobp, Trem2, and Cybb (Nelson et al., 2017; Weinhard et al., 2018b). Microglia phagocytic cups target neural progenitor cells for phagocytic clearance in females more than in males (Nelson et al., 2017). Increased expression of phagocytic markers correlates, but has not yet been mechanistically linked, to lower synaptic marker density in females prior to puberty (Weinhard et al., 2018b).

Cells of the innate and acquired immune system have also been linked to female behavioral development. Temporary depletion of microglia in the first postnatal week, using liposomal clodronate, leads to decreased anxiety-like behavior, decreased behavioral despair, and decreased acute stress responsivity in adult females (Nelson and Lenz, 2017b; VanRyzin et al., 2016), suggesting that microglia contribute to the normal development of these behaviors. Although most studies of neuroimmune function and brain development have focused on innate immune cells, there is increasing evidence that cells of the acquired immune system, such as T cells, also contribute to brain and behavioral function, possibly via signaling in the meninges and across the blood brain barrier (Kipnis, 2016). T-cells have recently been implicated in female-typical brain development. Female T-cell receptor knockout mice lack T-cells congenitally, and show both increased volume of the bed nucleus of the stria terminalis (BNST) and amygdala relative to wildtype females, as well as decreased hippocampal volume and decreased anxiety-like behavior as adults (Rilett et al., 2015). The brain regions impacted by congenital T-cell deficiency are crucial to sociosexual behavior, the stress response, learning, and anxiety behavior, but the mechanisms through which T-cells impact behavioral development are unknown.

Female response to early life perturbations

Given that immune cells contribute to normal brain development in females, it becomes obvious that early life perturbations that impact neuroimmune function may program brain development and lifelong behavior. Animal studies have used various early life immune challenge paradigms to model this risk and determine the underlying mechanisms through which immune activation shifts behavioral development (Estes and McAllister, 2016; Meyer, 2014; Patterson, 2009). To challenge the developing immune system, many studies have used the bacterial endotoxin, lipopolysaccharide (LPS), delivered either systemically to pregnant dams, locally into the uterus, or systemically to offspring, but other commonly used challenges are the viral mimetic, Poly I:C, live bacterial infection (E. coli), or exogenous cytokine treatment. Likewise, other early life perturbations, such as perinatal stress exposure, early life overnutrition, maternal obesity, maternal or neonatal ethanol exposure, maternal allergic asthma, and exposure to environmental pollutant exposure and neonatal pain or injury, influence female neural and behavioral development in part by eliciting neuroinflammation or perturbing normal immune responses in the brain, as will be discussed below.

Making hard and fast conclusions about the effects of early life inflammation on brain development is challenging given the large numbers of studies performed, differences in inflammogens used, timing of challenge, and the endpoints assessed. This has led to a call for early life inflammation studies to be performed with greater rigor to facilitate comparisons across studies (Kentner et al., 2018). In general, females show a mild but measurable acute pro-inflammatory response within the brain in response to perinatal inflammatory challenges, including increased gene expression for pro-inflammatory cytokines and changes in microglia staining density, number, and expression of phagocytic markers (for thorough recent reviews of sex-specific effects of early life inflammation and mechanisms of inflammation-induced brain development, refer to (Bilbo et al., 2018; Nelson and Lenz, 2017a; Sominsky et al., 2018)). However, in some cases, the female neuroimmune response to early life perturbations is not in the ‘expected’ pro-inflammatory direction. For example, neonatal overnutrition decreases inflammatory gene expression in the developing hypothalamus of females, but eventually leads to adult increases in both microglia number, and inflammatory gene expression in the adult hypothalamus (Ziko et al., 2014). Clearly, it is crucial to assess females at different developmental stages following early life perturbations before we can make any broader conclusions about how neuroimmune function is impacted by experience.

Careful studies of female microglial function after inflammation have yielded some insight into the mechanisms through which early life inflammation impacts brain development. In response to prenatal Poly I:C exposure, electron microscopy analysis of microglia in females show that they increase association with myelinated axons (Hui et al., 2018). Interestingly, a study of myelination after intrauterine inflammation showed decreased myelin basic protein in the brains of female offspring (Makinson et al., 2017), suggesting a possible link between inflammation-induced activation of microglia and decreased myelination. Exposure to allergic asthma in utero leads to altered DNA methylation in microglia, specifically in gene modules that have been associated with neurodevelopmental disorders in humans (Vogel Ciernia et al., 2018). Female microglia also have a faster baseline maturation rate than males at the transcriptomic level, and their maturation is not accelerated by immune challenge with LPS, as it is in males, which may reflect female resilience to early life inflammation (Hanamsagar et al., 2017). These studies suggest that the normal developmental functions of female microglia are disrupted by early life perturbations. This may be a cause of behavioral changes seen in females following early life immune activation, though few studies to date have mechanistically linked microglial dysfunction after inflammation with shifts in the trajectory of behavioral development.

After various early life inflammation exposures, females show deficits in memory tasks, motor function, social interaction and/or social preferences, increased anxiety-like behavior, and sensory gating or response inhibition deficits, and vulnerability to arthritis (Aavani et al., 2015; de Theije et al., 2015; Henderson et al., 2017; Hui et al., 2018; Lin et al., 2012; Makinson et al., 2017; Meyer et al., 2008; Ratnayake et al., 2013; Smith et al., 2007; Vogel Ciernia et al., 2018; Zhang et al., 2012). Yet, in many other cases, females show resilience, especially relative to males. For example, both prenatal exposure to LPS or, later, LPS challenge in the second postnatal week, leads to decreases in juvenile social play in males, but does not impact play in females (Hoffman et al., 2016; Taylor et al., 2012). Following early prenatal stress exposure, males but not females show hyperactivity, anhedonia, and heightened hypothalamic-pituitary-adrenal (HPA) axis responses to stress (Bronson and Bale, 2014; Mueller and Bale, 2008). Similarly, postnatal overfeeding leads to long-term changes in satiety signaling in males that are not seen in females (Ziko et al., 2017). Interestingly, the female placenta does not mount nearly as robust an inflammatory response to stress as the male placenta and shows epigenetic differences from males that lead to stress-buffering effects, which may indicate that the placenta is a key factor in female resilience (Nugent and Bale, 2015; Nugent et al., 2018). The maternal microbiome may also play a role here, in that eliminating the dam’s microbiome in mice can lead to a sex-specific impact on the microglial transcriptome, with a minimal early effect in females (Thion et al., 2018).

A single inflammatory exposure is often not sufficient to produce long-term behavioral consequences, but isolated early life inflammatory exposures may ‘prime’ the immune system to react more robustly to subsequent ‘second hits’ of inflammatory events (stress, toxin exposure, immune challenge) later in life. In the context of multiple hits of early life inflammation, females again display resilience. For example, prenatal diesel particulate exposure coupled with early life stress, leads to elevated pro-inflammatory mediators in the male brain (Bolton et al., 2013); but in the female brain, the same double hit leads to elevations in the anti-inflammatory cytokine, IL-10 (Bolton et al., 2013). This suggests that the immune system response in the female brain may actually buffer against later behavioral effects via anti-inflammatory pathways. Indeed, while both males and females showed elevated anxiety-like behavior following this double hit, females show no evidence of learning deficits, whereas males do (Bolton et al., 2013). The same group has demonstrated that females exposed to both air pollution and high-fat diet prenatally do not display metabolic changes and insulin resistance, whereas males do (Bolton et al., 2014). Both early life exposure to intrauterine inflammation and prenatal alcohol (Makinson et al., 2017; Terasaki and Schwarz, 2016) similarly lead to enhanced inflammatory responses to later life immune challenge in males, but not in females. Thus, female resilience appears to be a recurrent theme across diverse early life inflammation studies.

There are of course exceptions to this generality. For example, female rats show a larger inflammatory response than males in adulthood following neonatal inflammatory injury (LaPrairie and Murphy, 2007). Similarly, females show a robust potentiation of experimental arthritis induction in adulthood following prenatal ethanol exposure, accompanied by T cell proliferation and peripheral inflammation (Zhang et al., 2012). These data match with the known female susceptibility to both pain and autoimmune conditions (Fairweather et al., 2008; Rosen et al., 2017). These ‘exceptions’ to female resilience lead to an interesting question that deserves careful research attention: To what extent do females show a ‘quantitative’ difference in the inflammatory response following early life immune activation, versus a ‘qualitative’ difference relative to males? This qualitative difference could lead females to be resilient to certain perturbations but more vulnerable to others. There are data from the adult sex differences literature to support this idea. For example, in adult rodents, females and males both mount robust mechanical pain hypersensitivity, but in females, this pain response is mediated by T cells, and in males, it is mediated by microglia (Sorge et al., 2015). In a prenatal immune activation model that activates the toll-like receptor (TLR)7 pathway and leads to equal changes in microglia density and morphology in males and females, female mice show many fewer gene expression changes in the striatum than males (a ‘quantitative’ sex difference), and male and female mice show highly divergent gene expression responses in the brain (a ‘qualitative’ sex difference) (Missig et al., 2019). Thus, future research must look for both quantitative and qualitative differences between the sexes and across windows of development to best understand female-specific neurobiology, both in the context of normal function, and in the context of resilience and risk to neurological dysfunction.

Early life inflammation is a risk factor for neurodevelopmental disorders, such as autism spectrum disorder, schizophrenia, attention deficit hyperactivity disorder (ADHD), and Tourette’s syndrome (Estes and McAllister, 2016). Most of these same disorders affect females at a much lower rate than males (McCarthy et al., 2017). In the case of autism spectrum disorder, there is an hypothesis that being female is protective, in that females with autism typically have much higher genetic load than affected males (Jacquemont et al., 2014; Robinson et al., 2013). It is possible that blunted female neuroimmune responses to early life inflammatory perturbations could potentially underlie this apparent protection. Increased expression of microglial and astrocyte-specific genes is evident in the postmortem brains of autistic individuals, as well as in males relative to females irrespective of autism diagnosis (Werling et al., 2016). Thus, lower levels of glial-related gene expression in females could be the source of this buffering. This is a provocative hypothesis, but it has yet to be rigorously tested experimentally. For example, might recapitulating the female neuroimmune system in a genetic animal model of neurodevelopmental disorders confer protection against early life perturbations that induce behavioral changes that resemble human disorders? It would be a major breakthrough for neurodevelopmental disorder prevention or treatment if we were able to pinpoint what female-specific factor mediates this protection, or more generally, the factors that contribute to female resilience to early life inflammatory events.

3. The dynamic female brain and its neuroimmune changes across the ovarian cycle

By the time females pass adolescence, their brains have mostly structurally matured (Toga et al., 2006). Neuron numbers and pattern of connectivity, and microglia and astrocyte numbers and morphology are broadly similar at peri-adolescence to those in young adults (Schwarz et al., 2012; Stiles and Jemigan, 2010). At this phase of life, female sex hormones start to play a major role in brain dynamics and neuroimmune function. 17β-estradiol (E2) is the predominant circulating estrogen in females, and directly influences brain function through the estrogen receptors, ERα & ERβ, expressed on glia & neurons, including in the hippocampus (Azcoitia et al., 1999; Gonzalez et al., 2007; Santagati et al., 1994). In rodents, the estrous cycle lasts approximately four days, with a proestrous (follicular) phase where the ovarian follicles mature, characterized by high levels of E2, progesterone, prolactin, luteinizing hormone (LH) and follicle-stimulating hormone (FSH); an estrous (ovulatory) phase where the ovum is released and the sex hormones plummet; and metestrous and diestrous (luteal) phases where if the ovum is fertilized the corpus luteum secretes progesterone to maintain the pregnancy, or is resorbed if not (Goldman et al., 2007). In humans, this cycle is approximately 20-35 days long and differs primarily in that the endometrium is shed in menses rather than being resorbed. Remarkably, the hormonal changes associated with this cyclicity can fundamentally remodel the brain.

There is some early evidence that neuronal proliferation, or at least numbers of BrdU-incorporated cells, is dependent upon cycle stage and ovarian hormones, with rats having increased dentate gyrus proliferation and cell survival in the proestrous phase, when E2 levels are high, relative to the other phases (Tanapat et al., 1999). However, perhaps most remarkable, is that dendritic spine density is altered by up to 30% across the estrous cycle in the rodent hypothalamus and CA1 hippocampus (Frankfurt et al., 1990; Woolley et al., 1990). In the estrous phase when E2 and progesterone fall to their lowest levels, dendritic spines are robustly pruned. In the metestrous / diestrous phases, dendritic spine formation and maintenance is supported and the synaptic spines maintained and strengthened so that by proestrus spine density can be 30% higher in the CA1 and ventromedial hypothalamus than is seen in estrus (Frankfurt et al., 1990; Woolley et al., 1990). This dynamic remodeling is tied to sex hormone levels with high E2 and progesterone encouraging spine maintenance and sex hormone removal, such as with ovariectomy, leading to reduced spine density (Gould et al., 1990).

The synapse is considered the structural neuronal substrate from which information is transmitted. Thus, it is not surprising that, practically, this dynamic neuronal and spine remodeling may mean females use different cognitive strategies at different stages of the cycle to solve complex tasks (Korol et al., 2004; Paris and Frye, 2008). In the proestrous phase, when E2 and progesterone are high and dendritic spines dense, female rats are more likely to use a place strategy (using recall of learnt external spatial orientation) to solve spontaneous alternation and T maze tasks. In the estrous phase, when sex hormone levels are low, rats are more likely to use a response strategy (using recall of learnt body movements) (Korol et al., 2004). In both tasks, exploration and motivation are similar and the strategies used are equally effective in facilitating task learning (Korol et al., 2004). Pregnancy, when progesterone and its metabolites are elevated, can also improve performance in object recognition and place recognition tasks relative to non-pregnant states (Paris and Frye, 2008). Such dynamic changes may be part of a strategy whereby different life stages call for different cognitive priorities. Whether such dynamic remodeling of dendritic spines contributes to any real or perceived changes in cognitive priorities and attention in human pregnancy and lactation is yet to be determined (Barha and Galea, 2017).

Although sex hormones such as estrogen play an important role in this cyclical dynamic brain remodeling, they likely do so through interaction with the neuroimmune system. Thus, ovarian cycle stage can robustly influence neuroimmune health. Females are more vulnerable to exacerbations of several neuroimmune disorders at certain stages of the ovarian cycle. For example, females with disorders as diverse as inflammatory bowel syndrome or Parkinson’s disease exhibit worsening symptoms at menses, when ovarian hormones are low (Mulak et al., 2014); and oophorectomy prior to menopause can elevate the risk of Parkinson’s developing (Gillies et al., 2014). In rhesus macaques, vaccination responses have been linked to ovarian hormones, with ovariectomy reducing the T-cell cytokine and IgG response to Vaccina Ankara vaccination; E2 replacement partially restores this response (Engelmann et al., 2011). High antibody titres after vaccination in females may be associated with better protection, but also with more frequent side-effects (Furman et al., 2014; Klein et al., 2010; Vermeiren et al., 2013). In rodents, too, sex hormones have been implicated in peripheral mechanisms of differential immunomodulation throughout the ovarian cycle. As such, ovariectomy worsens peripheral diabetes symptoms and the inflammatory effects associated with diabetes (Stubbins et al., 2012), as well as the allergic reaction of the lung to ovalbumin; and E2 treatment reverses, while progesterone exacerbates this effect (de Oliveira et al., 2010).

There has been limited investigation of natural fluctuations in immune reactivity across the ovarian cycle (i.e. from studies other than with ovariectomy or E2 replacement), and even less work on central cyclical immune changes. Yet there is some human evidence that circulating inflammatory markers change across the menstrual cycle, with C-reactive protein (CRP) being low in the follicular phase (when sex hormones are high) (Gaskins et al., 2012; Lorenz et al., 2017). In rodent models, central cytokines can be differentially sensitive across the ovarian cycle. Hypothalamic IL-1 levels are unresponsive to foot-shock stress during metestrus but increase at the other stages (Arakawa et al., 2014). This effect is likely related to high progesterone at this time attenuating the hypothalamic immune response (Arakawa et al., 2014). Likewise, proestrus and estrus see an exacerbated infarct size in a mouse focal ischemia model; an effect that is IL-4-dependent (Xiong et al., 2015).

Although evidence for cycle-stage variations in brain immune cell populations is limited, there is work to suggest microglia and astrocytes are sensitive to substantial variations in sex hormones. Microglia express estrogen receptors (ERα and ERβ) and this expression is significantly reduced by peripheral immune challenge with LPS (Sierra et al., 2008). Experimental ovariectomy, i.e. withdrawal of the sex hormones as occurs with menopause (and to a lesser extent in the luteal phase), leads to inflammation associated with microglial and astrocyte activity (Benedusi et al., 2012; Siani et al., 2017). These changes are somewhat reversible with E2 replacement and are similar in rodents and humans (Sarvari et al., 2012; Sarvari et al., 2014). LPS-induced activation of microglia is modified by compounds that act at the estrogen receptor, with E2, tamoxifen, and raloxifene (selective estrogen receptor modulators) suppressing microglial activation in ovariectomized rats (Tapia-Gonzalez et al., 2008) in a mechanism that may involve E2-mediated inhibition of the inwardly rectifying K+ channel, Kir2.1 (Wu et al., 2016). Exogenous E2 mimetics given long-term after ovariectomy can even reduce microglial and astrocyte numbers (Lei et al., 2003). Notably, females are resistant to the neuroimmune and cognitive deficits imparted by galactic cosmic radiation and this resistance is associated with microglial resilience to the challenge (Krukowski et al., 2018). Paradoxically, female microglia display higher expression of inflammatory, apoptotic, and LPS-response genes than male microglia do (Thion et al., 2018) and therefore have stronger innate and adaptive responses to acute immune stimuli (Klein and Flanagan, 2016). Potentially this greater efficiency in the central immune response offers some protection against long-term negative effects of immune challenge in females, but may also underlie why females are resistant to the antinociceptive effects of morphine compared with males (Doyle et al., 2017). In states where microglia are highly active, such as juxtaposing beta-amyloid plaques in the APP23 Alzheimer’s mouse, reductions in endogenous E2 further activate the microglia; replacing estrogen reverses this effect (Vegeto et al., 2006). However, early work suggests chronically elevated estrogen may not be beneficial, with chronic estrogen replacement therapy after ovariectomy leading to impaired cognitive performance when the treatment is coupled with chronic inflammation (Marriott et al., 2002). As discussed in the next section, microglia also play an important role in modulating synaptic plasticity through pregnancy.

It is clear the female brain is highly sensitive to hormonal fluctuations across the ovarian cycle and at life stages of hormone withdrawal. However, our understanding of these processes is limited by the lack of research in this area. This sensitivity may lead to resilience to immune challenges and stress at phases when E2 is low and progesterone high. Such resilience is important to consider when scheduling treatments such as surgery or vaccinations to maximize safety and effectiveness. When our understanding reaches an appropriate level of sophistication, we can also envisage titrating pharmacological treatments across the cycle to exploit and avoid desensitizing this natural resilience.

4. Neuroimmunological changes in pregnancy

As the female matures, pregnancy is one significant life stage during which dynamic neuroimmunological changes play an important role. While it is well established that the peripheral immune system undergoes dramatic changes in pregnancy to ensure the fetus is tolerated by the maternal immune system (Aagaard-Tillery et al., 2006; Mor and Cardenas, 2010; Trowsdale and Betz, 2006), in recent years attention has turned to the neuroimmunological changes that occur in pregnancy. However, to date research in this area is relatively limited.

In adults, microglia play a key role in immune surveillance. They act as macrophages, eliminating microbes, cellular debris and dead neurons from the central nervous system and are the primary source of cytokine secretion in the brain, hence facilitating neuroinflammatory processes (Colonna and Butovsky, 2017). Microglia are also important in sculpting neuronal networks by dynamically modulating synaptic plasticity (Benarroch, 2013; Colonna and Butovsky, 2017; Schafer et al., 2012; Walker et al., 2014; Yirmiya and Goshen, 2011). However, it remains to be established if microglia are involved in remodelling the maternal brain in pregnancy. In late pregnancy, there is a reduction in the number of microglia in the brain, specifically in the medial prefrontal cortex, nucleus accumbens, basolateral amygdala and dorsal hippocampus, which seems to be a result of reduced proliferation, rather than increased apoptosis (Haim et al., 2017). In accordance, neuroimmune responses of the maternal brain to immune/inflammatory challenge are suppressed, especially in late pregnancy (Sherer et al., 2017). For example, up-regulation in gene expression for the cytokines, IL-1β and IL-6 in response to bacterial endotoxin LPS administration is markedly attenuated in the prefrontal cortex, preoptic area, hypothalamus and hippocampus of late pregnant rats, compared with non-pregnant females (Sherer et al., 2017). The functional role of these microglial changes in late pregnancy is not clear, however they may contribute to neuroplasticity in the maternal brain that promotes changes in maternal motivation, cognitive function and mood post-partum (Figure 1) (Galea et al., 2014; Hillerer et al., 2014; Kinsley and Lambert, 2008; Leuner and Sabihi, 2016), although this requires further study.

Fever plays an important role in the body’s defence against infection as the increase in body temperature above the normal range serves to provide an inhospitable environment in which microbes are less able to multiply. However, despite fever being considered as an important component of the immune response, in late pregnancy the febrile response to immune challenge is markedly suppressed (Harre et al., 2006; Mouihate et al., 2002; Mouihate et al., 2005). This change is likely to be adaptive, as prolonged exposure to fever/hyperthermia can negatively impact fetal development and offspring health (Brucato et al., 2017; Edwards, 2006; Fisher et al., 2010; Lowe et al., 2008), however it may come at a cost-compromising the mother’s ability to fight infection (Martin et al., 1995). The mechanisms underlying reduced febrile responses to immune challenge may involve suppression of pro-inflammatory and enhancement of anti-inflammatory processes. Indeed, in response to LPS administration, production of pro-inflammatory prostaglandins is reduced in late pregnancy (Imai-Matsumura et al., 2002), evidently as a result of reduced cyclo-oxygenase-2 (COX-2; one of the enzymes responsible for responsible for formation of prostaglandins) induction in the hypothalamus (Mouihate et al., 2002)); while the response of the anti-inflammatory cytokine, IL-1 receptor antagonist (IL-1Ra) is augmented (Ashdown et al., 2007). It is not known what induces the change in febrile responses in pregnancy. While ovarian hormones influence fever development across the estrous cycle (Mouihate et al., 1998), increased levels of E2 and progesterone in late pregnancy do not appear to mediate attenuated febrile responses before term (Finley et al., 2015).

Hormonal regulation of neuroimmune adaptations during pregnancy

Changes in the hormonal milieu in pregnancy are recognised as potentially underpinning the neuroimmune alterations described above (Figure 1). Pregnancy is characterised by considerably higher circulating concentrations of E2 and progesterone (Bridges, 1984; Brunton and Russell, 2010). Both estrogen and progesterone have central anti-inflammatory actions in pregnancy (Kipp et al., 2007). Indeed, treating virgin female rats with a combination of E2 and progesterone to simulate pregnancy results in reduced febrile responses to LPS, concomitant with reduced COX-2 expression in the hypothalamus (Mouihate and Pittman, 2003). Moreover, as mentioned earlier, microglia express receptors for estrogen (Habib and Beyer, 2015; Vegeto et al., 2001) and estrogen has been shown to suppress proliferation and activation of microglia, inhibit pro-inflammatory cytokine secretion and promote anti-inflammatory cytokine secretion (Bruce-Keller et al., 2000; Dimayuga et al., 2005; Vegeto et al., 2001). Hence increased circulating sex steroids in pregnancy may contribute to neuroimmunological alterations.

In addition to increases in circulating sex steroids, oxytocin has also been proposed as a potential candidate in modulating neuroimmune responses in pregnancy. In support, oxytocin has been reported to inhibit LPS-induced microglial activation and consequently reduce the release of pro-inflammatory mediators in vitro and in vivo (Yuan et al., 2016), and attenuate febrile responses to centrally administered IL-1α (Poulin and Pittman, 1993). However, given oxytocin release in the supraoptic nucleus (SON), paraventricular nucleus (PVN), mediolateral septum and dorsal hippocampus is similar between virgin females and late pregnant rats (Landgraf et al., 1992; Neumann et al., 1993) and is only increased in the ventral septal area (VSA) in late pregnancy (Landgraf 1992), it is unclear whether this modest increase in central oxytocin is physiologically relevant with respect to suppressing neuroimmunological responses. Nevertheless, immune challenge is known to activate centrally projecting oxytocin neurons (Matsunaga et al., 2000) and stimulate oxytocin release in the VSA and SON (Landgraf et al., 1990; Landgraf et al., 1995), so this may be important, although it is not yet known whether this central oxytocin response to immune challenge is altered in pregnancy.

Finally, given their potent anti-inflammatory actions we should also consider a role for glucocorticoids in suppressing neuroimmune responses in pregnancy. Basal concentrations of corticosterone in the circulation increase progressively from around mid-gestation in the rat (Atkinson and Waddell, 1995). Microglia express glucocorticoid receptors (Tanaka et al., 1997) and glucocorticoids inhibit microglial proliferation and suppress secretion of pro-inflammatory cytokines from microglia (Drew and Chavis, 2000; Tanaka et al., 1997). Thus, although HPA axis responses to immune challenge are suppressed in late pregnancy (discussed below) and glucocorticoids exert less potent anti-inflammatory actions in females than in males (Duma et al., 2010), elevated baseline glucocorticoid levels may contribute to attenuating neuroinflammatory processes.

There is still a lot we do not understand about the neuroimmune adaptations that occur in pregnancy and this is an area that warrants more research. Of particular interest is the potential that these changes may predispose women to postpartum mood disorders. Certainly, altered microglial activity and immune signalling have been implicated in anxiety and depression (Hodes et al., 2015; Kohler et al., 2018; Remus and Dantzer, 2016; Reus et al., 2015; Steiner et al., 2011; Yirmiya et al., 2015; Young et al., 2014). Whether neuroimmunological adaptations in pregnancy and the subsequent restoration in the postpartum period (Sherer et al., 2017) interact with the dramatic withdrawal of pregnancy-related hormones at birth, which has also been proposed to confer vulnerability to puerperal mood disorders (Bloch et al., 2000; Brunton et al., 2008), is not known but is an area that requires investigation.

Neuroendocrine responses to inflammatory challenge in pregnancy

Sex differences in HPA axis responses to stress, including immune challenge, are well established in rodents, with females typically displaying greater responses than males (Frederic et al., 1993; Seale et al., 2004). However, in late pregnancy, responsivity of the HPA axis and the magnocellular oxytocin system to a range of physical and psychological stressors is markedly attenuated, compared with the non-pregnant state (Brunton and Russell, 2008). It is generally considered that maintaining quiescence of the oxytocin neurons serves to minimise the risk of preterm birth and limiting activation of the HPA axis should conserve energy stores and protect the foetuses from adverse programming by exposure to excessive levels of maternal glucocorticoids (Brunton et al., 2014).

Of particular relevance here is that this phenomenon is also seen in response to acute immune/inflammatory challenges in late pregnancy (Brunton et al., 2005). In non-pregnant rats, systemic administration of IL-1β is a potent activator of the HPA axis, activating corticotropin-releasing hormone (CRH) neurons in the medial parvocellular PVN (mpPVN) (reflected by Fos induction), up-regulating mpPVN Crh gene expression and stimulating adrenocorticotropic hormone (ACTH) and corticosterone secretion (Brunton et al., 2012; Brunton et al., 2005; Buller et al., 2001; Ericsson et al., 1994; Sapolsky et al., 1987). Systemic IL-1β also activates magnocellular oxytocin neurons in the SON and PVN, increasing their electrical activity and stimulating oxytocin secretion from the posterior pituitary (Brunton et al., 2012; Brunton et al., 2006; Buller et al., 2001). In contrast, in late pregnancy both the HPA axis and the neurohypophysial oxytocin system are substantially less responsive to stimulation by acute inflammatory challenges such as LPS or IL-1β administration, compared with non-pregnant rats (Brunton et al., 2005; Brunton et al., 2006). Hence, in late pregnancy IL-1β fails to stimulate the HPA axis and has little effect on the electrical or secretory activity of the magnocellular oxytocin neurons (Brunton et al., 2012; Brunton et al., 2006). Moreover, centrally administered IL-1β depletes CRH content in the median eminence of virgin but not pregnant rats (Nakamura et al., 1998).

Central mechanisms involved in restraining the HPA axis and oxytocin system

Endogenous Opioids

Endogenous opioids are involved in restraining HPA axis and oxytocin responses to IL-1β in late pregnancy. As such, prior intravenous administration of the opioid receptor antagonist, naloxone, not only restores ACTH, corticosterone and oxytocin secretory responses to IL-1β, but in the hypothalamus increases the firing rate of oxytocin neurons and permits IL-1β-induced up-regulation of Crh mRNA in the mpPVN as well as Fos expression in the SON and PVN (Brunton et al., 2005; Brunton et al., 2006).

Brainstem noradrenergic neurons located in the nucleus tractus solitarius (NTS) and ventrolateral medulla (VLM) play a crucial role in integrating cytokine signals from the periphery and relaying them to the hypothalamus (Buller et al., 2001; Ek et al., 1998; Ericsson et al., 1997; Ericsson et al., 1994; Ericsson et al., 1995; Rivest et al., 2000). These neurons project to the parvocellular CRH neurons in the PVN and magnocellular oxytocin neurons in the PVN and SON (Buller et al., 2001; Buller et al., 2004; Ericsson et al., 1994; Xu et al., 1999) and mediate HPA axis and oxytocin neuron responses to IL-1β (Buller et al., 2001; Ericsson et al., 1994; Melik Parsadaniantz et al., 1995). Systemic IL-1β increases Fos expression in the A2 region of the NTS to a similar extent in late pregnant and non-pregnant rats (Brunton et al., 2005). However, despite triggering noradrenaline release in the PVN of non-pregnant female rats, IL-1β fails to stimulate noradrenaline release in the PVN of late pregnant rats, unless naloxone is retrodialysed directly into the PVN (Brunton et al., 2005). These data indicate that in late pregnancy endogenous opioids act presynaptically on noradrenergic afferents to the PVN and inhibit IL-1β-induced noradrenaline release (Brunton et al., 2005). The source of the endogenous opioids is likely to be the A2 NTS neurons themselves, since mRNA expression for both proenkephalin-A (Penk) and μ-opioid receptor (Oprm1) are up-regulated in the A2 region of the NTS in late pregnant rats (Brunton et al., 2005). Given noradrenergic NTS neurons also relay signals from the birth canal to drive oxytocin secretion at parturition (Douglas et al., 2001; Herbison et al., 1997; Meddle et al., 2000), it is thought that limiting activation of this pathway in pregnancy by endogenous opioids ensures premature activation of the oxytocin neurons and hence preterm birth is avoided.

Allopregnanolone

The steroid milieu dramatically changes in pregnancy. As mentioned above, concentrations of E2 and progesterone in the blood are increased (Bridges, 1984; Brunton and Russell, 2010). In addition, concentrations of the neuroactive steroid metabolite of progesterone, allopregnanolone is also substantially increased in the blood and in the brain (Concas et al., 1998). Gene expression for both of the allopregnanolone-synthesising enzymes (5α-reductase; Srd5a and 3α-hydroxysteroid dehydrogenase; Akrlc4) is increased in the NTS in late pregnant rats, as is enzyme activity in the hypothalamus (Brunton et al., 2009). This is expected to lead to increased local allopregnanolone production in pregnancy (Paul and Purdy, 1992), which is considered to be important with respect to HPA axis and oxytocin neuron regulation. Indeed, administration of the 5α-reductase inhibitor, finasteride (at a dose shown to reduce central allopregnanolone content by 90%) (Concas et al., 1998) restores HPA axis responses to IL-1β in late pregnant rats; while treating virgin females with allopregnanolone to mimic pregnancy levels, suppresses HPA axis responses to IL-1β (Brunton et al., 2009). The same treatment regimens alter responsiveness of the magnocellular oxytocin neurons to an inflammatory challenge in a similar manner, revealing a role for allopregnanolone in also restraining activity of the oxytocin system in late pregnancy (Brunton et al., 2012).

Interestingly, treating virgin rats with either of the allopregnanolone precursors, progesterone or dihydroprogesterone (DHP) alone is not sufficient to suppress HPA axis or oxytocin responses to IL-1β (Brunton et al., 2012; Brunton et al., 2009), indicating that conversion of progesterone/DHP into allopregnanolone is of critical importance in suppressing these responses. This is likely achieved locally through up-regulation of the allopregnanolone synthesising enzymes in the NTS in pregnancy (Brunton et al., 2009).

Allopregnanolone and induction of endogenous opioid tone

Allopregnanolone plays a key role in the induction of the inhibitory endogenous opioid tone over the HPA axis and the oxytocin system in late pregnancy (Brunton et al., 2012; Brunton et al., 2009). As mentioned above, pregnancy is associated with increased Penk and Oprm1 gene transcription in the NTS (Brunton et al., 2009). This up-regulation of opioid-related gene expression in the NTS can be induced in virgin females by exogenous allopregnanolone administration; while blocking allopregnanolone synthesis in pregnant rats with finasteride, reduces Penk and Oprm1 gene expression to virgin levels (Brunton et al., 2009). Moreover, in non-pregnant females allopregnanolone administration induces endogenous opioid inhibition over the HPA axis, as naloxone administration reverses allopregnanolone-induced suppression of ACTH secretory responses to IL-1β (Brunton et al., 2009).

Thus, in summary, increased allopregnanolone production in pregnancy is of critical importance in inducing and sustaining an inhibitory endogenous opioid mechanism that limits excitation of both the HPA axis and the neurohypophysial oxytocin system by acute exposure to inflammatory stimuli, through inhibiting the excitatory noradrenergic input to the CRH and oxytocin neurons in the hypothalamus. This mechanism is predicted to minimise the risk of premature activation of the oxytocin neurons and consequently premature uterine contractions and preterm birth. Indeed, sustained inhibition of allopregnanolone production during the last week of pregnancy results in preterm birth and markedly reduces survival rates of the neonates (Paris et al., 2011b). Furthermore, preventing activation of the maternal HPA axis in pregnancy is also expected to conserve energy and limit exposure of the fetuses to excessive levels of maternal glucocorticoids, which can adversely program the offspring, predisposing them to disease (Brunton et al., 2014; Maccari et al., 2014). However, in spite of this, repeated immune challenge during pregnancy leads to preterm birth and fetal programming of the offspring (Paris et al., 2011a), indicating this inherent defence mechanism can be breached by repeated insults.

5. Female cancer effects on brain

Later life stages can also change the female brain in specific ways. As a woman ages she has increased risk of developing sex hormone-dependent diseases like cancer, including breast cancer, the most common malignancy in women. Cancer is associated with debilitating neurological and behavioral comorbidities, including self-reported fatigue (~90%), depression (~18%), anxiety (~10%), and cognitive impairments (~35%), occurring before, during, and long after successful treatment (Ahles et al., 2012; Ancoli-Israel et al., 2014; Pitman et al., 2018; Shankar et al., 2016). These behavioral comorbidities are associated with inflammation in clinical cancer research and neuroinflammation in preclinical cancer research, as previously reviewed (Santos and Pyter, 2018; Schrepf et al., 2015; Vichaya et al., 2015), suggesting that the underlying mechanisms include immune dysregulation. Indeed, non-CNS tumors (Schrepf et al., 2015), chemotherapy (reviewed in (Santos and Pyter, 2018)), and radiation (Ramanan et al., 2008) each independently induce inflammatory signaling in the brain (e.g., IL-1β, IL-6, Tnfα, iNOS, COX-2), in part by activation of microglia (ionized calcium binding adaptor molecule 1 (Iba1+), cluster of differentiation 68 (CD68+), or Cd11b mRNA) and astrocytes (Gibson et al., 2019), in models of cancer and cancer treatments, with negative behavioral outcomes. However, these recent reviews do not consider the role of sex or sex hormones in the behavioral or neuroimmune consequences of cancer.

Based on meta-analyses, people with breast cancer have a relatively low prevalence of major depression compared with other cancers. However, depression symptoms based on diagnostic interviews or self-report are relatively higher for cancers affecting females (breast, ovary, uterine, etc.) than for other types of cancer (Krebber et al., 2014). Similarly, females report more psychological distress but less diagnosed depression and anxiety when male and female cancers are directly compared (van’t Spijker et al., 1997). Finally, female cancer patients and female partners of cancer patients perceive more stress and lower quality of life than women in healthy couples, whereas men do not show this difference (Hagedoorn et al., 2000). These statistics indicate that behavioral comorbidities due to cancer may by modulated by sex-specific mechanisms.

In fact, approximately 70% of women diagnosed with breast cancer are post-menopausal (Danforth, 1991). Menopause, occurring in women at around age 50, is the complete and permanent cessation of menstrual cycles due to the natural depletion of ovarian oocytes resulting in a near complete depletion of estrogen (MedlinePlus, 2011) and is a significant risk factor for a variety of diseases. In Australian women with non-metastatic breast cancer, those without ovaries have higher depression and anxiety scores compared with those without ovariectomy (Sayakhot et al., 2011). Conversely, in breast cancer survivors, antidepressant treatment is associated with significantly higher prevalence and severity of menopausal symptoms (Gupta et al., 2006). Indeed, quality of life issues related to menopause and cancer treatment are difficult to differentiate, given that menopause often overlaps with cancer diagnosis (Schultz et al., 2005). Furthermore, 75% of breast tumors are responsive to estrogen and progesterone, such that these hormones promote tumor growth and progression. Thus, in addition to standard chemotherapy and radiation treatments, breast cancer patients are often treated with adjuvant endocrine therapy to inhibit tumorigenic endocrine (primarily estrogen) activity in tumor cells (Rugo et al., 2016). Overall, there is a lack of understanding of how menopause and long-term endocrine, chemotherapy, and radiation treatments may influence the brain to cause cancer behavioral comorbidities.

Studies of treatment-free breast cancer patients report no significant differences in cognition between pre-versus post-menopausal women (Torino et al., 2012; van Dam et al., 1998) , nor do breast cancer patients grouped based on lower versus average cognitive performance vary by menstrual status (Ahles et al., 2008). These findings indicate that endocrine therapy and chemotherapy may be necessary to precipitate otherwise insignificant behavioral comorbidities. Currently, most cancer studies are not designed to consider hormonal status as a potential modifier of behavioral comorbidities. For this review, we explore how these cancer-related endocrine pathways may influence neuroimmunology to confer negative behavioral consequences. Of note, most research on the interaction between hormones and cancer behavioral comorbidities are focused on cognitive function.

While most breast cancer patients are diagnosed after menopause, chemotherapy treatment also causes ovarian failure, amenorrhea, and reduced reproductive hormones in the majority of pre-menopausal women (Mehta and Graves, 1992; Reyno et al., 1992). Thus, “chemotherapy-induced menopause” may have similar negative effects on cognition as natural menopause (Zec and Trivedi, 2002). Furthermore, hot flashes, which are linked to estrogen deprivation, are correlated with anxiety in healthy women (Freeman, 2014). In fact, early chemotherapy-induced menopause may render pre-menopausal breast cancer patients more vulnerable to behavioral issues associated with reduced estrogen signalling in the brain than post-menopausal patients due to their extended estrogen deprivation period (Roy et al., 1996). Some studies indicate that cognitive performance following chemotherapy varies according to pre-treatment menopausal status (Conroy et al., 2013; Jenkins et al., 2006). For example, post-menopausal breast cancer patients report significantly greater difficulty with concentration and memory then pre-menopausal patients. However, when treatment modalities were considered, a reverse trend was observed, such that radiation was harsher on cognitive functioning in post-menopausal women and chemotherapy was harsher in premenopausal women (Schagen et al., 1999). Incomplete reporting on whether or not radiation was combined with chemotherapy or hormone therapy limits the conclusions. Of note, anxiety was reported in 38% of these patients and was associated with the self-reported cognitive problems (Schagen et al., 1999), suggesting that some of these conclusions focused on cognitive function may be extrapolated to mood. In contrast, other studies indicate that chemotherapy-induced menopause confers no negative consequences on cognitive function (Schagen et al., 2006; Vearncombe et al., 2011).

The primary recommendation for adjuvant endocrine therapy for pre-menopausal cancer patients is selective estrogen receptor modulators (SERMs; historically tamoxifen) which dysregulate estrogen signalling in tumor cells, while leaving estrogen signalling in other tissues intact (e.g., brain). In contrast, for the majority of breast cancer patients (i.e., postmenopausal), estrogen modulator therapy consists of aromatase inhibitors (AIs) alone or in combination with tamoxifen. AIs and SERMs have different mechanisms of action on estrogen receptors and AIs inhibit signalling both in the brain and periphery. AIs are prescribed for 5 years post-chemotherapy/radiation, but due to their effectiveness at tumor inhibition, prescription of these drugs has recently been extended for 10+ years. In contrast to chemotherapy, findings on cognitive effects of estrogen modulators are relatively sparse, underpowered, and inconclusive (Agrawal et al., 2010). The lack of understanding of how endocrine therapy impacts cancer-associated behavior is partially due to few studies differentiating between estrogen modulator treatment and chemotherapy or differentiating among the various estrogen modulator mechanisms of action (Jenkins et al., 2004). The effects of tamoxifen and AIs on cognitive function in breast cancer patients is reviewed elsewhere (Zwart et al., 2015).

Thus, hormonal status, history of hormone replacement therapy, and age all modify endocrine therapy effects on behavior. Estrogen modulators impair cognitive function even when given without chemotherapy (Ahles et al., 2010; Bender et al., 2001; Jenkins et al., 2004; Jenkins et al., 2008), although tamoxifen may be more detrimental (but possibly, reversible) than AI (Bakoyiannis et al., 2016; Phillips et al., 2010; Schilder et al., 2010). Post-menopausal breast cancer patients treated only with endocrine therapy (no chemotherapy) display poorer processing speed, verbal memory, and verbal ability than those without any treatment or healthy controls (Bender et al., 2007). However, the opposite effect is observed in premenopausal women, suggesting that prior hormonal status modulates the negative effects of endocrine therapy on behavior. Another well-designed study indicates that AIs confer both acute and long-term impairments in visual working memory, in particular (Bender et al., 2015). The potential more moderate effects of AIs on the brain/behavior compared with SERMS is unexpected given that AIs deprive the brain of estrogens produced in both the periphery and in the brain, whereas tamoxifen only inhibits peripheral estrogen synthesis. Objective cognitive results do not consistently correspond to self-reported results in these studies. Clearly, additional prospective studies are necessary to draw more robust conclusions (Wu and Amidi, 2017). Of note, some reports indicate that depression may explain cognitive impairment in breast cancer survivors more than endocrine therapy or chemotherapy (Seliktar et al., 2015). Finally, powerful clinical studies have indicated that endocrine therapy in otherwise healthy, post-menopausal women increases their risk of developing cancer and stroke and has adverse effects on cognition (Anderson et al., 2004; Rossouw et al., 2002; Shumaker et al., 2004). This work indicates that the effects of reproductive hormone modulation on the brain in postmenopausal women are complicated by their age, uterine/ovarian status, and duration since their last menstrual period. Thus, nuanced effects of hormonal status and endocrine therapy may modulate behavior in cancer patients and requires consistent reporting in clinical studies.

The majority of behavioral comorbidity studies in cancer patients are not designed to differentiate between chemotherapy with or without endocrine therapy. However, those that are suggest that cognitive problems are worse in patients receiving both therapies than one or the other (Castellon et al., 2004; Collins et al., 2009); but see (Buchanan et al., 2015). The various estrogen-modulating factors associated with breast cancer likely impact neurobiological mechanisms underlying these behaviors (Ahles and Saykin, 2007). Estrogen significantly modulates cognitive behavior (McEwen and Alves, 1999) potentially through observed neuroprotective and antioxidant actions (Unfer et al., 2006) and by helping to maintain telomere length (Lee et al., 2005). Estrogen is also anti-inflammatory, reducing proinflammatory cytokines in the brain (Brown et al., 2010; Duckles and Krause, 2011; Vegeto et al., 2008). As neuroinflammation is associated with cognitive decline, negative mood, and fatigue, natural or pharmacologically-induced reductions in estrogen in cancer patients may exacerbate neuroinflammation, and thus, behavioral consequences (Cribbs et al., 2012).

Indeed, numerous female rodent models of solid tumors or cancer treatments report increased inflammation in the brain and deficits in cognitive performance or elevations in affective-like behaviors (Lamkin et al., 2011; McGinnis et al., 2017; Pyter et al., 2009). However, very few cancer modelling studies have assessed the role of estrogen in these neurobehavioral results through ovariectomy, hormone replacement, or estrogen modulator treatments. One relevant study demonstrates that long-term olfactory recognition memory impairments in mice receiving cranial radiation therapy are dependent upon estrous stage (Perez et al., 2018). Cancer anorexia is also hypothesized to be influenced by sex-related hormones (Varma et al., 2001). Other studies not directly related to cancer modelling demonstrate that the SERM, raloxifene, combined with E2 treatment reduces anxiety- and depressive-like behavior in ovariectomized rats (Karahancer et al., 2008; Walf and Frye, 2010). Finally, cognitive performance improves with AI treatment in ovariectomized mice (Aydin et al., 2008; Meng et al., 2011), suggesting that cancer-typical estrogen modulation in much needed models of post-menopausal cancer may similarly improve behavioral impairments.

Results gleaned from healthy rodents used to assess the effects of endocrine therapies on neuroinflammation may also be relevant in the context of cancer. The anti-inflammatory mechanisms of SERMs in the brain have been reviewed elsewhere (Arevalo et al., 2011), but, briefly, SERMs influence various brain glial cells through modulation of ER signalling in their nuclei, their membranes or cytoplasm, or through ER-independent mechanisms. Indeed, ERα and ERβ in the brain are increased under several pathological inflammatory conditions (see (Arevalo et al., 2011)). Most relevant to tumors, microglia ERβ activity is responsible for the neural loss induced by an optic glioma tumor in mice (Toonen et al., 2017). Relevant to cancer treatment, tamoxifen decreases the microglia inflammatory response induced by cranial irradiation (Liu et al., 2010). SERMs also suppress in vitro LPS-induced microglial activation and neuronal cell death via an ER-dependent pathway (Ishihara et al., 2015). Long-term second-line (more specific) SERM treatment of aged ovariectomized mice successfully reduces the number of astrocytes and microglia in the hippocampus similar to E2 (Lei et al., 2003), suggesting that these more specific SERMs may confer the benefits of estrogen on the brain while inhibiting estrogen signalling in tumors outside the brain. Furthermore, some SERMs can mitigate neuroinflammation, neurotransmitter transport, and gliosis via their actions on astrocytes (Acaz-Fonseca et al., 2014). These astrocyte studies focus primarily on in vitro and in vivo models of traumatic brain injury (Johann and Beyer, 2013). Finally, nonnuclear ER stimulation reduces leukocyte infiltration and neuroinflammation in the brain after ischemic stroke in ovariectomized mice (Selvaraj et al., 2018). Of note, raloxifene has moderate pro-inflammatory effects on non-pathological brain tissue (Tapia-Gonzalez et al., 2008), suggesting potential negative inflammatory consequences of this SERM in patients asymptomatic for mental health issues. Thus, female-specific sex hormones particularly influence cancer development and the cognitive consequences that ensue, yet our understanding of the neuroimmune interactions that are important for this influence remains poor. There is an immediate need for improved basic and clinical research in this field to ensure we have optimal treatments to assist vulnerable females to move forward into healthy aging.

6. The aging female brain: the neuroimmune phenotype and its role in cognitive function

By the year 2050 approximately 1/5th of the world’s population will be over the age of 65, a dramatic doubling of today’s aging populace (He et al., 2016). This is alarming because advanced age is the single strongest risk factor for developing Alzheimer’s disease, Parkinson’s dementia, and other persistent cognitive impairments precipitated by surgery, stroke, viral or bacterial infection, and other immune insults (Alzheimer’s Association, 2016; Inouye et al., 2014; Moller et al., 1998; Mozaffarian et al., 2015; Nguyen et al., 2014; Reeve et al., 2014; Wofford et al., 1996). Current treatments for many of these conditions are ineffective. Fortunately, in the last ten years numerous clinical and basic research studies have strongly implicated a critical role played by heightened neuroinflammation, owed to a sensitized microglial or altered astrocytic phenotype, in the pathogenesis of these conditions, thus providing a therapeutic target on which to focus (Barrientos et al., 2012; Cao et al., 2010; Chisholm and Sohrabji, 2016; Edison and Brooks, 2018; Ferreira et al., 2014; Frank et al., 2010; Jones et al., 2018; McKenzie et al., 2017; Rajendran and Paolicelli, 2018; Ransohoff, 2016; Rollins et al., 2018; Spencer et al., 2018; Spencer et al., 2017; Tucsek et al., 2014).

Despite the fact that over half of the aging population is comprised of females, the vast majority of basic research conducted on matters of aging, neuroinflammation, and cognitive decline has been conducted using only male subjects. This disproportionate focus on the male brain is unjustified and troubling because women exhibit increased susceptibility to cognitive impairments following an immune insult as early as middle age (Phillips Bute et al., 2003). Furthermore, aged women are reportedly twice as likely as men to exhibit long-lasting post-operative cognitive dysfunction (Kotekar et al., 2014), have poorer outcomes following stroke than men (Choleris et al., 2018), are more likely to suffer from Alzheimer’s disease and other dementias (Bachman et al., 1992; Borenstein et al., 2014; Hebert et al., 2013; Launer et al., 1999; Sun et al., 2014), and women with mild cognitive impairments show deteriorating cognitive functions at a rate twice as fast as men (Lin et al., 2015). The mechanisms underlying these sex differences are not fully understood and are under-studied, but there is growing evidence that this vulnerability is markedly influenced by changes in sex steroid hormone levels associated with advanced age (Choleris et al., 2018; Driscoll et al., 2005; Dubal and Rogine, 2017).

Menopause is associated with a precipitous decline in female sex hormones, including E2. Relevant to this discussion, it has been reported that normal E2-ER interactions in astrocytes and microglia of young adult rodents decrease NF-κB transcriptional activity and inhibit the release of proinflammatory compounds in response to inflammatory stimuli, while dysregulation of either ligand or receptor results in the opposite (Cerciat et al., 2010; Ghisletti et al., 2005; Spence and Voskuhl, 2012; Spence et al., 2013; Vegeto et al., 2006). Menopause-associated reductions in estrogen are strongly associated with significant declines in cognition (Abu-Taha et al., 2009; Dubal and Wise, 2002). Thus, there is reason to believe that menopause may be an important factor further driving neuroinflammation and cognitive decline in older women.

The use of a naturally menopausal aged rodent model is not possible because aging female rodents enter a state of persistent estrus (also termed estropause), producing constant intermediate estrogen levels (Berkley et al., 2007; Chakraborty and Gore, 2004; Huang and Meites, 1975; Meites and Lu, 1994), which does not resemble the ovarian function of aged women. Therefore, surgical removal of the ovaries is necessary in rodent models to achieve true estrogen depletion. In studies modeling various aging-related diseases this way, many have reported greater inflammatory risk and disease progression in ovariectomized rodents compared to those with intact ovaries (Berchtold et al., 2001; Burnham et al., 2016; Chen et al., 2016; Crain et al., 2013; da Palma et al., 2016; De Melo et al., 2016). Importantly, these studies were all performed on young adult, not aged, female rodents. Rendering rodents menopausal in the absence of advanced age disregards the robust aging-related alterations to the glial phenotype that have been widely reported (Barrientos et al., 2015; Costello et al., 2016; Niraula et al., 2017), and therefore likely underestimates the inflammatory response to a challenge and cognitive impairments that would result when both aging and menopause are combined.

Recent work has made several advances in this regard. In a series of informative papers, the Liposits laboratory found that ovariectomizing middle-aged rats provokes an upregulation of microglial activation markers (Iba1, CD68, and CD80, CD11b, and CD18), and recognition receptor molecules (TLR3, TLR9) in the hippocampus compared to ovary-intact middle-aged controls (young adult controls were not examined). E2 replacement ten days following ovariectomy attenuated the expression of many these markers, and increased expression of CD200R, indicating a shift towards a protective microglial phenotype (Sarvari et al., 2014). Response to an inflammatory stimulus was not examined. Follow-up studies demonstrated that the hippocampal transcriptome is extensively altered with ovariectomy (Sarvari et al., 2017), and that long-term treatment with the ERβ-specific agonist, diarylpropinontrile (DPN), contributes to processes involved in the regulation of transcription, translation, neurogenesis, neuromodulation, and neuroprotection in the hippocampus, suggesting that selective activation of ERβ may be an effective therapeutic approach to mitigate the neuroinflammatory phenotype caused by estrogen depletion in menopause (Sarvari et al., 2016). In another important study (Kireev et al., 2014), basal expression of various inflammatory markers in the dentate gyrus of the hippocampus was measured in female rats that were either intact (non-cycling), ovariectomized, or ovariectomized and supplemented with E2 at middle age (12 months) and allowed to age to 24 months. Findings revealed that, compared to intact young adult females, non-cycling intact aged rats exhibited increased expression of IL-1β, TNFα, and IL-6, GFAP, iNOS, and NFκB1. Twelve-month long estrogen depletion exacerbated these responses, while estrogen treatment (administered to mimic normal cycling levels) robustly attenuated them to levels indistinguishable from those of young adult controls. These findings further support the anti-neuroinflammatory role of estrogen, and underscore the importance of cycling estrogen levels for the health of the aging brain.

In another informative study, researchers ovariectomized mice at 5 months of age and allowed them to age to 12 or 22 months (Benedusi et al., 2012). Results revealed that ovariectomy dramatically increased ERα and ERβ mRNA expression in the hippocampus at the 22-month time point. In addition, expression of the proinflammatory cytokines and chemokines TNFα, IL-1β, and MIP2 was also significantly increased in the 22-month-old ovariectomized mice. Furthermore, the morphology of astrocytes and microglia took on a more reactive phenotype with ovariectomy. This study further demonstrated that the inflammatory response to an LPS challenge in middle-aged mice that had been surgically-induced menopausal for 7 months was robustly exacerbated compared to mice that had been menopausal for only 1 month (Benedusi et al., 2012). Taken together, these results suggest that the duration of estrogen depletion, in combination with advanced age, significantly alters the neuroinflammatory phenotype. These findings lend support for studies that claim that hormone replacement therapy only exerts beneficial effects when initiated near the onset of menopause (Henderson and Rocca, 2012; Rocca et al., 2011). Whether an ERβ-specific hormonal replacement therapeutic approach confers protection against developing Alzheimer’s disease (or other inflammatory-mediated cognitive impairments) in aged menopausal women, without also conferring detrimental side effects, remains to be examined.

As mentioned previously, fever is an important, beneficial, and adaptive physiological response to immune challenges and stressors, with important implications for mortality and cognition. Early work (Wachulec et al., 1997) showed that a peripheral LPS injection produced a significant increase in core body temperature in both male and female young adult rats. This response was significantly blunted in aged rats, with no sex differences evident in either age group. It should be noted that body temperatures were measured for only 8 hr, and estrogen levels were not depleted in the females to mimic aging-associated menopause (Wachulec et al., 1997). Follow-up studies demonstrated that central administration of prostaglandin or IL-1β produced comparable increases in core body temperatures in young and old rats, suggesting that the brain’s ability to generate fever is not impaired, but perhaps peripheral fever-causing signals to the brain are weakened (Plata-Salaman et al., 1998; Satinoff et al., 1999). A more recent study, with an extended measurement window, demonstrated a similar pattern of results during the first 24 hr following an E. coli infection (i.e., a hyperthermic response in young adults, and a hypothermic response in aged). Body temperature measurements extending beyond the first day however, revealed a striking hyperthermic response in aged rats that was both higher in magnitude and much longer in duration than that of young adult rats (Barrientos et al., 2009). Unfortunately, this study was conducted only in males, so these extended responses remain to be examined in aged females. Given that fever is mediated by activated microglia, and estrogen-depleted aged females show greater basal glial-activation markers and a greater proinflammatory response in the brain, it would not be too surprising to find that the fever response was also exaggerated in aged females.

In summary, despite the fact that for decades aging women have exhibited a greater susceptibility to cognitive impairments than men, our understanding of the neurobiological underpinnings of this disparity has been inadequate. Recent, albeit limited, preclinical research has demonstrated that the combination of advanced age and depletion of circulating estrogen levels plays a prominent role in exacerbating the neuroinflammatory milieu in the female brain. This research has advanced our understanding of important factors that contribute to this phenotype (e.g., age at time of menopause onset, time since menopause onset, and rhythmicity of estrogen). An exacerbated neuroinflammatory phenotype presumably drives cognitive decline, as has been shown in males, but whether this occurs in a similar manner to that seen in males remains to be investigated. Female-specific aging research is necessary to determine if the neuroinflammatory phenotype is restricted to the hippocampus, or if it is more widespread thus impacting other types of cognitive function (e.g., executive function). Furthermore, the magnitude, duration, and rate of cognitive decline also need to be fully characterized in these models.

1.7. Conclusions

Of the approximately 7.6 billion humans alive in the world at the time of writing, around 3.8 billion (give or take a few million) are female (Orzack et al., 2015). In addition, most of the world’s adult mammalian livestock and all of our egg-producing livestock are female. Yet female-only studies accounted for 20% or fewer of the papers published in 2009 in all life-sciences-related fields except reproduction (Beery and Zucker, 2011). This blinkered approach to research has resulted in a fundamental limitation in our understanding of the female brain with problematic health and economic consequences. Of a sample of 10 drugs withdrawn from the US pharmaceutical market between 1997 and 2001, 8 had greater adverse health effects in females due to pharmacodynamic or usage differences between males and females (Liu and Mager, 2016).

Specific female-centered research is essential now for answering several important challenges such as we have discussed here. Female-specific resilience and vulnerability to early life immune challenges means that it is now important to identify the meaning of quantitative and qualitative changes in gene expression after such challenges. This understanding could lead to new tools to mimic mechanisms by which females are sometimes relatively protected. Focused study on the importance of sex hormones in neuroimmune function is now needed for personalised pharmaceutical treatments, potentially advancing our capacity to tailor drug doses and delivery systems to accommodate sex hormone fluctuations. Moreover, females often spend decades taking hormonal contraception, and the impact of such chronic pharmaceutical hormone exposure could dramatically impact neuroimmune function in ways that have not been carefully considered in the context of individual differences in risk and resilience or the response to other pharmaceutical treatments or subsequent life events. Further understanding of a woman’s adaptive neuroimmune responses to pregnancy is needed for the prevention of pre-term birth by strategically enhancing these. Likewise, enhancing our understanding of the impact of neuroimmunological adaptations in pregnancy and post-partum on mood is important for strategies to prevent and treat pregnancy-related mood disorders. In the study of female-specific cancer effects on the brain, attention and systematic modelling of a more clinically-relevant physiological environment is imperative. For example, most breast cancer patients are post-menopausal and middle aged, whereas most neurobiological work in this field is performed in young adult rodents with functioning ovaries (i.e., higher circulating estrogens). In addition, basic research to address the strong potential for exposure to estrogen modulator treatments, which are commonly administered for 5-10+ years to estrogen-responsive cancer patients, to impact cognition and mood is long overdue. Much can also be gained regarding how estrogen modulates behavioral comorbidities in this context by reporting menopausal status, or better yet, circulating estrogen concentrations in the clinical research in this field. Aged females are at greatest risk for developing Alzheimer’s disease and other dementias. Neuroinflammation is widely regarded as playing a role in these diseases and, as we have reviewed, estrogen depletion in the context of aging exacerbates neuroinflammatory responses. A critical question in the aging literature that remains unanswered is whether early neuroinflammation triggers important neural changes that initiate neurodegenerative disease, or if other early disease modifications cause neuroinflammation, which in turn accelerate disease progression. Understanding prodromal modifications in the aged female brain will undoubtedly lead to exciting discoveries and more effective targets for treatments of Alzheimer’s disease.

Studying the female brain may pose some additional challenges; those associated with hormonal cycles and behavioral variability, yet to a scientific community capable of developing such magnificent technological leaps as the human genome project, Brainbow, and optogenetics, these challenges should not be insurmountable. Understanding something as dynamic, vulnerable, and resilient as the female brain presents one of the next great frontiers for future scientific endeavors.

Highlights.

  • Female brains are highly dynamic, remodelling throughout the normal ovarian cycle

  • They also demonstrate unique changes across various life stages

  • Females uniquely respond to early life challenges, pregnancy, disease and old-age

  • The female brain is under-researched but the field is, encouragingly, growing.

Acknowledgements:

This work was supported by grants from the National Institute on Aging, USA (RO1AG028271 and R21AG058109) to RMB; the Biotechnology and Biological Sciences Research Council, UK to PJB; the National Institute of Mental Health (R21MH105826) and NARSAD Young Investigator, USA Award to KML; the National Institute of Cancer, USA (R01CA216290 and R21AG058109) to LMP; and a National Health and Medical Research Count Career Development Fellowship II, Australia (APP1128646) to SJS. Graphic illustration funded by The Ohio State University Neurological Research Institute.

Footnotes

Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

7. References

  1. Aagaard-Tillery KM, Silver R, and Dalton J (2006). Immunology of normal pregnancy. Semin Fetal Neonatal Med 11, 279–295. [DOI] [PubMed] [Google Scholar]
  2. Aavani T, Rana SA, Hawkes R, and Pittman QJ (2015). Maternal immune activation produces cerebellar hyperplasia and alterations in motor and social behaviors in male and female mice. Cerebellum 14, 491–505. [DOI] [PubMed] [Google Scholar]
  3. Abu-Taha M, Rius C, Hermenegildo C, Noguera I, Cerda-Nicolas JM, Issekutz AC, Jose PJ, Cortijo J, Morcillo EJ, and Sanz MJ (2009). Menopause and ovariectomy cause a low grade of systemic inflammation that may be prevented by chronic treatment with low doses of estrogen or losartan. J Immunol 183, 1393–1402. [DOI] [PubMed] [Google Scholar]
  4. Acaz-Fonseca E, Sanchez-Gonzalez R, Azcoitia I, Arevalo MA, and Garcia-Segura LM (2014). Role of astrocytes in the neuroprotective actions of 17β-estradiol and selective estrogen receptor modulators. Molecular and cellular endocrinology 389, 48–57. [DOI] [PubMed] [Google Scholar]
  5. Agrawal K, Onami S, Mortimer JE, and Pal SK (2010). Cognitive changes associated with endocrine therapy for breast cancer. Maturitas 67, 209–214. [DOI] [PMC free article] [PubMed] [Google Scholar]
  6. Ahles TA, Root JC, and Ryan EL (2012). Cancer- and cancer treatment-associated cognitive change: an update on the state of the science. J Clin Oncol 30, 3675–3686. [DOI] [PMC free article] [PubMed] [Google Scholar]
  7. Ahles TA, and Saykin AJ (2007). Candidate mechanisms for chemotherapy-induced cognitive changes. Nat Rev Cancer 7, 192–201. [DOI] [PMC free article] [PubMed] [Google Scholar]
  8. Ahles TA, Saykin AJ, McDonald BC, Furstenberg CT, Cole BF, Hanscom BS, Mulrooney TJ, Schwartz GN, and Kaufman PA (2008). Cognitive function in breast cancer patients prior to adjuvant treatment. Breast Cancer Res Treat 110, 143–152. [DOI] [PMC free article] [PubMed] [Google Scholar]
  9. Ahles TA, Saykin AJ, McDonald BC, Li Y, Furstenberg CT, Hanscom BS, Mulrooney TJ, Schwartz GN, and Kaufman PA (2010). Longitudinal assessment of cognitive changes associated with adjuvant treatment for breast cancer: impact of age and cognitive reserve. J Clin Oncol 28, 4434–4440. [DOI] [PMC free article] [PubMed] [Google Scholar]
  10. Alzheimer’s Association (2016). 2016 Alzheimer’s disease facts and figures. Alzheimer’s & dementia : the journal of the Alzheimer’s Association 12, 1–80. [DOI] [PubMed] [Google Scholar]
  11. Ancoli-Israel S, Liu L, Rissling M, Natarajan L, Neikrug AB, Palmer BW, Mills PJ, Parker BA, Sadler GR, and Maglione J (2014). Sleep, fatigue, depression, and circadian activity rhythms in women with breast cancer before and after treatment: a 1-year longitudinal study. Supportive care in cancer : official journal of the Multinational Association of Supportive Care in Cancer 22, 2535–2545. [DOI] [PMC free article] [PubMed] [Google Scholar]
  12. Anderson GL, Limacher M, Assaf AR, Bassford T, Beresford SA, Black H, Bonds D, Brunner R, Brzyski R, Caan B, et al. (2004). Effects of conjugated equine estrogen in postmenopausal women with hysterectomy: the Women’s Health Initiative randomized controlled trial. Jama 291, 1701–1712. [DOI] [PubMed] [Google Scholar]
  13. Arakawa K, Arakawa H, Hueston CM, and Deak T (2014). Effects of the estrous cycle and ovarian hormones on central expression of interleukin-1 evoked by stress in female rats. Neuroendocrinology 100, 162–177. [DOI] [PubMed] [Google Scholar]
  14. Arevalo MA, Santos-Galindo M, Lagunas N, Azcoitia I, and Garcia-Segura LM (2011). Selective estrogen receptor modulators as brain therapeutic agents. J Mol Endocrinol 46, R1–9. [DOI] [PubMed] [Google Scholar]
  15. Ashdown H, Poole S, Boksa P, and Luheshi GN (2007). Interleukin-1 receptor antagonist as a modulator of gender differences in the febrile response to lipopolysaccharide in rats. Am J Physiol Regul Integr Comp Physiol. 292, R1667–R1674. [DOI] [PubMed] [Google Scholar]
  16. Atkinson HC, and Waddell BJ (1995). The hypothalamic-pituitary-adrenal axis in rat pregnancy and lactation: circadian variation and interrelationship of plasma adrenocorticotropin and corticosterone. Endocrinology 136, 512–520. [DOI] [PubMed] [Google Scholar]
  17. Aydin M, Yilmaz B, Alcin E, Nedzvetsky VS, Sahin Z, and Tuzcu M (2008). Effects of letrozole on hippocampal and cortical catecholaminergic neurotransmitter levels, neural cell adhesion molecule expression and spatial learning and memory in female rats. Neuroscience 151, 186–194. [DOI] [PubMed] [Google Scholar]
  18. Azcoitia I, Sierra A, and Garcia-Segura LM (1999). Localization of estrogen receptor beta-immunoreactivity in astrocytes of the adult rat brain. Glia 26, 260–267. [PubMed] [Google Scholar]
  19. Bachman DL, Wolf PA, Linn R, Knoefel JE, Cobb J, Belanger A, D’Agostino RB, and White LR (1992). Prevalence of dementia and probable senile dementia of the Alzheimer type in the Framingham Study. Neurology 42, 115–119. [DOI] [PubMed] [Google Scholar]
  20. Bakker J, and Baum MJ (2008). Role for estradiol in female-typical brain and behavioral sexual differentiation. Frontiers in neuroendocrinology 29, 1–16. [DOI] [PMC free article] [PubMed] [Google Scholar]
  21. Bakoyiannis I, Tsigka EA, Perrea D, and Pergialiotis V (2016). The Impact of Endocrine Therapy on Cognitive Functions of Breast Cancer Patients: A Systematic Review. Clin Drug Investig 36, 109–118. [DOI] [PubMed] [Google Scholar]
  22. Barha CK, and Galea LA (2017). The maternal ‘baby brain’ revisited. Nature neuroscience 20, 134–135. [DOI] [PubMed] [Google Scholar]
  23. Barrientos RM, Frank MG, Watkins LR, and Maier SF (2012). Aging-related changes in neuroimmune-endocrine function: implications for hippocampal-dependent cognition. Hormones and behavior 62, 219–227. [DOI] [PMC free article] [PubMed] [Google Scholar]
  24. Barrientos RM, Kitt MM, Watkins LR, and Maier SF (2015). Neuroinflammation in the normal aging hippocampus. Neuroscience 309, 84–99. [DOI] [PMC free article] [PubMed] [Google Scholar]
  25. Barrientos RM, Watkins LR, Rudy JW, and Maier SF (2009). Characterization of the sickness response in young and aging rats following E. coli infection. Brain Behav Immun 23, 450–454. [DOI] [PMC free article] [PubMed] [Google Scholar]
  26. Beery AK, and Zucker I (2011). Sex bias in neuroscience and biomedical research. Neuroscience and biobehavioral reviews 35, 565–572. [DOI] [PMC free article] [PubMed] [Google Scholar]
  27. Benarroch EE (2013). Microglia: Multiple roles in surveillance, circuit shaping, and response to injury. Neurology 81, 1079–1088. [DOI] [PubMed] [Google Scholar]
  28. Bender CM, Merriman JD, Gentry AL, Ahrendt GM, Berga SL, Brufsky AM, Casillo FE, Dailey MM, Erickson KI, Kratofil FM, et al. (2015). Patterns of change in cognitive function with anastrozole therapy. Cancer 121, 2627–2636. [DOI] [PMC free article] [PubMed] [Google Scholar]
  29. Bender CM, Paraska KK, Sereika SM, Ryan CM, and Berga SL (2001). Cognitive function and reproductive hormones in adjuvant therapy for breast cancer: a critical review. J Pain Symptom Manage 21, 407–424. [DOI] [PubMed] [Google Scholar]
  30. Bender CM, Sereika SM, Brufsky AM, Ryan CM, Vogel VG, Rastogi P, Cohen SM, Casillo FE, and Berga SL (2007). Memory impairments with adjuvant anastrozole versus tamoxifen in women with early-stage breast cancer. Menopause 14, 995–998. [DOI] [PMC free article] [PubMed] [Google Scholar]
  31. Benedusi V, Meda C, Della Torre S, Monteleone G, Vegeto E, and Maggi A (2012). A lack of ovarian function increases neuroinflammation in aged mice. Endocrinology 153, 2777–2788. [DOI] [PMC free article] [PubMed] [Google Scholar]
  32. Berchtold NC, Kesslak JP, Pike CJ, Adlard PA, and Cotman CW (2001). Estrogen and exercise interact to regulate brain-derived neurotrophic factor mRNA and protein expression in the hippocampus. The European journal of neuroscience 14, 1992–2002. [DOI] [PubMed] [Google Scholar]
  33. Berkley KJ, McAllister SL, Accius BE, and Winnard KP (2007). Endometriosis-induced vaginal hyperalgesia in the rat: effect of estropause, ovariectomy, and estradiol replacement. Pain 132 Suppl 1, S150–159. [DOI] [PMC free article] [PubMed] [Google Scholar]
  34. Bilbo SD, Block CL, Bolton JL, Hanamsagar R, and Tran PK (2018). Beyond infection - Maternal immune activation by environmental factors, microglial development, and relevance for autism spectrum disorders. Exp Neurol 299, 241–251. [DOI] [PMC free article] [PubMed] [Google Scholar]
  35. Blecher SR, and Erickson RP (2007). Genetics of sexual development: a new paradigm. Am J Med Genet A 143A, 3054–3068. [DOI] [PubMed] [Google Scholar]
  36. Bloch M, Schmidt PJ, Danaceau M, Murphy J, Nieman L, and Rubinow DR (2000). Effects of gonadal steroids in women with a history of postpartum depression. Am J Psychiatry 157, 924–930. [DOI] [PubMed] [Google Scholar]
  37. Bolton JL, Auten RL, and Bilbo SD (2014). Prenatal air pollution exposure induces sexually dimorphic fetal programming of metabolic and neuroinflammatory outcomes in adult offspring. Brain Behav Immun 37, 30–44. [DOI] [PubMed] [Google Scholar]
  38. Bolton JL, Huff NC, Smith SH, Mason SN, Foster WM, Auten RL, and Bilbo SD (2013). Maternal stress and effects of prenatal air pollution on offspring mental health outcomes in mice. Environ Health Perspect 121, 1075–1082. [DOI] [PMC free article] [PubMed] [Google Scholar]
  39. Borenstein AR, Wu Y, Bowen JD, McCormick WC, Uomoto J, McCurry SM, Schellenberg GD, and Larson EB (2014). Incidence rates of dementia, Alzheimer disease, and vascular dementia in the Japanese American population in Seattle, WA: the Kame Project. Alzheimer Dis Assoc Disord 28, 23–29. [DOI] [PMC free article] [PubMed] [Google Scholar]
  40. Bridges RS (1984). A quantitative analysis of the roles of dosage, sequence, and duration of estradiol and progesterone exposure in the regulation of maternal behavior in the rat. Endocrinology 114, 930–940. [DOI] [PubMed] [Google Scholar]
  41. Brock O, Baum MJ, and Bakker J (2011). The development of female sexual behavior requires prepubertal estradiol. J Neurosci 31, 5574–5578. [DOI] [PMC free article] [PubMed] [Google Scholar]
  42. Bronson SL, and Bale TL (2014). Prenatal stress-induced increases in placental inflammation and offspring hyperactivity are male-specific and ameliorated by maternal antiinflammatory treatment. Endocrinology 155, 2635–2646. [DOI] [PMC free article] [PubMed] [Google Scholar]
  43. Brown CM, Mulcahey TA, Filipek NC, and Wise PM (2010). Production of proinflammatory cytokines and chemokines during neuroinflammation: novel roles for estrogen receptors alpha and beta. Endocrinology 151, 4916–4925. [DOI] [PMC free article] [PubMed] [Google Scholar]
  44. Brucato M, Ladd-Acosta C, Li M, Caruso D, Hong X, Kaczaniuk J, Stuart EA, Fallin MD, and Wang X (2017). Prenatal exposure to fever is associated with autism spectrum disorder in the boston birth cohort. Autism Res 10, 1878–1890. [DOI] [PMC free article] [PubMed] [Google Scholar]
  45. Bruce-Keller AJ, Keeling JL, Keller JN, Huang FF, Camondola S, and Mattson MP (2000). Antiinflammatory effects of estrogen on microglial activation. Endocrinology 141, 3646–3656. [DOI] [PubMed] [Google Scholar]
  46. Brunton PJ, Bales J, and Russell JA (2012). Allopregnanolone and induction of endogenous opioid inhibition of oxytocin responses to immune stress in pregnant rats. Journal of neuroendocrinology 24, 690–700. [DOI] [PubMed] [Google Scholar]
  47. Brunton PJ, McKay AJ, Ochedalski T, Piastowska A, Rebas E, Lachowicz A, and Russell JA (2009). Central opioid inhibition of neuroendocrine stress responses in pregnancy in the rat is induced by the neurosteroid allopregnanolone. J Neurosci 29, 6449–6460. [DOI] [PMC free article] [PubMed] [Google Scholar]
  48. Brunton PJ, Meddle SL, Ma S, Ochedalski T, Douglas AJ, and Russell JA (2005). Endogenous opioids and attenuated hypothalamic-pituitary-adrenal axis responses to immune challenge in pregnant rats. J Neurosci 25, 5117–5126. [DOI] [PMC free article] [PubMed] [Google Scholar]
  49. Brunton PJ, and Russell JA (2008). Attenuated hypothalamo-pituitary-adrenal axis responses to immune challenge during pregnancy: the neurosteroid opioid connection. The Journal of physiology 586, 369–375. [DOI] [PMC free article] [PubMed] [Google Scholar]
  50. Brunton PJ, and Russell JA (2010). Endocrine induced changes in brain function during pregnancy. Brain research 1364, 198–215. [DOI] [PubMed] [Google Scholar]
  51. Brunton PJ, Russell JA, and Douglas AJ (2008). Adaptive responses of the maternal hypothalamic-pituitary-adrenal axis during pregnancy and lactation. Journal of neuroendocrinology 20, 764–776. [DOI] [PubMed] [Google Scholar]
  52. Brunton PJ, Russell JA, and Hirst JJ (2014). Allopregnanolone in the brain: protecting pregnancy and birth outcomes. Progress in neurobiology 113, 106–136. [DOI] [PubMed] [Google Scholar]
  53. Brunton PJ, Sabatier N, Leng G, and Russell JA (2006). Suppressed oxytocin neuron responses to immune challenge in late pregnant rats: a role for endogenous opioids. The European journal of neuroscience 23, 1241–1247. [DOI] [PubMed] [Google Scholar]
  54. Buchanan ND, Dasari S, Rodriguez JL, Lee Smith J, Hodgson ME, Weinberg CR, and Sandler DP (2015). Post-treatment Neurocognition and Psychosocial Care Among Breast Cancer Survivors. American journal of preventive medicine 49, S498–508. [DOI] [PMC free article] [PubMed] [Google Scholar]
  55. Buller K, Xu Y, Dayas C, and Day T (2001). Dorsal and ventral medullary catecholamine cell groups contribute differentially to systemic interleukin-1beta-induced hypothalamic pituitary adrenal axis responses. Neuroendocrinology 73, 129–138. [DOI] [PubMed] [Google Scholar]
  56. Buller KM, Allen T, Wilson LD, Munro F, and Day TA (2004). A critical role for the parabrachial nucleus in generating central nervous system responses elicited by a systemic immune challenge. J Neuroimmunol 152, 20–32. [DOI] [PubMed] [Google Scholar]
  57. Burnham V, Sundby C, Laman-Maharg A, and Thornton J (2016). Luteinizing hormone acts at the hippocampus to dampen spatial memory. Hormones and behavior 89, 55–63. [DOI] [PubMed] [Google Scholar]
  58. Cao XZ, Ma H, Wang JK, Liu F, Wu BY, Tian AY, Wang LL, and Tan WF (2010). Postoperative cognitive deficits and neuroinflammation in the hippocampus triggered by surgical trauma are exacerbated in aged rats. Progress in neuro-psychopharmacology & biological psychiatry 34, 1426–1432. [DOI] [PubMed] [Google Scholar]
  59. Carrel L, and Willard HF (2005). X-inactivation profile reveals extensive variability in X-linked gene expression in females. Nature 434, 400–404. [DOI] [PubMed] [Google Scholar]
  60. Castellon SA, Ganz PA, Bower JE, Petersen L, Abraham L, and Greendale GA (2004). Neurocognitive performance in breast cancer survivors exposed to adjuvant chemotherapy and tamoxifen. J Clin Exp Neuropsychol 26, 955–969. [DOI] [PubMed] [Google Scholar]
  61. Cerciat M, Unkila M, Garcia-Segura LM, and Arevalo MA (2010). Selective estrogen receptor modulators decrease the production of interleukin-6 and interferon-gamma-inducible protein-10 by astrocytes exposed to inflammatory challenge in vitro. Glia 58, 93–102. [DOI] [PubMed] [Google Scholar]
  62. Chakraborty TR, and Gore AC (2004). Aging-related changes in ovarian hormones, their receptors, and neuroendocrine function. Exp Biol Med (Maywood) 229, 977–987. [DOI] [PubMed] [Google Scholar]
  63. Chen CH, Huang TH, Cheng TL, Chang CF, Wang CZ, Wu MH, and Kang L (2016). Exercise training ameliorates glucosamine-induced insulin resistance in ovariectomized rats. Menopause. [DOI] [PubMed] [Google Scholar]
  64. Chisholm NC, and Sohrabji F (2016). Astrocytic response to cerebral ischemia is influenced by sex differences and impaired by aging. Neurobiol Dis 85, 245–253. [DOI] [PMC free article] [PubMed] [Google Scholar]
  65. Choleris E, Galea LAM, Sohrabji F, and Frick KM (2018). Sex differences in the brain: Implications for behavioral and biomedical research. Neuroscience and biobehavioral reviews 85, 126–145. [DOI] [PMC free article] [PubMed] [Google Scholar]
  66. Collins B, Mackenzie J, Stewart A, Bielajew C, and Verma S (2009). Cognitive effects of chemotherapy in post-menopausal breast cancer patients 1 year after treatment. Psychooncology 18, 134–143. [DOI] [PubMed] [Google Scholar]
  67. Colonna M, and Butovsky O (2017). Microglia Function in the Central Nervous System During Health and Neurodegeneration. Annu Rev Immunol 35, 441–468. [DOI] [PMC free article] [PubMed] [Google Scholar]
  68. Concas A, Mostallino MC, Porcu P, Follesa P, Barbaccia ML, Trabucchi M, Purdy RH, Grisenti P, and Biggio G (1998). Role of brain allopregnanolone in the plasticity of gamma-aminobutyric acid type A receptor in rat brain during pregnancy and after delivery. Proceedings of the National Academy of Sciences of the United States of America 95, 13284–13289. [DOI] [PMC free article] [PubMed] [Google Scholar]
  69. Conroy SK, McDonald BC, Ahles TA, West JD, and Saykin AJ (2013). Chemotherapy-induced amenorrhea: a prospective study of brain activation changes and neurocognitive correlates. Brain Imaging Behav 7, 491–500. [DOI] [PMC free article] [PubMed] [Google Scholar]
  70. Costello DA, Keenan K, McManus RM, Falvey A, and Lynch MA (2016). The age-related neuroinflammatory environment promotes macrophage activation, which negatively impacts synaptic function. Neurobiology of aging 43, 140–148. [DOI] [PubMed] [Google Scholar]
  71. Crain JM, Nikodemova M, and Watters JJ (2013). Microglia express distinct M1 and M2 phenotypic markers in the postnatal and adult central nervous system in male and female mice. J Neurosci Res 91, 1143–1151. [DOI] [PMC free article] [PubMed] [Google Scholar]
  72. Cribbs DH, Berchtold NC, Perreau V, Coleman PD, Rogers J, Tenner AJ, and Cotman CW (2012). Extensive innate immune gene activation accompanies brain aging, increasing vulnerability to cognitive decline and neurodegeneration: a microarray study. Journal of neuroinflammation 9, 179. [DOI] [PMC free article] [PubMed] [Google Scholar]
  73. Cunningham CL, Martinez-Cerdeno V, and Noctor SC (2013). Microglia regulate the number of neural precursor cells in the developing cerebral cortex. J Neurosci 33, 4216–4233. [DOI] [PMC free article] [PubMed] [Google Scholar]
  74. da Palma RK, Moraes-Silva IC, da Silva Dias D, Shimojo GL, Conti FF, Bernardes N, Barboza CA, Sanches IC, da Rosa Araujo AS, Irigoyen MC, et al. (2016). Resistance or aerobic training decreases blood pressure and improves cardiovascular autonomic control and oxidative stress in hypertensive menopausal rats. J Appl Physiol (1985) 121, 1032–1038. [DOI] [PubMed] [Google Scholar]
  75. Danforth DN (1991). How subsequent pregnancy affects outcome in women with a prior breast cancer. Oncology (Williston Park) 5, 23–30; discussion 30-21, 35. [PubMed] [Google Scholar]
  76. Davalos D, Grutzendler J, Yang G, Kim JV, Zuo Y, Jung S, Littman DR, Dustin ML, and Gan WB (2005). ATP mediates rapid microglial response to local brain injury in vivo. Nature neuroscience 8, 752–758. [DOI] [PubMed] [Google Scholar]
  77. De Melo VU, Saldanha RR, Dos Santos CR, De Campos Cruz J, Lira VA, Santana-Filho VJ, and Michelini LC (2016). Ovarian Hormone Deprivation Reduces Oxytocin Expression in Paraventricular Nucleus Preautonomic Neurons and Correlates with Baroreflex Impairment in Rats. Frontiers in physiology 7, 461. [DOI] [PMC free article] [PubMed] [Google Scholar]
  78. de Oliveira AP, Peron JP, Damazo AS, Franco AL, Domingos HV, Oliani SM, Oliveira-Filho RM, Vargaftig BB, and Tavares-de-Lima W (2010). Female sex hormones mediate the allergic lung reaction by regulating the release of inflammatory mediators and the expression of lung E-selectin in rats. Respir Res 11, 115. [DOI] [PMC free article] [PubMed] [Google Scholar]
  79. de Theije CG, van den Elsen LW, Willemsen LE, Milosevic V, Korte-Bouws GA, Lopes da Silva S, Broersen LM, Korte SM, Olivier B, Garssen J, et al. (2015). Dietary long chain n-3 polyunsaturated fatty acids prevent impaired social behaviour and normalize brain dopamine levels in food allergic mice. Neuropharmacology 90, 15–22. [DOI] [PubMed] [Google Scholar]
  80. Dimayuga FO, Reed JL, Carnero GA, Wang C, Dimayuga ER, Dimayuga VM, Perger A, Wilson ME, Keller JN, and Bruce-Keller AJ (2005). Estrogen and brain inflammation: effects on microglial expression of MHC, costimulatory molecules and cytokines. J Neuroimmunol 161, 123–136. [DOI] [PubMed] [Google Scholar]
  81. Douglas AJ, Scullion S, Antonijevic IA, Brown D, Russell JA, and Leng G (2001). Uterine contractile activity stimulates supraoptic neurons in term pregnant rats via a noradrenergic pathway. Endocrinology 142, 633–644. [DOI] [PubMed] [Google Scholar]
  82. Doyle HH, Eidson LN, Sinkiewicz DM, and Murphy AZ (2017). Sex Differences in Microglia Activity within the Periaqueductal Gray of the Rat: A Potential Mechanism Driving the Dimorphic Effects of Morphine. J Neurosci 37, 3202–3214. [DOI] [PMC free article] [PubMed] [Google Scholar]
  83. Drew PD, and Chavis JA (2000). Inhibition of microglial cell activation by cortisol. Brain research bulletin 52, 391–396. [DOI] [PubMed] [Google Scholar]
  84. Driscoll I, Hamilton DA, Yeo RA, Brooks WM, and Sutherland RJ (2005). Virtual navigation in humans: the impact of age, sex, and hormones on place learning. Hormones and behavior 47, 326–335. [DOI] [PubMed] [Google Scholar]
  85. Dubal DB, and Rogine C (2017). Apolipoprotein E epsilon4 and Risk Factors for Alzheimer Disease-Let’s Talk About Sex. JAMA Neurol 74, 1167–1168. [DOI] [PubMed] [Google Scholar]
  86. Dubal DB, and Wise PM (2002). Estrogen and neuroprotection: from clinical observations to molecular mechanisms. Dialogues in clinical neuroscience 4, 149–161. [DOI] [PMC free article] [PubMed] [Google Scholar]
  87. Duckles SP, and Krause DN (2011). Mechanisms of cerebrovascular protection: oestrogen, inflammation and mitochondria. Acta Physiol (Oxf) 203, 149–154. [DOI] [PMC free article] [PubMed] [Google Scholar]
  88. Duma D, Collins JB, Chou JW, and Cidlowski JA (2010). Sexually dimorphic actions of glucocorticoids provide a link to inflammatory diseases with gender differences in prevalence. Science signaling 3, ra74. [DOI] [PMC free article] [PubMed] [Google Scholar]
  89. Edison P, and Brooks DJ (2018). Role of Neuroinflammation in the Trajectory of Alzheimer’s Disease and in vivo Quantification Using PET. J Alzheimers Dis 64, S339–S351. [DOI] [PubMed] [Google Scholar]
  90. Edwards MJ (2006). Review: Hyperthermia and fever during pregnancy. Birth Defects Res A Clin Mol Teratol. 76, 507–516. [DOI] [PubMed] [Google Scholar]
  91. Ek M, Kurosawa M, Lundeberg T, and Ericsson A (1998). Activation of vagal afferents after intravenous injection of interleukin-1beta: Role of endogenous prostaglandins. Journal of Neuroscience 18, 9471–9479. [DOI] [PMC free article] [PubMed] [Google Scholar]
  92. Engelmann F, Barron A, Urbanski H, Neuringer M, Kohama SG, Park B, and Messaoudi I (2011). Accelerated immune senescence and reduced response to vaccination in ovariectomized female rhesus macaques. Age (Dordr) 33, 275–289. [DOI] [PMC free article] [PubMed] [Google Scholar]
  93. Ericsson A, Arias C, and Sawchenko PE (1997). Evidence for an intramedullary prostaglandin-dependent mechanism in the activation of stress-related neuroendocrine circuitry by intravenous interleukin-1. J Neurosci 17, 7166–7179. [DOI] [PMC free article] [PubMed] [Google Scholar]
  94. Ericsson A, Kovacs KJ, and Sawchenko PE (1994). A functional anatomical analysis of central pathways subserving the effects of interleukin-1 on stress-related neuroendocrine neurons. Journal of Neuroscience 14, 897–913. [DOI] [PMC free article] [PubMed] [Google Scholar]
  95. Ericsson A, Liu C, Hart RP, and Sawchenko PE (1995). Type 1 interleukin-1 receptor in the rat brain: Distribution, regulation, and relationship to sites of IL-1-induced cellular activation. Journal of Comparative Neurology 361, 681–698. [DOI] [PubMed] [Google Scholar]
  96. Estes ML, and McAllister AK (2016). Maternal immune activation: Implications for neuropsychiatric disorders. Science 353, 772–777. [DOI] [PMC free article] [PubMed] [Google Scholar]
  97. Fairweather D, Frisancho-Kiss S, and Rose NR (2008). Sex differences in autoimmune disease from a pathological perspective. Am J Pathol 173, 600–609. [DOI] [PMC free article] [PubMed] [Google Scholar]
  98. Ferreira ST, Clarke JR, Bomfim TR, and De Felice FG (2014). Inflammation, defective insulin signaling, and neuronal dysfunction in Alzheimer’s disease. Alzheimer’s & dementia : the journal of the Alzheimer’s Association 10, S76–83. [DOI] [PubMed] [Google Scholar]
  99. Finley C, Zhang C, and Fewell JE (2015). Sex steroid levels near the term of pregnancy do not alter lipopolysaccharide-induced fever in oophorectomized rats. Experimental physiology 100, 323–330. [DOI] [PubMed] [Google Scholar]
  100. Fisher RE, Karrow NA, Quinton M, Finegan EJ, Miller SP, Atkinson JL, and Boermans HJ (2010). Endotoxin exposure during late pregnancy alters ovine offspring febrile and hypothalamic-pituitary-adrenal axis responsiveness later in life. Stress (Amsterdam, Netherlands) 13, 334–342. [DOI] [PubMed] [Google Scholar]
  101. Frank MG, Barrientos RM, Watkins LR, and Maier SF (2010). Aging sensitizes rapidly isolated hippocampal microglia to LPS ex vivo. J Neuroimmunol 226, 181–184. [DOI] [PMC free article] [PubMed] [Google Scholar]
  102. Frankfurt M, Gould E, Woolley CS, and McEwen BS (1990). Gonadal steroids modify dendritic spine density in ventromedial hypothalamic neurons: a Golgi study in the adult rat. Neuroendocrinology 51, 530–535. [DOI] [PubMed] [Google Scholar]
  103. Frederic F, Oliver C, Wollman E, Delhaye-Bouchaud N, and Mariani J (1993). IL-1 and LPS induce a sexually dimorphic response of the hypothalamo-pituitary-adrenal axis in several mouse strains. Eur Cytokine Netw 4, 321–329. [PubMed] [Google Scholar]
  104. Freeman EW (2014). Hot flushes and the menopause: how long should they be expected to last? Maturitas 78, 153–154. [DOI] [PubMed] [Google Scholar]
  105. Furman D, Hejblum BP, Simon N, Jojic V, Dekker CL, Thiebaut R, Tibshirani RJ, and Davis MM (2014). Systems analysis of sex differences reveals an immunosuppressive role for testosterone in the response to influenza vaccination. Proceedings of the National Academy of Sciences of the United States of America 111, 869–874. [DOI] [PMC free article] [PubMed] [Google Scholar]
  106. Galea LA, Leuner B, and Slattery DA (2014). Hippocampal plasticity during the peripartum period: influence of sex steroids, stress and ageing. Journal of neuroendocrinology 26, 641–648. [DOI] [PMC free article] [PubMed] [Google Scholar]
  107. Gartler SM, and Riggs AD (1983). Mammalian X-chromosome inactivation. Annu Rev Genet 17, 155–190. [DOI] [PubMed] [Google Scholar]
  108. Gaskins AJ, Wilchesky M, Mumford SL, Whitcomb BW, Browne RW, Wactawski-Wende J, Perkins NJ, and Schisterman EF (2012). Endogenous reproductive hormones and C-reactive protein across the menstrual cycle: the BioCycle Study. American journal of epidemiology 175, 423–431. [DOI] [PMC free article] [PubMed] [Google Scholar]
  109. Ghisletti S, Meda C, Maggi A, and Vegeto E (2005). 17beta-estradiol inhibits inflammatory gene expression by controlling NF-kappaB intracellular localization. Mol Cell Biol 25, 2957–2968. [DOI] [PMC free article] [PubMed] [Google Scholar]
  110. Gibson EM, Nagaraja S, Ocampo A, Tam LT, Wood LS, Pallegar PN, Greene JJ, Geraghty AC, Goldstein AK, Ni L, et al. (2019). Methotrexate Chemotherapy Induces Persistent Tri-glial Dysregulation that Underlies Chemotherapy-Related Cognitive Impairment. Cell 176, 43–55 e13. [DOI] [PMC free article] [PubMed] [Google Scholar]
  111. Gillies GE, Pienaar IS, Vohra S, and Qamhawi Z (2014). Sex differences in Parkinson’s disease. Frontiers in neuroendocrinology 35, 370–384. [DOI] [PMC free article] [PubMed] [Google Scholar]
  112. Goldman JM, Murr AS, and Cooper RL (2007). The rodent estrous cycle: characterization of vaginal cytology and its utility in toxicological studies. Birth Defects Res B Dev Reprod Toxicol 80, 84–97. [DOI] [PubMed] [Google Scholar]
  113. Gonzalez M, Cabrera-Socorro A, Perez-Garcia CG, Fraser JD, Lopez FJ, Alonso R, and Meyer G (2007). Distribution patterns of estrogen receptor alpha and beta in the human cortex and hippocampus during development and adulthood. The Journal of comparative neurology 503, 790–802. [DOI] [PubMed] [Google Scholar]
  114. Gould E, Woolley CS, Frankfurt M, and McEwen BS (1990). Gonadal steroids regulate dendritic spine density in hippocampal pyramidal cells in adulthood. J Neurosci 10, 1286–1291. [DOI] [PMC free article] [PubMed] [Google Scholar]
  115. Gregg C, Zhang J, Butler JE, Haig D, and Dulac C (2010). Sex-specific parent-of-origin allelic expression in the mouse brain. Science 329, 682–685. [DOI] [PMC free article] [PubMed] [Google Scholar]
  116. Gupta P, Sturdee DW, Palin SL, Majumder K, Fear R, Marshall T, and Paterson I (2006). Menopausal symptoms in women treated for breast cancer: the prevalence and severity of symptoms and their perceived effects on quality of life. Climacteric 9, 49–58. [DOI] [PubMed] [Google Scholar]
  117. Habib P, and Beyer C (2015). Regulation of brain microglia by female gonadal steroids. The Journal of steroid biochemistry and molecular biology 146, 3–14. [DOI] [PubMed] [Google Scholar]
  118. Hagedoorn M, Buunk BP, Kuijer RG, Wobbes T, and Sanderman R (2000). Couples dealing with cancer: role and gender differences regarding psychological distress and quality of life. Psychooncology 9, 232–242. [DOI] [PubMed] [Google Scholar]
  119. Hagemeyer N, Hanft KM, Akriditou MA, Unger N, Park ES, Stanley ER, Staszewski O, Dimou L, and Prinz M (2017). Microglia contribute to normal myelinogenesis and to oligodendrocyte progenitor maintenance during adulthood. Acta Neuropathol 134, 441–458. [DOI] [PMC free article] [PubMed] [Google Scholar]
  120. Haim A, Julian D, Albin-Brooks C, Brothers HM, Lenz KM, and Leuner B (2017). A survey of neuroimmune changes in pregnant and postpartum female rats. Brain Behav Immun 59, 67–78. [DOI] [PubMed] [Google Scholar]
  121. Hanamsagar R, Alter MD, Block CS, Sullivan H, Bolton JL, and Bilbo SD (2017). Generation of a microglial developmental index in mice and in humans reveals a sex difference in maturation and immune reactivity. Glia 65, 1504–1520. [DOI] [PMC free article] [PubMed] [Google Scholar]
  122. Harre EM, Mouihate A, and Pittman QJ (2006). Attenuation of fever at near term: is interleukin-6-STAT3 signalling altered? J Neuroendocrinol. 18, 57–63. [DOI] [PubMed] [Google Scholar]
  123. He W, Goodkind D, and Kowal P (2016). An Aging World: 2015. C.B. U.S. Department of Commerce, and N.I.o.A. U.S. Department of Health and Human Services, eds. (Washington, DC: U.S. Government Publishing Office; ). [Google Scholar]
  124. Hebert LE, Weuve J, Scherr PA, and Evans DA (2013). Alzheimer disease in the United States (2010-2050) estimated using the 2010 census. Neurology 80, 1778–1783. [DOI] [PMC free article] [PubMed] [Google Scholar]
  125. Henderson VW, and Rocca WA (2012). Estrogens and Alzheimer disease risk: is there a window of opportunity? Neurology 79, 1840–1841. [DOI] [PubMed] [Google Scholar]
  126. Henderson YO, Nalloor R, Vazdarjanova A, Murphy AZ, and Parent MB (2017). Sex-dependent effects of early life inflammatory pain on sucrose intake and sucrose-associated hippocampal Arc expression in adult rats. Physiology & behavior 173, 1–8. [DOI] [PubMed] [Google Scholar]
  127. Herbison AE, Voisin DL, Douglas AJ, and Chapman C (1997). Profile of monoamine and excitatory amino acid release in rat supraoptic nucleus over parturition. Endocrinology 138, 33–40. [DOI] [PubMed] [Google Scholar]
  128. Hillerer KM, Jacobs VR, Fischer T, and Aigner L (2014). The maternal brain: an organ with peripartal plasticity. Neural plasticity 2014, 574159. [DOI] [PMC free article] [PubMed] [Google Scholar]
  129. Hodes GE, Kana V, Menard C, Merad M, and Russo SJ (2015). Neuroimmune mechanisms of depression. Nature neuroscience 18, 1386–1393. [DOI] [PMC free article] [PubMed] [Google Scholar]
  130. Hoffman JF, Wright CL, and McCarthy MM (2016). A Critical Period in Purkinje Cell Development Is Mediated by Local Estradiol Synthesis, Disrupted by Inflammation, and Has Enduring Consequences Only for Males. J Neurosci 36, 10039–10049. [DOI] [PMC free article] [PubMed] [Google Scholar]
  131. Huang HH, and Meites J (1975). Reproductive capacity of aging female rats. Neuroendocrinology 17, 289–295. [DOI] [PubMed] [Google Scholar]
  132. Hui CW, St-Pierre A, El Hajj H, Remy Y, Hebert SS, Luheshi GN, Srivastava LK, and Tremblay ME (2018). Prenatal Immune Challenge in Mice Leads to Partly Sex-Dependent Behavioral, Microglial, and Molecular Abnormalities Associated with Schizophrenia. Front Mol Neurosci 11, 13. [DOI] [PMC free article] [PubMed] [Google Scholar]
  133. Imai-Matsumura K, Matsumura K, Terao A, and Watanabe Y (2002). Attenuated fever in pregnant rats is associated with blunted syntheses of brain cyclooxygenase-2 and PGE2. Am J Physiol Regul Integr Comp Physiol. 283, R1346–R1353. [DOI] [PubMed] [Google Scholar]
  134. Inouye SK, Westendorp RG, and Saczynski JS (2014). Delirium in elderly people. Lancet 383, 911–922. [DOI] [PMC free article] [PubMed] [Google Scholar]
  135. Ishihara Y, Itoh K, Ishida A, and Yamazaki T (2015). Selective estrogen-receptor modulators suppress microglial activation and neuronal cell death via an estrogen receptor-dependent pathway. The Journal of steroid biochemistry and molecular biology 145, 85–93. [DOI] [PubMed] [Google Scholar]
  136. Jacquemont S, Coe BP, Hersch M, Duyzend MH, Krumm N, Bergmann S, Beckmann JS, Rosenfeld JA, and Eichler EE (2014). A higher mutational burden in females supports a “female protective model” in neurodevelopmental disorders. Am J Hum Genet 94, 415–425. [DOI] [PMC free article] [PubMed] [Google Scholar]
  137. Jenkins V, Shilling V, Deutsch G, Bloomfield D, Morris R, Allan S, Bishop H, Hodson N, Mitra S, Sadler G, et al. (2006). A 3-year prospective study of the effects of adjuvant treatments on cognition in women with early stage breast cancer. Br J Cancer 94, 828–834. [DOI] [PMC free article] [PubMed] [Google Scholar]
  138. Jenkins V, Shilling V, Fallowfield L, Howell A, and Hutton S (2004). Does hormone therapy for the treatment of breast cancer have a detrimental effect on memory and cognition? A pilot study. Psychooncology 13, 61–66. [DOI] [PubMed] [Google Scholar]
  139. Jenkins VA, Ambroisine LM, Atkins L, Cuzick J, Howell A, and Fallowfield LJ (2008). Effects of anastrozole on cognitive performance in postmenopausal women: a randomised, double-blind chemoprevention trial (IBIS II). The lancet oncology 9, 953–961. [DOI] [PubMed] [Google Scholar]
  140. Johann S, and Beyer C (2013). Neuroprotection by gonadal steroid hormones in acute brain damage requires cooperation with astroglia and microglia. The Journal of steroid biochemistry and molecular biology 137, 71–81. [DOI] [PubMed] [Google Scholar]
  141. Jones KA, Maltby S, Plank MW, Kluge M, Nilsson M, Foster PS, and Walker FR (2018). Peripheral immune cells infiltrate into sites of secondary neurodegeneration after ischemic stroke. Brain Behav Immun 67, 299–307. [DOI] [PubMed] [Google Scholar]
  142. Jost A, Vigier B, Prepin J, and Perchellet JP (1973). Studies on sex differentiation in mammals. Recent progress in hormone research 29, 1–41. [DOI] [PubMed] [Google Scholar]
  143. Karahancer M, Cirpan T, Kanit L, Terek MC, Dikmen Y, and Ozsener S (2008). The effects of raloxifen on depression and cognition in ovariectomized rats. Fertil Steril 89, 240–242. [DOI] [PubMed] [Google Scholar]
  144. Kentner AC, Bilbo SD, Brown AS, Hsiao EY, McAllister AK, Meyer U, Pearce BD, Pletnikov MV, Yolken RH, and Bauman MD (2018). Maternal immune activation: reporting guidelines to improve the rigor, reproducibility, and transparency of the model. Neuropsychopharmacology. [DOI] [PMC free article] [PubMed] [Google Scholar]
  145. Kinsley CH, and Lambert KG (2008). Reproduction-induced neuroplasticity: natural behavioural and neuronal alterations associated with the production and care of offspring. Journal of neuroendocrinology 20, 515–525. [DOI] [PubMed] [Google Scholar]
  146. Kipnis J (2016). Multifaceted interactions between adaptive immunity and the central nervous system. Science 353, 766–771. [DOI] [PMC free article] [PubMed] [Google Scholar]
  147. Kipp M, Karakaya S, Johann S, Kampmann E, Mey J, and Beyer C (2007). Oestrogen and Progesterone Reduce Lipopolysaccharide-Induced Expression of Tumour Necrosis Factor-alpha and Interleukin-18 in Midbrain Astrocytes. J Neuroendocrinol. 19, 819–822. [DOI] [PubMed] [Google Scholar]
  148. Kireev RA, Vara E, Vina J, and Tresguerres JA (2014). Melatonin and oestrogen treatments were able to improve neuroinflammation and apoptotic processes in dentate gyrus of old ovariectomized female rats. Age (Dordr) 36, 9707. [DOI] [PMC free article] [PubMed] [Google Scholar]
  149. Klein SL, and Flanagan KL (2016). Sex differences in immune responses. Nature reviews. Immunology 16, 626–638. [DOI] [PubMed] [Google Scholar]
  150. Klein SL, Jedlicka A, and Pekosz A (2010). The Xs and Y of immune responses to viral vaccines. The Lancet infectious diseases 10, 338–349. [DOI] [PMC free article] [PubMed] [Google Scholar]
  151. Kohler CA, Freitas TH, Stubbs B, Maes M, Solmi M, Veronese N, de Andrade NQ, Morris G, Fernandes BS, Brunoni AR, et al. (2018). Peripheral Alterations in Cytokine and Chemokine Levels After Antidepressant Drug Treatment for Major Depressive Disorder: Systematic Review and Meta-Analysis. Molecular neurobiology 55, 4195–4206. [DOI] [PubMed] [Google Scholar]
  152. Konkle AT, and McCarthy MM (2011). Developmental time course of estradiol, testosterone, and dihydrotestosterone levels in discrete regions of male and female rat brain. Endocrinology 152, 223–235. [DOI] [PMC free article] [PubMed] [Google Scholar]
  153. Korol DL, Malin EL, Borden KA, Busby RA, and Couper-Leo J (2004). Shifts in preferred learning strategy across the estrous cycle in female rats. Hormones and behavior 45, 330–338. [DOI] [PubMed] [Google Scholar]
  154. Kotekar N, Kuruvilla CS, and Murthy V (2014). Post-operative cognitive dysfunction in the elderly: A prospective clinical study. Indian J Anaesth 58, 263–268. [DOI] [PMC free article] [PubMed] [Google Scholar]
  155. Krebber AM, Buffart LM, Kleijn G, Riepma IC, de Bree R, Leemans CR, Becker A, Brug J, van Straten A, Cuijpers P, et al. (2014). Prevalence of depression in cancer patients: a meta-analysis of diagnostic interviews and self-report instruments. Psychooncology 23, 121–130. [DOI] [PMC free article] [PubMed] [Google Scholar]
  156. Krishnan S, Intlekofer KA, Aggison LK, and Petersen SL (2009). Central role of TRAF-interacting protein in a new model of brain sexual differentiation. Proceedings of the National Academy of Sciences of the United States of America 106, 16692–16697. [DOI] [PMC free article] [PubMed] [Google Scholar]
  157. Krukowski K, Grue K, Frias ES, Pietrykowski J, Jones T, Nelson G, and Rosi S (2018). Female mice are protected from space radiation-induced maladaptive responses. Brain Behav Immun 74, 106–120. [DOI] [PMC free article] [PubMed] [Google Scholar]
  158. Lamkin DM, Lutgendorf SK, Lubaroff D, Sood AK, Beltz TG, and Johnson AK (2011). Cancer induces inflammation and depressive-like behavior in the mouse: modulation by social housing. Brain, Behavior, and Immunity 25, 555–564. [DOI] [PMC free article] [PubMed] [Google Scholar]
  159. Landgraf R, Malkinson TJ, Veale WL, Lederis K, and Pittman QJ (1990). Vasopressin and oxytocin in rat brain in response to prostaglandin fever. The American journal of physiology 259, R1056–1062. [DOI] [PubMed] [Google Scholar]
  160. Landgraf R, Neumann I, Holsboer F, and Pittman QJ (1995). Interleukin-1 beta stimulates both central and peripheral release of vasopressin and oxytocin in the rat. Eur J Neurosci. 7, 592–598. [DOI] [PubMed] [Google Scholar]
  161. Landgraf R, Neumann I, Russell JA, and Pittman QJ (1992). Push-pull perfusion and microdialysis studies of central oxytocin and vasopressin release in freely moving rats during pregnancy, parturition, and lactation. Annals of the New York Academy of Sciences 652, 326–339. [DOI] [PubMed] [Google Scholar]
  162. LaPrairie JL, and Murphy AZ (2007). Female rats are more vulnerable to the long-term consequences of neonatal inflammatory injury. Pain 132 Suppl 1, S124–133. [DOI] [PMC free article] [PubMed] [Google Scholar]
  163. Launer LJ, Andersen K, Dewey ME, Letenneur L, Ott A, Amaducci LA, Brayne C, Copeland JR, Dartigues JF, Kragh-Sorensen P, et al. (1999). Rates and risk factors for dementia and Alzheimer’s disease: results from EURODEM pooled analyses. EURODEM Incidence Research Group and Work Groups. European Studies of Dementia. Neurology 52, 78–84. [DOI] [PubMed] [Google Scholar]
  164. Lee DC, Im JA, Kim JH, Lee HR, and Shim JY (2005). Effect of long-term hormone therapy on telomere length in postmenopausal women. Yonsei Med J 46, 471–479. [DOI] [PMC free article] [PubMed] [Google Scholar]
  165. Lei DL, Long JM, Hengemihle J, O’Neill J, Manaye KF, Ingram DK, and Mouton PR (2003). Effects of estrogen and raloxifene on neuroglia number and morphology in the hippocampus of aged female mice. Neuroscience 121, 659–666. [DOI] [PubMed] [Google Scholar]
  166. Lenz KM, Nugent BM, Haliyur R, and McCarthy MM (2013). Microglia are essential to masculinization of brain and behavior. J Neurosci 33, 2761–2772. [DOI] [PMC free article] [PubMed] [Google Scholar]
  167. Lenz KM, Nugent BM, and McCarthy MM (2012). Sexual differentiation of the rodent brain: dogma and beyond. Frontiers in neuroscience 6, 26. [DOI] [PMC free article] [PubMed] [Google Scholar]
  168. Lenz KM, Pickett LA, Wright CL, Davis KT, Joshi A, and McCarthy MM (2018). Mast Cells in the Developing Brain Determine Adult Sexual Behavior. J Neurosci 38, 8044–8059. [DOI] [PMC free article] [PubMed] [Google Scholar]
  169. Leuner B, and Sabihi S (2016). The birth of new neurons in the maternal brain: Hormonal regulation and functional implications. Frontiers in neuroendocrinology 41, 99–113. [DOI] [PMC free article] [PubMed] [Google Scholar]
  170. Lin KA, Choudhury KR, Rathakrishnan BG, Marks DM, Petrella JR, Doraiswamy PM, and Alzheimer’s Disease Neuroimaging I (2015). Marked gender differences in progression of mild cognitive impairment over 8 years. Alzheimers Dement (N Y) 1, 103–110. [DOI] [PMC free article] [PubMed] [Google Scholar]
  171. Lin YL, Lin SY, and Wang S (2012). Prenatal lipopolysaccharide exposure increases anxiety-like behaviors and enhances stress-induced corticosterone responses in adult rats. Brain Behav Immun 26, 459–468. [DOI] [PubMed] [Google Scholar]
  172. Liu JL, Tian DS, Li ZW, Qu WS, Zhan Y, Xie MJ, Yu ZY, Wang W, and Wu G (2010). Tamoxifen alleviates irradiation-induced brain injury by attenuating microglial inflammatory response in vitro and in vivo. Brain research 1316, 101–111. [DOI] [PubMed] [Google Scholar]
  173. Liu KA, and Mager NA (2016). Women’s involvement in clinical trials: historical perspective and future implications. Pharm Pract (Granada) 14, 708. [DOI] [PMC free article] [PubMed] [Google Scholar]
  174. Lorenz TK, Demas GE, and Heiman JR (2017). Partnered sexual activity moderates menstrual cycle-related changes in inflammation markers in healthy women: an exploratory observational study. Fertil Steril 107, 763–773 e763. [DOI] [PMC free article] [PubMed] [Google Scholar]
  175. Lowe GC, Luheshi GN, and Williams S (2008). Maternal infection and fever during late gestation are associated with altered synaptic transmission in the hippocampus of juvenile offspring rats. American journal of physiology 295, R1563–1571. [DOI] [PubMed] [Google Scholar]
  176. Maccari S, Krugers HJ, Morley-Fletcher S, Szyf M, and Brunton PJ (2014). The consequences of early-life adversity: neurobiological, behavioural and epigenetic adaptations. Journal of neuroendocrinology 26, 707–723. [DOI] [PubMed] [Google Scholar]
  177. Makinson R, Lloyd K, Rayasam A, McKee S, Brown A, Barila G, Grissom N, George R, Marini M, Fabry Z, et al. (2017). Intrauterine inflammation induces sex-specific effects on neuroinflammation, white matter, and behavior. Brain Behav Immun 66, 277–288. [DOI] [PMC free article] [PubMed] [Google Scholar]
  178. Marin-Teva JL, Dusart I, Colin C, Gervais A, van Rooijen N, and Mallat M (2004). Microglia promote the death of developing Purkinje cells. Neuron 41, 535–547. [DOI] [PubMed] [Google Scholar]
  179. Marriott LK, Hauss-Wegrzyniak B, Benton RS, Vraniak PD, and Wenk GL (2002). Long-term estrogen therapy worsens the behavioral and neuropathological consequences of chronic brain inflammation. Behavioral neuroscience 116, 902–911. [DOI] [PubMed] [Google Scholar]
  180. Martin SM, Malkinson TJ, Veale WL, and Pittman QJ (1995). Fever in pregnant, parturient, and lactating rats. The American journal of physiology 268, R919–923. [DOI] [PubMed] [Google Scholar]
  181. Matsunaga W, Miyata S, Takamata A, Bun H, Nakashima T, and Kiyohara T (2000). LPS-induced Fos expression in oxytocin and vasopressin neurons of the rat hypothalamus. Brain research 858, 9–18. [DOI] [PubMed] [Google Scholar]
  182. McCarthy MM (2008). Estradiol and the developing brain. Physiological reviews 88, 91–124. [DOI] [PMC free article] [PubMed] [Google Scholar]
  183. McCarthy MM, and Arnold AP (2011). Reframing sexual differentiation of the brain. Nature neuroscience 14, 677–683. [DOI] [PMC free article] [PubMed] [Google Scholar]
  184. McCarthy MM, Nugent BM, and Lenz KM (2017). Neuroimmunology and neuroepigenetics in the establishment of sex differences in the brain. Nat Rev Neurosci 18, 471–484. [DOI] [PMC free article] [PubMed] [Google Scholar]
  185. McEwen BS, and Alves SE (1999). Estrogen actions in the central nervous system. Endocrine reviews 20, 279–307. [DOI] [PubMed] [Google Scholar]
  186. McGinnis GJ, Friedman D, Young KH, Torres ER, Thomas CR, Gough MJ, and Raber J (2017). Neuroinflammatory and cognitive consequences of combined radiation and immunotherapy in a novel preclinical model. Oncotarget 8, 9155–9173. [DOI] [PMC free article] [PubMed] [Google Scholar]
  187. McKenzie JA, Spielman LJ, Pointer CB, Lowry JR, Bajwa E, Lee CW, and Klegeris A (2017). Neuroinflammation as a Common Mechanism Associated with the Modifiable Risk Factors for Alzheimer’s and Parkinson’s Diseases. Curr Aging Sci 10, 158–176. [DOI] [PubMed] [Google Scholar]
  188. Meddle SL, Leng G, Selvarajah JR, Bicknell RJ, and Russell JA (2000). Direct pathways to the supraoptic nucleus from the brainstem and the main olfactory bulb are activated at parturition in the rat. Neuroscience 101, 1013–1021. [DOI] [PubMed] [Google Scholar]
  189. MedlinePlus (2011). Menopause.
  190. Mehta RR, and Graves JM (1992). Breast tumor-derived factors stimulate reduction of estrone to estradiol in nonmalignant breast tissue. Breast Cancer Res Treat 23, 51–56. [DOI] [PubMed] [Google Scholar]
  191. Meites J, and Lu J (1994). Reproductive aging and neuroendocrine function In Oxford review of reproductive biology. Charlton H, ed. (New York: Oxford Press; ), pp. 215–239. [Google Scholar]
  192. Melik Parsadaniantz S, Gaillet S, Malaval F, Lenoir V, Batsche E, Barbanel G, Gardier A, Terlain B, Jacquot C, Szafarczyk A, et al. (1995). Lesions of the afferent catecholaminergic pathways inhibit the temporal activation of the CRH and POMC gene expression and ACTH release induced by human interleukin-1beta in the male ra. Neuroendocrinology 62, 586–595. [DOI] [PubMed] [Google Scholar]
  193. Meng FT, Ni RJ, Zhang Z, Zhao J, Liu YJ, and Zhou JN (2011). Inhibition of oestrogen biosynthesis induces mild anxiety in C57BL/6J ovariectomized female mice. Neurosci Bull 27, 241–250. [DOI] [PMC free article] [PubMed] [Google Scholar]
  194. Meyer U (2014). Prenatal poly(i:C) exposure and other developmental immune activation models in rodent systems. Biological psychiatry 75, 307–315. [DOI] [PubMed] [Google Scholar]
  195. Meyer U, Nyffeler M, Yee BK, Knuesel I, and Feldon J (2008). Adult brain and behavioral pathological markers of prenatal immune challenge during early/middle and late fetal development in mice. Brain Behav Immun 22, 469–486. [DOI] [PubMed] [Google Scholar]
  196. Missig G, Robbins JO, Mokler EL, McCullough KM, Bilbo SD, McDougle CJ, and Carlezon WA Jr. (2019). Sex-dependent neurobiological features of prenatal immune activation via TLR7. Mol Psychiatry. [DOI] [PMC free article] [PubMed] [Google Scholar]
  197. Miyamoto A, Wake H, Ishikawa AW, Eto K, Shibata K, Murakoshi H, Koizumi S, Moorhouse AJ, Yoshimura Y, and Nabekura J (2016). Microglia contact induces synapse formation in developing somatosensory cortex. Nat Commun 7, 12540. [DOI] [PMC free article] [PubMed] [Google Scholar]
  198. Moller JT, Cluitmans P, Rasmussen LS, Houx P, Rasmussen H, Canet J, Rabbitt P, Jolles J, Larsen K, Hanning CD, et al. (1998). Long-term postoperative cognitive dysfunction in the elderly ISPOCD1 study. ISPOCD investigators. International Study of Post-Operative Cognitive Dysfunction. Lancet 351, 857–861. [DOI] [PubMed] [Google Scholar]
  199. Mor G, and Cardenas I (2010). The immune system in pregnancy: a unique complexity. American journal of reproductive immunology (New York, N.Y.: 1989) 63, 425–433. [DOI] [PMC free article] [PubMed] [Google Scholar]
  200. Mouihate A, Chen X, and Pittman QJ (1998). Interleukin-1beta fever in rats: gender difference and estrous cycle influence. Am.J.Physiol 275, R1450–R1454. [DOI] [PubMed] [Google Scholar]
  201. Mouihate A, Clerget-Froidevaux MS, Nakamura K, Negishi M, Wallace JL, and Pittman QJ (2002). Suppression of fever at near term is associated with reduced COX-2 protein expression in rat hypothalamus. Am.J.Physiol Regul.Integr.Comp Physiol 283, R800–R805. [DOI] [PubMed] [Google Scholar]
  202. Mouihate A, Ellis S, Harre EM, and Pittman QJ (2005). Fever suppression in near term pregnant rats is dissociated from the LPS-activated signaling pathways. Am.J.Physiol Regul.Integr.Comp Physiol. [DOI] [PubMed] [Google Scholar]
  203. Mouihate A, and Pittman QJ (2003). Neuroimmune response to endogenous and exogenous pyrogens is differently modulated by sex steroids. Endocrinology 144, 2454–2460. [DOI] [PubMed] [Google Scholar]
  204. Mozaffarian D, Benjamin EJ, Go AS, Arnett DK, Blaha MJ, Cushman M, de Ferranti S, Despres JP, Fullerton HJ, Howard VJ, et al. (2015). Heart disease and stroke statistics−-2015 update: a report from the American Heart Association. Circulation 131, e29–322. [DOI] [PubMed] [Google Scholar]
  205. Mueller BR, and Bale TL (2008). Sex-specific programming of offspring emotionality after stress early in pregnancy. J Neurosci 28, 9055–9065. [DOI] [PMC free article] [PubMed] [Google Scholar]
  206. Mulak A, Tache Y, and Larauche M (2014). Sex hormones in the modulation of irritable bowel syndrome. World J Gastroenterol 20, 2433–2448. [DOI] [PMC free article] [PubMed] [Google Scholar]
  207. Nakamura H, Seto T, Hatta K, Matsuzaki I, Nagase H, Yoshida M, and Ogino K (1998). Central administration of interleukin-1 beta reduces natural killer cell activity in non-pregnant rats, but not in pregnant rats. Psychoneuroendocrinology 23, 651–659. [DOI] [PubMed] [Google Scholar]
  208. Nelson LH, and Lenz KM (2017a). The immune system as a novel regulator of sex differences in brain and behavioral development. J Neurosci Res 95, 447–461. [DOI] [PMC free article] [PubMed] [Google Scholar]
  209. Nelson LH, and Lenz KM (2017b). Microglia depletion in early life programs persistent changes in social, mood-related, and locomotor behavior in male and female rats. Behavioural brain research 316, 279–293. [DOI] [PMC free article] [PubMed] [Google Scholar]
  210. Nelson LH, Warden S, and Lenz KM (2017). Sex differences in microglial phagocytosis in the neonatal hippocampus. Brain Behav Immun 64, 11–22. [DOI] [PMC free article] [PubMed] [Google Scholar]
  211. Neumann I, Russell JA, and Landgraf R (1993). Oxytocin and vasopressin release within the supraoptic and paraventricular nuclei of pregnant, parturient and lactating rats: a microdialysis study. Neuroscience 53, 65–75. [DOI] [PubMed] [Google Scholar]
  212. Nguyen JC, Killcross AS, and Jenkins TA (2014). Obesity and cognitive decline: role of inflammation and vascular changes. Frontiers in neuroscience 8, 375. [DOI] [PMC free article] [PubMed] [Google Scholar]
  213. Nimmerjahn A, Kirchhoff F, and Helmchen F (2005). Resting microglial cells are highly dynamic surveillants of brain parenchyma in vivo. Science 308, 1314–1318. [DOI] [PubMed] [Google Scholar]
  214. Niraula A, Sheridan JF, and Godbout JP (2017). Microglia Priming with Aging and Stress. Neuropsychopharmacology 42, 318–333. [DOI] [PMC free article] [PubMed] [Google Scholar]
  215. Nugent BM, and Bale TL (2015). The omniscient placenta: Metabolic and epigenetic regulation of fetal programming. Frontiers in neuroendocrinology 39, 28–37. [DOI] [PMC free article] [PubMed] [Google Scholar]
  216. Nugent BM, O’Donnell CM, Epperson CN, and Bale TL (2018). Placental H3K27me3 establishes female resilience to prenatal insults. Nat Commun 9, 2555. [DOI] [PMC free article] [PubMed] [Google Scholar]
  217. Nugent BM, Wright CL, Shetty AC, Hodes GE, Lenz KM, Mahurkar A, Russo SJ, Devine SE, and McCarthy MM (2015). Brain feminization requires active repression of masculinization via DNA methylation. Nature neuroscience 18, 690–697. [DOI] [PMC free article] [PubMed] [Google Scholar]
  218. Orzack SH, Stubblefield JW, Akmaev VR, Colls P, Munne S, Scholl T, Steinsaltz D, and Zuckerman JE (2015). The human sex ratio from conception to birth. Proceedings of the National Academy of Sciences of the United States of America 112, E2102–2111. [DOI] [PMC free article] [PubMed] [Google Scholar]
  219. Paolicelli RC, Bolasco G, Pagani F, Maggi L, Scianni M, Panzanelli P, Giustetto M, Ferreira TA, Guiducci E, Dumas L, et al. (2011). Synaptic pruning by microglia is necessary for normal brain development. Science 333, 1456–1458. [DOI] [PubMed] [Google Scholar]
  220. Paris JJ, Brunton PJ, Russell JA, and Frye CA (2011a). Immune stress in late pregnant rats decreases length of gestation and fecundity, and alters later cognitive and affective behaviour of surviving pre-adolescent offspring. Stress (Amsterdam, Netherlands) 14, 652–664. [DOI] [PMC free article] [PubMed] [Google Scholar]
  221. Paris JJ, Brunton PJ, Russell JA, Walf AA, and Frye CA (2011b). Inhibition of 5alpha-reductase activity in late pregnancy decreases gestational length and fecundity and impairs object memory and central progestogen milieu of juvenile rat offspring. Journal of neuroendocrinology 23, 1079–1090. [DOI] [PMC free article] [PubMed] [Google Scholar]
  222. Paris JJ, and Frye CA (2008). Estrous cycle, pregnancy, and parity enhance performance of rats in object recognition or object placement tasks. Reproduction 136, 105–115. [DOI] [PMC free article] [PubMed] [Google Scholar]
  223. Patterson PH (2009). Immune involvement in schizophrenia and autism: etiology, pathology and animal models. Behavioural brain research 204, 313–321. [DOI] [PubMed] [Google Scholar]
  224. Paul SM, and Purdy RH (1992). Neuroactive steroids. FASEB Journal 6, 2311–2322. [PubMed] [Google Scholar]
  225. Perez EC, Rodgers SP, Inoue T, Pedersen SE, Leasure JL, and Gaber MW (2018). Olfactory Memory Impairment Differs by Sex in a Rodent Model of Pediatric Radiotherapy. Front Behav Neurosci 12, 158. [DOI] [PMC free article] [PubMed] [Google Scholar]
  226. Phillips Bute B, Mathew J, Blumenthal JA, Welsh-Bohmer K, White WD, Mark D, Landolfo K, and Newman MF (2003). Female gender is associated with impaired quality of life 1 year after coronary artery bypass surgery. Psychosomatic medicine 65, 944–951. [DOI] [PubMed] [Google Scholar]
  227. Phillips KA, Ribi K, Sun Z, Stephens A, Thompson A, Harvey V, Thurlimann B, Cardoso F, Pagani O, Coates AS, et al. (2010). Cognitive function in postmenopausal women receiving adjuvant letrozole or tamoxifen for breast cancer in the BIG 1–98 randomized trial. Breast 19, 388–395. [DOI] [PMC free article] [PubMed] [Google Scholar]
  228. Pitman A, Suleman S, Hyde N, and Hodgkiss A (2018). Depression and anxiety in patients with cancer. Bmj 361, k1415. [DOI] [PubMed] [Google Scholar]
  229. Plata-Salaman CR, Peloso E, and Satinoff E (1998). Interleukin-1beta-induced fever in young and old Long-Evans rats. The American journal of physiology 275, R1633–1638. [DOI] [PubMed] [Google Scholar]
  230. Poulin P, and Pittman QJ (1993). Possible involvement of brain oxytocin in modulating vasopressin antipyretic action. Am J Physiol. 265, t-6. [DOI] [PubMed] [Google Scholar]
  231. Prendergast BJ, Onishi KG, and Zucker I (2014). Female mice liberated for inclusion in neuroscience and biomedical research. Neuroscience and biobehavioral reviews 40, 1–5. [DOI] [PubMed] [Google Scholar]
  232. Pyter LM, Pineros V, Galang JA, McClintock MK, and Prendergast BJ (2009). Peripheral tumors induce depressive-like behaviors and cytokine production and alter hypothalamic-pituitary-adrenal axis regulation. Proceedings of the National Academy of Sciences of the United States of America 106, 9069–9074. [DOI] [PMC free article] [PubMed] [Google Scholar]
  233. Rajendran L, and Paolicelli RC (2018). Microglia-Mediated Synapse Loss in Alzheimer’s Disease. J Neurosci 38, 2911–2919. [DOI] [PMC free article] [PubMed] [Google Scholar]
  234. Ramanan S, Kooshki M, Zhao W, Hsu FC, and Robbins ME (2008). PPARalpha ligands inhibit radiation-induced microglial inflammatory responses by negatively regulating NF-kappaB and AP-1 pathways. Free Radic Biol Med 45, 1695–1704. [DOI] [PMC free article] [PubMed] [Google Scholar]
  235. Ransohoff RM (2016). How neuroinflammation contributes to neurodegeneration. Science 353, 777–783. [DOI] [PubMed] [Google Scholar]
  236. Ratnayake U, Quinn T, Walker DW, and Dickinson H (2013). Cytokines and the neurodevelopmental basis of mental illness. Frontiers in neuroscience 7, 180. [DOI] [PMC free article] [PubMed] [Google Scholar]
  237. Reeve A, Simcox E, and Turnbull D (2014). Ageing and Parkinson’s disease: why is advancing age the biggest risk factor? Ageing research reviews 14, 19–30. [DOI] [PMC free article] [PubMed] [Google Scholar]
  238. Remus JL, and Dantzer R (2016). Inflammation Models of Depression in Rodents: Relevance to Psychotropic Drug Discovery. Int J Neuropsychopharmacol 19. [DOI] [PMC free article] [PubMed] [Google Scholar]
  239. Reus GZ, Fries GR, Stertz L, Badawy M, Passos IC, Barichello T, Kapczinski F, and Quevedo J (2015). The role of inflammation and microglial activation in the pathophysiology of psychiatric disorders. Neuroscience 300, 141–154. [DOI] [PubMed] [Google Scholar]
  240. Reyno LM, Levine MN, Skingley P, Arnold A, and Abu Zahra H (1992). Chemotherapy induced amenorrhoea in a randomised trial of adjuvant chemotherapy duration in breast cancer. European journal of cancer 29A, 21–23. [DOI] [PubMed] [Google Scholar]
  241. Rilett KC, Friedel M, Ellegood J, MacKenzie RN, Lerch JP, and Foster JA (2015). Loss of T cells influences sex differences in behavior and brain structure. Brain Behav Immun 46, 249–260. [DOI] [PubMed] [Google Scholar]
  242. Rivest S, Lacroix S, Vallieres L, Nadeau S, Zhang J, and Laflamme N (2000). How the blood talks to the brain parenchyma and the paraventricular nucleus of the hypothalamus during systemic inflammatory and infectious stimuli. Experimental Biology & Medicine 223, 22–38. [DOI] [PubMed] [Google Scholar]
  243. Robinson EB, Lichtenstein P, Anckarsater H, Happe F, and Ronald A (2013). Examining and interpreting the female protective effect against autistic behavior. Proceedings of the National Academy of Sciences of the United States of America 110, 5258–5262. [DOI] [PMC free article] [PubMed] [Google Scholar]
  244. Rocca WA, Grossardt BR, and Shuster LT (2011). Oophorectomy, menopause, estrogen treatment, and cognitive aging: clinical evidence for a window of opportunity. Brain research 1379, 188–198. [DOI] [PMC free article] [PubMed] [Google Scholar]
  245. Rollins CPE, Gallino D, Kong V, Ayranci G, Devenyi GA, Germann J, and Chakravarty MM (2018). Contributions of a high-fat diet to Alzheimer’s disease-related decline: A longitudinal behavioural and structural neuroimaging study in mouse models. NeuroImage. Clinical. [DOI] [PMC free article] [PubMed] [Google Scholar]
  246. Rosen S, Ham B, and Mogil JS (2017). Sex differences in neuroimmunity and pain. J Neurosci Res 95, 500–508. [DOI] [PubMed] [Google Scholar]
  247. Rossouw JE, Anderson GL, Prentice RL, LaCroix AZ, Kooperberg C, Stefanick ML, Jackson RD, Beresford SA, Howard BV, Johnson KC, et al. (2002). Risks and benefits of estrogen plus progestin in healthy postmenopausal women: principal results From the Women’s Health Initiative randomized controlled trial. JAMA 288, 321–333. [DOI] [PubMed] [Google Scholar]
  248. Roy JA, Sawka CA, and Pritchard KI (1996). Hormone replacement therapy in women with breast cancer. Do the risks outweigh the benefits? J Clin Oncol 14, 997–1006. [DOI] [PubMed] [Google Scholar]
  249. Rugo HS, Rumble RB, Macrae E, Barton DL, Connolly HK, Dickler MN, Fallowfield L, Fowble B, Ingle JN, Jahanzeb M, et al. (2016). Endocrine Therapy for Hormone Receptor-Positive Metastatic Breast Cancer: American Society of Clinical Oncology Guideline. J Clin Oncol 34, 3069–3103. [DOI] [PubMed] [Google Scholar]
  250. Rummel C (2018). Perhaps woman are better astronauts? Brain Behav Immun 74, 47–48. [DOI] [PubMed] [Google Scholar]
  251. Santagati S, Melcangi RC, Celotti F, Martini L, and Maggi A (1994). Estrogen receptor is expressed in different types of glial cells in culture. Journal of neurochemistry 63, 2058–2064. [DOI] [PubMed] [Google Scholar]
  252. Santos JC, and Pyter LM (2018). Neuroimmunology of Behavioral Comorbidities Associated With Cancer and Cancer Treatments. Front Immunol 9, 1195. [DOI] [PMC free article] [PubMed] [Google Scholar]
  253. Sapolsky R, Rivier C, Yamamoto G, Plotsky P, and Vale W (1987). Interleukin-1 stimulates the secretion of hypothalamic corticotropin-releasing factor. Science 238, 522–524. [DOI] [PubMed] [Google Scholar]
  254. Sarvari M, Hrabovszky E, Kallo I, Solymosi N, Liko I, Berchtold N, Cotman C, and Liposits Z (2012). Menopause leads to elevated expression of macrophage-associated genes in the aging frontal cortex: rat and human studies identify strikingly similar changes. Journal of neuroinflammation 9, 264. [DOI] [PMC free article] [PubMed] [Google Scholar]
  255. Sarvari M, Kallo I, Hrabovszky E, Solymosi N, and Liposits Z (2014). Ovariectomy and subsequent treatment with estrogen receptor agonists tune the innate immune system of the hippocampus in middle-aged female rats. PloS one 9, e88540. [DOI] [PMC free article] [PubMed] [Google Scholar]
  256. Sarvari M, Kallo I, Hrabovszky E, Solymosi N, and Liposits Z (2017). Ovariectomy Alters Gene Expression of the Hippocampal Formation in Middle-Aged Rats. Endocrinology 158, 69–83. [DOI] [PubMed] [Google Scholar]
  257. Sarvari M, Kallo I, Hrabovszky E, Solymosi N, Rodolosse A, and Liposits Z (2016). Long-Term Estrogen Receptor Beta Agonist Treatment Modifies the Hippocampal Transcriptome in Middle-Aged Ovariectomized Rats. Frontiers in cellular neuroscience 10, 149. [DOI] [PMC free article] [PubMed] [Google Scholar]
  258. Satinoff E, Peloso E, and Plata-Salamn CR (1999). Prostaglandin E2-induced fever in young and old Long-Evans rats. Physiology & behavior 67, 149–152. [DOI] [PubMed] [Google Scholar]
  259. Sayakhot P, Vincent A, Deeks A, and Teede H (2011). Potential adverse impact of ovariectomy on physical and psychological function of younger women with breast cancer. Menopause 18, 786–793. [DOI] [PubMed] [Google Scholar]
  260. Schafer DP, Lehrman EK, Kautzman AG, Koyama R, Mardinly AR, Yamasaki R, Ransohoff RM, Greenberg ME, Barres BA, and Stevens B (2012). Microglia sculpt postnatal neural circuits in an activity and complement-dependent manner. Neuron 74, 691–705. [DOI] [PMC free article] [PubMed] [Google Scholar]
  261. Schagen SB, Muller MJ, Boogerd W, Mellenbergh GJ, and van Dam FS (2006). Change in cognitive function after chemotherapy: a prospective longitudinal study in breast cancer patients. J Natl Cancer Inst 98, 1742–1745. [DOI] [PubMed] [Google Scholar]
  262. Schagen SB, van Dam FS, Muller MJ, Boogerd W, Lindeboom J, and Bruning PF (1999). Cognitive deficits after postoperative adjuvant chemotherapy for breast carcinoma. Cancer 85, 640–650. [DOI] [PubMed] [Google Scholar]
  263. Schilder CM, Seynaeve C, Beex LV, Boogerd W, Linn SC, Gundy CM, Huizenga HM, Nortier JW, van de Velde CJ, van Dam FS, et al. (2010). Effects of tamoxifen and exemestane on cognitive functioning of postmenopausal patients with breast cancer: results from the neuropsychological side study of the tamoxifen and exemestane adjuvant multinational trial. J Clin Oncol 28, 1294–1300. [DOI] [PubMed] [Google Scholar]
  264. Schrepf A, Lutgendorf SK, and Pyter LM (2015). Pre-treatment effects of peripheral tumors on brain and behavior: neuroinflammatory mechanisms in humans and rodents. Brain Behav Immun 49, 1–17. [DOI] [PMC free article] [PubMed] [Google Scholar]
  265. Schultz PN, Klein MJ, Beck ML, Stava C, and Sellin RV (2005). Breast cancer: relationship between menopausal symptoms, physiologic health effects of cancer treatment and physical constraints on quality of life in long-term survivors. Journal of clinical nursing 14, 204–211. [DOI] [PubMed] [Google Scholar]
  266. Schwarz JM, Sholar PW, and Bilbo SD (2012). Sex differences in microglial colonization of the developing rat brain. Journal of neurochemistry 120, 948–963. [DOI] [PMC free article] [PubMed] [Google Scholar]
  267. Seale JV, Wood SA, Atkinson HC, Bate E, Lightman SL, Ingram CD, Jessop DS, and Harbuz MS (2004). Gonadectomy reverses the sexually diergic patterns of circadian and stress-induced hypothalamic-pituitary-adrenal axis activity in male and female rats. J.Neuroendocrinol. 16, 516–524. [DOI] [PubMed] [Google Scholar]
  268. Seliktar N, Polek C, Brooks A, and Hardie T (2015). Cognition in breast cancer survivors: hormones versus depression. Psychooncology 24, 402–407. [DOI] [PubMed] [Google Scholar]
  269. Selvaraj UM, Zuurbier KR, Whoolery CW, Plautz EJ, Chambliss KL, Kong X, Zhang S, Kim SH, Katzenellenbogen BS, Katzenellenbogen JA, et al. (2018). Selective Nonnuclear Estrogen Receptor Activation Decreases Stroke Severity and Promotes Functional Recovery in Female Mice. Endocrinology 159, 3848–3859. [DOI] [PMC free article] [PubMed] [Google Scholar]
  270. Semaan SJ, and Kauffman AS (2010). Sexual differentiation and development of forebrain reproductive circuits. Current opinion in neurobiology 20, 424–431. [DOI] [PMC free article] [PubMed] [Google Scholar]
  271. Shankar A, Dracham C, Ghoshal S, and Grover S (2016). Prevalence of depression and anxiety disorder in cancer patients: An institutional experience. Indian J Cancer 53, 432–434. [DOI] [PubMed] [Google Scholar]
  272. Sherer ML, Posillico CK, and Schwarz JM (2017). An examination of changes in maternal neuroimmune function during pregnancy and the postpartum period. Brain Behav Immun 66, 201–209. [DOI] [PMC free article] [PubMed] [Google Scholar]
  273. Shigemoto-Mogami Y, Hoshikawa K, Goldman JE, Sekino Y, and Sato K (2014). Microglia enhance neurogenesis and oligodendrogenesis in the early postnatal subventricular zone. J Neurosci 34, 2231–2243. [DOI] [PMC free article] [PubMed] [Google Scholar]
  274. Shumaker SA, Legault C, Kuller L, Rapp SR, Thal L, Lane DS, Fillit H, Stefanick ML, Hendrix SL, Lewis CE, et al. (2004). Conjugated equine estrogens and incidence of probable dementia and mild cognitive impairment in postmenopausal women: Women’s Health Initiative Memory Study. JAMA 291, 2947–2958. [DOI] [PubMed] [Google Scholar]
  275. Siani F, Greco R, Levandis G, Ghezzi C, Daviddi F, Demartini C, Vegeto E, Fuzzati-Armentero MT, and Blandini F (2017). Influence of Estrogen Modulation on Glia Activation in a Murine Model of Parkinson’s Disease. Frontiers in neuroscience 11, 306. [DOI] [PMC free article] [PubMed] [Google Scholar]
  276. Sierra A, Gottfried-Blackmore A, Milner TA, McEwen BS, and Bulloch K (2008). Steroid hormone receptor expression and function in microglia. Glia 56, 659–674. [DOI] [PubMed] [Google Scholar]
  277. Smith SE, Li J, Garbett K, Mirnics K, and Patterson PH (2007). Maternal immune activation alters fetal brain development through interleukin-6. J Neurosci 27, 10695–10702. [DOI] [PMC free article] [PubMed] [Google Scholar]
  278. Sominsky L, De Luca S, and Spencer SJ (2018). Microglia: Key players in neurodevelopment and neuronal plasticity. The international journal of biochemistry & cell biology 94, 56–60. [DOI] [PubMed] [Google Scholar]
  279. Sorge RE, Mapplebeck JC, Rosen S, Beggs S, Taves S, Alexander JK, Martin LJ, Austin JS, Sotocinal SG, Chen D, et al. (2015). Different immune cells mediate mechanical pain hypersensitivity in male and female mice. Nature neuroscience 18, 1081–1083. [DOI] [PMC free article] [PubMed] [Google Scholar]
  280. Spence RD, and Voskuhl RR (2012). Neuroprotective effects of estrogens and androgens in CNS inflammation and neurodegeneration. Frontiers in neuroendocrinology 33, 105–115. [DOI] [PMC free article] [PubMed] [Google Scholar]
  281. Spence RD, Wisdom AJ, Cao Y, Hill HM, Mongerson CR, Stapornkul B, Itoh N, Sofroniew MV, and Voskuhl RR (2013). Estrogen mediates neuroprotection and antiinflammatory effects during EAE through ERalpha signaling on astrocytes but not through ERbeta signaling on astrocytes or neurons. J Neurosci 33, 10924–10933. [DOI] [PMC free article] [PubMed] [Google Scholar]
  282. Spencer SJ, Basri B, Sominsky L, Soch A, Ayala MT, Reineck P, Gibson BC, and Barrientos RM (2018). High-fat diet worsens the impact of aging on microglial function and morphology in a region-specific manner. Neurobiology of aging 74, 121–134. [DOI] [PMC free article] [PubMed] [Google Scholar]
  283. Spencer SJ, D’Angelo H, Soch A, Watkins LR, Maier SF, and Barrientos RM (2017). High-fat diet and aging interact to produce neuroinflammation and impair hippocampal- and amygdalar-dependent memory. Neurobiology of aging 58, 88–101. [DOI] [PMC free article] [PubMed] [Google Scholar]
  284. Steiner J, Walter M, Gos T, Guillemin GJ, Bernstein HG, Sarnyai Z, Mawrin C, Brisch R, Bielau H, Meyer zu Schwabedissen L, et al. (2011). Severe depression is associated with increased microglial quinolinic acid in subregions of the anterior cingulate gyrus: evidence for an immune-modulated glutamatergic neurotransmission? Journal of neuroinflammation 8, 94. [DOI] [PMC free article] [PubMed] [Google Scholar]
  285. Stiles J, and Jernigan TL (2010). The basics of brain development. Neuropsychol Rev 20, 327–348. [DOI] [PMC free article] [PubMed] [Google Scholar]
  286. Stubbins RE, Najjar K, Holcomb VB, Hong J, and Nunez NP (2012). Oestrogen alters adipocyte biology and protects female mice from adipocyte inflammation and insulin resistance. Diabetes Obes Metab 14, 58–66. [DOI] [PMC free article] [PubMed] [Google Scholar]
  287. Sumida H, Nishizuka M, Kano Y, and Arai Y (1993). Sex differences in the anteroventral periventricular nucleus of the preoptic area and in the related effects of androgen in prenatal rats. Neuroscience letters 151, 41–44. [DOI] [PubMed] [Google Scholar]
  288. Sun Y, Lee HJ, Yang SC, Chen TF, Lin KN, Lin CC, Wang PN, Tang LY, and Chiu MJ (2014). A nationwide survey of mild cognitive impairment and dementia, including very mild dementia, in Taiwan. PloS one 9, e100303. [DOI] [PMC free article] [PubMed] [Google Scholar]
  289. Tanaka J, Fujita H, Matsuda S, Toku K, Sakanaka M, and Maeda N (1997). Glucocorticoid- and mineralocorticoid receptors in microglial cells: the two receptors mediate differential effects of corticosteroids. Glia 20, 23–37. [PubMed] [Google Scholar]
  290. Tanapat P, Hastings NB, Reeves AJ, and Gould E (1999). Estrogen stimulates a transient increase in the number of new neurons in the dentate gyrus of the adult female rat. J Neurosci 19, 5792–5801. [DOI] [PMC free article] [PubMed] [Google Scholar]
  291. Tapia-Gonzalez S, Carrero P, Pernia O, Garcia-Segura LM, and Diz-Chaves Y (2008). Selective oestrogen receptor (ER) modulators reduce microglia reactivity in vivo after peripheral inflammation: potential role of microglial ERs. The Journal of endocrinology 198, 219–230. [DOI] [PubMed] [Google Scholar]
  292. Taylor PV, Veenema AH, Paul MJ, Bredewold R, Isaacs S, and de Vries GJ (2012). Sexually dimorphic effects of a prenatal immune challenge on social play and vasopressin expression in juvenile rats. Biol Sex Differ 3, 15. [DOI] [PMC free article] [PubMed] [Google Scholar]
  293. Terasaki LS, and Schwarz JM (2016). Effects of Moderate Prenatal Alcohol Exposure during Early Gestation in Rats on Inflammation across the Maternal-Fetal-Immune Interface and Later-Life Immune Function in the Offspring. Journal of neuroimmune pharmacology : the official journal of the Society on NeuroImmune Pharmacology 11, 680–692. [DOI] [PMC free article] [PubMed] [Google Scholar]
  294. Thion MS, Low D, Silvin A, Chen J, Grisel P, Schulte-Schrepping J, Blecher R, Ulas T, Squarzoni P, Hoeffel G, et al. (2018). Microbiome Influences Prenatal and Adult Microglia in a Sex-Specific Manner. Cell 172, 500–516 e516. [DOI] [PMC free article] [PubMed] [Google Scholar]
  295. Toga AW, Thompson PM, and Sowell ER (2006). Mapping brain maturation. Trends in neurosciences 29, 148–159. [DOI] [PMC free article] [PubMed] [Google Scholar]
  296. Toonen JA, Solga AC, Ma Y, and Gutmann DH (2017). Estrogen activation of microglia underlies the sexually dimorphic differences in Nf1 optic glioma-induced retinal pathology. J Exp Med 214, 17–25. [DOI] [PMC free article] [PubMed] [Google Scholar]
  297. Torino F, Barnabei A, De Vecchis L, Appetecchia M, Strigari L, and Corsello SM (2012). Recognizing menopause in women with amenorrhea induced by cytotoxic chemotherapy for endocrine-responsive early breast cancer. Endocr Relat Cancer 19, R21–33. [DOI] [PubMed] [Google Scholar]
  298. Trowsdale J, and Betz AG (2006). Mother’s little helpers: mechanisms of maternal-fetal tolerance. Nat Immunol 7, 241–246. [DOI] [PubMed] [Google Scholar]
  299. Tucsek Z, Toth P, Sosnowska D, Gautam T, Mitschelen M, Koller A, Szalai G, Sonntag WE, Ungvari Z, and Csiszar A (2014). Obesity in aging exacerbates blood-brain barrier disruption, neuroinflammation, and oxidative stress in the mouse hippocampus: effects on expression of genes involved in beta-amyloid generation and Alzheimer’s disease. The journals of gerontology 69, 1212–1226. [DOI] [PMC free article] [PubMed] [Google Scholar]
  300. Unfer TC, Conterato GM, da Silva JC, Duarte MM, and Emanuelli T (2006). Influence of hormone replacement therapy on blood antioxidant enzymes in menopausal women. Clin Chim Acta 369, 73–77. [DOI] [PubMed] [Google Scholar]
  301. van Dam FS, Schagen SB, Muller MJ, Boogerd W, vd Wall E, Droogleever Fortuyn ME, and Rodenhuis S (1998). Impairment of cognitive function in women receiving adjuvant treatment for high-risk breast cancer: high-dose versus standard-dose chemotherapy. J Natl Cancer Inst 90, 210–218. [DOI] [PubMed] [Google Scholar]
  302. van’t Spijker A, Trijsburg RW, and Duivenvoorden HJ (1997). Psychological sequelae of cancer diagnosis: a meta-analytical review of 58 studies after 1980. Psychosomatic medicine 59, 280–293. [DOI] [PubMed] [Google Scholar]
  303. VanRyzin JW, Yu SJ, Perez-Pouchoulen M, and McCarthy MM (2016). Temporary Depletion of Microglia during the Early Postnatal Period Induces Lasting Sex-Dependent and Sex-Independent Effects on Behavior in Rats. eNeuro 3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  304. Varma M, Chai JK, Meguid MM, and Yang ZJ (2001). Gender differences in tumor-induced anorectic feeding pattern in Fischer-344 rats. Physiology & behavior 74, 29–35. [DOI] [PubMed] [Google Scholar]
  305. Vearncombe KJ, Rolfe M, Andrew B, Pachana NA, Wright M, and Beadle G (2011). Cognitive effects of chemotherapy-induced menopause in breast cancer. The Clinical neuropsychologist 25, 1295–1313. [DOI] [PubMed] [Google Scholar]
  306. Vegeto E, Belcredito S, Ghisletti S, Meda C, Etteri S, and Maggi A (2006). The endogenous estrogen status regulates microglia reactivity in animal models of neuroinflammation. Endocrinology 147, 2263–2272. [DOI] [PubMed] [Google Scholar]
  307. Vegeto E, Benedusi V, and Maggi A (2008). Estrogen anti-inflammatory activity in brain: a therapeutic opportunity for menopause and neurodegenerative diseases. Frontiers in neuroendocrinology 29, 507–519. [DOI] [PMC free article] [PubMed] [Google Scholar]
  308. Vegeto E, Bonincontro C, Pollio G, Sala A, Viappiani S, Nardi F, Brusadelli A, Viviani B, Ciana P, and Maggi A (2001). Estrogen prevents the lipopolysaccharide-induced inflammatory response in microglia. J Neurosci 21, 1809–1818. [DOI] [PMC free article] [PubMed] [Google Scholar]
  309. Vermeiren AP, Hoebe CJ, and Dukers-Muijrers NH (2013). High non-responsiveness of males and the elderly to standard hepatitis B vaccination among a large cohort of healthy employees. J Clin Virol 58, 262–264. [DOI] [PubMed] [Google Scholar]
  310. Vichaya EG, Chiu GS, Krukowski K, Lacourt TE, Kavelaars A, Dantzer R, Heijnen CJ, and Walker AK (2015). Mechanisms of chemotherapy-induced behavioral toxicities. Frontiers in neuroscience 9, 131. [DOI] [PMC free article] [PubMed] [Google Scholar]
  311. Vogel Ciernia A, Careaga M, LaSalle JM, and Ashwood P (2018). Microglia from offspring of dams with allergic asthma exhibit epigenomic alterations in genes dysregulated in autism. Glia 66, 505–521. [DOI] [PMC free article] [PubMed] [Google Scholar]
  312. Wachulec M, Peloso E, and Satinoff E (1997). Individual differences in response to LPS and psychological stress in aged rats. The American journal of physiology 272, R1252–1257. [DOI] [PubMed] [Google Scholar]
  313. Wakselman S, Bechade C, Roumier A, Bernard D, Triller A, and Bessis A (2008). Developmental neuronal death in hippocampus requires the microglial CD11b integrin and DAP12 immunoreceptor. J Neurosci 28, 8138–8143. [DOI] [PMC free article] [PubMed] [Google Scholar]
  314. Walf AA, and Frye CA (2010). Raloxifene and/or estradiol decrease anxiety-like and depressive-like behavior, whereas only estradiol increases carcinogen-induced tumorigenesis and uterine proliferation among ovariectomized rats. Behav Pharmacol 21, 231–240. [DOI] [PMC free article] [PubMed] [Google Scholar]
  315. Walker FR, Beynon SB, Jones KA, Zhao Z, Kongsui R, Cairns M, and Nilsson M (2014). Dynamic structural remodelling of microglia in health and disease: a review of the models, the signals and the mechanisms. Brain Behav Immun 37, 1–14. [DOI] [PubMed] [Google Scholar]
  316. Waters EM, and Simerly RB (2009). Estrogen induces caspase-dependent cell death during hypothalamic development. J Neurosci 29, 9714–9718. [DOI] [PMC free article] [PubMed] [Google Scholar]
  317. Weinhard L, di Bartolomei G, Bolasco G, Machado P, Schieber NL, Neniskyte U, Exiga M, Vadisiute A, Raggioli A, Schertel A, et al. (2018a). Microglia remodel synapses by presynaptic trogocytosis and spine head filopodia induction. Nat Commun 9, 1228. [DOI] [PMC free article] [PubMed] [Google Scholar]
  318. Weinhard L, Neniskyte U, Vadisiute A, di Bartolomei G, Aygun N, Riviere L, Zonfrillo F, Dymecki S, and Gross C (2018b). Sexual dimorphism of microglia and synapses during mouse postnatal development. Dev Neurobiol 78, 618–626. [DOI] [PMC free article] [PubMed] [Google Scholar]
  319. Werling DM, Parikshak NN, and Geschwind DH (2016). Gene expression in human brain implicates sexually dimorphic pathways in autism spectrum disorders. Nat Commun 7, 10717. [DOI] [PMC free article] [PubMed] [Google Scholar]
  320. Wofford JL, Loehr LR, and Schwartz E (1996). Acute cognitive impairment in elderly ED patients: etiologies and outcomes. Am J Emerg Med 14, 649–653. [DOI] [PubMed] [Google Scholar]
  321. Woolley CS, Gould E, Frankfurt M, and McEwen BS (1990). Naturally occurring fluctuation in dendritic spine density on adult hippocampal pyramidal neurons. J Neurosci 10, 4035–4039. [DOI] [PMC free article] [PubMed] [Google Scholar]
  322. Wu H, Luo J, Yu H, Rattner A, Mo A, Wang Y, Smallwood PM, Erlanger B, Wheelan SJ, and Nathans J (2014). Cellular resolution maps of X chromosome inactivation: implications for neural development, function, and disease. Neuron 81, 103–119. [DOI] [PMC free article] [PubMed] [Google Scholar]
  323. Wu LM, and Amidi A (2017). Cognitive impairment following hormone therapy: current opinion of research in breast and prostate cancer patients. Current opinion in supportive and palliative care 11, 38–45. [DOI] [PMC free article] [PubMed] [Google Scholar]
  324. Wu SY, Chen YW, Tsai SF, Wu SN, Shih YH, Jiang-Shieh YF, Yang TT, and Kuo YM (2016). Estrogen ameliorates microglial activation by inhibiting the Kir2.1 inward-rectifier K(+) channel. Sci Rep 6, 22864. [DOI] [PMC free article] [PubMed] [Google Scholar]
  325. Xiong X, Xu L, Wei L, White RE, Ouyang YB, and Giffard RG (2015). IL-4 Is Required for Sex Differences in Vulnerability to Focal Ischemia in Mice. Stroke 46, 2271–2276. [DOI] [PMC free article] [PubMed] [Google Scholar]
  326. Xu Y, Day TA, and Buller KM (1999). The central amygdala modulates hypothalamic-pituitary-adrenal axis responses to systemic interleukin-1beta administration. Neuroscience 94, 175–183. [DOI] [PubMed] [Google Scholar]
  327. Yirmiya R, and Goshen I (2011). Immune modulation of learning, memory, neural plasticity and neurogenesis. Brain Behav Immun 25, 181–213. [DOI] [PubMed] [Google Scholar]
  328. Yirmiya R, Rimmerman N, and Reshef R (2015). Depression as a microglial disease. Trends in neurosciences 38, 637–658. [DOI] [PubMed] [Google Scholar]
  329. Young JJ, Bruno D, and Pomara N (2014). A review of the relationship between proinflammatory cytokines and major depressive disorder. J Affect Disord 169, 15–20. [DOI] [PubMed] [Google Scholar]
  330. Yuan L, Liu S, Bai X, Gao Y, Liu G, Wang X, Liu D, Li T, Hao A, and Wang Z (2016). Oxytocin inhibits lipopolysaccharide-induced inflammation in microglial cells and attenuates microglial activation in lipopolysaccharide-treated mice. Journal of neuroinflammation 13, 77. [DOI] [PMC free article] [PubMed] [Google Scholar]
  331. Zec RF, and Trivedi MA (2002). The effects of estrogen replacement therapy on neuropsychological functioning in postmenopausal women with and without dementia: a critical and theoretical review. Neuropsychol Rev 12, 65–109. [DOI] [PubMed] [Google Scholar]
  332. Zhang X, Lan N, Bach P, Nordstokke D, Yu W, Ellis L, Meadows GG, and Weinberg J (2012). Prenatal alcohol exposure alters the course and severity of adjuvant-induced arthritis in female rats. Brain Behav Immun 26, 439–450. [DOI] [PMC free article] [PubMed] [Google Scholar]
  333. Ziko I, De Luca S, Dinan T, Barwood JM, Sominsky L, Cai G, Kenny R, Stokes L, Jenkins TA, and Spencer SJ (2014). Neonatal overfeeding alters hypothalamic microglial profiles and central responses to immune challenge long-term. Brain Behav Immun 41, 32–43. [DOI] [PubMed] [Google Scholar]
  334. Ziko I, Sominsky L, Nguyen T-X, Yam K-Y, De Luca S, Korosi A, and Spencer SJ (2017). Hyperleptinemia in Neonatally Overfed Female Rats Does Not Dysregulate Feeding Circuitry. Frontiers in endocrinology 8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  335. Zwart W, Terra H, Linn SC, and Schagen SB (2015). Cognitive effects of endocrine therapy for breast cancer: keep calm and carry on? Nature reviews. Clinical oncology 12, 597–606. [DOI] [PubMed] [Google Scholar]

RESOURCES