Abstract
Men and women differ in their vulnerability to a variety of stress-related illnesses, but the underlying neurobiological mechanisms are not well understood. This is likely due to a comparative dearth of neurobiological studies that assess male and female rodents at the same time, while human neuroimaging studies often don’t model sex as a variable of interest. These sex differences are often attributed to the actions of sex hormones, i.e. estrogens, progestogens and androgens. In this review, we summarize the results on sex hormone actions in the hippocampus and seek to bridge the gap between animal models and findings in humans. However, while effects of sex hormones on the hippocampus are largely consistent in animals and humans, methodological differences challenge the comparability of animal and human studies on stress effects. We summarise our current understanding of the neurobiological mechanisms that underlie sex-related differences in behavior and discuss implications for stress-related illnesses.
Keywords: Stress, Sex differences, Cognition, Translational
1. Introduction
It has long been known that men and women differ in the susceptibility and prevalence of a wide-range of stress-related illnesses such as psychiatric disorders, neurological and neurodegenerative disorders. While men show a higher prevalence in neurodevelopmental disorders like autism or ADHD (Faraone, 2015; Schuck et al., 2019), women show a higher prevalence in stress-related disorders like anxiety or depression in adulthood (Albert, 2015; Angst et al., 2002; Barnes et al., 2005; Breslau et al., 1995; Cahill, 2006; Irvine et al., 2012; Kessler et al., 1994; McLean et al., 2011; Olff, 2017; Seeman, 1997) with higher prevalence rates emerging during adolescence. Furthermore, an increased prevalence in stress-related disorders is observed during periods of drastic hormonal changes (puberty, pregnancy, postpartum, menopause) along the female life-span (see Slavich and Sacher, 2019 for review). Accordingly, sex hormones and their interaction with stress hormones seem to play an important role in the development of stress-related disorders. Many of these disorders have been linked to altered structure, function and neurogenic processes within the hippocampus, as well as hippocampus-dependent cognitive functions, all of which have been shown to differ between males and females. Thus, in order to understand the moderating influence of sex and stress hormones on the development of stress-related disorders, it is important to disentangle the effects of these hormones on hippocampal morphology and function. Since the complexity of this topic and a variety of different methodological approaches has led to a multitude of different research findings, this review aims to provide a thorough and timely overview of steroid actions on the hippocampus.
Most importantly, this review seeks to integrate findings from both the animal and human literature on the topic. While animal models allow us to study the neurobiological underpinnings of sex differences in behavior down to the molecular level, it is not possible to model all aspects of human behavior in rodents. This is particularly true for verbal functions, certain socio-emotional functions and fine-motor skills, as they only developed in primates/humans. Furthermore, rodent-models cannot cover stress-related disorders like depression, post-traumatic stress disorder (PTSD) or anxiety disorders in their entirety. While many animal models have been developed to assess stress-related disorders, they have traditionally used male rodents; although this has recently changed, partly due to altered regulations by funding agencies. However, the available studies have shown that there are large behavioral, neurochemical, endocrine and neurobiological sex differences in most of these models (Dalla et al., 2005, 2008; Dallak et al., 2008; Palanza, 2001). Accordingly, this review will compare the cellular and molecular findings in animals to neuroimaging findings in humans in order to work out parallels and discrepancies between these approaches. This overview can help to identify which of the factors observed to modulate hippocampal morphology in pre-clinical animal studies are also of behavioral, and possibly clinical relevance in humans.
2. Why sex hormones matter for sex-related disorders: Sex and sex hormone influences on the stress response
Cortisol is chronically elevated in patients with stress-related disorders, such as depression or PTSD (Travis et al., 2016; Szeszko et al., 2018; Keller et al., 2017; Bremner, 2006) and also cortisol reactivity to acute stress appears to be altered. In humans the cortisol awakening response (CAR) has been described as an indicator of the capacity of the HPA-axis to respond to stressors (Langelaan et al., 2006). While some studies show an increased CAR in patients under chronic stress (Wust et al., 2000; Schlotz et al., 2004), other studies demonstrate that the cortisol awakening response is blunted in patient groups with chronically elevated cortisol levels (Wessa et al., 2006; Roberts et al., 2004; Duan et al., 2013). Accordingly, the elevated CAR in participants who report chronic stress represents a healthy adaptation to the expected stress during the upcoming day. However, if the chronic stress continues, this adaptive capacity is lost, resulting in disorders like depression, PTSD or chronic fatigue syndrome. Likewise, it appears that chronic stress/chronically elevated cortisol levels affect acute stress reactivity. While some studies show that participants under chronic stress show an increased cortisol response to acute stress, most studies demonstrate blunted cortisol reactivity in patients under chronic stress (Zorn et al., 2017).
Accordingly, the question arises, whether the increased prevalence of stress-related disorders in females can be linked to sex differences in the endocrinological response to chronic and acute stressors. Indeed, animal studies show that females display increased basal and stress-induced levels of ACTH and corticosterone (CORT) compared with males (Hillerer et al., 2013; Luo et al., 2013; Romeo et al., 2004; Handa et al., 1994; Jezova et al., 1996). In humans however, the CAR, which is usually assessed as area under the curve, is stronger in women compared to men due to a delayed decrease in cortisol (Hollanders et al., 2017; Wust et al., 2000), but does not change along the menstrual cycle (Wolfram et al., 2011). Conversely, men show a higher cortisol response to psychosocial stress (mostly TSST) than women (Pulopulos et al., 2018; Stephens et al., 2016; Reschke-Hernandez et al., 2017; Kogler et al., 2017; Kirschbaum et al., 1992; Khaksari et al., 2005; Herbison et al., 2016; Childs et al., 2010). However, since the cortisol response to stress is attenuated in depressed patients and rates of depression are higher in women, it is possible that a higher proportion of depressed individuals in the female group have contributed to these findings. Indeed a recent study was not only able to show that attenuated cortisol responses in women are related to depressive symptoms, but that in men, the opposite pattern was observed. In men depressive symptoms predicted a higher cortisol response in stressful situations (Zorn et al., 2017; Powers et al., 2016). Accordingly, sex differences in the physiological stress response may be overestimated, if depression symptomology is not accounted for.
Several studies indicate that sex hormones contribute to the stress response difference between men and women. Firstly, sex differences in the stress response appear to emerge around puberty due to a reduction in cortisol elevations in girls (Stroud et al., 2017; Cameron et al., 2017). Some studies also indicate an increased cortisol response in girls compared to boys before puberty (Hollanders et al., 2017). Secondly, the sex difference in the cortisol response appears to be moderated by women’s menstrual cycle phase. While women in their follicular phase show reduced cortisol responses, women in their luteal cycle phase showed comparable cortisol reactivity to stress as men (Stephens et al., 2016; Childs et al., 2010; Montero-Lopez et al., 2018; Kirschbaum et al., 1999; Kajantie and Phillips, 2006). Likewise, high estrogen levels in women being associated with the increased basal levels of ACTH and CORT (Altemus et al., 2001; Marinari et al., 1976) and female rodents in proestrus show higher levels of basal and stress-induced CORT compared to other estrus phases (Carey et al., 1995; Figueiredo et al., 2002; Viau and Meaney, 1991). This finding is somewhat unexpected, given the fact that depressive symptoms are more common in the luteal phase of the menstrual cycle (see Sundstrom Poromaa and Gingnell, 2014 for a review). Indeed, premenstrual syndrome is associated with blunted cortisol reactivity to social or physical stress (Roca et al., 2003; Huang et al., 2015) as well as a blunted CAR (Roca et al., 2003) and the finding of increased cortisol response to stress during the luteal cycle phase is controversial, with some studies reporting no difference between cycle phases (Bouma et al., 2009; Abplanalp et al., 1977) and other studies reporting higher cortisol response during the follicular cycle phase (Nakajima et al., 2019; Maki et al., 2015; Hlavacova et al., 2008). Accordingly, sex and sex hormones are important factors to consider in the etiology of stress-related illnesses.
3. The role of the hippocampus in stress-related disorders
In animals, impaired hippocampal neurogenesis has been linked to an increase in anxiety-like and depressive-like behavior and ablation and/or altered neurogenesis has been associated with increased anxiety-related behavior (Kheirbek et al., 2012; Revest et al., 2009; Sah et al., 2012; Boldrini et al., 2012; Boldrini et al., 2009; Santarelli et al., 2003; Dias et al., 2014). Likewise, in human neuroimaging studies, stress-related disorders like major depressive disorder (MDD), bipolar disorder (BD), anxiety disorders and PTSD have reliably been associated with hippocampal volume loss (MDD: Zhao et al. (2008), Taylor et al. (2005), Spalletta et al. (2014), Sivakumar et al. (2015), Schuhmacher et al. (2012), Opel et al. (2014), MacMaster et al. (2014), Kong et al. (2013), Hastings et al. (2004), Gerritsen et al. (2011), Frodl et al. (2002, 2014); see Vythilingam et al., 2004; Schmaal et al., 2017 for review; BD: MacMaster et al. (2014), Redlich et al. (2014), Frazier et al. (2008, 2005), anxiety disorders: Yamasue et al. (2008), Moon et al. (2014), Irle et al. (2010), Coker et al. (2010): (Ahmed-Leitao et al., 2016; Keding and Herringa, 2015; Logue et al., 2018; Woon and Hedges, 2011. In the case of MDD, which has been most extensively studied, reduced hippocampal volumes have also been related to higher symptom severity (Lorenzetti et al., 2009; Jaworska et al., 2014; Axelson et al., 1993); see Dotson et al., 2009 for review), number of depressive episodes (Axelson et al., 1993; Wisse et al., 2015; Han et al., 2014), longer disorder duration (Sheline et al., 1999; McKinnon et al., 2009) and suicide attempts (Colle et al., 2015). These effects appear to be stronger in the left compared to the right hippocampus (Zhao et al., 2008; Sivakumar et al., 2015; MacMaster et al., 2014). Neither in MDD, nor in PTSD, a sex difference in volume reduction was observed (Woon and Hedges, 2011). However, volume reductions in BD appear to be stronger in females (Frazier et al., 2008, 2005), while no volume reduction was observed in female survivors of sexual abuse (Landre et al., 2010). Furthermore, smaller hippocampal volumes were also observed in schizophrenia (Pruessner et al., 2015; Pegues et al., 2003; Irle et al., 2011; Exner et al., 2008) and ADHD (Wang et al., 2018). Along with volume reductions, a reduced activation of the hippocampus during memory tasks and viewing of emotional pictures was observed in female patients with MDD (Milne et al., 2012; Holsen et al., 2011, 2012). Furthermore, women with PTSD displayed reduced hippocampal reactivity to fear compared to men (Felmingham et al., 2010). Accordingly, understanding the factors that lead to this altered hippocampal morphology and functioning is of uttermost importance for understanding the development of stress-related disorders.
4. Hippocampus-dependent cognitive functions and their role for mood and anxiety disorders
The role of the hippocampus in stress-related disorders has been linked to its function. As an important part of the limbic system the hippocampus has been linked to emotional processing in a wide range of tasks (Mackiewicz et al., 2006). Larger, more active hippocampi, particularly in women, have been related to emotion regulation ability (Kong et al., 2014). However, in both animal and human studies, the hippocampus is, above all, seen as a key area in cognition. The hippocampus forms part of a larger network of structures involved in various aspects of learning and memory (Orsini et al., 2015) and hippocampal neurogenesis and dendritic morphology has long been linked with hippocampal-dependent learning and memory (Leuner et al., 2006; Gould et al., 1999; Jessberger et al., 2009; Kee et al., 2007; Shors et al., 2001). Most prominently, the hippocampus has been implicated in spatial processing (navigation) on the one hand (Wegman et al., 2014; Sneider et al., 2018; Ohnishi et al., 2006; Iaria et al., 2008) and episodic memory processing on the other hand (Pletzer et al., 2019; Protopopescu et al., 2008). Specifically, the right hippocampus seems to play an important role in spatial navigation, while the left hippocampus is more involved in verbal memory (Ezzati et al., 2016). Reduced spatial learning and memory are associated with hippocampal impairments in several animal models (Lassalle et al., 2000; Morris et al., 1982) and are also seen in humans with damage to the hippocampus or in psychiatric disorders and AD (Corkin, 2002; Perl, 2010). Accordingly, it is not surprising that several neuropsychiatric disorders associated with hippocampal volume loss, are also characterized by cognitive deficits (McKinnon et al., 2009; Beinhoff et al., 2008; Gutierrez-Lobos et al., 2002; Scheff et al., 2006; Baum and Sex, hormones, 2005). Therefore, spatial and memory tasks are widely used in both animal and human studies to behaviorally assess hippocampal function.
The fact that cognitive training can also affect hippocampal morphology may even be of therapeutic relevance for stress-related disorders. As an example, hippocampal neurogenesis has been shown to be essential for spatial pattern separation and completion (Clelland et al., 2009; Creer et al., 2010; Sahay et al., 2011); although an optimal level is required i.e. too much is not necessarily a good thing (Akers et al., 2014; Jessberger et al., 2007; Scharfman and Myers, 2016) as has also been demonstrated using computational models (Aimone et al., 2009; Butz et al., 2006). On the flipside, hippocampal neurogenesis can be promoted through spatial training, although the effect is dependent on the developmental stage in which the immature neurons are exposed to the training and the sex of the individual (Epp et al., 2007).
5. Sex differences in hippocampus-dependent cognitive functions
As for the prevalence of stress-related disorders that are associated with altered hippocampal morphology, sex differences for hippo-campus-dependent functions are well-documented. Men and women differ in various aspects of cognition, social functioning and emotion (Hollanders et al., 2017; Andreano and Cahill, 2009; Halpern, 2000; Hamann and Canli, 2004; Merz and Wolf, 2017). Sex differences in spatial performance exist across a variety of species and tasks, including MWM and spatial perception tasks in rodents. Typically, males outperform females in spatial acquisition, spatial reference and working memory (Galea et al., 1996; Perrot-Sinal et al., 1996; Jonasson, 2005; Dabbs et al., 1998; Kaufman, 2007; Woolley et al., 2010; Endo et al., 1994; Markowska, 1999). In humans, sex differences in spatial performance have also been observed quite robustly and across cultures in spatial navigation and mental rotation tasks (Galea and Kimura, 1993; Saucier et al., 2002; Schoning et al., 2007; Silverman et al., 2000; Voyer and Bryden, 1990; Voyer et al., 1995). Vice versa, women outperform men in verbal memory tasks (Capitani et al., 2005; Capitani et al., 1998; de Frias et al., 2006; Gauthier et al., 2009; Hirnstein et al., 2014; Kimura and Seal, 2003; Soleman et al., 2013; Yonker et al., 2003). This advantage in verbal memory is accompanied by a female advantage in other aspects of memory (Pillemer et al., 2003) including the recognition of objects (Bettis and Jacobs, 2012), female faces (Lewin and Herlitz, 2002), locations (Voyer et al., 2007) or even odors (Oberg et al., 2002). Additionally, in both animals and humans, sex differences in cognition have also been linked to differential cognitive strategies in males and females (Galea and Kimura, 1993; Pletzer, 2014; Beiko et al., 2004; Grön et al., 2000; Williams et al., 1990). For example in the Morris water maze, it has been shown that males prefer extra-maze spatial/geometric cues, which are hippocampus-dependent, whereas females tend to rely on visible landmark cues, which are more dependent on the striatum (Cherney et al., 2008; Grissom et al., 2013). A stronger focus on local landmark cues by women has also repeatedly been demonstrated in spatial navigation in humans (see Harris et al., 2019 for review).
Regarding emotion recognition, women appear to particularly outperform men in the recognition of sad and fearful faces (Olderbak et al., 2019). Nevertheless, women appear to show greater expression of emotions (Hamann and Canli, 2004). Interestingly, it appears that in summary, a female advantage is more likely found in those aspects of behavior that cannot be studied in animals, like verbal functions, such that the neurobiological underpinnings of these behaviors are not as well understood as for those showing a male advantage, like spatial functions. Therefore, the molecular mechanisms that differ between males and females are likely to play key roles in the differences that have been observed regarding cognitive processes and the higher incidence of psychiatric disorders in women. In the following sections, we outline some of the molecular mechanisms, focussing on the hippo-campus that underpin these sex differences.
6. Preclinical studies on the molecular mechanisms of sex and stress hormone actions in the hippocampus – Studies on neurogenesis and dendritic morphology
Steroid hormones are potent regulators of adult hippocampal neurogenesis and dendritic morphology with its regulation being dependent on sex, age and reproductive experience (see Mahmoud et al., 2016 for review). Estrogens (i.e. 17α/β-estradiol, estrone), progestins (i.e progesterone) and androgens (i.e. testosterone) play an important role in regulating different stages of adult hippocampal neurogenesis i.e. proliferation, differentiation and survival of new neurons during developmental and adult hippocampal neurogenesis in males and females (Bowers et al., 2010; Waddell et al., 2013; Zhang et al., 2008). Similar to sex differences in adult hippocampal neurogenesis, there is a profound amount of data suggesting similar differences with respect to dendritic morphology that may underlie sex-differences in behavior (for review see Koss and Frick, 2017; Scharfman and MacLusky, 2017). The predominant evidence for a role of sex hormones in adult hippocampal neurogenesis comes from exogenous hormone manipulation studies in ovariectomised (OVX) and castrated (GDX) animals. In the following section, we review the available literature about the influence of acute and chronic treatment of estrogens, progestogens and testosterone on adult hippocampal neurogenesis and dendritic morphology in males and females.
6.1. Sex differences in hippocampal neurogenesis
During adolescence, the number of immature neurons in the dentate gyrus declines more rapidly in males and the number of immature neurons is higher in adult females compared to males (Siddiqui and Romeo, 2019). As outlined in a recent review, sex differences in hippocampus-dependent cognitive functions have also been linked to sex differences in hippocampal proliferation during learning (Yagi and Galea, 2019). For example, better spatial acquisition in males is concomitant with increased survival of new neurons during training; however these two factors were not correlated (Chow et al., 2013). When cell activation within the granule cell layer (GCL) was assessed via co-labelling of bromodeoxyuridine (BrdU) and EGR-1 again there was a greater activation of new cells in males than in females; however, performance was positively correlated in females, but not in males and the response to spatial memory retrieval was similar between the sexes (Chow et al., 2013). Moreover, better spatial learning is associated with greater activation of 20d neurons only in females, which may be interpreted such that (1) new neurons are more excitable in females than in males, (2) new neurons mature faster under the influence of estradiol, which regulates cell development/facilitates effects on neural excitability (Lee et al., 2004; Smith and McMahon, 2006) and (3) may depend on an increased perceived task difficulty in females, which increases involvement of the hippocampus (Beylin et al., 2001). Fast acquisition of trace eyeblink conditioning, which is a hippocampal-dependent task, is correlated with greater cell survival (in terms of percentage) in females (Dalla et al., 2009).
6.2. Sex differences in dendritic morphology
Sex differences in hippocampal morphology, vary across the life span and different effects have been reported for different hippocampal subfields (Bartesaghi et al., 2003; Shors et al., 2001). For instance, while young adult males show stronger dendritic branching in CA1 than young adult females, this sex differences disappears during aging (Markham et al., 2005). Since sex differences in dendritic morphology of the adult hippocampus appear to be largely dependent on the estrus cycle in females and other endocrinological factors like stress, these changes across the life span are likely also dependent on hormonal variations (Markham et al., 2005). Accordingly, the following sections will focus on the modulation of dendritic morphology by sex and stress hormones.
6.3. Estrogens
Estrogens, with its most important endogenous specimen 17α-estradiol, are among the most important sexual hormones, enzymatically derived via transformation of androgens, which is catalysed by the enzyme aromatase. Synthetic estrogens used in animal studies are mostly 17β-estradiol, estrogen benzoate, estrone or conjugated estrogens like sodium estrogen sulfate or sodium equilin sulfate. Estrogens exert its neuronal and behavioral action via the specific estrogen receptors ERα/β, by classical and non-classical mechanisms (see Fuentes and Silveyra, 2019; Galea et al., 2017 for review). The effects of estrogens on hippocampal neurogenesis and dendritic morphology have been investigated by observing natural estrogen fluctuations in female animals (e.g. along the estrous cycle), by surgical intervention (ovariectomy) as well as by pharmacological intervention (exogenous estrogen administration, inhibition of estrogen synthesis). Overall results demonstrate a positive effect of estrogens on hippocampal neurogenesis and dendritic morphology in females. Studies in males are less common, but seem to demonstrate rather little effect, but perhaps an increase in cell survival (Barker and Galea, 2008; Spritzer and Galea, 2007).
6.3.1. Estrogenic effects on hippocampal neurogenesis
Generally it seems that estrogen levels are positively correlated with cell proliferation and negatively correlated with cell death (Pawluski et al., 2009 for review). However, its effects are highly dependent on a variety of factors including temporal dynamics, dose, age, sex and species (Bowers et al., 2010; Barker and Galea, 2008; Chan et al., 2014; Galea, 2008; Galea et al., 2013 for review). The first hint about a possible involvement of sex steroids in regulating adult hippocampal neurogenesis came from observations of Galea and Ormerod. They demonstrated that female meadow voles express decreased levels of cell proliferation during breading season, when estrogen levels are naturally high (Galea and McEwen, 1999; Ormerod and Galea, 2001). In contrast, rats during pro-estrus/phases of high estrogen, have been shown to have increased levels of c-Fos and zif268 activation, cell proliferation, 14d cell survival and neural excitability in the DG (McClure et al., 2013; Rummel et al., 2010; Tanapat et al., 1999; Warren et al., 1995; Yagi et al., 2017). Further corroborating the first findings about a possible involvement of estrogen in regulating hippocampal neurogenesis, lowering estrogen levels via removal of ovaries, significantly reduced cell proliferation, which could be rescued by estrogen replacement (Fowler et al., 2008; Perez-Martin et al., 2005) (see Pawluski et al., 2009; Galea, 2008 for review). However, the aforementioned effect of OVX on cell proliferation is dependent on the time elapsed since OVX. In detail, OVX decreases cell proliferation when assessed at 6-7d (Tanapat et al., 1999), but this appears to be a short-lasting consequence as long-term OVX (21/27d) has no effect on cell proliferation in rats and mice (Banasr et al., 2001; Green and Galea, 2008; Lagace et al., 2007; Tanapat et al., 2005). Together, these findings suggest that the female brain may have a compensatory mechanism that restores cell proliferation after longer periods of OVX, i.e. by an increase of extra-gonadal aromatization (Zhao et al., 2005) and synthesis of estrogen in hippocampal neurons (Fester et al., 2012; Hojo et al., 2008). Indeed, in vitro studies demonstrate that hippocampal granule cells in hippocampal dispersion cultures require local estrogen to proliferate, as inhibition of its synthesis with the specific aromatase inhibitor letrozole decreases cell proliferation, whereas application of exogenous estrogen had no effect (Fester et al., 2006).
Similar to OVX, the effects of exogenous estrogen administration are largely treatment duration-, time- and dose-dependent. Thus, acute treatment with estrogen has been shown to have a dose-dependent nonlinear effect (Tanapat et al., 2005; Barha et al., 2009), with single injections of low (di-estrus range; 0.3 µg) and high doses (pro-estrus range; 10 µg) restoring cell proliferation in OVX rats, whereas medium/supraphysiological doses (1 µg/50 µg) are ineffective (Tanapat et al., 2005; Barha et al., 2009). The acute effect of estrogen seems to have a rapid within hour effect that is persistent, but only if it occurs within a critical period after OVX. Respectively, acute estrogen replacement in high/pro-estrus doses (10 µg) is able to restore cell proliferation to the level of SHAM controls in young adult rats 1 week after OVX, whereas treatment 28d after OVX was ineffective in this manner (Tanapat et al., 2005). These findings suggest that long-term OVX reduces the cells sensitivity, or ability, to respond to the proliferative effect of acute estrogen treatment after long-term deprivation. Time-dependent effects of acute estrogen treatment are rather transient, occurring rapidly with an increase in cell proliferation followed by a decrease after longer timepoints. In more detail, 17β-estradiol, estradiol benzoate and estrone increase cell proliferation 30 min, 2 h and 4 h post-administration in young adult, adult and middle-aged OVX rats (Ormerod and Galea, 2001; Tanapat et al., 1999; Tanapat et al., 2005; Barha et al., 2009; Barha and Galea, 2011; Ormerod et al., 2003), whereas a decrease in cell proliferation has been observed 48 h after estradiol benzoate administration (Ormerod and Galea, 2001; Ormerod et al., 2003). The observed time-dependent and transient effects of acute estrogen may be a result of stimulating different populations of proliferative cells i.e. cells in a progressive steady state and a replenishing cell population (Nowakowski et al., 2008). In this context, stimulating one population of cells would decrease the rate of division in the other population to keep the amount of proliferating cells constant, which in fact may also explain why longer times of acute estrogen administration do not increase cell proliferation.
Similar to the temporal effects of OVX, chronic estrogen treatment seems to play a minor role in influencing hippocampal cell proliferation in females, as 21d treatment with either release pellets, injections or cyclic regimen of injections every four days for one week post OVX showed no effect on cell proliferation in rats (Chan et al., 2014; Tanapat et al., 2005).However, results may also vary dependent on methodological differences (i.e. continuous exposure via pellet vs. pulsatile exposure via subcutaneous injections), as 15d continuing estrogen treatment had a positive effect on cell proliferation in female rats, when administered one week after OVX (Barker and Galea, 2008). Another important point to consider when investigating the effects of estrogen on adult hippocampal neurogenesis is the timeline of estrogen administration relative to the examination of cell proliferation and relative to the length of estrogen exposure.
Overall, the effects of estrogen on stages of neurogenesis apart from proliferation i.e. cell survival are significantly less well studied. Regarding the latter, the outcome seems to be largely dependent on whether the proliferative marker (i.e. BrdU) is injected before or after the hormonal treatment. Injecting BrdU prior to estrogen treatment will reveal the effect of estrogen independent of cell proliferation, whereas BrdU injections after hormonal application include the effects of estrogen on proliferation and survival. Thus, the hormone-related environment during which the cells are generated determines the outcome of chronic estrogen on cell survival. In this context, a decreased survival was observed in cells produced prior to treatment with estradiol benzoate (with BrdU injected prior to treatment) (Barker and Galea, 2008; Chan et al., 2014), whereas an increase in cell survival has been shown in populations produced after initiation of 17β-estradiol treatment (with BrdU injected following hormonal treatment) (McClure et al., 2013). This is most likely attributed to the estrogen-induced increase in cell proliferation, as the increase in cell survival is only seen, when respective cells are produced in an estrogen-enriched environment. Other forms of estrogen seem to negatively impact cell survival under chronic conditions, as high estrone for 20d decreases new neuron survival in the DG, revealing that cells that proliferate under estrone do not survive in an estrogen-rich environment (McClure et al., 2013).
Unfortunately, the aforementioned studies do not include males, which limits our understanding of the effect of estrogen treatment mostly to females. However, the few studies performed point towards a sex-dependent effect of estrogen in hippocampal neurogenesis. Thus, acute treatment with high doses of estradiol benzoate (100 µg) in intact neonates (postnatal day (PND) 0) increases cell proliferation in females, with no effect observed in males, despite an increased level of cell proliferation under basal conditions. In contrast, administering the aromatase inhibitor, formestane, or the estrogen receptor antagonist, tamoxifen, significantly decreased the number of new cells in males but not females. Given the fact that the increased rate of proliferation induced by neonatal estradiol persisted until at least 3 weeks of age, there appears to be an organizational effect (Bowers et al., 2010). Although interpreting the results of the latter study needs caution, as it has to be kept in mind that endogenous estrogen synthesis may have influenced the outcome of exogenous estrogen application, the results in neonates are mirrored by those in adult rats revealing that chronic estradiol benzoate treatment had no significant effect on cell proliferation or survival in males (Barker and Galea, 2008; Spritzer and Galea, 2007). In contrast short-term treatment has been shown to increase survival in males after a 3–5 day treatment with estradiol benzoate in voles and mice (Ormerod et al., 2004; Saravia et al., 2007; Zhang et al., 2010), an effect that seems to be limited to the axon extension phase of the cell development as 29d treatment did not affect neurogenesis (Ormerod et al., 2004; Saravia et al., 2007). Therefore, taken as a whole, the general consensus is that neurogenesis in males is not, or only minimally, affected by exogenous estrogen exposure.
The molecular mechanism mediating the effect of estrogen on adult hippocampal neurogenesis may be largely attributed to the action of estrogen receptors (ERs), ERα and ERβ that are located in the CA1/3 region and DG of the hippocampus of males and females (Fowler et al., 2005; Mazzucco et al., 2006; Perez-Martin et al., 2003; Shima et al., 2003; Shughrue et al., 1997) (and see Duarte-Guterman et al., 2015 for review), as activation of ERα/ERβ with the specific agonists propyl pyrazole triol (PPT) and diarylproprionitrile (DPN) increases cell proliferation (Mazzucco et al., 2006), whereas the use of a selective ER antagonist blocks the effect of estrogen in OVX females (Nagy et al., 2006). However, treatment with the above mentioned agonists is not able to restore cell proliferation to levels seen with estrogen, suggesting that modulation of neurogenesis by estrogen occurs not only via ERα/β, but also via the G-protein coupled estrogen receptor GPER, which is expressed in the DG, CA1,2,3 in male and female rats (Brailoiu et al., 2007). Indeed, treatment with the GPER antagonist G15 or the GPER agonist G1 has been shown to have a proliferative/anti-proliferative effect (Duarte-Guterman et al., 2015). Furthermore, there seems to be a mediation of estrogen effects via adrenal hormones, as adrenalectomy (ADX) eliminates the estradiol benzoate induced increase in cell proliferation seen after 48 h of estrogen exposure in OVX rats (Ormerod et al., 2003). Whether these ERα and ERβ-mediated effects are via membrane estradiol signalling or transcriptional or a combination of both remains to be fully elucidated. In a recent study using the streptozocin-induced diabetic mouse model, ERα and ERβ agonists were shown to increase the membrane ERα and ERβ protein levels in the hippocampus, but not in the nucleus and subsequently CREB and BDNF levels (Tang et al., 2019). Moreover, sex-differences in the molecular mechanism via which estradiol potentiates glutamatergic signalling and within the hippocampus have been observed. While this occurs in both sexes, at both the pre-synapse and post-synapse, it appears to occur through the oppose receptors in males and females (Oberlander and Woolley, 2016). In keeping, the mechanism through which estradiol facilitates long-term potentiation also differs between males and females (Jain et al., 2019).
6.3.2. Estrogenic effects on dendritic morphology
The effects of estrogen on hippocampal dendritic morphology are wide-ranging, not only affecting dendritic density, but also spine type. Thus, whereas males tend to express higher numbers of thin spines, females have been shown to have more mushroom spines, an effect that is even more pronounced during phases of high estrogen (i.e. pro-estrus) compared to phases of lower estrogen (i.e. estrus) (Gonzalez-Burgos et al., 2005; Vierk et al., 2012) (see Sheppard et al., 2019 for review). This is an interesting finding given the fact that thin and mushroom spines differ in their functional characteristics with the latter being more mature/stable spines, with a larger postsynaptic density and increased number of AMPA receptors. In contrast, thin spines are typically more dynamic/transient spines (Ashby et al., 2006; Harris et al., 1992; Holtmaat et al., 2005; Shinohara et al., 2008; Zuo et al., 2005) (see Bourne and Harris, 2007; Kasai et al., 2010 for review). Such differences may also explain differences in behavioral performances between males and females or females during different phases of the estrus cycle.
Estrogen treatment is known for its rapid effect on dendritic spines. Accordingly, dendritic spine density has been shown to fluctuate in CA1 pyramidal cells across the estrus cycle, with highest spine density levels observed during phases of high estrogen i.e. pro-estrus (McEwen and Woolley, 1994; Woolley et al., 1990). In keeping with the changes observed in hippocampal neurogenesis, the density of spines in the CA1 region of the hippocampus gradually decrease after OVX (Gould et al., 1990), an effect that remains stable for up to 40d (Woolley and McEwen, 1993). The reduction can be rescued by exogenous estrogen administration in rats (Gould et al., 1990; Woolley and McEwen, 1993; Jacome et al., 2016; MacLusky et al., 2005) and mice (Mukai et al., 2007; Phan et al., 2012; Tuscher et al., 2016), or after treatment with specific ER agonists. In more detail, in vivo studies revealed and increase in the number of dendritic spines in CA1 after the administration of the ERα agonist PPT (Phan et al., 2011) or the GPER agonist G1 (Gabor et al., 2015), whereas the antagonist DPN had the opposite effect (Phan et al., 2011). Similar findings of increased spine density have been reported in the CA1 region of male rats following 17β-estradiol administration in male rats (Jacome et al., 2016). Importantly, the results observed in vivo are mirrored by in vitro studies using hippocampal sections from female and male rats that were exposed to exogenous estrogen application or application of an ERα agonist (Mukai et al., 2007; Phan et al., 2015; Tsurugizawa et al., 2005). However, the effects observed in vivo and in vitro may rather be dependent on local estrogen synthesis in spines than estrogen from the periphery, at least in females, as revealed by studies using the aromatase inhibitor letrozole, which induces a non-steroidogenic, reversible inhibition of the local estrogen synthesis in hippocampal spines. Hence, inhibition of local hippocampal estrogen synthesis leads to a loss of spines in intact and OVX females, as well as in hippocampal cell cultures from female rats (Fester et al., 2012; Brandt et al., 2013; Kretz et al., 2004), with the degree of spine loss being even higher in cycling females (Zhou et al., 2010). The fact that there was no effect observed in males/male cell culture (Fester et al., 2012; Brandt et al., 2013; Kretz et al., 2004; Zhou et al., 2010), suggests a sex-specific difference in sexual steroid-induced synaptogenesis. The observed gender differences after letrozole treatment may be either a result of a dramatic impairment of LTPs in females, which play an important role in spine formation (Yuste and Bonhoeffer, 2001; Yuste and Bonhoeffer, 2004), or may depend on a GnRH-regulatory mechanisms, which may stimulate estrogen synthesis in hippocampal neurons and spine synapse density in a dose-dependent manner in females. Indeed, treatment with the GnRH antagonist antide has been shown to abolish the effect of local estrogen synthesis in female rats (Zhou et al., 2010). Although, the exact mechanisms are unknown, and there are no direct neuronal connections present that could account for the observed phenomenon, there may be an indirect pathway via the median eminence.
6.4. Progestogens
The second group of sex hormones are progestogens. In contrast to estrogens, their precursor-stage is not testosterone, but pregnenolone, which is enzymatically converted into progesterone via the 3β-hydroxysteroide dehydrogenase (3β-HSD). The most important endogenous progestogen in humans is progesterone (synthetic progestogens are termed progestins). In addition, its metabolite allopregnanolone, which is also produced in hippocampal neurons plays an important role in a variety of processes.
Progestogens act via the progesterone membrane receptors PR-A/B expressed throughout reproductive tissue and the brain (see Brinton et al., 2008 for review). Studies on the effects of progesterone on molecular mechanisms in the hippocampus are rare and have yielded inconsistent results. While some studies demonstrate facilitating effects on estrogenic mechanisms, other studies demonstrate counteracting effects on estrogenic mechanisms. The results appear to be dose- and time-dependent, which may account for some of the inconsistencies in the literature.
6.4.1. Progestogenic effects on hippocampal neurogenesis
In addition to estrogens, progestins like progesterone are important regulators that can modulate the effect of estrogen in influencing adult hippocampal neurogenesis in adult females (Tanapat et al., 2005). In this context it seems that progesterone may antagonize the effect of estrogen on cell proliferation, as a single dose of progesterone given 24 h after estrogen administration diminishes the estrogen-induced increase in cell proliferation in female rats (Tanapat et al., 2005; Liu et al., 2010). However, the interaction of progesterone and estrogen to affect neurogenesis differs from acute to chronic administration. Respectively, acute progesterone increases cell proliferation in the SVZ of OVX rats after 1 h (Bali et al., 2012), whereas chronic progesterone plus or minus estrogen for 21d (1/4mg/kg) one week after OVX has no significant effect on cell proliferation in rats, while decreasing the survival of cells that were built prior to hormonal treatment (Chan et al., 2014). Due to a dearth of studies the importance of progesterone in regulating hippocampal cell proliferation or cell survival in males is largely unknown, but some results in rats pointing towards a stimulating effect on cell proliferation, with no effect seen on survival after a 7 day treatment with progesterone (Barha et al., 2011). Despite this, and like the case for estrogens, the results are largely dependent on the species and timepoint of hormonal treatment relative to BrdU administration. Male mice were shown to display increased levels of surviving cells, when progesterone treatment took place 5–7 days post BrdU, whereas no effect was observed 10–12 or 15–17 days post BrdU (Zhang et al., 2010). In summary, the few studies available to date suggest a sex-difference regarding the effect of progesterone on neurogenesis, with progesterone increasing it in males and decreasing it in females, however there is need for more detailed studies to prove this assumption.
6.4.2. Progestogenic effects on dendritic morphology
The effects of progesterone on dendritic morphology in the hippo-campus are significantly less well studied compared to estrogen. The few available studies suggest a potential involvement of progesterone with initially potentiating the effect of estrogen on spine formation, while observation of later timepoints rather suggests a downregulation in number (Gould et al., 1990; Woolley and McEwen, 1993).
6.5. Androgens
The third group of sex hormones, known for their regulatory function in adult hippocampal neurogenesis are androgens, which particularly play an important role in males and have mostly been studied in males. The most important androgens are testosterone and dihydrotestosterone (DHT). While testosterone is enzymatically derived from androstenedione via 17β-HSD, the pharmacologically more active DHT results from another reductive step out of testosterone via the 5αdehydrogenase; with both acting via the androgen receptor AR (for review see Hajszan et al., 2007).
The most frequently used synthetic androgens used in animal studies are testosterone propionate, androsterone and 3α-androstandione. Studies in animals include surgical intervention (gonadectomy) as well as pharmacological intervention (testosterone/DHT administration). Overall androgens appear to facilitate hippocampal neurogenesis and dendritic spine formation in males, with rather no effect in females.
6.5.1. Androgenic effects on hippocampal neurogenesis
Although GDX does not affect cell proliferation in the male DG, it significantly decreases the number of newly generated neurons that survive to maturity (Spritzer and Galea, 2007; Hamson et al., 2013; Ormerod and Galea, 2003; Spritzer et al., 2011). Interestingly, the timing of castration during development may play a role, as the negative effect on cell survival was only observed when GDX takes place in adulthood (Spritzer and Galea, 2007), whereas GDX prior to puberty rather increased survival of new neurons and enhanced differentiation of cells into neurons (Allen et al., 2014). However caution is required when interpreting these results, as they are highly likely to be influenced by methodological differences i.e. species (i.e. rodents vs. rhesus macaques) and the timing of neurogenesis measurement (i.e. 37d vs. 2y after GDX). There is also conflicting evidence regarding the ability of androgen administration to rescue the GDX-induced deficit in cell survival in male rats, as 30d administration of testosterone or DHT, but only at doses of 0.25 mg/day or higher, was able to rescue cell survival in male rats (Spritzer and Galea, 2007; Hamson et al., 2013), whereas others have shown that testosterone supplementation does not affect survival in GDX males (Spritzer et al., 2011; Buwalda et al., 2010; Carrier and Kabbaj, 2012). Thus, the neurogenesis promoting effect of testosterone may rather be attributed to the necessity of a stable optimal hormone level than high levels of testosterone. Indeed, androgens may also have deleterious effects on hippocampal neurogenesis, as seen after a seven day treatment with finasteride, which lead to a diminished cell proliferation one day post treatment in intact male mice (Romer et al., 2010). Furthermore, the duration of androgen administration is a critical factor in promoting the survival of cells, with short-term treatments (up to 21d) rather decreasing (Spritzer et al., 2011; Carrier and Kabbaj, 2012; Brännvall et al., 2005) and long-term treatments (30–90d) rather increasing (Spritzer and Galea, 2007; Hamson et al., 2013) the number of new neurons in the DG of GDX rats. The effect of testosterone in females is virtually unknown, with only one study directly comparing males and females showing that testosterone increases proliferation in the SVZ of male rats, with no effect in females (Farinetti et al., 2015).
The molecular mechanism behind the effects of testosterone to mediate neuronal survival are most likely regulated via an AR-mediated mechanism, as treatment with high-AR-affinity testosterone metabolite DHT, led to an increase in neurogenesis, while this enhancement was blocked by the AR antagonist flutamide. Moreover, mutations of ARs have been shown to be associated with an inhibition of neurogenesis in male rats (Hamson et al., 2013). Although the expression of ARs has been revealed in the hippocampus of male and female rats, with higher density in males (Xiao and Jordan, 2002), they seem to be absent in the DG of most rodents (see Mahmoud et al., 2016; Galea et al., 2013 for review), suggesting a lack of direct actions of androgens in the DG, but rather a molecular mechanism initiated outside of it. Given the presence of ARs in the CA3 region of the hippocampus and the fact that granule neurons contact thorny excrescences located on CA3 pyramidal neurons, it is likely that ARs in CA3 contribute to the neurogenesis promoting effect of testosterone in the DG. In addition, a retrograde transport survival mechanism that is secreted from CA3 has been suggested (Hatanaka et al., 2009).
6.5.2. Androgenic effects on dendritic morphology
Similar to estrogen, testosterone and its metabolite DHT seem to increase spine density in the CA1 region as revealed in hippocampal slice culture from male rats (Ooishi et al., 2012), with DHT rather increasing the number of large and middle head spines, while testosterone rather increasing the number of small spines, which may have functional consequences as discussed above for estrogen.
6.6. Stress and stress hormones
Although acute and chronic stress are generally known for their negative effect on adult hippocampal neurogenesis, like described for the sex steroids above, this is dependent on the age-, sex-, stressor- and BrdU-timing (Gould et al., 1997; Gould et al., 1998; Pham et al., 2003; Tanapat et al., 2001; Torner et al., 2009; Uno et al., 1989) (and see Abrous et al., 2005; Cameron and Schoenfeld, 2018; Gobinath et al., 2015 for review). In general, there are significant stress-induced sex-differences on diverse phases of hippocampal neurogenesis, which are mainly due to differences in the stress response between males and females, who do not only differ in their stress sensitivity but also with respect to coping mechanism (Dalla et al., 2008; Bowman et al., 2001) and see Goel et al., 2014 for review). Unfortunately, most of the studies available to date only include one sex when assessing the effects of acute or chronic stress exposure on neurogenetic processes like adult hippocampal neurogenesis, dendritic morphology or synaptic plasticity. To examine how stress alters plasticity in the male and female hippo-campus and how these changes in neuroplasticity in fact translate to sex-differences in behavior and susceptibility for mood related-disorders like depression and anxiety, there is certain need for studies investigating sex-differences in those processes by direct comparison of males and females. In the following section, we summarise the current literature about the effects of stress during different developmental windows (i.e. prenatal and early life, adolescence and adulthood) on sex-differences in adult hippocampal neurogenesis and dendritic morphology.
6.6.1. Early-life (adolescent) stress and stress hormones – effects on hippocampal neurogenesis
Prenatal stress (PNS), which is usually achieved by exposure of the dam to stressful physiological or psychological events during different stages of pregnancy has been shown to have wide-ranging consequences on offspring brain development and behavior (for review see Weinstock, 2016). Although there is a general lack of studies assessing the effect of PNS on hippocampal neurogenesis, the few available datasets suggest deleterious effects on hippocampal neurogenesis in juvenile, adolescent and adult rodents (Belnoue et al., 2013; Lemaire et al., 2000; Mandyam et al., 2008; Rayen et al., 2015) and pre-pubertal rhesus monkeys (Coe et al., 2003), with effects being more prominent in the ventral hippocampus (Zuena et al., 2008), which has been linked with stress regulation and anxiety (Fanselow and Dong, 2010). Further complicating an analysis of sex-differences of PNS on hippocampal neurogenesis, there are even fewer studies performed in females. However, it appears that males are more susceptible to perturbations induced by PNS than females. In more detail, RS of the dam 3 times per day for 45 min from gestation day (GD) 15–20 and twice per day on GD21 resulted in a markedly reduced hippocampal cell proliferation and a trend towards a decreased survival in male rats (Rayen et al., 2015), when assessed during adulthood (i.e. on postnatal day (PND) 69–76; 3w post BrdU). The results in females are rather controversial, most likely attributed to the use of different PNS paradigms, with some studies showing no effect on cell proliferation (Zuena et al., 2008) and others showing a similar decrease in cell proliferation (Mandyam et al., 2008) as observed in males. Additionally, the limited nesting and bedding material paradigm was shown to cause a rapid increase in neurogenesis as observed by increased proliferation and differentiation in the DG at PND9 in both sexes. However, when assessed at PND150, both the early life stress paradigm was revealed to cause a long-lasting decrease in DG volume as well as long-term survival of developmentally-born neurons in both sexes. Males were also shown to display reduced neuronal survival, without changes in proliferation or differentiation, whereas females did not display such changes. These changes in survival correlated with impaired learning and memory in males, whereas such impairments were less pronounced in females (Naninck et al., 2014). Taken together, these findings suggests that the paradigm is more suited for use in male rodents than females. While not performed in the same study, social instability stress in adolescence has been shown to differentially affect hippocampal neurogenesis, but not spatial memory in males and females. Thus, exposure to the stressor leads to spatial memory impairments in adulthood in males and females. However, male rats initially (PND 33) displayed increased Ki67 immunoreactive cells and a greater survival, as assessed using DCX as a marker (PND 46), whereas females showed a reduction in both proliferation and survival (McCormick et al., 2012; McCormick et al., 2010). Thus, given the fact that any disturbances of developmental neurogenesis may relate to sex-dependent differences in the outcome of psychiatric disease later in life, there is urgent need for studies directly comparing males and females regarding different stages of hippocampal neurogenesis during different developmental windows after PNS in one experimental outline.
While our understanding of the effects of PNS on hippocampal neurogenesis is rather limited, the effects of early life stress (ELS) are well-studied in both males and females. ELS can be achieved by disturbances of the mother-infant-interaction, which includes maternal deprivation/separation or exposure of the dam to high CORT, thus decreasing maternal care, limited bedding or exposure to psychological stressors like predator odor. Although the exact mechanisms are unknown, adverse early life events seem to negatively affect hippocampal neurogenesis due to a persistent rise of HPA axis activity during a very sensitive developmental window (Sapolsky and Meaney, 1986). The outcome of ELS on sex-differences in hippocampal neurogenesis is dependent on various factors that have to be considered when interpreting the results i.e. stress paradigm/species used in the studies and the timepoint of the assessment of hippocampal neurogenesis after ELS exposure. Thus, while early maternal deprivation for 24 h at PND3 increases proliferation in the DG of PND21 male rats, it decreases it in females (Oomen et al., 2009), whereas maternal separation for longer periods (i.e. 2–6 h/day for up to 21 days) or high maternal exogenous CORT administration (40 mg/kg/day) leads to a decreased cell proliferation in males with no effect in females, when investigated on PND15, PND 22, juvenile age or adulthood (Baek et al., 2011; Baek et al., 2012; Brummelte et al., 2006; Hulshof et al., 2011; Lajud et al., 2012; Mirescu et al., 2004; Oreland et al., 2010; Suri et al., 2013). However, there are other studies in rats and mice revealing an increased level of cell proliferation after similar maternal separation periods or after exposure to limited nesting stress, when examined on PND19, during late postnatal life and young adulthood (Suri et al., 2013; Feng et al., 2014; Naninck et al., 2015). Taken as a whole, these studies indicate that ELS may transiently endow animals with a potential adaptive advantage in a stressful environment, but with long-term deleterious effects across the life span. Moreover, it is likely that the decreased cell proliferation observed in females during this critical period of brain development could be one factor contributing to the increased vulnerability of females to develop psychiatric disorders such as depression in later life. The sex-dependent effects of ELS are not limited to differences in hippocampal cell proliferation, but also include changes in immature neuron production and cell survival. In more detail, maternal separation for 24 h on PND3 increases immature neuron production in male rats, but decreases it in females (Oomen et al., 2009). However, the observed increase in males may be a short-term-restricted adaptive change in response to stress exposure, as it was only observed when immature neuron production was investigated at the time of weaning. In contrast, the number of DCX positive cells in the ventral hippocampus were shown to be significantly diminished after maternal separation when assessed during adulthood, with no effect in females (Oomen et al., 2011; Oomen et al., 2010), suggesting that males express more dynamic, long-lasting changes in hippocampal plasticity in response to ELS. Nevertheless, the effects of ELS on immature neuron production may be largely dependent on the stress protocol and species used, as maternal exposure to high CORT (postpartum day (PPD) 2–23; in rats) or limited nesting and bedding material (PPD2-9; in mice) revealed contrary results, with adult males showing either no effect or an increased density of immature neurons, while in females the opposite picture occurred (Naninck et al., 2014; Gobinath et al., 2016). The effects of ELS on cell survival are rather conflicting and as discussed above, dependent on species, as maternal deprivation, early weaning or limited nesting material diminished the number of BrdU+ and BrdU +/NeuN+ cells in adult male mice, with mostly no effects in females (Naninck et al., 2014; Kikusui et al., 2009; Leslie et al., 2011), whereas maternal separation did significantly affect cell survival in male rats when assessed in adulthood (Hulshof et al., 2011; Mirescu et al., 2004). Overall, it is likely, that the different changes observed after ELS may be an adaptive mechanism to enable the individual to cope with a potential future adversity in life.
PNS and ELS rely on the disturbance of the mother-infant-relationship and stressors during adolescence are mostly effective when a social stress component is used i.e. social isolation stress (Perani and Slattery, 2014; Hillerer et al., 2012). Thus, social isolation for one or three weeks reduces cell proliferation in the rostral hippocampus and reduced the number of immature neurons throughout the hippocampus in adolescent non-human primates (Cinini et al., 2014). Although the latter study did not assess stress-induced sex-differences in hippocampal neurogenesis, studies in rodents suggest, that in contrast to PNS and ELS, which seem to affect males more than females, stress during adolescence induces predominantly deleterious effects in females. Thus, social instability stress (1 h isolation/d and change of cage partner on PND30-45) reduces hippocampal cell proliferation and survival in female rats on PND49 (McCormick et al., 2010), whereas the same stress protocol induced an increase in cell proliferation in males on PND46 and in adulthood (McCormick et al., 2012). Similar sex-dependent effects were observed after chronic restraint stress (RS) (1 h/d from PND30-52), when rats were perfused on PND70 (Barha et al., 2011).
6.6.2. Early-life stress (adolescent) and stress hormones – effects on dendritic morphology
Although less well studied, the available data suggest that acute and/or chronic stress can also affect dendritic morphology and synaptic plasticity of the hippocampus throughout different developmental stages. However, given the overall lack of studies in females regarding this topic, sex-dependent effects are largely unknown, illustrating the need for more research in this field. Similar to the effects of ELS on hippocampal neurogenesis, different stressors during early life have been shown to have deleterious effects on dendritic morphology in different rodent species. Thus, maternal separation or chronic RS results in a diminished dendritic complexity in CA3 pyramidal neurons in mice and rats (Leslie et al., 2011; Galea et al., 1997), with males expressing a reduced number of branching points and dendritic length particularly in apical dendrites, whereas the same effect was more pronounced in basal dendrites in females (Galea et al., 1997).
6.6.3. Stress during adulthood – effect on hippocampal neurogenesis
Given the importance of long-term consequences of acute and chronic stress during adulthood on physiological and mental health, there have been numerous studies dealing with this topic during the last decades. Although these studies have tremendously increased our understanding of stress-induced alterations in hippocampal plasticity in males and females, the various different experimental outlines makes a direct comparison of results virtually impossible. Nevertheless, the following section reviews the available data on sex-dependent effects of acute and chronic stress during adulthood on hippocampal neurogenesis.
The effects of stress on adult hippocampal neurogenesis are largely dependent on the duration of stress exposure as well as the sex of the individual. Although different forms of acute stress may induce one specific picture in males and females, the opposite may occur after chronic stress exposure. Accordingly, acute foot-shock stress (Shors et al., 2007) or predator stress (Falconer and Galea, 2003) was shown to decrease the number of proliferating cells in males and females, whereas chronic exposure to the same paradigms increases the number of BrdU+ cells in females, while not affected proliferation in males (Westenbroek et al., 2004). Although various stress paradigms like acute psychosocial- (social dominance), restraint- or footshock stress did not reveal significant effects of stress on cell proliferation in male rats (Pham et al., 2003; Hanson et al., 2011; Thomas et al., 2007), extended time courses of daily RS, inescapable stress, chronic mild stress (CMS) and social stressors were shown to diminish the rate of proliferation up to 24% (Hillerer et al., 2013; Pham et al., 2003; Shors et al., 2007; Westenbroek et al., 2004; Czeh et al., 2002; Ferragud et al., 2010; Malberg and Duman, 2003) with either no effect on survival (Hillerer et al., 2013; Heine et al., 2004) or a distinct, up to 47% decrease in cell survival (Pham et al., 2003; Lee et al., 2006). While most chronic stress procedures do not appear to effect cell proliferation in female rodents and primates (Hillerer et al., 2013; Uno et al., 1989; Galea et al., 1997; Shors et al., 2007; Westenbroek et al., 2004); there seem to be long-term consequences of chronic stress exposure in females, as cell survival was shown to be either decreased (6/16d post BrdU) (Hillerer et al., 2013; Kuipers et al., 2006) or increased (2w post BrdU) (Westenbroek et al., 2004) after RS/footshock stress in rats.
The observed effects of acute and chronic stress on hippocampal neurogenesis in males and females are predominantly attributed to stress-induced alterations of the HPA axis and the rise in CORT levels. Thus, both acute and chronic exogenous CORT administration have been shown to decrease cell proliferation in both, male and female rodents (mice and rats) (Tanapat et al., 2001; Shors et al., 2007; Falconer and Galea, 2003; Heine et al., 2004; Brummelte and Galea, 2010; Cameron and Gould, 1994; Murray et al., 2008; Yap et al., 2006). However, an increase in CORT is not necessarily associated with changes in cell proliferation as seen after various acute or chronic stress procedures in female rats (Shors et al., 2007; Falconer and Galea, 2003; Westenbroek et al., 2004).
One disadvantage of all of the studies discussed above is that they do not directly assess and compare the effect of the experimental manipulation on male and female hippocampal neurogenesis in the same study. A few years ago, we were the first group to perform a direct and detailed evaluation of sex-dependent and chronic stress-induced changes in adult hippocampal neurogenesis, revealing significant differences between males and females regarding hippocampal cell proliferation, astroglial and neuronal differentiation, cell survival and stem cell quiescence. In more detail, we revealed that chronic RS (2 h/d for 12 days) induced a significant decrease in hippocampal cell proliferation and an increase in stem cell quiescence (BrdU+/SOX2+/PCNA–cells) in male rats with no effect on those parameters in females. Interestingly, chronic stress significantly diminished hippocampal cell survival in females, but not males. Further analysis of astroglial and neuronal differentiation patterns revealed that the reduced cell survival was solely attributed to a profound decrease in the number of cells differentiating into neurons (BrdU+/GFAP-/NeuN+cells) (Hillerer et al., 2013), a result that might be of clinical relevance, given the known increased susceptibility for women to suffer from stress-related disorders like major depression and the fact that there seems to be a link between a reduced neurogenesis and depression.
6.6.4. Stress and stress hormones during adulthood – effects on dendritic morphology
Overall, studies in rodents and non-human primates suggest that stress during adulthood is associated with alterations in dendritic atrophy in CA3 pyramidal neurons in males and females (Uno et al., 1989; Galea et al., 1997; Magarinos and McEwen, 1995). By directly comparing male and female rats in one experimental outline, Shors et al. were able to show that there are not only basal sex differences in hippocampal dendritic morphology, but moreover that phases of acute stress differentially affect males and females with regard to dendritic spine density in the hippocampus. In more detail, females during proestrus had a greater density of spines in the CA1 region of the hippo-campus, when compared to males. Interestingly, spine density was affected in opposite directions after exposure to acute intermittent foot-shock stress, with males showing an increased density of spines, while females expressing reduced levels of spines in the CA1 region of the hippocampus (Shors et al., 2001), suggesting that males and females response to stress is opponent with respect to neural anatomy in the hippocampus, when exposed to the same stressful event. A similar effect was observed in cycling and masculinized females. Here they were able to show that cycling females exposed to acute RS tended to possess fewer spines on apical and basal dendrites in the CA1 area of the hippocampus, whereas the masculinized females possessed significantly more spines after the stressor (Dalla et al., 2009). Although there are some studies suggesting a negative effect of acute stress on dendritic morphology in males (Chen et al., 2008; Sebastian et al., 2013), the bulk of available data suggest that the neuroendocrine background of males may be rather associated with an increase in dendritic complexity after exposure to acute high levels of stress hormones. Thus, a rapid increase in CA1 and CA3 pyramidal spines was also observed in vitro when hippocampal slice cultures from males were exposed to high doses of acute CORT (100, 500 or 1000 nM) or stressful high concentrations of DEX (100 nM) (Komatsuzaki et al., 2005; Yoshiya et al., 2013); an effect that was particularly observed within thin and mushroom spines, but not stubby or filopodium spines (Komatsuzaki et al., 2005). In contrast, chronic exposure to stress hormones induces dendritic atrophy in both sexes. Chronic DEX (0,2mg/kg for 5d) or CORT (10 mg/kg for 21d) administration or exposure to repeated stress (social defeat) were all shown to decrease the number of spines, the number of branching points, reduces the surface area and length of apical and basolateral CA1/3 dendrites in male rats (Kole et al., 2004; McKittrick et al., 2000; Silva-Gomez et al., 2013; Woolley et al., 1990), which was even more pronounced in the ventral than the dorsal hippocampus (Silva-Gomez et al., 2013). Although the effects of stress on hippocampal dendritic morphology occur rapidly, typically within hours, they can be reversible, as they were shown to be abolished 10 days after the cessation of stress and after a 1 month recovery period (Conrad et al., 1999; Sousa et al., 2000).
In summary it seems that chronic stress during adulthood alters hippocampal functioning in a sex-dependent manner with shorter-term alterations in males and long-term consequences in females.
7. Neuroimaging studies on the effects of sex and stress hormones in the human hippocampus
In the following we will present an overview of neuroimaging studies on the human hippocampus, assessing structural aspects (grey matter volumes, grey matter density, microstructural integrity) on the one hand and brain activation (BOLD-response) on the other hand. It is important to point out, that – unlike animal studies – human neuroimaging studies usually capture the whole brain and have thus identified sex differences and hormonal influences on a wide range of areas and brain network properties that are not captured by this review. After summarizing results on sex differences in these aspects, we will focus primarily on the effects of steroid actions on hippocampal volumes and brain activation.
With the emergence of studies investigating short-lived intra-individual changes in gray matter volumes, the question arises, how these changes are to be interpreted. It is yet unclear, whether an increase in grey matter volumes in a certain brain area represents an increase in cell bodies, i.e. neurogenesis, an increase in synaptic spines, i.e. synaptogenesis or an increase in the size of cell bodies, i.e. cell swelling. In animal studies, sex hormone influences have been demonstrated on either of these processes (Barker and Galea, 2008; Fester et al., 2012; Rutkowsky et al., 2011). While the functional implications of an increased number of cells or spines are apparent, it has only recently been demonstrated that even cell swelling has an impact on brain function by influencing the excitability of neurons (Chiang et al., 2019). The BOLD-signal on the other hand has been linked to glutamate signalling (Attwell et al., 2010). In this section, we summarize the small number of studies investigating sex hormone influences on hippocampal volumes and brain activation.
7.1. Sex differences in hippocampal volumes in healthy individuals
The brains of men and women differ in several aspects. Female brains are overall smaller, but show a larger proportion of gray matter than males (Cosgrove et al., 2007). Of greater interest to sex differences in behavior however, are regional gray matter volumes, i.e. sex differences in the size of brain areas proportional to overall brain size. A recent meta-analysis of whole-brain voxel-based morphometry (VBM) analyses revealed larger inferior and middle frontal areas, planum temporale and thalami in women, but larger amygdalae, hippocampi and parahippocamapl gyri, as well as larger cerebelli in men (Ruigrok et al., 2014). However, sex differences in the size of the human hippocampus are disputed as it appears that in younger populations, up until early adolescence hippocampi are larger in girls than in boys (Tan et al., 2016; Gur et al., 2002). This suggests a strong remodelling of male and female hippocampi during puberty due to its activation via sex hormones (Cosgrove et al., 2007). Indeed, some studies report a stronger puberty-related increase in hippocampal volumes in boys compared to girls (Suzuki et al., 2005; Bramen et al., 2011). However, without controlling for pubertal stage, non-significant sex-differences in age-related trajectories (Neufang et al., 2009; Dennison et al., 2013) and stronger age-related increases in girls (Giedd et al., 1997; Giedd et al., 1996) have also been reported. Studies extracting hippocampal volumes directly, rather than testing for differences at the whole-brain level often also report larger hippocampal volumes in women after puberty (Filipek et al., 1994; Garcia-Falgueras et al., 2006; Satterthwaite et al., 2014), although sometimes only in certain hippocampal subfields (Persson et al., 2014; Maller et al., 2006; Riggins et al., 2018; Meyer et al., 2017). However, a recent meta-analysis arrives at the conclusion that irrespective of age, hippocampal volumes do not differ between males and females after correcting for total brain size (Gur et al., 2002; Babson et al., 2017; Perlaki et al., 2014; McHugh et al., 2007; Jancke et al., 2015; Bhatia et al., 1993). In contrast, a large-scale, world-wide, study including almost 16.000 subjects reported slightly larger volumes in males compared to females (Guadalupe et al., 2017).
A variety of methodological issues need to be considered when comparing male and female hippocampi at different age groups: (i) Whole-brain analyses rely on normalization techniques on the one hand and smoothing kernels on the other hand. Normalization fits both male and female individual brains to a standard template. However, given the pronounced differences between male and female brain morphology, it is unclear whether normalization works equally well for all brain structures in men and women. As a subcortical structure, the hippocampus lies in close proximity to the amygdala, the basal ganglia, but also to the parahippocampal gyrus. Unlike for the hippocampus, studies of various methodology consistently show that all these structures are larger in men compared to women, even after correcting for total brain size (Pulopulos et al., 2018; Stephens et al., 2016; Reschke-Hernandez et al., 2017; Kogler et al., 2017; Kirschbaum et al., 1992; Khaksari et al., 2005; Herbison et al., 2016; Childs et al., 2010; Powers et al., 2016; Stroud et al., 2017; Cameron et al., 2017). In whole-brain studies, larger volumes for men compared to women are usually found in large subcortical clusters including the basal ganglia and amygdala and extending to the parahippocampus and cerebellum (Pletzer et al., 2010; Pletzer, 2019). If these clusters also include the hippocampus, it is possible that normalization and smoothing techniques transfer some of the differences in the basal ganglia and amygdala to the hippo-campus. In line with this argument, we recently found sex differences favouring men in the hippocampi at the whole brain level, while in the same sample extracted hippocampal volumes did not differ between men and women (Pletzer, 2019). (ii) Studies extracting hippocampal volumes vary greatly in methodology. While some rely on normalization, others work on individual brain morphology. Furthermore, some assess regional hippocampal volume differences by calculating relative hippocampal volumes via division of absolute volumes by total brain size (Satterthwaite et al., 2014), while others correct for total brain size via covariate analyses. Since, compared to cortical gyri, the hippocampus is rather small relative to total brain size, these methods may give markedly different estimates for regional hippocampal volumes. Especially with regard to developmental trajectories these different statistical procedures may arrive at different results, if cortical areas develop at a different rate than the hippocampus. (iii) Female hippocampal volumes vary with changes in hormonal status, e.g. along the menstrual cycle (see below), due to current or previous hormonal contraceptive use (Pletzer et al., 2010; Pletzer, 2019; Pletzer et al., 2015), due to current or previous pregnancies (Hoekzema et al., 2017) and due to menopause and hormone replacement therapy (see below). Differences in hippocampal volumes between males and females are hard to interpret, if these factors are not adequately controlled for.
7.2. Sex differences in hippocampal activation in healthy individuals
Regarding cognitive processing, sex differences in hippocampal activation appear to be task- and strategy dependent. During spatial navigation and spatial memory, stronger right hippocampal activation was observed in men, while hippocampal activation in women was more left-lateralized (Persson et al., 2013; Frings et al., 2006). Accordingly, a more verbal strategy has been suggested for women, while a more spatial strategy has been suggested for men. However, it has repeatedly demonstrated that women utilize landmark-information in the environment more than men (Harris et al., 2019; Lawton, 2001; Lawton et al., 1996; Lawton, 1994), which may also contribute to sex differences in hippocampal/parahippocampal activation (Sneider et al., 2011).
Vice versa, women show stronger activation of the hippocampus in verbal memory tasks (Jacobs et al., 2016). Women also show stronger hippocampal activation (and limbic activation in general) during working memory tasks (Hill et al., 2014). Accordingly, women recruit the (left) hippocampus more strongly during memory tasks, while men recruit the (right) hippocampus more during spatial tasks.
Regarding emotion processing, women show stronger activation for negatively valenced words, faces and memories compared to men (Young et al., 2013; Victor et al., 2017; Hofer et al., 2007). Stronger activation for emotional words was found in the left hippocampus, while stronger activation for emotional pictures was found in the right hippocampus (Bellace et al., 2013). A meta-analysis of 56 neuroimaging using emotion-eliciting stimuli revealed distinct brain networks for men and women, confirming stronger hippocampal activation in women compared to men (Filkowski et al., 2017). Results regarding sex differences during fear conditioning are inconsistent, with some studies demonstrating stronger hippocampal activation to conditioned stimuli in women (Merz et al., 2010; Benson et al., 2014), other studies report no sex differences in the hippocampus (Merz et al., 2013; Lebron-Milad et al., 2012). In line with the increased BOLD-response during verbal and emotional tasks, women show stronger glutamate concentrations in the hippocampus (Hadel et al., 2013).
Flexibility of dynamic functional connectivity of the hippocampus was lower in women compared to men (Nini et al., 2017). Women show stronger resting state functional connectivity between the amygdala and the hippocampus (Kogler et al., 2016). Increased connectivity between amygdala and hippocampus during emotion processing was related to decreased dysphoric mood in women (Mareckova et al., 2016).
7.3. Estrogens
Estrogen-dependent modulation of hippocampal volumes and activation has mostly been studied in women. Evidence comes from menstrual cycle studies focusing on the pre-ovulatory estradiol peak, studies on post-menopausal or post-partum loss of estradiol and estrogen replacement therapy during menopause, as well as a limited number of studies using an estrogen administration protocol in pre-menopausal women. Studies discussing changes in male-to-female transsexuals will not be discussed here, since they receive both, estrogen and anti-androgen treatment, complicating the interpretation of results (Seiger et al., 2016; Kim and Jeong, 2014).
7.3.1. Estrogenic effects on hippocampal volumes
With one exception (den Heijer et al., 2003), the studies performed to date demonstrate a positive effect of estrogens on hippocampal volumes and differential effects on brain activation during cognitive vs. emotional processing. Along the menstrual cycle, hippocampal grey matter volumes increase during the pre-ovulatory estradiol peak compared to other cycle phases (Protopopescu et al., 2008; Barth et al., 2016; Lisofsky et al., 2015), an effect that has recently been linked to estradiol (Pletzer et al., 2018). Hippocampal gray matter volumes decrease during menopause (Goto et al., 2011), but increase with estrogen only treatment in both peri-/postmenopausal women (Yue et al., 2007; Pintzka and Haberg, 2015; Lord et al., 2010; Lord et al., 2008; Hu et al., 2006; Eberling et al., 2003) (see Maki and Dumas, 2009 for a review) and pre-menopausal women (Albert et al., 2017). Furthermore, long-term use of combined oral contraceptives containing the potent estrogen ethinylestradiol is associated with larger hippocampal GM volumes (Pletzer et al., 2015; Pletzer et al., 2019). Also in males, genotypes associated with higher estradiol levels, showed larger hippocampal gray matter volumes (Bayer et al., 2013), and females with Turner syndrome, who typically show reduced estradiol levels, had smaller hippocampi (Kesler et al., 2004). Furthermore, the fact that up until menopause the age related decline in hippocampal volumes is steeper in men than in women (Raz et al., 2004; Nordin et al., 2017; Malykhin et al., 2017; Li et al., 2014; Golomb et al., 1993) (for review see Murphy et al., 1996) has been attributed to the neuroprotective effects of estradiol (see Pruessner et al., 2008 for a review). In a recent study, circulating estradiol levels were not related to hippocampal volumes in either men or women (Pletzer, 2019), suggesting that changes in estradiol levels are more relevant to hippocampal morphology than absolute levels. However a positive association between left hippocampal volumes and serum estradiol levels was observed in female patients with prolactinomas (Yao et al., 2017).
7.3.2. Estrogenic effects on hippocampal activation
Furthermore, hippocampal activation increases during the preovulatory cycle phase during cognitive tasks (Pletzer et al., 2019; Wei et al., 2018) as well as to food stimuli (Frank et al., 2010), while the BOLD-response to negatively valenced emotional stimuli seems to decrease during this high-estradiol cycle phase (Jacobs et al., 2015; Goldstein et al., 2005; Goldstein et al., 2010; Bayer et al., 2014). The latter finding has been interpreted as a hormonal capacity to regulate stress circuitry (Jacobs et al., 2015). During menopause, estrogen therapy users showed stronger hippocampal activation and better performance during a variety of memory tasks (Maki and Resnick, 2000; Maki et al., 2011; Braden et al., 2017; Berent-Spillson et al., 2010; Shafir et al., 2012) (see Maki and Dumas, 2009 for a review). Likewise, estrogen administration in pre-menopausal women increased hippocampal activation during a recognition memory tasks (Bayer et al., 2018). Furthermore, estrogen receptor agonists increased hippocampal brain activation in schizophrenic patients during an emotion recognition task (Ji et al., 2016), as well during a probabilistic learning task (Kindler et al., 2015), while aromatase inhibitors decreased hippocampal brain activation and memory performance in breast cancer patients (Bayer et al., 2015; Apple et al., 2017). Postmenopausal women show reduced hippocampal activation compared to age-matched pre-menopausal women in a verbal memory task (Jacobs et al., 2016). However, during a working memory task, when the hippo-campus was deactivated, post-menopausal women showed less hippocampal activation than age-matched pre-menopausal women (Shafir et al., 2012; Jacobs et al., 2017).
Finally, some studies have also evaluated the effects of estrogen modulation on hippocampal connectivity, particularly on the connectivity between the hippocampus and pre-frontal cortex. Post-menopausal women, who showed decreased working memory performance, demonstrated increased fronto-hippocampal connectivity compared to age-matched pre-menopausal controls (Jacobs et al., 2017). Results regarding the effects of estrogen-blockade therapy (tamoxifen) on fronto-hippocampal connectivity in breast-cancer survivors are mixed. One study reports increased fronto-hippocampal connectivity after tamoxifen treatment to be related to more cognitive deficits (Apple et al., 2018), while another study reports decreased fronto-hippocampal connectivity during tamoxifen treatment to be related with working memory deficits (Chen et al., 2017). Along the menstrual cycle fronto-hippocampal connectivity changes in a task-dependent manner during the pre-ovulatory phase, but these changes intensify during the luteal phase (Pletzer et al., accepted). Postpartum women, who experience a rapid loss of estradiol, showed a decrease in hippocampal connectivity to the ACC, which was related to decreased memory performance (Shin et al., 2018; Shin et al., 2018). Likewise, hippocampal connectivity to the ACC was decreased in the resting state after treatment with a gonadotropin releasing hormone agonist, resulting in reduced estradiol levels (Fisher et al., 2017). These changes in hippocampal-ACC connectivity were related to depressed mood (Fisher et al., 2017).
7.4. Progestogens
Compared to studies focussing on estrogen-dependent effects on the hippocampus, studies regarding progesterone are rare and yield less consistent results. The reason for this lack of studies may be that in natural designs, progesterone never varies alone, but always in combination with estradiol. Likewise, many hormone replacement therapy regimens, as well as hormonal contraceptives contain progestins in combination with estrogens.
Along the menstrual cycle, both structural and functional studies indicate that progesterone increases during the luteal cycle phase reverse the estradiol-dependent effects during the pre-ovulatory cycle phase (Pletzer et al., 2019; Protopopescu et al., 2008; Barth et al., 2016; Pletzer et al., 2018). Our most recent study indeed demonstrates a significant estradiol-progesterone interaction in the modulation of hippocampal brain activation during cognitive tasks (Pletzer et al., 2019), suggesting positive effects of estradiol only in the absence of progesterone. However, focussing on arousal circuitry, increased hippocampal activation was observed during the luteal cycle phase (Andreano and Cahill, 2010), suggesting that like estradiol, progesterone has opposite effects on hippocampal activation in cognitive and emotional tasks. In a meta-analysis of neuroimaging studies until 2013, Lisofsky and colleagues found larger luteal compared to early follicular hippocampal activations in the left hemisphere, but larger follicular compared to luteal activations in the right hemisphere (Lisofsky et al., 2015). Our own study found a decrease in hippocampal activation after ovulation only in the left, but not in the right hemisphere (Pletzer et al., 2019). Thus, studies suggest differential effects of progesterone on the left and right hippocampus. Combining these seemingly contradicting finding, it is important to note, that the majority of studies included in the Lisofsky meta-analysis focused on emotional stimuli or response inhibition, while our own study focused on cognitive tasks.
As outlined above, menstrual cycle dependent changes in frontohippocampal connectivity intensify during the luteal cycle phase, suggesting a role of progesterone, rather than estradiol, in that respect (Pletzer et al., 2019). During the resting state, progesterone increases fronto-hippocampal connectivity along the menstrual cycle (Arelin et al., 2015).
Similarly, studies focusing on peri-menopausal hormone replacement therapy yield less consistent results than the above reported estrogen replacement therapy studies, when using combined hormone replacement therapy, i.e. HRT containing an estrogen + a progestin (Coker et al., 2010; Hu et al., 2006; Resnick et al., 2009). In fact the majority of these studies demonstrate a reduction in hippocampal volumes in combined HRT users (Coker et al., 2010; Resnick et al., 2009). Studies of progesterone only administration demonstrate larger hippocampal brain activation during face processing (van Wingen et al., 2007) and verbal processing (Berent-Spillson et al., 2015). However, while face recognition performance was reduced, verbal memory performance was improved following progesterone administration.
7.5. Androgens
In an effort to explain the presumed sex differences in hippocampal volumes and activation, studies on the testosterone-dependent modulation of hippocampal volumes and activation have focused on either testosterone increases during puberty, circulating testosterone levels or testosterone administration, and clinical conditions of androgen excess (e.g. congenital adrenal hyperplasia - CAH) or androgen deficiency (e.g. Klinefelter syndrome). The vast majority of these studies report larger hippocampal volumes and activation in higher testosterone stages.
7.5.1. Androgenic effects on hippocampal volume
During puberty, testosterone increases were predictive of increases in hippocampal volumes in both males and females (Neufang et al., 2009; Wierenga et al., 2018). In fact, these studies show that testosterone was a better predictor of hippocampal volumes than pubertal stage or age (Neufang et al., 2009; Wierenga et al., 2018; Herting et al., 2014). Circulating testosterone levels were positively related to hippocampal volumes in young adult women (Pletzer, 2019) and elderly men (Lee et al., 2017), while no association was observed in young adult men (Pletzer, 2019) (Panizzon et al., 2012). However, patients with Klinefelter syndrome (XXY caryotype), who typically show lower circulating testosterone levels, also have lower hippocampal volumes compared to controls (Skakkebaek et al., 2014). Treatment with oxandrolone, a synthetic hormone analog of testosterone, increased hippocampal volumes in Klinefelter patients (Foland-Ross et al., 2019). However, CAH seems to be more strongly associated with larger amygdalae than hippocampi (see Mueller, 2013 for a review).
7.5.2. Androgenic effects on hippocampal activation
In keeping, cerebral blood flow in the hippocampus was related to circulating testosterone levels in elderly men (Moffat and Resnick, 2007). Testosterone administration in healthy young women during their early follicular phase increased hippocampal activation and performance during a navigation task (Pintzka et al., 2016), the recognition of male, but not female faces (van Wingen et al., 2008). A recent meta-analysis focusing on affective stimuli, identified positive effects of both exogenous testosterone administration (to women only) and circulating endogenous testosterone levels on brain activation in bilateral amygdala/parahippocampal regions including the hippocampus (Heany et al., 2016). Specifically, since negative associations of endogenous testosterone levels to parahippocampal/amygdala activations were also observed, this meta-analysis identified the right cornu ammonis (CA) hippocampus as the area, in which activations were positively related to testosterone levels (Sundstrom Poromaa and Gingnell, 2014). Patients with Klinefelter syndrome show reduced hippocampal activation during a word generation task (Steinman et al., 2009) while patients with familial male-precocious puberty (FMPP), which is characterized by early excess testosterone secretion, show increased hippocampal activation to fearful faces (Mueller et al., 2009).
Focussing on hippocampal connectivity, the structural covariance between the hippocampus and the anterior cingulate cortex (ACC) was positively related to testosterone levels in boys, and this covariance in turn related negatively to executive functions (Nguyen et al., 2017).
7.6. Effects of stress and stress hormones on hippocampal volumes and brain activation
Regarding cortisol effects on the hippocampus, an important distinction has to be made between chronically elevated cortisol levels (e.g. chronic stress) and the short-lived cortisol response to an acute stressor. Cortisol is chronically elevated in patients with Cushing’s syndrome (Crapo, 1979) and in patients with stress-related disorders, such as MDD (Travis et al., 2016; Szeszko et al., 2018; Keller et al., 2017; Bremner, 2006). Furthermore, patients with chronic pain, rheumatoid arthroitis, or multiple sclerosis receive hydrocortisone treatments over prolonged time periods (Gold et al., 2010; Coluccia et al., 2008). Studies focusing on chronic stress usually assess diurnal cortisol, e.g. from 24 h urine or repeated salivary sampling, or hair cortisol. Some studies have also assessed the cortisol awakening response (CAR). However, while some studies show an increased CAR in patients under chronic stress (Wust et al., 2000; Schlotz et al., 2004), other studies demonstrate that the cortisol awakening response is blunted in patient groups with chronically elevated cortisol levels (Wessa et al., 2006; Roberts et al., 2004; Duan et al., 2013). The CAR has been described as an indicator of the HPA-axis’ capacity to respond to stressors (Langelaan et al., 2006). Accordingly, the elevated CAR in participants who report chronic stress represents a healthy adaptation to the expected stress during the upcoming day. However, if the chronic stress continues, this adaptive capacity is lost, resulting in disorders like depression, PTSD or chronic fatigue syndrome. Likewise, it appears that chronic stress/chronically elevated cortisol levels affect acute stress reactivity. While some studies show that participants experiencing chronic stress show an increased cortisol response to acute stress, most studies demonstrate blunted cortisol reactivity in patients under chronic stress (Zorn et al., 2017).
7.6.1. Stress effects on gray matter volumes
Chronically elevated cortisol levels have been linked to reduced hippocampal volumes in a variety of studies including different patient groups and healthy subjects of different sex and ages (Travis et al., 2016; Babson et al., 2017; Wolf et al., 2002; Watanabe et al., 2017; Vachon-Presseau et al., 2013; Sudheimer et al., 2014; Starkman et al., 2003; Jin et al., 2016; Davis et al., 2017; Chen et al., 2016; Burkhardt et al., 2015; Brown et al., 2004) and have been linked to declarative memory deficits (Coluccia et al., 2008; Starkman et al., 2003). Likewise, increased diurnal cortisol levels are related to reduced hippocampal activation (Cunningham-Bussel et al., 2009). While it was originally assumed that high cortisol causes the hippocampal volume reduction, newer studies discuss the hippocampus as an important mediator in stress reactivity. Accordingly, participants with smaller hippocampi show reduced cortisol awakening response (Valli et al., 2016; Seo et al., 2019; Pruessner et al., 2007; Dedovic et al., 2010; Buchanan et al., 2004) and reduced stress reactivity of the HPA-axis (Frodl et al., 2014; Valli et al., 2016; Pruessner et al., 2007; Knoops et al., 2010). Likewise, hippocampal damage abolishes the cortisol response to psychosocial stress (Buchanan et al., 2009). Accordingly, smaller hippocampal volume has been discussed as a vulnerability factor for developing stress related disorders like MDD (Dedovic et al., 2010), PTSD (Szeszko et al., 2018) or psychosis (Valli et al., 2016; Collip et al., 2013). Also in patients with head injuries or patients receiving hydrocortisone treatment, smaller hippocampi were related to more depressive symptoms (Gold et al., 2010; Jorge et al., 2007).
Recent results indicate that effects of MDD on the hippocampus are probably explained by childhood adversities (Opel et al., 2014), since differences between MDD patients and controls disappeared if childhood maltreatment was controlled for. Multiple studies indeed show that childhood adversities, ranging from low birth weight or social status to severe maltreatment or abuse, are associated with chronically-elevated cortisol levels (Sheridan et al., 2013; Frodl and O'Keane, 2013; Engert et al., 2010; Dahmen et al., 2018; Bremner, 2005), but lower hippocampal volumes (Dahmen et al., 2018; Samplin et al., 2013; Frodl et al., 2017; Everaerd et al., 2012; Calem et al., 2017). While there seem to be no sex differences in MDD effects on hippocampal volumes, effects of childhood adversities are consistently stronger in males (Samplin et al., 2013; Everaerd et al., 2012; Calem et al., 2017; Helpman et al., 2017). Following the idea that smaller hippocampal volumes are not so much a result of chronically elevated cortisol levels, but represent a vulnerability factor for HPA-axis dysregulation due to severe stress, the fact that pre-adolescent girls have larger hippocampi and a larger cortisol response than boys may contribute to this finding (Whittle et al., 2011). Both factors may protect girls up until adolescence against the adverse effects of childhood maltreatment. In contrast, the smaller increase in hippocampal volumes in girls compared to boys during puberty may represent a vulnerability factor explaining the increasing prevalence rates of stress-related disorders in girls during adolescence (Kessler, 2003).
Relatedly it has been discussed that the age-related decline in hippocampal volumes is related to accumulated effects of glucocorticoids over the life time on the one hand (see Pruessner et al., 2008 for a review) and the fact that cortisol levels increase with age on the other hand (Seeman et al., 1997; Lupien et al., 1998). Indeed hippocampal atrophy was stronger in elderly participants with higher cortisol levels (Valli et al., 2016). However, a series of studies failed to show an associated between age-related volume decline in the hippocampus and increased cortisol levels (Brown et al., 2004; Stomby et al., 2016; Cox et al., 2017; Cox et al., 2015). It is thus again possible, that smaller hippocampal volumes represent a risk factor for cortisol effects during aging.
However, even short term elevations in cortisol due to hydro-cortisone treatment have been associated with a reduction in hippocampal volumes within the same participant (Tessner et al., 2007; Brown et al., 2015). A recent study suggests that both high and low levels of stress-induced cortisol release are associated with smaller hippocampi, suggesting an inverse u-shaped relationship between hippocampal volumes and the cortisol response to stress (Admon et al., 2017).
7.6.2. Stress effects on hippocampal activation
Naturally, while most studies on chronic stress have focused on hippocampal volumes, studies on acute stress have focused on hippocampal brain activation. Short-term elevations in cortisol are induced either by pharmacological treatment (hydrocortisone) or using psycho-physiological stress paradigms. Those paradigms include the Trier social stress test (TSST), which is mostly performed outside the scanner, the Montreal imaging stress task (MIST), which can be performed inside the scanner and the cold pressure stress test (CPT), which may or may not include a social evaluative component (see Noack et al., 2019 for a review of stress paradigms in the scanner). Importantly, a common finding in studies using the TSST or MIST is that only a certain proportion of participants respond to the task with elevated cortisol, while up to 50% do not show elevated cortisol levels and are classified as non-responders. It is possible that chronic stress, as discussed before, contributes to the non-responders blunted cortisol response to stress. This is relevant since associations between cortisol or subjective stress ratings and brain activation or memory performance are (mostly) only observed in group of responders, but not among non-responders (Khalili-Mahani et al., 2010; Dedovic et al., 2009). These studies have assessed (i) brain activity during acutely elevated cortisol either at rest or in some paradigm, or (ii) brain activity in some emotional or cognitive paradigm following acutely elevated cortisol.
Regarding brain activity during acute stress, studies found hippocampal deactivation, which was correlated to the cortisol response (Dedovic et al., 2009; Brown et al., 2013; Klaassen et al., 2013; Kukolja et al., 2011; Liu et al., 2012; Lovallo et al., 2010; Mareckova et al., 2017; Pruessner et al., 2008). A minimum in hippocampal activation was observed after about 30 min of cortisol exposure (Lovallo et al., 2010). Accordingly, even stressors applied before entering the scanner are effective during scanning. This fact has been used to study the effects of cortisol elevation on memory retrieval. Patients who received a hydrocortisone injection or performed the TSST prior to scanning, showed reduced hippocampal activation during a memory retrieval task (Khalili-Mahani et al., 2010; Wolf, 2009; Kukolja et al., 2008). Relatedly, during acutely elevated cortisol, the hippocampus showed stronger connectivity to the default mode network, which usually deactivates during cognitive tasks (Thomason et al., 2013). This reduced hippocampal activation resulted in impaired retrieval performance for contents learned prior to cortisol exposure (Khalili-Mahani et al., 2010; Wolf, 2009; Kukolja et al., 2008). Interestingly, also during the encoding phase, elevated cortisol results in reduced hippocampal activation (Weerda et al., 2010; Vogel et al., 2015; Vogel et al., 2018; van Stegeren et al., 2010; Oei et al., 2007; Merz et al., 2018; Henckens et al., 2009). However, elevated cortisol during learning results in better recall of the learned materials (Wolf, 2009). Recent studies show that this reduced hippocampal activation during memory tasks is accompanied by increased striatal involvement, suggesting a strategy shift from hippocampus dependent declarative strategies to more procedural striatum-dependent strategies during stress (Vogel et al., 2015; Totterman et al., 1989; Schwabe and Wolf, 2012; Schwabe et al., 2013). This shift in learning strategy may explain why acute stress enhances memory performance when applied during encoding, but decreases memory performance when applied during retrieval. In the latter case, cortisol seems to block access to previously formed hippocampus dependent memories, while in the former case cortisol seems to support a strategy of memory formation that is more easily accessible during retrieval.
Note, however, that a series of studies, specifically focussing on emotional material, have also reported a cortisol-dependent increase in hippocampal activation during the encoding phase (van Stegeren, 2009; Bos et al., 2014). These studies suggest that enhanced memory after stress during encoding is related to enhanced hippocampal activation, potentially mediated also via stronger involvement of the amygdala (de Voogd et al., 2017). It is thus possible that hippocampal activation in response to cortisol during encoding follows an inverted u-shaped function, with low and high cortisol causing increased hippocampal activation, while moderate cortisol elevations reduce hippocampal activation.
It is important to point out that – with the exception of MDD samples, which are primarily female – the majority of studies reviewed in this paragraph have been performed on male subjects and that information regarding sex differences or the impact of sex hormones on stress effects on hippocampal activation is limited. While a number of studies have identified sex differences in the cortisol response to psychosocial stress, only few studies have explicitly focused on sex differences in stress effects on the brain. Such studies are however, of uttermost importance to understand sex differences in stress related disorders.
Given the attenuated cortisol response in women, and the fact that reduced hippocampal activation is observed in response to elevated cortisol, one could expect stronger hippocampal activation during psychosocial stress in women. As outlined above, even when no specific stressor is applied, hippocampal activation during verbal and emotional tasks is higher in women compared to men (see above). Assuming that an MRI scan induces a mild stress reaction in scanner naïve participants, it is possible that these sex differences are in part attributable to sex differences in the cortisol response. Data regarding sex differences in hippocampal activation during psychosocial stressors are however limited and inconsistent. While some studies report higher activation in men (Kogler et al., 2017; Seo et al., 2011), others report higher activation in women (Kogler et al., 2015; Abercrombie et al., 2011), particularly women during their luteal cycle phase (Chung et al., 2016; Chung et al., 2016). Furthermore, in women stronger hippocampal activation was related to increased subjective stress ratings (Kogler et al., 2015; Chung et al., 2016). Following the rationale, that hippocampal deactivation during acute stress relates to enhanced memory performance when administered during memory encoding or consolidation, but impaired memory performance when administered during retrieval, it can be predicted that these effects should be abolished in women, due to their reduced cortisol response. While indeed the memory enhancing effects of acute stress during encoding were observed more strongly in men compared to women, the situation is less consistent with regards to memory retrieval (Merz and Wolf, 2017). While there is some evidence to stronger retrieval deficits in men (Hidalgo et al., 2015; Bentz et al., 2013), most studies also observe retrieval deficits in women, but focus on memory for emotional material or fear conditioning (Merz and Wolf, 2017; Quas et al., 2018). Results are mixed suggesting a role of emotional valence (Merz and Wolf, 2017; Quas et al., 2018), but also – most importantly – women’s hormonal status (Maki et al., 2015).
8. Effects of sex and stress hormones on the hippocampus: Comparing animal and human studies
In summary, even though animal and human studies show some differences regarding sex differences in the stress response, there are striking parallels regarding the effects of sex hormones on hippocampal morphology and activity (summarised in Table 1). with the consensus from both animal and human studies, is that sex differences in hippocampal structure and function occur across the lifespan and depend on hormonal modulation and its interactions with stress hormones (stressful events).
Table 1.
Summary of the effects of sex (a)/stress (b) hormones on hippocampal plasticity/morphology and behavior (cognition/mood). M = MALE, F = FEMALE, OVX = ovariectomy, GDX = gonadectomy, T = Testosterone, DHT = Dihydrotestosterone, E = Estrogen, P = Progestogen, ER = estrogen receptor, PR = progesterone receptor, ERT = estrogen replacement therepy, HRT = combined hormone replacement therapy, OC = combined oral contraception; WM = working memory.
a) | Hippocampus | Behavior | |||||||
---|---|---|---|---|---|---|---|---|---|
Neurogenesls (NG) | Dendrltlc Morphology | Gray matter (GM) | Actlvatlon (ACT) | Cognition | Mood | ||||
Sex | Animal studies | Human studies | Animal studies | Human studies | Animal studies | Human studies | |||
ESTROGENS... | ... INCREASE NG IN FEMALES; NO EFFECT IN MALES | ... INCREASE DM IN FEMALES; NO EFFECT IN MALES | ... INCREASE GM IN FEMALES | .... INCREASE ACT. DURING COGNITION DECREASE ACT.DURING EMOTION | ... SEEM TO IMPROVE COGNITION; CYCLE DATA | ... SEEMS TO IMPROVE COGNITION; CYCLE DATA INCONSISTENT | ... SEEM TO BE ANXIOL YTIC AND ANTIDEPRESSIVE | ... SEEM TO BE ANTIDEPRESSIVE | |
high systemic E/E Increase | F |
↑ Cell proliferation
↑Cell survival 14d |
↑mushroom spines
↑density in CA1 |
Pre-ovulatory: ↑ |
↑during cognition
↓during arousal |
↓spatial performance (proestrous)
↑ object memory (proestrous) ↑following OVX |
? |
↓anxiety
↑extinction recall |
|
M | Genetic high E: ↑ | ||||||||
Exogenous E/ER agonist | F | acute: ↑ cell survival
↑cell prolif. lw post OVX ↑ cell prolif. 4w post OVX Chronic: ↑ cell proliferation 15d ↑ cell proliferation 21d |
↑ density in CA1 | ERT: ↑
OC:↑ |
ERT: ↑ during cognition
Agonists: ↑ during cognition |
acute 17β-estradiol ↑memory acute estrone: ↓memory |
ERT: ↑memory
OC: ↑memory |
systemic ↓ anxiety ↓depression local ↓ anxiety |
ERT: ↓ depresion |
M | Acute: ↑ cell survival
↔ cell proliferation Chronic: ↔ cell survival ↔ cell proliferation |
AcuteL ↑ density in CA1 | |||||||
low systemic E/F withdrawal | F | OVX Short term: ↓cell proliferation OVX Long term: ↔ cell proliferation |
OVX: ↓ density in CA1 up to 40d after OVX |
Turner: ↓
Menopause: ↓ |
Menopause: ↓during memory |
OVX: ↓ | Menopause: ↓ memory |
postpartum: ↑depression OVX: ↑ depression |
Postpartum/Menopause/Premenstrual: ↑ depression |
ER antagonist/Aromatase inhibitors | F | ER antagonists: ↔
Synthesis inhibition: ↑ |
↓density in CA1 | ↓ during memory | ↓ memory | ↔ depression | |||
M | ↓ cell proliferation | ↔ density in CA1 | |||||||
PROGESTOGENS... |
... DECREASE NG IN F
... INCREASE NG IN M |
... INITIALLY POTENTIATETHE EFFECT OFE ON DM; DOWNREGULATE AT LATER TIMEPOINTS | .... PROBABLY DECREASE GM IN FEMALES |
.... DECREASE ACT. DURING COGNITION
... INCREASEACT. DURING EMOTION |
... SEEM TO COUNTERACT THE EFFECTS OF OESTROGENS | RESULTS INCONSISTENT | ... SEEM TO BE ANXIOLYTIC AND ANTIDEPRESSIVE | ... SEEM TO BE ANXIOLYTIC | |
High systemic P/P increase | F | ↓ |
↓during cognition
↑during arousal |
? | |||||
Exogenous P/PR agonist | F | Acute: ↓ cell prolif. 24h post E ↑cell prolif. lh post Ponly Chronic: ↔ cell proliferation 21d ↓cell survival 21d |
Combined HRT: ↓ | P only: ↑ | ↑ | ? |
↓anxiety
↑ extinction recall ↓depression |
↓anxiety | |
M |
↑ cell proliferation
↔ cell survival |
↓ depression | |||||||
ANDROGENS... | ... INCREASE NG IN THE LONG TERM IN M; SHORT-TERM EFFECTS AND EFFECTS IN F. UNCLEAR | ... INCREASE DM IN MALES; EFFECT IN FEMALES UNKNOWN | ... INCREASE GM IN MALES AND FEMALES | ... INCREASE ACT. IN MALES AND FEMALES | ... INCREASE SPATIAL COGNITION IN MALES | ... PROBABLY IMPROVE COGNITION AT OPTIMAL LEVELS | ... ACT ANXIOLYTIC AND ANTIDEPRESSIVE | ... PROBABLY ACT ANTIDEPRESSIVE AT OPTIMAL LEVELS | |
Systemic high T/T increase | F | Puberty: ↑
Circulating T: ↑ |
↑ during arousal | ? | |||||
M | Puberty: ↑
Circulating T: ↑ in older men ↔in voung men |
Circulating T: ↑ in older men FMPP: ↑ during face proc. |
? | ↑ depression | |||||
Exogenous T/DHT | F | ↔ cell proliferation in the SVZ |
↑ during cognition
↑ during arousal |
↓ anxiety | ↓ depression | ||||
M | Short-term: ↓ cell survival ↔ cell proliferation Long Term: ↑ cell survival ↑cell proliferation |
↑ density in CA1
↑ small spines (T) ↑ large/middle head spines (DHT) |
↑ | ↑ spatial WW |
↓ anxiety
↓ depression |
↓
depression |
|||
Systemic low T/T loss | M | GDX: ↓ cell survival ↔ cell proliferation |
Klinefelter: ↓ | Klinefelter: ↓ during word gen. |
↓ spatial WW | Aging: ↓ memory |
↑ anxiety
↑ depression |
↑ depression | |
Blockade of DHT synthesis | M | ↓ cell proliferation 7d | |||||||
b) | Pre-/peri-adolescent stress | Post-adolescent stress | |||||||
Neurogenesis (NG) | Dendritic Morphology (DM) | Gray matter (GM) | Neurogenesis (NG) | Dendritic Morphology | Gray matter (GM) | Activation (ACT) | |||
Sex | Animal studies | Human studies | Animal studies | Human studies | |||||
NOTE: LACK OF STUDIES DIRECTLY COMPARING M AND F | ... MOSTLY DECREASE NG IN MALES; RESULTS IN FEMALES INCONSISTENT |
... DECREASE DM IN FEMALES
... ACUTELY INCREASE DM IN MALES ... CHRONICALLY DECREASE DM IN MALES |
... MOSTLY DECREASE NG IN FEMALES- RESULTS IN MALES INCONSISTENT | ... MOSTLY DECREASE DM IN FEMALES; RESULTS IN MALES INCONSISTENT | ... DECREASE GM |
... DECREASE ACT. DURING COGNITION
...INCREASE ACT. DURING EMOTION |
|||
F | Prenatal stress / High maternal CORT |
↓/↔ cell proliferation (depends on PNS model)
↔ cell prolif. PND15/22 juvenile age and adulthood ↓ immature neurons |
Acute Physical: ↓cell proliferation Chronic Physical: ↑ cell proliferation ↑/↓ cell survival 6,16d/2w post BrdU ↔ stem cell quiesc. ↓ neuronal differentiation Chronic Social: ↔ cell proliferation |
Acute: ↓ nb. of apical and basal dendrites in CA1 ↓ density in CA1 |
Acute: ↓ Chronic: ↓ |
Acute: ↓ during encoding of cognitive material ↑ during encoding of emotional material ↓ during retrieval Chronic: ↓ |
|||
Early Life Stress | Acute: ↓ cell prolif. PND21 ↓ immature neurons PND21 ↔ immature neurons adult Chronic: ↔ cell prolif. PND15/22 juvenile and adult ↔ cell survival |
Chronic: ↓ dendritic length and branching points in CA3 (basal dendrites) | Chronic: ↔ | ||||||
Adolescence |
Chronic:
↓ cell prolif. PND49, adult |
Chronic: ↓ | |||||||
M | Prenatal stress / High maternal CORT |
↓ cell prolif. PND 69-76
↓ cell prolif. PND15/22, juvenile and adulthood ↓ cell survival PND 69-76 (after pregn. RS) ↑ immature neuron prod. |
Acute Physical: ↓ cell proliferation Acute Social: ↔ cell proliferation Chronic Physical: ↔ cell proliferation ↔/↓ cell survival ↑ stem cell quiesc. ↔ neuronal differentiation Chronic Social: ↓ cell proliferation |
Acute: ↑ density in CA1 ↑ thin and mushroom spines in CA1/3 Chtronic: ↓dendritic length and branching points in CAl/3 ↓ density in CA1/3 |
|||||
Early Life Stress | Acute: ↑ cell prolif. on PND21
↑ immature neurons PND21 ↓ immature neuron prod. in adulthood Chronic: ↓ cell prolif. PND15/22, juvenile and adult ↑ cell prolif. PND19, adult ↓/↔ cell survival |
Chronic:
↓ dendritic length and branching points in CA3 (apical dendrites) |
Chronic: ↓ | ||||||
Adolescence |
Chronic:
↑ cell prolif. PND46 |
Chronic: ↔ |
8.1. Estrogens
Both preclinical animal and human neuroimaging studies demonstrate positive effects of estrogen on the hippocampus. The estrogendependent increase in neurogenesis and cell survival may play a role in the observations of increased hippocampal gray matter in humans. Likewise the increase in hippocampal spine density observed in animal study, may play a role in findings of increased hippocampal activation in humans. The consistencies regarding the effects of estrogen are most pronounced in females, possibly in part because females have been more extensively assessed in both animal and human studies. While preclinical studies suggest rather no effect of estrogen in females, some human studies show positive effects of estrogen on the hippocampus also in males.
8.2. Progestogens
Similarly, both animal and human studies appear to struggle with disentangling the effects of gestagens on the hippocampus. In both designs, gestagens have hardly been studied in isolation, but rather in combination with estrogens. It appears that some effects of estrogen are even increased due to gestagens, while gestagens counteract the effects of estrogen in other situations. Accordingly, a better understanding of gestagen effects on the hippocampus should be the focus of future studies, particularly to understand the precise dose- and time-dependent mechanisms that may lead to a facilitation or counteraction of estrogen effects. Thereby, it will be important to also take into account the differences in hormonal fluctuations along the very short estrous cycle in animals and the much longer menstrual cycle in humans.
8.3. Androgens
Regarding androgens, an important difference between preclinical and human studies is that preclinical studies have studied androgens mostly in males, while several human studies have studied testosterone effects in females, who usually have low testosterone levels. Human studies in males usually use a correlational design, while animal studies allow for a more causative approach. Furthermore, animal studies have also taken into account the effects of the physiologically more active androgen DHT, while human studies have focussed more strongly on testosterone. A striking parallel between these studies is that most of them report positive effects of androgens on the hippocampus in terms of morphology and activity. While those effects appear to be partially restricted to males in animal studies, human studies also report effects in females – sometimes even exclusively in females. However, these differences may be due to the differences in experimental design between studies.
8.4. Stress and stress hormones
Stress effects on the hippocampus are complex and appear to depend on the timing of stress exposure, as well as the duration of stress exposure. While there are striking consistencies between animal and human studies regarding the effects of estrogens and androgens, some discrepancies emerge regarding stress effects (see Table 1). These discrepancies are likely due to differences in study design. For ethical reasons, effects of stress on pre-pubescent girls and boys have not been studied systematically in humans. While chronic stress can be systematically modelled in animal studies, studies in humans concern mostly clinical populations. Furthermore, animal and human studies differ in the way, acute stress is induced and measured with more immediate approaches in humans, but more delayed measurement of the stress effects in animals. Furthermore, human studies focus more strongly on cortisol increases and subjective stress ratings in their evaluation of the stress response, the latter of which are not available in animal studies. Accordingly, a potential focus of prospective studies in animals could be the more direct pharmacological induction of stress via corticosteroid elevation, while human studies could extend their analyses to the group of endocrinological non-responders.
Despite these methodological differences, some parallels can be observed. In both animal and human studies chronically elevated cortisol levels appear to negatively affect hippocampal morphology. While preclinical studies make a pretty convincing case for these effects to be more severe in males if they appear pre-puberty, but more severe in females if they appear during or after puberty, this dissociation is less clear in humans. However, there is some indication that also in humans, childhood adversities show more severe effects in males, while the prevalence in stress-related disorders rises during adolescence in females. Acute elevations of cortisol have shown mostly negative effects on the hippocampus in animal and human studies, with the exception that hippocampal activation during processing of emotional material appears to be elevated in humans during stress exposure. While animal studies prominently report differential effects of stress on the hippo-campus of males and females, only few studies have focused on sex differences in stress-effects on the hippocampus in humans. However, direct comparisons between males and females are rare in all species. While the problem in animal research is that studies mostly focus on only one sex, the problem in human studies is that many studies employ mixed-sex samples without modelling sex as a variable of interest.
9. Effects of sex steroids on the hippocampus – behavioral implications
After summarizing the effects of sex steroids on hippocampal morphology and activity, the question arises regarding the behavioral implications of these results. As outlined in the introduction the hippo-campus has been linked to spatial abilities and memory, as well as mood disorders like anxiety and depression. In order to provide an idea regarding the behavioral implications of the above-mentioned findings, the following sections shall provide a short overview of sex hormone actions on hippocampus-dependent cognitive functions as well as mood symptoms. However, this summary is by no means exhaustive, since there is a much larger number of behavioral than neurobiological studies. Also, the results of behavioral studies appear to be less consistent, since – especially in humans – behavior is influenced by a multitude of social, biological and psychological factors that are harder to control experimentally.
Regarding hippocampal-dependent cognitive functions, the question arises whether hormones that increase aspects of hippocampal morphology, also lead to improved performance in spatial cognition and memory tasks. Likewise, since many stress-related disorders, particularly mood and anxiety disorders are associated with a reduction in hippocampal volumes and activation, the question arises, whether such hormones can also result in a reduction of depressive symptoms and anxiety. While there are some indications supporting this assumption, it is important to point out that particularly in the human literature, many studies regarding sex hormone actions on cognition report null or opposite findings (see Sundstrom Poromaa and Gingnell, 2014 for review). This problematic may largely arise from methodological issues in both experimental design and hormone assessment. Accordingly, the following paragraphs will focus less on correlational approaches and summarize mostly those studies that implicate a causative role of sex hormones in cognitive functions and mood.
9.1. Estrogens
9.1.1. Estrogenic effects on hippocampus-dependent cognitive functions
Cognitive performance is correlated with the fluctuation in ovarian hormones levels across the oestrous cycle in humans and rodents. In keeping, administration of 17β-estradiol has been shown to have positive effects on cognition, whereas estrone has negative effects on cognition (Barha et al., 2009; Lymer et al., 2017). These effects appear to be mediated via rapid actions of ERs, which are important for memory consolidation (Frick, 2015). Moreover, changes in learning performance across the oestrus cycle may be related to changes in neurogenesis (Hampson, 1990; Warren and Juraska, 1997). Thus, hippocampus-dependent tasks are negatively affected during reproductive phases with high estrogen at pro-oestrous: females in pro-oestrous perform worse in spatial tasks, such as the MWM, compared to oestrous females (Warren and Juraska, 1997; Frye, 1995). These findings may be related to the higher level of stress reactivity that occurs during pro-oestrous (Viau and Meaney, 1991; Conrad et al., 2004). In contrast, pro-oestrous has been shown to be associated with increased object memory (Cordeira et al., 2018) showing that the effects of estrus cycle are complex and perhaps task dependent. This may be in part related to some studies only distinguishing between pro-oestrous/oestrous and diestrus/met-estrus. Indeed, it has been shown that OVX leads to a decrease in cognitive function in young adult humans and rodents in a variety of tasks and oestrogen replacement can reverse these negative effects in a species, dose and temporal manner (Duarte-Guterman et al., 2015) (see Galea et al., 2013 for a review). In humans, effects of estradiol variations along the menstrual cycle on cognition are less clear, with some studies demonstrating positive effects, other studies demonstrating negative effects (see Sundstrom Poromaa and Gingnell, 2014 for review). A recent study suggests positive effects in participants with low baseline dopamine, but negative effects in participants with high baseline dopamine (Jacobs and D'Esposito, 2011). Exogenous estradiol administration to ageing females may decrease the risk of AD, as well as AD-associated cognitive decline (Hogervorst et al., 2005; Hogervorst et al., 2000; Maki, 2013). Likewise, rapid hormone loss during menopause in aging women has been associated with cognitive decline and ERT has been discussed to reverse these effects in a time-sensitive manner (Galea et al., 2017; Duarte-Guterman et al., 2015; Hamson et al., 2016 for review).
9.1.2. Estrogenic effects on mood and anxiety
Rodents typically show decreased anxiety-related behavior during pro-estrus when estrogen levels are high and exogenous administration of estrogen has anxiolytic and antidepressant-like effects in females (Walf and Frye, 2007; Walf and Frye, 2007; Walf and Frye, 2009). Moreover, OVX induces depressive-like symptoms in female rodents and this outcome can be prevented through estrogen replacement (Bekku and Yoshimura, 2005) with similar findings observed following hormone withdrawal after hormone-stimulated pregnancy (Green and Galea, 2008; Craft et al., 2010; Galea et al., 2001; Suda et al., 2008). Additionally, lower estradiol levels have been associated with an increased prevalence of such disorders in humans (Baum and Sex, hormones, 2005; Rosario et al., 2011; Wieck, 2011). In humans, estrogen therapy is used to treat post-partum depression (Moses-Kolko et al., 2009), as well as mood symptoms during menopause (Toffol et al., 2015). Furthermore, oral contraceptives containing the potent estrogen ethinyl-estradiol are used in the treatment of pre-menstrual dysphoric symptoms (for review see Lete and Lapuente, 2016). These effects may be mediated via ER-dependent hippocampal actions, since bilateral injection of 17β-estradiol into the dorsal hippocampus has been shown to be anxiolytic (Walf and Frye, 2006; Walf et al., 2006; Frye and Rhodes, 2002). However, altered hippocampal neurogenesis is not necessarily predictive for the behavioral outcome, as inhibition of estrogen synthesis via chronic letrozole treatment increases the number of immature neurons in the DG without affecting depressive-like behavior in middle-aged female mice (Chaiton et al., 2019).
9.2. Progestogens
9.2.1. Progestogenic effects on hippocampus-dependent cognitive functions
Progestogens, like estrogens, have repeatedly been linked to cognitive functioning with the consensus showing that progesterone improves memory; both object and spatial (see Frick and Kim, 2018 for review). As stated above, OVX leads to spatial memory impairment and this can be reversed by estrogen in combination with progesterone (Harburger et al., 2009; Frye et al., 2007). Interestingly, as with its effects on hippocampal morphology, these effects appear to be age-dependent as the positive effects are lost in aged rats (Bimonte-Nelson et al., 2004).
9.2.2. Progestogenic effects on mood and anxiety
Progesterone has also been implicated in anxiety- and depressive-like behavior in rodents. Thus, it has been shown that exogeneous progesterone administration in female rats facilitates extinction recall to a similar degree as estrogen (Milad et al., 2009). Similar findings have been described for allopregnanolone, the metabolite of progesterone, which also displays anxiolytic properties in a variety of tests, especially following OVX (Milad et al., 2009; Pibiri et al., 2008; Toufexis et al., 2004; Sovijit et al., 2019). Also in humans, the anxiolytic properties of progestogens and the metabolite allopregnanolone have been extensively studied (Le Melledo and Baker, 2004). There are also reports supporting a role for progesterone in depressive-like behavior. Thus, exogenous progesterone administration to progesterone knockout mice resulted in an antidepressant-like effect in the forced swim test independent of sex (Frye, 2011). However, it is clear that more studies are warranted to further assess the role of progestins on behavior.
9.3. Androgens
9.3.1. Androgenic effects on hippocampus-dependent cognitive functions
Androgens have also been shown to influence spatial memory, but only when given for a limited duration. In more detail, short-term testosterone supplementation can influence early (7d), but not late (15d), spatial memory whereas testosterone-induced increase in hippocampal neurogenesis occurs after 30d showing it to be unrelated to its behavioral effect (Spritzer and Galea, 2007). The aforementioned findings appear to be mediated through an AR-dependent mechanism within the CA1 since flutamide has been shown to impair MWM acquisition (Edinger and Frye, 2007; Naghdi et al., 2001). While OVX leads to a decline in cognitive function in women, the loss of androgens may contribute to cognitive decline in older men with AD, since AD has been associated with reduced brain and serum levels of testosterone (Rosario et al., 2011; Moffat et al., 2004); even before diagnosis of AD (Moffat et al., 2004). In keeping, castration reduces accuracy in spatial working memory, whereas testosterone administration improves spatial working memory in rodents (Gibbs and Johnson, 2008; Spritzer et al., 2008). In humans, positive effects of testosterone on spatial cognition and memory have been discussed, but results remain inconsistent with some studies demonstrating effects only in females, other studies demonstrating effects only in males and some studies suggesting an inverse u-shaped relationship of testosterone to cognition (Celec et al., 2015).
9.3.2. Androgenic effects on mood and anxiety
In contrast, there is evidence purporting to a correlation in age-related testosterone decline and major depressive disorder (McIntyre et al., 2006; Shores et al., 2009; Shores et al., 2005; Shores et al., 2004) and testosterone replacement therapy can reverse depressive symptoms in hypogonadal men (Shores et al., 2009; Zarrouf et al., 2009). Moreover, exogenous testosterone administration results in anxiolytic effects in male mice (Aikey et al., 2002), as well as reversing the anxiogenic and pro-depressant-like effects of castration (Carrier and Kabbaj, 2012; Wainwright et al., 2016; Carrier and Kabbaj, 2012). These effects can be blocked by flutamide suggesting that the actions of testosterone are mediated via ARs (Fernandez-Guasti and Martinez-Mota, 2005; Hodosy et al., 2012). In human men, both high and low testosterone levels appear to increase depression rates, while testosterone administration seems to be effective in the treatment of depressive symptoms (Amanatkar et al., 2014). While less studied, there is also evidence that androgen administration may have similar effects in females (Carrier and Kabbaj, 2012; Carrier and Kabbaj, 2012; Buddenberg et al., 2009; Frye and Walf, 2009). Finally, there is clinical evidence as well that testosterone may be effective to reverse depression in premenopausal women and woman with treatment-resistant depression (Goldstat et al., 2003; Miller et al., 2009).
10. Conclusions and outlook
In the present review, we have summarised the role of sex- and stress-hormones on hippocampal morphology and functioning in both preclinical and clinical studies. These studies reveal a strong agreement between animal and human studies regarding the influences of sex hormones on hippocampal morphology and functioning. Indeed, preclinical neurogenesis findings closely resemble those observed regarding hormonal influences on human neuroimaging results regarding grey matter suggesting the former may, at least in part, underlie the latter. Likewise, pre-clinical results regarding dendritic complexity may point to a mechanism underlying the human functional imaging results. While estrogens and androgens appear to exert positive effects on the hippocampus, the effects of gestagens are less well understood and require further investigation. Stress hormones appear to exert negative effects on the hippocampus depending on sex and timing of the stressor. While findings on behavioral implications are less consistent, these hormonal influences on the hippocampus are, at least partially, also reflected in cognitive and emotional changes due to sex and stress-hormone actions. Nevertheless, studies directly linking hormonal influences on the hippocampus to associated behavioral changes are scarce Importantly, the majority of studies have been performed in only one sex, rending sex/gender differences difficult. Accordingly, the effects of estrogen are well studied in females, but not males, while the effects of androgens are well studied in males, but not females. Regarding stress effects on the hippocampus, most studies have been performed in either males or females, with few direct comparisons between the sexes. While the results clearly indicate sex differences in stress effects on the hippocampus, more studies are required to gain further insight into the interactive effects between sex and stress hormones. Due to the opposite effects of estradiol and glucocorticoids on the hippocampus, a protective effect of estradiol against stress effects on the hippocampus has repeatedly been discussed and was recently reviewed (Ycaza Herrera and Mather, 2015). A similar effect of testosterone has also been proposed, but has not been studied extensively (for review see Toufexis et al., 2014). In both cases, timing of estrogen/androgen exposure appears to be critical, since stress also down-regulates the HPG-axis (for review see Ycaza Herrera and Mather, 2015; Toufexis et al., 2014). Importantly, a comparison of stress and sex hormone interactions on hippocampal morphology between clinical and pre-clinical studies depends on the comparability of stress-models between animals and humans. The fact that sex differences in the stress response differ between animal and human models questions their said comparability. One contributing factor may be that rodents and humans differ in respect to the glucocorticoid that mainly carries the stress response (corticosterone in rodents, cortisol in humans). However, the striking parallels observed across species regarding sex hormone actions on the hippocampus, suggest that the discrepancies in stress effects are – at least partly – of a methodological nature. Accordingly, the further development of comparable translational stress and behavioral paradigms that can be used in both animal and human research is an important step towards a comprehensive understanding of potentially protective effects of estrogens and androgens against stress-effects on the brain. Such an understanding is essential for developing sex-specific treatment options against stress-related disorders.
Acknowledgements
This research was in part funded by the Austrian Science Fund (P28261, P32276).
Footnotes
Appendix A: Supplementary material Supplementary data to this article can be found online at https://doi.org/10.1016/j.yfrne.2019.100796.
References
- Abercrombie HC, Jahn AL, Davidson RJ, Kern S, Kirschbaum C, Halverson J. Cortisol's effects on hippocampal activation in depressed patients are related to alterations in memory formation. J Psychiatr Res. 2011;45:15–23. doi: 10.1016/j.jpsychires.2010.10.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Abplanalp JM, Livingston L, Rose RM, Sandwisch D. Cortisol and growth hormone responses to psychological stress during the menstrual cycle. Psychosom Med. 1977;39:158–177. doi: 10.1097/00006842-197705000-00002. [DOI] [PubMed] [Google Scholar]
- Abrous DN, Koehl M, Le Moal M. Adult neurogenesis: from precursors to network and physiology. Physiol Rev. 2005;85:523–569. doi: 10.1152/physrev.00055.2003. [DOI] [PubMed] [Google Scholar]
- Admon R, Treadway MT, Valeri L, Mehta M, Douglas S, Pizzagalli DA. Distinct trajectories of cortisol response to prolonged acute stress are linked to affective responses and hippocampal gray matter volume in healthy females. J Neurosci. 2017;37:7994–8002. doi: 10.1523/JNEUROSCI.1175-17.2017. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ahmed-Leitao F, Spies G, van den Heuvel L, Seedat S. Hippocampal and amygdala volumes in adults with posttraumatic stress disorder secondary to childhood abuse or maltreatment: a systematic review. Psychiatry Res Neuroimag. 2016;256:33–43. doi: 10.1016/j.pscychresns.2016.09.008. [DOI] [PubMed] [Google Scholar]
- Aikey JL, Nyby JG, Anmuth DM, James PJ. Testosterone rapidly reduces anxiety in male house mice (Mus musculus) Horm Behav. 2002;42:448–460. doi: 10.1006/hbeh.2002.1838. [DOI] [PubMed] [Google Scholar]
- Aimone JB, Wiles J, Gage FH. Computational influence of adult neurogenesis on memory encoding. Neuron. 2009;61:187–202. doi: 10.1016/j.neuron.2008.11.026. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Akers KG, Martinez-Canabal A, Restivo L, Yiu AP, De Cristofaro A, Hsiang HL, Wheeler AL, Guskjolen A, Niibori Y, Shoji H, Ohira K, et al. Hippocampal neurogenesis regulates forgetting during adulthood and infancy. Science. 2014;344:598–602. doi: 10.1126/science.1248903. [DOI] [PubMed] [Google Scholar]
- Albert PR. Why is depression more prevalent in women? J Psychiatry Neurosci. 2015;40:219–221. doi: 10.1503/jpn.150205. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Albert K, Hiscox J, Boyd B, Dumas J, Taylor W, Newhouse P. Estrogen enhances hippocampal gray-matter volume in young and older postmenopausal women: a prospective dose-response study. Neurobiol Aging. 2017;56:1–6. doi: 10.1016/j.neurobiolaging.2017.03.033. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Allen KM, Fung SJ, Rothmond DA, Noble PL, Weickert CS. Gonadectomy increases neurogenesis in the male adolescent rhesus macaque hippocampus. Hippocampus. 2014;24:225–238. doi: 10.1002/hipo.22217. [DOI] [PubMed] [Google Scholar]
- Altemus M, Redwine LS, Leong YM, Frye CA, Porges SW, Carter CS. Responses to laboratory psychosocial stress in postpartum women. Psychosom Med. 2001;63:814–821. doi: 10.1097/00006842-200109000-00015. [DOI] [PubMed] [Google Scholar]
- Amanatkar HR, Chibnall JT, Seo BW, Manepalli JN, Grossberg GT. Impact of exogenous testosterone on mood: a systematic review and meta-analysis of randomized placebo-controlled trials. Ann Clin Psychiatry. 2014;26:19–32. [PubMed] [Google Scholar]
- Andreano JM, Cahill L. Sex influences on the neurobiology of learning and memory. Learn Mem. 2009;16:248–266. doi: 10.1101/lm.918309. [DOI] [PubMed] [Google Scholar]
- Andreano JM, Cahill L. Menstrual cycle modulation of medial temporal activity evoked by negative emotion. Neuroimage. 2010;53:1286–1293. doi: 10.1016/j.neuroimage.2010.07.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Angst J, Gamma A, Gastpar M, Lepine JP, Mendlewicz J, Tylee A. Gender differences in depression. Epidemiological findings from the European DEPRES I and II studies. Eur Arch Psychiatry Clin Neurosci. 2002;252:201–209. doi: 10.1007/s00406-002-0381-6. [DOI] [PubMed] [Google Scholar]
- Apple AC, Ryals AJ, Alpert KI, Wagner LI, Shih PA, Dokucu M, Cella D, Penedo FJ, Voss JL, Wang L. Subtle hippocampal deformities in breast cancer survivors with reduced episodic memory and self-reported cognitive concerns. Neuroimage Clin. 2017;14:685–691. doi: 10.1016/j.nicl.2017.03.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Apple AC, Schroeder MP, Ryals AJ, Wagner LI, Cella D, Shih PA, Reilly J, Penedo FJ, Voss JL, Wang L. Hippocampal functional connectivity is related to self-reported cognitive concerns in breast cancer patients undergoing adjuvant therapy. Neuroimage Clin. 2018;20:110–118. doi: 10.1016/j.nicl.2018.07.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Arelin K, Mueller K, Barth C, Rekkas PV, Kratzsch J, Burmann I, Villringer A, Sacher J. Progesterone mediates brain functional connectivity changes during the menstrual cycle-a pilot resting state MRI study. Front Neurosci. 2015;9:44. doi: 10.3389/fnins.2015.00044. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ashby MC, Maier SR, Nishimune A, Henley JM. Lateral diffusion drives constitutive exchange of AMPA receptors at dendritic spines and is regulated by spine morphology. J Neurosci. 2006;26:7046–7055. doi: 10.1523/JNEUROSCI.1235-06.2006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Attwell D, Buchan AM, Charpak S, Lauritzen M, Macvicar BA, Newman EA. Glial and neuronal control of brain blood flow. Nature. 2010;468:232–243. doi: 10.1038/nature09613. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Axelson DA, Doraiswamy PM, McDonald WM, Boyko OB, Tupler LA, Patterson LJ, Nemeroff CB, Ellinwood EH, Jr, Krishnan KR. Hypercortisolemia and hippocampal changes in depression. Psychiatry Res. 1993;47:163–173. doi: 10.1016/0165-1781(93)90046-j. [DOI] [PubMed] [Google Scholar]
- Babson KA, Woodward SH, Schaer M, Sephton SE, Kaloupek DG. Salivary cortisol and regional brain volumes among veterans with and without posttraumatic stress disorder. Biol Psychiatry Cogn Neurosci Neuroimaging. 2017;2:372–379. doi: 10.1016/j.bpsc.2016.11.007. [DOI] [PubMed] [Google Scholar]
- Baek SB, Bahn G, Moon SJ, Lee J, Kim KH, Ko IG, Kim SE, Sung YH, Kim BK, Kim TS, Kim CJ, et al. The phosphodiesterase type-5 inhibitor, tadalafil, improves depressive symptoms, ameliorates memory impairment, as well as suppresses apoptosis and enhances cell proliferation in the hippocampus of maternal-separated rat pups. Neurosci Lett. 2011;488:26–30. doi: 10.1016/j.neulet.2010.10.074. [DOI] [PubMed] [Google Scholar]
- Baek SS, Jun TW, Kim KJ, Shin MS, Kang SY, Kim CJ. Effects of postnatal treadmill exercise on apoptotic neuronal cell death and cell proliferation of maternal-separated rat pups. Brain Dev. 2012;34:45–56. doi: 10.1016/j.braindev.2011.01.011. [DOI] [PubMed] [Google Scholar]
- Bali N, Arimoto JM, Iwata N, Lin SW, Zhao L, Brinton RD, Morgan TE, Finch CE. Differential responses of progesterone receptor membrane component-1 (Pgrmc1) and the classical progesterone receptor (Pgr) to 17beta-estradiol and progesterone in hippocampal subregions that support synaptic remodeling and neurogenesis. Endocrinology. 2012;153:759–769. doi: 10.1210/en.2011-1699. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Banasr M, Hery M, Brezun JM, Daszuta A. Serotonin mediates oestrogen stimulation of cell proliferation in the adult dentate gyrus. Eur J Neurosci. 2001;14:1417–1424. doi: 10.1046/j.0953-816x.2001.01763.x. [DOI] [PubMed] [Google Scholar]
- Barha CK, Lieblich SE, Galea LA. Different forms of oestrogen rapidly upre-gulate cell proliferation in the dentate gyrus of adult female rats. J Neuroendocrinol. 2009;21:155–166. doi: 10.1111/j.1365-2826.2008.01809.x. [DOI] [PubMed] [Google Scholar]
- Barha CK, Brummelte S, Lieblich SE, Galea LA. Chronic restraint stress in adolescence differentially influences hypothalamic-pituitary-adrenal axis function and adult hippocampal neurogenesis in male and female rats. Hippocampus. 2011;21:1216–1227. doi: 10.1002/hipo.20829. [DOI] [PubMed] [Google Scholar]
- Barha CK, Ishrat T, Epp JR, Galea LA, Stein DG. Progesterone treatment normalizes the levels of cell proliferation and cell death in the dentate gyrus of the hippocampus after traumatic brain injury. Exp Neurol. 2011;231:72–81. doi: 10.1016/j.expneurol.2011.05.016. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Barha CK, Galea LA. Motherhood alters the cellular response to estrogens in the hippocampus later in life. Neurobiol Aging. 2011;32:2091–2095. doi: 10.1016/j.neurobiolaging.2009.12.004. [DOI] [PubMed] [Google Scholar]
- Barker JM, Galea LA. Repeated estradiol administration alters different aspects of neurogenesis and cell death in the hippocampus of female, but not male, rats. Neuroscience. 2008;152:888–902. doi: 10.1016/j.neuroscience.2007.10.071. [DOI] [PubMed] [Google Scholar]
- Barnes LL, Wilson RS, Bienias JL, Schneider JA, Evans DA, Bennett DA. Sex differences in the clinical manifestations of Alzheimer disease pathology. Arch Gen Psychiatry. 2005;62:685–691. doi: 10.1001/archpsyc.62.6.685. [DOI] [PubMed] [Google Scholar]
- Bartesaghi R, Guidi S, Severi S, Contestabile A, Ciani E. Sex differences in the hippocampal dentate gyrus of the guinea-pig before puberty. Neuroscience. 2003;121:327–339. doi: 10.1016/s0306-4522(03)00434-2. [DOI] [PubMed] [Google Scholar]
- Barth C, Steele CJ, Mueller K, Rekkas VP, Arelin K, Pampel A, Burmann I, Kratzsch J, Villringer A, Sacher J. In-vivo dynamics of the human hippocampus across the menstrual cycle. Sci Rep. 2016;6 doi: 10.1038/srep32833. 32833. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Baum LW. Sex, hormones, and Alzheimer's disease. J Gerontol Ser A, Biol Sci Med Sci. 2005;60:736–743. doi: 10.1093/gerona/60.6.736. [DOI] [PubMed] [Google Scholar]
- Bayer J, Rune G, Kutsche K, Schwarze U, Kalisch R, Buchel C, Sommer T. Estrogen and the male hippocampus: genetic variation in the aromatase gene predicting serum estrogen is associated with hippocampal gray matter volume in men. Hippocampus. 2013;23:117–121. doi: 10.1002/hipo.22059. [DOI] [PubMed] [Google Scholar]
- Bayer J, Schultz H, Gamer M, Sommer T. Menstrual-cycle dependent fluctuations in ovarian hormones affect emotional memory. Neurobiol Learn Mem. 2014;110:55–63. doi: 10.1016/j.nlm.2014.01.017. [DOI] [PubMed] [Google Scholar]
- Bayer J, Rune G, Schultz H, Tobia MJ, Mebes I, Katzler O, Sommer T. The effect of estrogen synthesis inhibition on hippocampal memory. Psychoneuroendocrinology. 2015;56:213–225. doi: 10.1016/j.psyneuen.2015.03.011. [DOI] [PubMed] [Google Scholar]
- Bayer J, Glascher J, Finsterbusch J, Schulte LH, Sommer T. Linear and inverted U-shaped dose-response functions describe estrogen effects on hippocampal activity in young women. Nat Commun. 2018;9 doi: 10.1038/s41467-018-03679-x. 1220. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Beiko J, Lander R, Hampson E, Boon F, Cain DP. Contribution of sex differences in the acute stress response to sex differences in water maze performance in the rat. Behav Brain Res. 2004;151:239–253. doi: 10.1016/j.bbr.2003.08.019. [DOI] [PubMed] [Google Scholar]
- Beinhoff U, Tumani H, Brettschneider J, Bittner D, Riepe MW. Gender-specificities in Alzheimer's disease and mild cognitive impairment. J Neurol. 2008;255:117–122. doi: 10.1007/s00415-008-0726-9. [DOI] [PubMed] [Google Scholar]
- Bekku N, Yoshimura H. Animal model of menopausal depressive-like state in female mice: prolongation of immobility time in the forced swimming test following ovariectomy. Psychopharmacology. 2005;183:300–307. doi: 10.1007/s00213-005-0179-0. [DOI] [PubMed] [Google Scholar]
- Bellace M, Williams JM, Mohamed FB, Faro SH. An fMRI study of the activation of the hippocampus by emotional memory. Int J Neurosci. 2013;123:121–127. doi: 10.3109/00207454.2012.742894. [DOI] [PubMed] [Google Scholar]
- Belnoue L, Grosjean N, Ladeveze E, Abrous DN, Koehl M. Prenatal stress inhibits hippocampal neurogenesis but spares olfactory bulb neurogenesis. PLoS ONE. 2013;8:e72972. doi: 10.1371/journal.pone.0072972. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Benson S, Kattoor J, Kullmann JS, Hofmann S, Engler H, Forsting M, Gizewski ER, Elsenbruch S. Towards understanding sex differences in visceral pain: enhanced reactivation of classically-conditioned fear in healthy women. Neurobiol Learn Mem. 2014;109:113–121. doi: 10.1016/j.nlm.2013.12.014. [DOI] [PubMed] [Google Scholar]
- Bentz D, Michael T, Wilhelm FH, Hartmann FR, Kunz S, von Rohr IR, de Quervain DJ. Influence of stress on fear memory processes in an aversive differential conditioning paradigm in humans. Psychoneuroendocrinology. 2013;38:1186–1197. doi: 10.1016/j.psyneuen.2012.12.018. [DOI] [PubMed] [Google Scholar]
- Berent-Spillson A, Persad CC, Love T, Tkaczyk A, Wang H, Reame NK, Frey KA, Zubieta JK, Smith YR. Early menopausal hormone use influences brain regions used for visual working memory. Menopause. 2010;17:692–699. doi: 10.1097/gme.0b013e3181cc49e9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Berent-Spillson A, Briceno E, Pinsky A, Simmen A, Persad CC, Zubieta JK, Smith YR. Distinct cognitive effects of estrogen and progesterone in menopausal women. Psychoneuroendocrinology. 2015;59:25–36. doi: 10.1016/j.psyneuen.2015.04.020. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bettis T, Jacobs LF. Sex differences in object recognition are modulated by object similarity. Behav Brain Res. 2012;233:288–292. doi: 10.1016/j.bbr.2012.04.028. [DOI] [PubMed] [Google Scholar]
- Beylin AV, Gandhi CC, Wood GE, Talk AC, Matzel LD, Shors TJ. The role of the hippocampus in trace conditioning: temporal discontinuity or task difficulty? Neurobiol. Learn Mem. 2001;76:447–461. doi: 10.1006/nlme.2001.4039. [DOI] [PubMed] [Google Scholar]
- Bhatia S, Bookheimer SY, Gaillard WD, Theodore WH. Measurement of whole temporal lobe and hippocampus for MR volumetry: normative data. Neurology. 1993;43:2006–2010. doi: 10.1212/wnl.43.10.2006. [DOI] [PubMed] [Google Scholar]
- Bimonte-Nelson HA, Singleton RS, Williams BJ, Granholm AC. Ovarian hormones and cognition in the aged female rat: II. progesterone supplementation reverses the cognitive enhancing effects of ovariectomy. Behav Neurosci. 2004;118:707–714. doi: 10.1037/0735-7044.118.4.707. [DOI] [PubMed] [Google Scholar]
- Boldrini M, Underwood MD, Hen R, Rosoklija GB, Dwork AJ, John Mann J, Arango V. Antidepressants increase neural progenitor cells in the human hippocampus. Neuropsychopharm: Off Publ Am College Neuropsychopharm. 2009;34:2376–2389. doi: 10.1038/npp.2009.75. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Boldrini M, Hen R, Underwood MD, Rosoklija GB, Dwork AJ, Mann JJ, Arango V. Hippocampal angiogenesis and progenitor cell proliferation are increased with antidepressant use in major depression. Biol Psychiatry. 2012;72:562–571. doi: 10.1016/j.biopsych.2012.04.024. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bos PA, Montoya ER, Terburg D, van Honk J. Cortisol administration increases hippocampal activation to infant crying in males depending on childhood neglect. Hum Brain Mapp. 2014;35:5116–5126. doi: 10.1002/hbm.22537. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bouma EM, Riese H, Ormel J, Verhulst FC, Oldehinkel AJ. Adolescents' cortisol responses to awakening and social stress; effects of gender, menstrual phase and oral contraceptives. The TRAILS study Psychoneuroendocrinology. 2009;34:884–893. doi: 10.1016/j.psyneuen.2009.01.003. [DOI] [PubMed] [Google Scholar]
- Bourne J, Harris KM. Do thin spines learn to be mushroom spines that remember? Curr Opin Neurobiol. 2007;17:381–386. doi: 10.1016/j.conb.2007.04.009. [DOI] [PubMed] [Google Scholar]
- Bowers JM, Waddell J, McCarthy MM. A developmental sex difference in hippocampal neurogenesis is mediated by endogenous oestradiol. Biol Sex Differ. 2010;1:8. doi: 10.1186/2042-6410-1-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bowman RE, Zrull MC, Luine VN. Chronic restraint stress enhances radial arm maze performance in female rats. Brain Res. 2001;904:279–289. doi: 10.1016/s0006-8993(01)02474-x. [DOI] [PubMed] [Google Scholar]
- Braden BB, Dassel KB, Bimonte-Nelson HA, O'Rourke HP, Connor DJ, Moorhous S, Sabbagh MN, Caselli RJ, Baxter LC. Sex and post-menopause hormone therapy effects on hippocampal volume and verbal memory. Neuropsychol Dev Cogn B Aging Neuropsychol Cogn. 2017;24:227–246. doi: 10.1080/13825585.2016.1182962. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Brailoiu E, Dun SL, Brailoiu GC, Mizuo K, Sklar LA, Oprea TI, Prossnitz ER, Dun NJ. Distribution and characterization of estrogen receptor G protein- coupled receptor 30 in the rat central nervous system. J Endocrinol. 2007;193:311–321. doi: 10.1677/JOE-07-0017. [DOI] [PubMed] [Google Scholar]
- Bramen JE, Hranilovich JA, Dahl RE, Forbes EE, Chen J, Toga AW, Dinov ID, Worthman CM, Sowell ER. Puberty influences medial temporal lobe and cortical gray matter maturation differently in boys than girls matched for sexual maturity. Cereb Cortex. 2011;21:636–646. doi: 10.1093/cercor/bhq137. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Brandt N, Vierk R, Rune GM. Sexual dimorphism in estrogen-induced synaptogenesis in the adult hippocampus. Int J Dev Biol. 2013;57:351–356. doi: 10.1387/ijdb.120217gr. [DOI] [PubMed] [Google Scholar]
- Brännvall K, Bogdanovic N, Korhonen L, Lindholm D. 19-Nortestosterone influences neural stem cell proliferation and neurogenesis in the rat brain. Eur J Neurosci. 2005;21:871–878. doi: 10.1111/j.1460-9568.2005.03942.x. [DOI] [PubMed] [Google Scholar]
- Bremner JD. Effects of traumatic stress on brain structure and function: relevance to early responses to trauma. J Trauma Dissoc. 2005;6:51–68. doi: 10.1300/J229v06n02_06. [DOI] [PubMed] [Google Scholar]
- Bremner JD. Traumatic stress: effects on the brain. Dialogues Clin Neurosci. 2006;8:445–461. doi: 10.31887/DCNS.2006.8.4/jbremner. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Breslau N, Schultz L, Peterson E. Sex differences in depression: a role for preexisting anxiety. Psychiatry Res. 1995;58:1–12. doi: 10.1016/0165-1781(95)02765-o. [DOI] [PubMed] [Google Scholar]
- Brinton RD, Thompson RF, Foy MR, Baudry M, Wang J, Finch CE, Morgan TE, Pike CJ, Mack WJ, Stanczyk FZ, Nilsen J. Progesterone receptors: form and function in brain. Front Neuroendocrinol. 2008;29:313–339. doi: 10.1016/j.yfrne.2008.02.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Brown ES, Woolston DJ, Frol A, Bobadilla L, Khan DA, Hanczyc M, Rush AJ, Fleckenstein J, Babcock E, Cullum CM. Hippocampal volume, spectroscopy, cognition, and mood in patients receiving corticosteroid therapy. Biol Psychiatry. 2004;55(5):538–545. doi: 10.1016/j.biopsych.2003.09.010. [DOI] [PubMed] [Google Scholar]
- Brown ES, Lu H, Denniston D, Uh J, Thomas BP, Carmody TJ, Auchus RJ, Diaz-Arrastia R, Tamminga C. A randomized, placebo-controlled proof-of-concept, crossover trial of phenytoin for hydrocortisone-induced declarative memory changes. J Affect Disord. 2013;150:551–558. doi: 10.1016/j.jad.2013.01.038. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Brown ES, Jeon-Slaughter H, Lu H, Jamadar R, Issac S, Shad M, Denniston D, Tamminga C, Nakamura A, Thomas BP. Hippocampal volume in healthy controls given 3-day stress doses of hydrocortisone. Neuropsychopharmacology. 2015;40:1216–1221. doi: 10.1038/npp.2014.307. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Brummelte S, Galea LA. Chronic high corticosterone reduces neurogenesis in the dentate gyrus of adult male and female rats. Neuroscience. 2010;168:680–690. doi: 10.1016/j.neuroscience.2010.04.023. [DOI] [PubMed] [Google Scholar]
- Brummelte S, Pawluski JL, Galea LA. High post-partum levels of corticosterone given to dams influence postnatal hippocampal cell proliferation and behavior of offspring: a model of post-partum stress and possible depression. Horm Behav. 2006;50:370–382. doi: 10.1016/j.yhbeh.2006.04.008. [DOI] [PubMed] [Google Scholar]
- Buchanan TW, Kern S, Allen JS, Tranel D, Kirschbaum C. Circadian regulation of cortisol after hippocampal damage in humans. Biol Psychiatry. 2004;56:651–656. doi: 10.1016/j.biopsych.2004.08.014. [DOI] [PubMed] [Google Scholar]
- Buchanan TW, Tranel D, Kirschbaum C. Hippocampal damage abolishes the cortisol response to psychosocial stress in humans. Horm Behav. 2009;56:44–50. doi: 10.1016/j.yhbeh.2009.02.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Buddenberg TE, Komorowski M, Ruocco LA, Silva MA, Topic B. Attenuating effects of testosterone on depressive-like behavior in the forced swim test in healthy male rats. Brain Res Bull. 2009;79:182–186. doi: 10.1016/j.brainresbull.2009.02.008. [DOI] [PubMed] [Google Scholar]
- Burkhardt T, Ludecke D, Spies L, Wittmann L, Westphal M, Flitsch J. Hippocampal and cerebellar atrophy in patients with Cushing's disease. Neurosurg Focus. 2015;39:E5. doi: 10.3171/2015.8.FOCUS15324. [DOI] [PubMed] [Google Scholar]
- Butz M, Lehmann K, Dammasch IE, Teuchert-Noodt G. A theoretical network model to analyse neurogenesis and synaptogenesis in the dentate gyrus. Neural Netw : Off J Int Neural Netw Soc. 2006;19:1490–1505. doi: 10.1016/j.neunet.2006.07.007. [DOI] [PubMed] [Google Scholar]
- Buwalda B, van der Borght K, Koolhaas JM, McEwen BS. Testosterone decrease does not play a major role in the suppression ofhippocampal cell proliferation following social defeat stress in rats. Physiol Behav. 2010;101:719–725. doi: 10.1016/j.physbeh.2010.08.010. [DOI] [PubMed] [Google Scholar]
- Cahill L. Why sex matters for neuroscience. Nat Rev Neurosci. 2006;7:477–484. doi: 10.1038/nrn1909. [DOI] [PubMed] [Google Scholar]
- Calem M, Bromis K, McGuire P, Morgan C, Kempton MJ. Meta-analysis of associations between childhood adversity and hippocampus and amygdala volume in non-clinical and general population samples. Neuroimage Clin. 2017;14:471–479. doi: 10.1016/j.nicl.2017.02.016. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Cameron HA, Gould E. Adult neurogenesis is regulated by adrenal steroids in the dentate gyrus. Neuroscience. 1994;61:203–209. doi: 10.1016/0306-4522(94)90224-0. [DOI] [PubMed] [Google Scholar]
- Cameron CA, McKay S, Susman EJ, Wynne-Edwards K, Wright JM, Weinberg J. Cortisol stress response variability in early adolescence: attachment, affect and sex. J Youth Adolesc. 2017;46:104–120. doi: 10.1007/s10964-016-0548-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Cameron HA, Schoenfeld TJ. Behavioral and structural adaptations to stress. Front Neuroendocrinol. 2018 doi: 10.1016/j.yfrne.2018.02.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Capitani E, Laiacona M, Basso A. Phonetically cued word-fluency, gender differences and aging: a reappraisal. Cortex. 1998;34:779–783. doi: 10.1016/s0010-9452(08)70781-0. [DOI] [PubMed] [Google Scholar]
- Capitani E, Barbarotto R, Laiacona M. Gender differences and the brain representation of semantic knowledge. Brain Lang. 2005;95:56–57. [Google Scholar]
- Carey MP, Deterd CH, de Koning J, Helmerhorst F, de Kloet ER. The influence of ovarian steroids on hypothalamic-pituitary-adrenal regulation in the female rat. J Endocrinol. 1995;144:311–321. doi: 10.1677/joe.0.1440311. [DOI] [PubMed] [Google Scholar]
- Carrier N, Kabbaj M. Testosterone and imipramine have antidepressant effects in socially isolated male but not female rats. Horm Behav. 2012;61:678–685. doi: 10.1016/j.yhbeh.2012.03.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Carrier N, Kabbaj M. Extracellular signal-regulated kinase 2 signaling in the hippocampal dentate gyrus mediates the antidepressant effects of testosterone. Biol Psychiatry. 2012;71:642–651. doi: 10.1016/j.biopsych.2011.11.028. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Celec P, Ostatnikova D, Hodosy J. On the effects of testosterone on brain behavioral functions. Front Neurosci. 2015;9:12. doi: 10.3389/fnins.2015.00012. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Chaiton JA, Wong SJ, Galea LA. Chronic aromatase inhibition increases ventral hippocampal neurogenesis in middle-aged female mice. Psychoneuroendocrinology. 2019;106:111–116. doi: 10.1016/j.psyneuen.2019.04.003. [DOI] [PubMed] [Google Scholar]
- Chan M, Chow C, Hamson DK, Lieblich SE, Galea LA. Effects of chronic oestradiol, progesterone and medroxyprogesterone acetate on hippocampal neuro-genesis and adrenal mass in adult female rats. J Neuroendocrinol. 2014;26:386–399. doi: 10.1111/jne.12159. [DOI] [PubMed] [Google Scholar]
- Chen Y, Dube CM, Rice CJ, Baram TZ. Rapid loss of dendritic spines after stress involves derangement ofspine dynamics by corticotropin-releasing hormone. J Neurosci. 2008;28:2903–2911. doi: 10.1523/JNEUROSCI.0225-08.2008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Chen X, He X, Tao L, Li J, Wu J, Zhu C, Yu F, Zhang L, Zhang J, Qiu B, Yu Y, et al. The working memory and dorsolateral prefrontal-hippocampal functional connectivity changes in long-term survival breast cancer patients treated with tamoxifen. Int J Neuropsychopharmacol. 2017;20:374–382. doi: 10.1093/ijnp/pyx008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Chen R, Muetzel RL, El Marroun H, Noppe G, van Rossum EF, Jaddoe VW, Verhulst FC, White T, Fang F, Tiemeier H. No association between hair cortisol or cortisone and brain morphology in children. Psychoneuroendocrinology. 2016;74:101–110. doi: 10.1016/j.psyneuen.2016.08.023. [DOI] [PubMed] [Google Scholar]
- Cherney ID, Brabec CM, Runco DV. Mapping out spatial ability: sex differences in way-finding navigation. Percept Mot Skills. 2008;107:747–760. doi: 10.2466/pms.107.3.747-760. [DOI] [PubMed] [Google Scholar]
- Chiang CC, Shivacharan RS, Wei X, Gonzalez-Reyes LE, Durand DM. Slow periodic activity in the longitudinal hippocampal slice can self-propagate non-sy-naptically by a mechanism consistent with ephaptic coupling. J Physiol. 2019;597:249–269. doi: 10.1113/JP276904. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Childs E, Dlugos A, De Wit H. Cardiovascular, hormonal, and emotional responses to the TSST in relation to sex and menstrual cycle phase. Psychophysiology. 2010;47:550–559. doi: 10.1111/j.1469-8986.2009.00961.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Chow C, Epp JR, Lieblich SE, Barha CK, Galea LA. Sex differences in neurogenesis and activation of new neurons in response to spatial learning and memory. Psychoneuroendocrinology. 2013;38:1236–1250. doi: 10.1016/j.psyneuen.2012.11.007. [DOI] [PubMed] [Google Scholar]
- Chung KC, Peisen F, Kogler L, Radke S, Turetsky B, Freiherr J, Derntl B. The influence of menstrual cycle and androstadienone on female stress reactions: an fMRI study. Front Hum Neurosci. 2016;10:44. doi: 10.3389/fnhum.2016.00044. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Chung KC, Peisen F, Kogler L, Radke S, Turetsky B, Freiherr J, Derntl B. Corrigendum: The influence of menstrual cycle and androstadienone on female stress reactions: an fMRI study. Front Hum Neurosci. 2016;10:293. doi: 10.3389/fnhum.2016.00293. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Cinini SM, Barnabe GF, Galvao-Coelho N, de Medeiros MA, Perez-Mendes P, Sousa MB, Covolan L, Mello LE. Social isolation disrupts hippocampal neurogenesis in young non-human primates. Front Neurosci. 2014;8:45. doi: 10.3389/fnins.2014.00045. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Clelland CD, Choi M, Romberg C, Clemenson GD, Jr, Fragniere A, Tyers P, Jessberger S, Saksida LM, Barker RA, Gage FH, Bussey TJ. A functional role for adult hippocampal neurogenesis in spatial pattern separation. Science. 2009;325:210–213. doi: 10.1126/science.1173215. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Coe CL, Kramer M, Czeh B, Gould E, Reeves AJ, Kirschbaum C, Fuchs E. Prenatal stress diminishes neurogenesis in the dentate gyrus of juvenile rhesus monkeys. Biol Psychiatry. 2003;54:1025–1034. doi: 10.1016/s0006-3223(03)00698-x. [DOI] [PubMed] [Google Scholar]
- Coker LH, Espeland MA, Rapp SR, Legault C, Resnick SM, Hogan P, Gaussoin S, Dailey M, Shumaker SA. Postmenopausal hormone therapy and cognitive outcomes: the Women's Health Initiative Memory Study (WHIMS) J Steroid Biochem Mol Biol. 2010;118:304–310. doi: 10.1016/j.jsbmb.2009.11.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Colle R, Chupin M, Cury C, Vandendrie C, Gressier F, Hardy P, Falissard B, Colliot O, Ducreux D, Corruble E. Depressed suicide attempters have smaller hippocampus than depressed patients without suicide attempts. J Psychiatr Res. 2015;61:13–18. doi: 10.1016/j.jpsychires.2014.12.010. [DOI] [PubMed] [Google Scholar]
- Collip D, Habets P, Marcelis M, Gronenschild E, Lataster T, Lardinois M, Nicolson NA, Myin-Germeys I. Hippocampal volume as marker of daily life stress sensitivity in psychosis. Psychol Med. 2013;43:1377–1387. doi: 10.1017/S003329171200219X. [DOI] [PubMed] [Google Scholar]
- Coluccia D, Wolf OT, Kollias S, Roozendaal B, Forster A, de Quervain DJ. Glucocorticoid therapy-induced memory deficits: acute versus chronic effects. J Neurosci. 2008;28:3474–3478. doi: 10.1523/JNEUROSCI.4893-07.2008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Conrad CD, LeDoux JE, Magarinos AM, McEwen BS. Repeated restraint stress facilitates fear conditioning independently of causing hippocampal CA3 dendritic atrophy. Behav Neurosci. 1999;113:902–913. doi: 10.1037//0735-7044.113.5.902. [DOI] [PubMed] [Google Scholar]
- Conrad CD, Jackson JL, Wieczorek L, Baran SE, Harman JS, Wright RL, Korol DL. Acute stress impairs spatial memory in male but not female rats: influence of estrous cycle. Pharmacol Biochem Behav. 2004;78:569–579. doi: 10.1016/j.pbb.2004.04.025. [DOI] [PubMed] [Google Scholar]
- Cordeira J, Kolluru SS, Rosenblatt H, Kry J, Strecker RE, McCarley RW. Learning and memory are impaired in the object recognition task during metestrus/diestrus and after sleep deprivation. Behav Brain Res. 2018;339:124–129. doi: 10.1016/j.bbr.2017.11.033. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Corkin S. What's new with the amnesic patient H.M.? Nat Rev Neurosci. 2002;3:153–160. doi: 10.1038/nrn726. [DOI] [PubMed] [Google Scholar]
- Cosgrove KP, Mazure CM, Staley JK. Evolving knowledge of sex differences in brain structure, function, and chemistry. Biol Psychiatry. 2007;62:847–855. doi: 10.1016/j.biopsych.2007.03.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Cox SR, MacPherson SE, Ferguson KJ, Royle NA, Maniega SM, Hernandez Mdel C, Bastin ME, MacLullich AM, Wardlaw JM, Deary IJ. Does white matter structure or hippocampal volume mediate associations between cortisol and cognitive ageing? Psychoneuroendocrinology. 2015;62:129–137. doi: 10.1016/j.psyneuen.2015.08.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Cox SR, Valdes Hernandez MDC, Kim J, Royle NA, MacPherson SE, Ferguson KJ, Munoz Maniega S, Anblagan D, Aribisala BS, Bastin ME, Park J, Starr JM, Deary IJ, MacLullich AMJ, Wardlaw JM. Associations between hippocampal morphology, diffusion characteristics, and salivary cortisol in older men. Psychoneuroendocrinology. 2017;78:151–158. doi: 10.1016/j.psyneuen.2017.01.027. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Craft RM, Kostick ML, Rogers JA, White CL, Tsutsui KT. Forced swim test behavior in postpartum rats. Pharmacol Biochem Behav. 2010;96:402–412. doi: 10.1016/j.pbb.2010.06.012. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Crapo L. Cushing's syndrome: a review of diagnostic tests. Metabolism. 1979;28:955–977. doi: 10.1016/0026-0495(79)90097-0. [DOI] [PubMed] [Google Scholar]
- Creer DJ, Romberg C, Saksida LM, van Praag H, Bussey TJ. Running enhances spatial pattern separation in mice. PNAS. 2010;107:2367–2372. doi: 10.1073/pnas.0911725107. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Cunningham-Bussel AC, Root JC, Butler T, Tuescher O, Pan H, Epstein J, Weisholtz DS, Pavony M, Silverman ME, Goldstein MS, Altemus M, Cloitre M, Ledoux J, McEwen B, Stern E, Silbersweig D. Diurnal cortisol amplitude and fronto-limbic activity in response to stressful stimuli. Psychoneuroendocrinology. 2009;34:694–704. doi: 10.1016/j.psyneuen.2008.11.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Czeh B, Welt T, Fischer AK, Erhardt A, Schmitt W, Muller MB, Toschi N, Fuchs E, Keck ME. Chronic psychosocial stress and concomitant repetitive transcranial magnetic stimulation: effects on stress hormone levels and adult hippocampal neurogenesis. Biol Psychiatry. 2002;52:1057–1065. doi: 10.1016/s0006-3223(02)01457-9. [DOI] [PubMed] [Google Scholar]
- Dabbs JM, Chang EL, Strong RA, Milun R. Spatial ability, navigation strategy, and geographic knowledge among men and women. Evol Hum Behav. 1998;19:89–98. [Google Scholar]
- Dahmen B, Puetz VB, Scharke W, von Polier GG, Herpertz-Dahlmann B, Konrad K. Effects of early-life adversity on hippocampal structures and associated HPA axis functions. Dev Neurosci. 2018;40:13–22. doi: 10.1159/000484238. [DOI] [PubMed] [Google Scholar]
- Dalla C, Antoniou K, Drossopoulou G, Xagoraris M, Kokras N, Sfikakis A, Papadopoulou-Daifoti Z. Chronic mild stress impact: are females more vulnerable? Neuroscience. 2005;135:703–714. doi: 10.1016/j.neuroscience.2005.06.068. [DOI] [PubMed] [Google Scholar]
- Dalla C, Antoniou K, Kokras N, Drossopoulou G, Papathanasiou G, Bekris S, Daskas S, Papadopoulou-Daifoti Z. Sex differences in the effects of two stress paradigms on dopaminergic neurotransmission. Physiol Behav. 2008;93:595–605. doi: 10.1016/j.physbeh.2007.10.020. [DOI] [PubMed] [Google Scholar]
- Dalla C, Whetstone AS, Hodes GE, Shors TJ. Stressful experience has opposite effects on dendritic spines in the hippocampus of cycling versus masculinized females. Neurosci Lett. 2009;449:52–56. doi: 10.1016/j.neulet.2008.10.051. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Dalla C, Papachristos EB, Whetstone AS, Shors TJ. Female rats learn trace memories better than male rats and consequently retain a greater proportion of new neurons in their hippocampi. PNAS. 2009;106:2927–2932. doi: 10.1073/pnas.0809650106. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Dallak MM, Mikhailidis DP, Haidara MA, Bin-Jaliah IM, Tork OM, Rateb MA, Yassin HZ, Al-Refaie ZA, Ibrahim IM, Elawa SM, Rashed LA, et al. Oxidative stress as a common mediator for apoptosis induced-cardiac damage in diabetic rats. Open Cardiovasc Med J. 2008;2:70–78. doi: 10.2174/1874192400802010070. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Davis EG, Humphreys KL, McEwen LM, Sacchet MD, Camacho MC, MacIsaac JL, Lin DTS, Kobor MS, Gotlib IH. Accelerated DNA methylation age in adolescent girls: associations with elevated diurnal cortisol and reduced hippocampal volume. Transl Psychiatry. 2017;7:e1223. doi: 10.1038/tp.2017.188. [DOI] [PMC free article] [PubMed] [Google Scholar]
- de Frias CM, Nilsson LG, Herlitz A. Sex differences in cognition are stable over a 10-year period in adulthood and old age. Neuropsychol Dev Cogn B Aging Neuropsychol Cogn. 2006;13:574–587. doi: 10.1080/13825580600678418. [DOI] [PubMed] [Google Scholar]
- de Voogd LD, Klumpers F, Fernandez G, Hermans EJ. Intrinsic functional connectivity between amygdala and hippocampus during rest predicts enhanced memory under stress. Psychoneuroendocrinology. 2017;75:192–202. doi: 10.1016/j.psyneuen.2016.11.002. [DOI] [PubMed] [Google Scholar]
- Dedovic K, Rexroth M, Wolff E, Duchesne A, Scherling C, Beaudry T, Lue SD, Lord C, Engert V, Pruessner JC. Neural correlates of processing stressful information: an event-related fMRI study. Brain Res. 2009;1293:49–60. doi: 10.1016/j.brainres.2009.06.044. [DOI] [PubMed] [Google Scholar]
- Dedovic K, Engert V, Duchesne A, Lue SD, Andrews J, Efanov SI, Beaudry T, Pruessner JC. Cortisol awakening response and hippocampal volume: vulnerability for major depressive disorder? Biol. Psychiatry. 2010;68:847–853. doi: 10.1016/j.biopsych.2010.07.025. [DOI] [PubMed] [Google Scholar]
- den Heijer T, Geerlings MI, Hofman A, de Jong FH, Launer LJ, Pols HA, Breteler MM. Higher estrogen levels are not associated with larger hippocampi and better memory performance. Arch Neurol. 2003;60:213–220. doi: 10.1001/archneur.60.2.213. [DOI] [PubMed] [Google Scholar]
- Dennison M, Whittle S, Yucel M, Vijayakumar N, Kline A, Simmons J, Allen NB. Mapping subcortical brain maturation during adolescence: evidence of hemisphere- and sex-specific longitudinal changes. Dev Sci. 2013;16:772–791. doi: 10.1111/desc.12057. [DOI] [PubMed] [Google Scholar]
- Dias GP, Bevilaqua MC, da Luz AC, Fleming RL, de Carvalho LA, Cocks G, Beckman D, Hosken LC, de Sant'Anna Machado W, Correa-e-Castro AC, Mousovich-Neto F, et al. Hippocampal biomarkers of fear memory in an animal model of generalized anxiety disorder. Behav Brain Res. 2014;263:34–45. doi: 10.1016/j.bbr.2014.01.012. [DOI] [PubMed] [Google Scholar]
- Dotson VM, Davatzikos C, Kraut MA, Resnick SM. Depressive symptoms and brain volumes in older adults: a longitudinal magnetic resonance imaging study. J.Psychiatry Neurosci. 2009;34:367–375. [PMC free article] [PubMed] [Google Scholar]
- Duan H, Yuan Y, Zhang L, Qin S, Zhang K, Buchanan TW, Wu J. Chronic stress exposure decreases the cortisol awakening response in healthy young men. Stress. 2013;16:630–637. doi: 10.3109/10253890.2013.840579. [DOI] [PubMed] [Google Scholar]
- Duarte-Guterman P, Yagi S, Chow C, Galea LA. Hippocampal learning, memory, and neurogenesis: effects of sex and estrogens across the lifespan in adults. Horm Behav. 2015;74:37–52. doi: 10.1016/j.yhbeh.2015.05.024. [DOI] [PubMed] [Google Scholar]
- Duarte-Guterman P, Lieblich SE, Chow C, Galea LA. Estradiol and GPER activation differentially affect cell proliferation but not GPER expression in the hippocampus of adult female rats. PLoS ONE. 2015;10:e0129880. doi: 10.1371/journal.pone.0129880. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Eberling JL, Wu C, Haan MN, Mungas D, Buonocore M, Jagust WJ. Preliminary evidence that estrogen protects against age-related hippocampal atrophy. Neurobiol Aging. 2003;24:725–732. doi: 10.1016/s0197-4580(02)00056-8. [DOI] [PubMed] [Google Scholar]
- Edinger KL, Frye CA. Androgens' performance-enhancing effects in the inhibitory avoidance and water maze tasks may involve actions at intracellular androgen receptors in the dorsal hippocampus. Neurobiol Learn Mem. 2007;87:201–208. doi: 10.1016/j.nlm.2006.08.008. [DOI] [PubMed] [Google Scholar]
- Endo Y, Mizuno T, Fujita K, Funabashi T, Kimura F. Soft-diet feeding during development enhances later learning abilities in female rats. Physiol Behav. 1994;56:629–633. doi: 10.1016/0031-9384(94)90217-8. [DOI] [PubMed] [Google Scholar]
- Engert V, Buss C, Khalili-Mahani N, Wadiwalla M, Dedovic K, Pruessner JC. Investigating the association between early life parental care and stress responsivity in adulthood. Dev Neuropsychol. 2010;35:570–581. doi: 10.1080/87565641.2010.494752. [DOI] [PubMed] [Google Scholar]
- Epp JR, Spritzer MD, Galea LA. Hippocampus-dependent learning promotes survival of new neurons in the dentate gyrus at a specific time during cell maturation. Neuroscience. 2007;149:273–285. doi: 10.1016/j.neuroscience.2007.07.046. [DOI] [PubMed] [Google Scholar]
- Everaerd D, Gerritsen L, Rijpkema M, Frodl T, van Oostrom I, Franke B, Fernandez G, Tendolkar I. Sex modulates the interactive effect of the serotonin transporter gene polymorphism and childhood adversity on hippocampal volume. Neuropsychopharmacology. 2012;37:1848–1855. doi: 10.1038/npp.2012.32. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Exner C, Nehrkorn B, Martin V, Huber M, Shiratori K, Rief W. Sex-dependent hippocampal volume reductions in schizophrenia relate to episodic memory deficits. J Neuropsychiatry Clin Neurosci. 2008;20:227–230. doi: 10.1176/jnp.2008.20.2.227. [DOI] [PubMed] [Google Scholar]
- Ezzati A, Katz MJ, Zammit AR, Lipton ML, Zimmerman ME, Sliwinski MJ, Lipton RB. Differential association of left and right hippocampal volumes with verbal episodic and spatial memory in older adults. Neuropsychologia. 2016;93:380–385. doi: 10.1016/j.neuropsychologia.2016.08.016. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Falconer EM, Galea LA. Sex differences in cell proliferation, cell death and defensive behavior following acute predator odor stress in adult rats. Brain Res. 2003;975:22–36. doi: 10.1016/s0006-8993(03)02542-3. [DOI] [PubMed] [Google Scholar]
- Fanselow MS, Dong HW. Are the dorsal and ventral hippocampus functionally distinct structures? Neuron. 2010;65:7–19. doi: 10.1016/j.neuron.2009.11.031. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Faraone SV. Attention deficit hyperactivity disorder and premature death. Lancet. 2015;385:2132–2133. [Google Scholar]
- Farinetti A, Tomasi S, Foglio B, Ferraris A, Ponti G, Gotti S, Peretto P, Panzica GC. Testosterone and estradiol differentially affect cell proliferation in the subventricular zone of young adult gonadectomized male and female rats. Neuroscience. 2015;286:162–170. doi: 10.1016/j.neuroscience.2014.11.050. [DOI] [PubMed] [Google Scholar]
- Felmingham K, Williams LM, Kemp AH, Liddell B, Falconer E, Peduto A, Bryant R. Neural responses to masked fear faces: sex differences and trauma exposure in posttraumatic stress disorder. J Abnorm Psychol. 2010;119:241–247. doi: 10.1037/a0017551. [DOI] [PubMed] [Google Scholar]
- Feng M, Sheng G, Li Z, Wang J, Ren K, Jin X, Jiang K. Postnatal maternal separation enhances tonic GABA current of cortical layer 5 pyramidal neurons in juvenile rats and promotes genesis of GABAergic neurons in neocortical molecular layer and subventricular zone in adult rats. Behav Brain Res. 2014;260:74–82. doi: 10.1016/j.bbr.2013.11.040. [DOI] [PubMed] [Google Scholar]
- Fernandez-Guasti A, Martinez-Mota L. Anxiolytic-like actions of testosterone in the burying behavior test: role of androgen and GABA-benzodiazepine receptors. Psychoneuroendocrinology. 2005;30:762–770. doi: 10.1016/j.psyneuen.2005.03.006. [DOI] [PubMed] [Google Scholar]
- Ferragud A, Haro A, Sylvain A, Velazquez-Sanchez C, Hernandez-Rabaza V, Canales JJ. Enhanced habit-based learning and decreased neurogenesis in the adult hippocampus in a murine model of chronic social stress. Behav Brain Res. 2010;210:134–139. doi: 10.1016/j.bbr.2010.02.013. [DOI] [PubMed] [Google Scholar]
- Fester L, Ribeiro-Gouveia V, Prange-Kiel J, von Schassen C, Bottner M, Jarry H, Rune GM. Proliferation and apoptosis of hippocampal granule cells require local oestrogen synthesis. J Neurochem. 2006;97:1136–1144. doi: 10.1111/j.1471-4159.2006.03809.x. [DOI] [PubMed] [Google Scholar]
- Fester L, Prange-Kiel J, Zhou L, Blittersdorf BV, Bohm J, Jarry H, Schumacher M, Rune GM. Estrogen-regulated synaptogenesis in the hippocampus: sexual dimorphism in vivo but not in vitro. J Steroid Biochem Mol Biol. 2012;131:24–29. doi: 10.1016/j.jsbmb.2011.11.010. [DOI] [PubMed] [Google Scholar]
- Figueiredo HF, Dolgas CM, Herman JP. Stress activation of cortex and hippocampus is modulated by sex and stage of estrus. Endocrinology. 2002;143:2534–2540. doi: 10.1210/endo.143.7.8888. [DOI] [PubMed] [Google Scholar]
- Filipek PA, Richelme C, Kennedy DN, Caviness VS., Jr The young adult human brain: an MRI-based morphometric analysis. Cereb Cortex. 1994;4:344–360. doi: 10.1093/cercor/4.4.344. [DOI] [PubMed] [Google Scholar]
- Filkowski MM, Olsen RM, Duda B, Wanger TJ, Sabatinelli D. Sex differences in emotional perception: meta analysis of divergent activation. Neuroimage. 2017;147:925–933. doi: 10.1016/j.neuroimage.2016.12.016. [DOI] [PubMed] [Google Scholar]
- Fisher PM, Larsen CB, Beliveau V, Henningsson S, Pinborg A, Holst KK, Jensen PS, Svarer C, Siebner HR, Knudsen GM, Frokjaer VG. Pharmacologically induced sex hormone fluctuation effects on resting-state functional connectivity in a risk model for depression: a randomized trial. Neuropsychopharmacology. 2017;42:446–453. doi: 10.1038/npp.2016.208. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Foland-Ross LC, Ross JL, Reiss AL. Androgen treatment effects on hippocampus structure in boys with Klinefelter syndrome. Psychoneuroendocrinology. 2019;100:223–228. doi: 10.1016/j.psyneuen.2018.09.039. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Fowler CD, Johnson F, Wang Z. Estrogen regulation of cell proliferation and distribution of estrogen receptor-alpha in the brains of adult female prairie and meadow voles. J Comp Neurol. 2005;489:166–179. doi: 10.1002/cne.20638. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Fowler CD, Liu Y, Wang Z. Estrogen and adult neurogenesis in the amygdala and hypothalamus. Brain Res Rev. 2008;57:342–351. doi: 10.1016/j.brainresrev.2007.06.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Frank TC, Kim GL, Krzemien A, Van Vugt DA. Effect of menstrual cycle phase on corticolimbic brain activation by visual food cues. Brain Res. 2010;1363:81–92. doi: 10.1016/j.brainres.2010.09.071. [DOI] [PubMed] [Google Scholar]
- Frazier JA, Chiu S, Breeze JL, Makris N, Lange N, Kennedy DN, Herbert MR, Bent EK, Koneru VK, Dieterich ME, Hodge SM, et al. Structural brain magnetic resonance imaging of limbic and thalamic volumes in pediatric bipolar disorder. Am J Psychiatry. 2005;162:1256–1265. doi: 10.1176/appi.ajp.162.7.1256. [DOI] [PubMed] [Google Scholar]
- Frazier JA, Hodge SM, Breeze JL, Giuliano AJ, Terry JE, Moore CM, Kennedy DN, Lopez-Larson MP, Caviness VS, Seidman LJ, Zablotsky B, et al. Diagnostic and sex effects on limbic volumes in early-onset bipolar disorder and schizophrenia. Schizophr Bull. 2008;34:37–46. doi: 10.1093/schbul/sbm120. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Frick KM. Molecular mechanisms underlying the memory-enhancing effects of estradiol. Horm Behav. 2015;74:4–18. doi: 10.1016/j.yhbeh.2015.05.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Frick KM, Kim J. Mechanisms underlying the rapid effects of estradiol and progesterone on hippocampal memory consolidation in female rodents. Horm Behav. 2018;104:100–110. doi: 10.1016/j.yhbeh.2018.04.013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Frings L, Wagner K, Unterrainer J, Spreer J, Halsband U, Schulze-Bonhage A. Gender-related differences in lateralization of hippocampal activation and cognitive strategy. NeuroReport. 2006;17:417–421. doi: 10.1097/01.wnr.0000203623.02082.e3. [DOI] [PubMed] [Google Scholar]
- Frodl T, Meisenzahl EM, Zetzsche T, Born C, Groll C, Jager M, Leinsinger G, Bottlender R, Hahn K, Moller HJ. Hippocampal changes in patients with a first episode of major depression. Am J Psychiatry. 2002;159:1112–1118. doi: 10.1176/appi.ajp.159.7.1112. [DOI] [PubMed] [Google Scholar]
- Frodl T, Carballedo A, Frey EM, O'Keane V, Skokauskas N, Morris D, Gill M, Hughes MM, Harkin A, Connor T. Expression of glucocorticoid inducible genes is associated with reductions in cornu ammonis and dentate gyrus volumes in patients with major depressive disorder. Dev Psychopathol. 2014;26:1209–1217. doi: 10.1017/S0954579414000972. [DOI] [PubMed] [Google Scholar]
- Frodl T, Janowitz D, Schmaal L, Tozzi L, Dobrowolny H, Stein DJ, Veltman DJ, Wittfeld K, van Erp TGM, Jahanshad N, Block A, et al. Childhood adversity impacts on brain subcortical structures relevant to depression. J Psychiatr Res. 2017;86:58–65. doi: 10.1016/j.jpsychires.2016.11.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Frodl T, O'Keane V. How does the brain deal with cumulative stress? A review with focus on developmental stress, HPA axis function and hippocampal structure in humans. Neurobiol Dis. 2013;52:24–37. doi: 10.1016/j.nbd.2012.03.012. [DOI] [PubMed] [Google Scholar]
- Frye CA. Estrus-associated decrements in a water maze task are limited to acquisition. Physiol Behav. 1995;57:5–14. doi: 10.1016/0031-9384(94)00197-d. [DOI] [PubMed] [Google Scholar]
- Frye CA. Progesterone attenuates depressive behavior ofyounger and older adult C57/BL6, wildtype, and progesterone receptor knockout mice. Pharmacol Biochem Behav. 2011;99:525–531. doi: 10.1016/j.pbb.2011.05.024. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Frye CA, Duffy CK, Walf AA. Estrogens and progestins enhance spatial learning of intact and ovariectomized rats in the object placement task. Neurobiol Learn Mem. 2007;88:208–216. doi: 10.1016/j.nlm.2007.04.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Frye CA, Rhodes ME. Enhancing effects of estrogen on inhibitory avoidance performance may be in part independent of intracellular estrogen receptors in the hippocampus. Brain Res. 2002;956:285–293. doi: 10.1016/s0006-8993(02)03559-x. [DOI] [PubMed] [Google Scholar]
- Frye CA, Walf AA. Depression-like behavior of aged male and female mice is ameliorated with administration of testosterone or its metabolites. Physiol Behav. 2009;97:266–269. doi: 10.1016/j.physbeh.2009.02.022. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Fuentes N, Silveyra P. Estrogen receptor signaling mechanisms. Adv Protein Chem Struct Biol. 2019;116:135–170. doi: 10.1016/bs.apcsb.2019.01.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gabor C, Lymer J, Phan A, Choleris E. Rapid effects of the G-protein coupled oestrogen receptor (GPER) on learning and dorsal hippocampus dendritic spines in female mice. Physiol Behav. 2015;149:53–60. doi: 10.1016/j.physbeh.2015.05.017. [DOI] [PubMed] [Google Scholar]
- Galea LA. Gonadal hormone modulation of neurogenesis in the dentate gyrus of adult male and female rodents. Brain Res Rev. 2008;57:332–341. doi: 10.1016/j.brainresrev.2007.05.008. [DOI] [PubMed] [Google Scholar]
- Galea LAM, Frick KM, Hampson E, Sohrabji F, Choleris E. Why estrogens matter for behavior and brain health. Neurosci Biobehav Rev. 2017;76:363–379. doi: 10.1016/j.neubiorev.2016.03.024. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Galea LA, Kavaliers M, Ossenkopp KP. Sexually dimorphic spatial learning in meadow voles Microtus pennsylvanicus and deer mice Peromyscus maniculatus. J Exp Biol. 1996;199:195–200. doi: 10.1242/jeb.199.1.195. [DOI] [PubMed] [Google Scholar]
- Galea L, Kimura D. Sex differences in route-learning. Pers Individ Dif. 1993;14:53–65. [Google Scholar]
- Galea LA, McEwen BS, Tanapat P, Deak T, Spencer RL, Dhabhar FS. Sex differences in dendritic atrophy of CA3 pyramidal neurons in response to chronic restraint stress. Neuroscience. 1997;81:689–697. doi: 10.1016/s0306-4522(97)00233-9. [DOI] [PubMed] [Google Scholar]
- Galea LA, McEwen BS. Sex and seasonal differences in the rate of cell proliferation in the dentate gyrus of adult wild meadow voles. Neuroscience. 1999;89:955–964. doi: 10.1016/s0306-4522(98)00345-5. [DOI] [PubMed] [Google Scholar]
- Galea LA, Wide JK, Barr AM. Estradiol alleviates depressive-like symptoms in a novel animal model of post-partum depression. Behav Brain Res. 2001;122:1–9. doi: 10.1016/s0166-4328(01)00170-x. [DOI] [PubMed] [Google Scholar]
- Galea LA, Wainwright SR, Roes MM, Duarte-Guterman P, Chow C, Hamson DK. Sex, hormones and neurogenesis in the hippocampus: hormonal modulation of neurogenesis and potential functional implications. J Neuroendocrinol. 2013;25:1039–1061. doi: 10.1111/jne.12070. [DOI] [PubMed] [Google Scholar]
- Garcia-Falgueras A, Junque C, Gimenez M, Caldu X, Segovia S, Guillamon A. Sex differences in the human olfactory system. Brain Res. 2006;1116:103–111. doi: 10.1016/j.brainres.2006.07.115. [DOI] [PubMed] [Google Scholar]
- Gauthier CT, Duyme M, Zanca M, Capron C. Sex and performance level effects on brain activation during a verbal fluency task: a functional magnetic resonance imaging study. Cortex. 2009;45:164–176. doi: 10.1016/j.cortex.2007.09.006. [DOI] [PubMed] [Google Scholar]
- Gerritsen L, Comijs HC, van der Graaf Y, Knoops AJ, Penninx BW, Geerlings MI. Depression, hypothalamic pituitary adrenal axis, and hippocampal and en-torhinal cortex volumes-the SMART Medea study. Biol Psychiatry. 2011;70:373–380. doi: 10.1016/j.biopsych.2011.01.029. [DOI] [PubMed] [Google Scholar]
- Gibbs RB, Johnson DA. Sex-specific effects of gonadectomy and hormone treatment on acquisition of a 12-arm radial maze task by Sprague Dawley rats. Endocrinology. 2008;149:3176–3183. doi: 10.1210/en.2007-1645. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Giedd JN, Vaituzis AC, Hamburger SD, Lange N, Rajapakse JC, Kaysen D, Vauss YC, Rapoport JL. Quantitative MRI of the temporal lobe, amygdala, and hippocampus in normal human development: ages 4–18 years. J Comp Neurol. 1996;366:223–230. doi: 10.1002/(SICI)1096-9861(19960304)366:2<223::AID-CNE3>3.0.CO;2-7. [DOI] [PubMed] [Google Scholar]
- Giedd JN, Castellanos FX, Rajapakse JC, Vaituzis AC, Rapoport JL. Sexual dimorphism of the developing human brain. Prog Neuro-Psychopharmacol Biol Psychiatry. 1997;21:1185–1201. doi: 10.1016/s0278-5846(97)00158-9. [DOI] [PubMed] [Google Scholar]
- Gobinath AR, Mahmoud R, Galea LA. Influence of sex and stress exposure across the lifespan on endophenotypes of depression: focus on behavior, glucocorticoids, and hippocampus. Front Neurosci. 2015;8:420. doi: 10.3389/fnins.2014.00420. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gobinath AR, Workman JL, Chow C, Lieblich SE, Galea LA. Maternal postpartum corticosterone and fluoxetine differentially affect adult male and female offspring on anxiety-like behavior, stress reactivity, and hippocampal neurogenesis. Neuropharmacology. 2016;101:165–178. doi: 10.1016/j.neuropharm.2015.09.001. [DOI] [PubMed] [Google Scholar]
- Goel N, Workman JL, Lee TT, Innala L, Viau V. Sex differences in the HPA axis. Comprehens Physiol. 2014;4:1121–1155. doi: 10.1002/cphy.c130054. [DOI] [PubMed] [Google Scholar]
- Gold SM, Kern KC, O'Connor MF, Montag MJ, Kim A, Yoo YS, Giesser BS, Sicotte NL. Smaller cornu ammonis 2–3/dentate gyrus volumes and elevated cortisol in multiple sclerosis patients with depressive symptoms. Biol Psychiatry. 2010;68:553–559. doi: 10.1016/j.biopsych.2010.04.025. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Goldstat R, Briganti E, Tran J, Wolfe R, Davis SR. Transdermal testosterone therapy improves well-being, mood, and sexual function in premenopausal women. Menopause. 2003;10:390–398. doi: 10.1097/01.GME.0000060256.03945.20. [DOI] [PubMed] [Google Scholar]
- Goldstein JM, Jerram M, Poldrack R, Ahern T, Kennedy DN, Seidman LJ, Makris N. Hormonal cycle modulates arousal circuitry in women using functional magnetic resonance imaging. J Neurosci. 2005;25:9309–9316. doi: 10.1523/JNEUROSCI.2239-05.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Goldstein JM, Jerram M, Abbs B, Whitfield-Gabrieli S, Makris N. Sex differences in stress response circuitry activation dependent on female hormonal cycle. J Neurosci. 2010;30:431–438. doi: 10.1523/JNEUROSCI.3021-09.2010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Golomb J, de Leon MJ, Kluger A, George AE, Tarshish C, Ferris SH. Hippocampal atrophy in normal aging. An association with recent memory impairment Arch Neurol. 1993;50:967–973. doi: 10.1001/archneur.1993.00540090066012. [DOI] [PubMed] [Google Scholar]
- Gonzalez-Burgos I, Alejandre-Gomez M, Cervantes M. Spine-type densities of hippocampal CA1 neurons vary in proestrus and estrus rats. Neurosci Lett. 2005;379:52–54. doi: 10.1016/j.neulet.2004.12.043. [DOI] [PubMed] [Google Scholar]
- Goto M, Abe O, Miyati T, Inano S, Hayashi N, Aoki S, Mori H, Kabasawa H, Ino K, Yano K, Iida K, et al. 3 Tesla MRI detects accelerated hippocampal volume reduction in postmenopausal women. J Magn Reson Imaging. 2011;33:48–53. doi: 10.1002/jmri.22328. [DOI] [PubMed] [Google Scholar]
- Gould E, Woolley CS, Frankfurt M, McEwen BS. Gonadal steroids regulate dendritic spine density in hippocampal pyramidal cells in adulthood. J Neurosci. 1990;10:1286–1291. doi: 10.1523/JNEUROSCI.10-04-01286.1990. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gould E, McEwen BS, Tanapat P, Galea LA, Fuchs E. Neurogenesis in the dentate gyrus of the adult tree shrew is regulated by psychosocial stress and NMDA receptor activation. J Neurosci. 1997;17:2492–2498. doi: 10.1523/JNEUROSCI.17-07-02492.1997. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gould E, Tanapat P, McEwen BS, Flugge G, Fuchs E. Proliferation of granule cell precursors in the dentate gyrus of adult monkeys is diminished by stress. Proc Natl Acad Sci U S A. 1998;95:3168–3171. doi: 10.1073/pnas.95.6.3168. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gould E, Beylin A, Tanapat P, Reeves A, Shors TJ. Learning enhances adult neurogenesis in the hippocampal formation. Nat Neurosci. 1999;2:260–265. doi: 10.1038/6365. [DOI] [PubMed] [Google Scholar]
- Green AD, Galea LA. Adult hippocampal cell proliferation is suppressed with estrogen withdrawal after a hormone-simulated pregnancy. Horm Behav. 2008;54:203–211. doi: 10.1016/j.yhbeh.2008.02.023. [DOI] [PubMed] [Google Scholar]
- Grissom EM, Hawley WR, Hodges KS, Fawcett-Patel JM, Dohanich GP. Biological sex influences learning strategy preference and muscarinic receptor binding in specific brain regions of prepubertal rats. Hippocampus. 2013;23:313–322. doi: 10.1002/hipo.22085. [DOI] [PubMed] [Google Scholar]
- Gron G, Wunderlich AP, Spitzer M, Tomzcak R, Riepe MW. Brain activation during human navigation: gender-different neural networks as substrates of performance. Am Nat. 2000;127:74–88. doi: 10.1038/73980. [DOI] [PubMed] [Google Scholar]
- Guadalupe T, Mathias SR, vanErp TGM, Whelan CD, Zwiers MP, Abe Y, Abramovic L, Agartz I, Andreassen OA, Arias-Vasquez A, Aribisala BS, et al. Human subcortical brain asymmetries in 15,847 people worldwide reveal effects of age and sex. Brain Imaging Behav. 2017;11:1497–1514. doi: 10.1007/s11682-016-9629-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gur RC, Gunning-Dixon F, Bilker WB, Gur RE. Sex differences in temporolimbic and frontal brain volumes of healthy adults. Cereb Cortex. 2002;12:998–1003. doi: 10.1093/cercor/12.9.998. [DOI] [PubMed] [Google Scholar]
- Gutierrez-Lobos K, Scherer M, Anderer P, Katschnig H. The influence of age on the female/male ratio of treated incidence rates in depression. BMC Psychiatry. 2002;2:3. doi: 10.1186/1471-244X-2-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hadel S, Wirth C, Rapp M, Gallinat J, Schubert F. Effects of age and sex on the concentrations of glutamate and glutamine in the human brain. J Magn Reson Imaging. 2013;38:1480–1487. doi: 10.1002/jmri.24123. [DOI] [PubMed] [Google Scholar]
- Hajszan T, Milner TA, Leranth C. Sex steroids and the dentate gyrus. Prog Brain Res. 2007;163:399–415. doi: 10.1016/S0079-6123(07)63023-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Halpern DF. Sex Differences in Cognitive Abilities. L Erlbaum Associates; Mahwah, N.J: 2000. [Google Scholar]
- Hamann S, Canli T. Individual differences in emotion processing. Curr Opin Neurobiol. 2004;14:233–238. doi: 10.1016/j.conb.2004.03.010. [DOI] [PubMed] [Google Scholar]
- Hampson E. Variations in sex-related cognitive abilities across the menstrual cycle. Brain Cogn. 1990;14:26–43. doi: 10.1016/0278-2626(90)90058-v. [DOI] [PubMed] [Google Scholar]
- Hamson DK, Wainwright SR, Taylor JR, Jones BA, Watson NV, Galea LA. Androgens increase survival of adult-born neurons in the dentate gyrus by an androgen receptor-dependent mechanism in male rats. Endocrinology. 2013;154:3294–3304. doi: 10.1210/en.2013-1129. [DOI] [PubMed] [Google Scholar]
- Hamson DK, Roes MM, Galea LA. Sex hormones and cognition: neuroendo-crine influences on memory and learning. Compr Physiol. 2016;6:1295–1337. doi: 10.1002/cphy.c150031. [DOI] [PubMed] [Google Scholar]
- Han KM, Choi S, Jung J, Na KS, Yoon HK, Lee MS, Ham BJ. Cortical thickness, cortical and subcortical volume, and white matter integrity in patients with their first episode of major depression. J Affect Disord. 2014;155:42–48. doi: 10.1016/j.jad.2013.10.021. [DOI] [PubMed] [Google Scholar]
- Handa RJ, Burgess LH, Kerr JE, O'Keefe JA. Gonadal steroid hormone receptors and sex differences in the hypothalamo-pituitary-adrenal axis. Horm Behav. 1994;28:464–476. doi: 10.1006/hbeh.1994.1044. [DOI] [PubMed] [Google Scholar]
- Hanson ND, Owens MJ, Boss-Williams KA, Weiss JM, Nemeroff CB. Several stressors fail to reduce adult hippocampal neurogenesis. Psychoneuroendocrinology. 2011;36:1520–1529. doi: 10.1016/j.psyneuen.2011.04.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Harburger LL, Saadi A, Frick KM. Dose-dependent effects of post-training estradiol plus progesterone treatment on object memory consolidation and hippo-campal extracellular signal-regulated kinase activation in young ovariectomized mice. Neuroscience. 2009;160:6–12. doi: 10.1016/j.neuroscience.2009.02.024. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Harris KM, Jensen FE, Tsao B. Three-dimensional structure of dendritic spines and synapses in rat hippocampus (CA1) at postnatal day 15 and adult ages: implications for the maturation of synaptic physiology and long-term potentiation. J Neurosci. 1992;12:2685–2705. doi: 10.1523/JNEUROSCI.12-07-02685.1992. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Harris T, Scheuringer A, Pletzer B. Perspective and strategy interactively modulate sex differences in a 3D navigation task. Biol Sex Differ. 2019;10:17. doi: 10.1186/s13293-019-0232-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hastings RS, Parsey RV, Oquendo MA, Arango V, Mann JJ. Volumetric analysis of the prefrontal cortex, amygdala, and hippocampus in major depression. Neuropsychopharmacology. 2004;29:952–959. doi: 10.1038/sj.npp.1300371. [DOI] [PubMed] [Google Scholar]
- Hatanaka Y, Mukai H, Mitsuhashi K, Hojo Y, Murakami G, Komatsuzaki Y, Sato R, Kawato S. Androgen rapidly increases dendritic thorns of CA3 neurons in male rat hippocampus. Biochem Biophys Res Commun. 2009;381:728–732. doi: 10.1016/j.bbrc.2009.02.130. [DOI] [PubMed] [Google Scholar]
- Heany SJ, van Honk J, Stein DJ, Brooks SJ. A quantitative and qualitative review of the effects of testosterone on the function and structure of the human social-emotional brain. Metab Brain Dis. 2016;31:157–167. doi: 10.1007/s11011-015-9692-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Heine VM, Maslam S, Joels M, Lucassen PJ. Prominent decline of newborn cell proliferation, differentiation, and apoptosis in the aging dentate gyrus, in absence of an age-related hypothalamus-pituitary-adrenal axis activation. Neurobiol Aging. 2004;25:361–375. doi: 10.1016/S0197-4580(03)00090-3. [DOI] [PubMed] [Google Scholar]
- Helpman L, Zhu X, Suarez-Jimenez B, Lazarov A, Monk C, Neria Y. Sex differences in trauma-related psychopathology: a critical review of neuroimaging literature (2014–2017) Curr Psychiatry Rep. 2017;19:104. doi: 10.1007/s11920-017-0854-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Henckens MJ, Hermans EJ, Pu Z, Joels M, Fernandez G. Stressed memories: how acute stress affects memory formation in humans. J Neurosci. 2009;29:10111–10119. doi: 10.1523/JNEUROSCI.1184-09.2009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Herbison CE, Henley D, Marsh J, Atkinson H, Newnham JP, Matthews SG, Lye SJ, Pennell CE. Characterization and novel analyses of acute stress response patterns in a population-based cohort of young adults: influence of gender, smoking, and BMI. Stress. 2016;19:139–150. doi: 10.3109/10253890.2016.1146672. [DOI] [PubMed] [Google Scholar]
- Herting MM, Gautam P, Spielberg JM, Kan E, Dahl RE, Sowell ER. The role of testosterone and estradiol in brain volume changes across adolescence: a longitudinal structural MRI study. Hum Brain Mapp. 2014;35:5633–5645. doi: 10.1002/hbm.22575. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hidalgo V, Pulopulos MM, Puig-Perez S, Espin L, Gomez-Amor J, Salvador A. Acute stress affects free recall and recognition of pictures differently depending on age and sex. Behav Brain Res. 2015;292:393–402. doi: 10.1016/j.bbr.2015.07.011. [DOI] [PubMed] [Google Scholar]
- Hill AC, Laird AR, Robinson JL. Gender differences in working memory networks: a BrainMap meta-analysis. Biol Psychol. 2014;102:18–29. doi: 10.1016/j.biopsycho.2014.06.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hillerer KM, Neumann ID, Slattery DA. From stress to postpartum mood and anxiety disorders: how chronic peripartum stress can impair maternal adaptations. Neuroendocrinology. 2012;95:22–38. doi: 10.1159/000330445. [DOI] [PubMed] [Google Scholar]
- Hillerer KM, Neumann ID, Couillard-Despres S, Aigner L, Slattery DA. Sex-dependent regulation of hippocampal neurogenesis under basal and chronic stress conditions in rats. Hippocampus. 2013;23:476–487. doi: 10.1002/hipo.22107. [DOI] [PubMed] [Google Scholar]
- Hirnstein M, Coloma Andrews L, Hausmann M. Gender-stereotyping and cognitive sex differences in mixed- and same-sex groups. Arch Sex Behav. 2014;43:1663–1673. doi: 10.1007/s10508-014-0311-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hlavacova N, Wawruch M, Tisonova J, Jezova D. Neuroendocrine activation during combined mental and physical stress in women depends on trait anxiety and the phase of the menstrual cycle. Ann N Y Acad Sci. 2008;1148:520–525. doi: 10.1196/annals.1410.030. [DOI] [PubMed] [Google Scholar]
- Hodosy J, Zelmanova D, Majzunova M, Filova B, Malinova M, Ostatnikova D, Celec P. The anxiolytic effect of testosterone in the rat is mediated via the androgen receptor. Pharmacol Biochem Behav. 2012;102:191–195. doi: 10.1016/j.pbb.2012.04.005. [DOI] [PubMed] [Google Scholar]
- Hoekzema E, Barba-Muller E, Pozzobon C, Picado M, Lucco F, Garcia-Garcia D, Soliva JC, Tobena A, Desco M, Crone EA, Ballesteros A, et al. Pregnancy leads to long-lasting changes in human brain structure. Nat Neurosci. 2017;20:287–296. doi: 10.1038/nn.4458. [DOI] [PubMed] [Google Scholar]
- Hofer A, Siedentopf CM, Ischebeck A, Rettenbacher MA, Verius M, Felber S, Wolfgang Fleischhacker W. Sex differences in brain activation patterns during processing of positively and negatively valenced emotional words. Psychol Med. 2007;37:109–119. doi: 10.1017/S0033291706008919. [DOI] [PubMed] [Google Scholar]
- Hogervorst E, Williams J, Budge M, Riedel W, Jolles J. The nature of the effect of female gonadal hormone replacement therapy on cognitive function in postmenopausal women: a meta-analysis. Neuroscience. 2000;101:485–512. doi: 10.1016/s0306-4522(00)00410-3. [DOI] [PubMed] [Google Scholar]
- Hogervorst E, Bandelow S, Moffat SD. Increasing testosterone levels and effects on cognitive functions in elderly men and women: a review. Curr Drug Targets CNS Neurol Disord. 2005;4:531–540. doi: 10.2174/156800705774322049. [DOI] [PubMed] [Google Scholar]
- Hojo Y, Murakami G, Mukai H, Higo S, Hatanaka Y, Ogiue-Ikeda M, Ishii H, Kimoto T, Kawato S. Estrogen synthesis in the brain-role in synaptic plasticity and memory. Mol Cell Endocrinol. 2008;290:31–43. doi: 10.1016/j.mce.2008.04.017. [DOI] [PubMed] [Google Scholar]
- Hollanders JJ, van der Voorn B, Rotteveel J, Finken MJJ. Is HPA axis reactivity in childhood gender-specific? A systematic review. Biol Sex Differ. 2017;8:23. doi: 10.1186/s13293-017-0144-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Holsen LM, Spaeth SB, Lee JH, Ogden LA, Klibanski A, Whitfield-Gabrieli S, Goldstein JM. Stress response circuitry hypoactivation related to hormonal dysfunction in women with major depression. J Affect Disord. 2011;131:379–387. doi: 10.1016/j.jad.2010.11.024. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Holsen LM, Lee JH, Spaeth SB, Ogden LA, Klibanski A, Whitfield-Gabrieli S, Sloan RP, Goldstein JM. Brain hypoactivation, autonomic nervous system dysregulation, and gonadal hormones in depression: a preliminary study. Neurosci Lett. 2012;514:57–61. doi: 10.1016/j.neulet.2012.02.056. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Holtmaat AJ, Trachtenberg JT, Wilbrecht L, Shepherd GM, Zhang X, Knott GW, Svoboda K. Transient and persistent dendritic spines in the neocortex in vivo. Neuron. 2005;45:279–291. doi: 10.1016/j.neuron.2005.01.003. [DOI] [PubMed] [Google Scholar]
- Hu L, Yue Y, Zuo PP, Jin ZY, Feng F, You H, Li ML, Ge QS. Evaluation of neuroprotective effects of long-term low dose hormone replacement therapy on postmenopausal women brain hippocampus using magnetic resonance scanner. Chin Med Sci J. 2006;21:214–218. [PubMed] [Google Scholar]
- Huang Y, Zhou R, Wu M, Wang Q, Zhao Y. Premenstrual syndrome is associated with blunted cortisol reactivity to the TSST. Stress. 2015;18:160–168. doi: 10.3109/10253890.2014.999234. [DOI] [PubMed] [Google Scholar]
- Hulshof HJ, Novati A, Sgoifo A, Luiten PG, den Boer JA, Meerlo P. Maternal separation decreases adult hippocampal cell proliferation and impairs cognitive performance but has little effect on stress sensitivity and anxiety in adult Wistar rats. Behav Brain Res. 2011;216:552–560. doi: 10.1016/j.bbr.2010.08.038. [DOI] [PubMed] [Google Scholar]
- Iaria G, Lanyon LJ, Fox CJ, Giaschi D, Barton JJ. Navigational skills correlate with hippocampal fractional anisotropy in humans. Hippocampus. 2008;18:335–339. doi: 10.1002/hipo.20400. [DOI] [PubMed] [Google Scholar]
- Irle E, Ruhleder M, Lange C, Seidler-Brandler U, Salzer S, Dechent P, Weniger G, Leibing E, Leichsenring F. Reduced amygdalar and hippocampal size in adults with generalized social phobia. J Psychiatry Neurosci. 2010;35:126–131. doi: 10.1503/jpn.090041. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Irle E, Lange C, Ruhleder M, Exner C, Siemerkus J, Weniger G. Hippocampal size in women but not men with schizophrenia relates to disorder duration. Psychiatry Res. 2011;192:133–139. doi: 10.1016/j.pscychresns.2010.12.009. [DOI] [PubMed] [Google Scholar]
- Irvine K, Laws KR, Gale TM, Kondel TK. Greater cognitive deterioration in women than men with Alzheimer's disease: a meta analysis. J Clin Exp Neuropsychol. 2012;34:989–998. doi: 10.1080/13803395.2012.712676. [DOI] [PubMed] [Google Scholar]
- Jacobs E, D'Esposito M. Estrogen shapes dopamine-dependent cognitive processes: implications for women's health. J Neurosci. 2011;31:5286–5293. doi: 10.1523/JNEUROSCI.6394-10.2011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Jacobs EG, Holsen LM, Lancaster K, Makris N, Whitfield-Gabrieli S, Remington A, Weiss B, Buka S, Klibanski A, Goldstein JM. 17beta-estradiol differentially regulates stress circuitry activity in healthy and depressed women. Neuropsychopharmacology. 2015;40:566–576. doi: 10.1038/npp.2014.203. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Jacobs EG, Weiss BK, Makris N, Whitfield-Gabrieli S, Buka SL, Klibanski A, Goldstein JM. Impact of sex and menopausal status on episodic memory circuitry in early midlife. J Neurosci. 2016;36:10163–10173. doi: 10.1523/JNEUROSCI.0951-16.2016. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Jacobs EG, Weiss B, Makris N, Whitfield-Gabrieli S, Buka SL, Klibanski A, Goldstein JM. Reorganization of functional networks in verbal working memory circuitry in early midlife: the impact of sex and menopausal status. Cereb Cortex. 2017;27:2857–2870. doi: 10.1093/cercor/bhw127. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Jacome LF, Barateli K, Buitrago D, Lema F, Frankfurt M, Luine VN. Gonadal hormones rapidly enhance spatial memory and increase hippocampal spine density in male rats. Endocrinology. 2016;157:1357–1362. doi: 10.1210/en.2015-1959. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Jain A, Huang GZ, Woolley CS. Latent sex differences in molecular signaling that underlies excitatory synaptic potentiation in the hippocampus. J Neurosci. 2019;39:1552–1565. doi: 10.1523/JNEUROSCI.1897-18.2018. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Jancke L, Merillat S, Liem F, Hanggi J. Brain size, sex, and the aging brain. Hum Brain Mapp. 2015;36:150–169. doi: 10.1002/hbm.22619. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Jaworska N, MacMaster FP, Yang XR, Courtright A, Pradhan S, Gaxiola I, Cortese F, Goodyear B, Ramasubbu R. Influence of age of onset on limbic and paralimbic structures in depression. Psychiatry Clin Neurosci. 2014;68:812–820. doi: 10.1111/pcn.12197. [DOI] [PubMed] [Google Scholar]
- Jessberger S, Nakashima K, Clemenson GD, Jr, Mejia E, Mathews E, Ure K, Ogawa S, Sinton CM, Gage FH, Hsieh J. Epigenetic modulation of seizure-induced neurogenesis and cognitive decline. J Neurosci: Off J Soc Neurosci. 2007;27:5967–5975. doi: 10.1523/JNEUROSCI.0110-07.2007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Jessberger S, Clark RE, Broadbent NJ, Clemenson GD, Jr, Consiglio A, Lie DC, Squire LR, Gage FH. Dentate gyrus-specific knockdown of adult neuro-genesis impairs spatial and object recognition memory in adult rats. Learn Memory. 2009;16:147–154. doi: 10.1101/lm.1172609. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Jezova D, Jurankova E, Mosnarova A, Kriska M, Skultetyova I. Neuroendocrine response during stress with relation to gender differences. Acta Neurobiol Exp (Wars) 1996;56:779–785. doi: 10.55782/ane-1996-1183. [DOI] [PubMed] [Google Scholar]
- Ji E, Weickert CS, Lenroot R, Kindler J, Skilleter AJ, Vercammen A, White C, Gur RE, Weickert TW. Adjunctive selective estrogen receptor modulator increases neural activity in the hippocampus and inferior frontal gyrus during emotional face recognition in schizophrenia. Transl Psychiatry. 2016;6:e795. doi: 10.1038/tp.2016.59. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Jin RO, Mason S, Mellon SH, Epel ES, Reus VI, Mahan L, Rosser RL, Hough CM, Burke HM, Mueller SG, Wolkowitz OM. Cortisol/DHEA ratio and hippocampal volume: a pilot study in major depression and healthy controls. Psychoneuroendocrinology. 2016;72:139–146. doi: 10.1016/j.psyneuen.2016.06.017. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Jonasson Z. Meta-analysis of sex differences in rodent models of learning and memory: a review of behavioral and biological data. Neurosci Biobehav Rev. 2005;28:811–825. doi: 10.1016/j.neubiorev.2004.10.006. [DOI] [PubMed] [Google Scholar]
- Jorge RE, Acion L, Starkstein SE, Magnotta V. Hippocampal volume and mood disorders after traumatic brain injury. Biol Psychiatry. 2007;62:332–338. doi: 10.1016/j.biopsych.2006.07.024. [DOI] [PubMed] [Google Scholar]
- Kajantie E, Phillips DI. The effects of sex and hormonal status on the physiological response to acute psychosocial stress. Psychoneuroendocrinology. 2006;31:151–178. doi: 10.1016/j.psyneuen.2005.07.002. [DOI] [PubMed] [Google Scholar]
- Kasai H, Fukuda M, Watanabe S, Hayashi-Takagi A, Noguchi J. Structural dynamics of dendritic spines in memory and cognition. Trends Neurosci. 2010;33:121–129. doi: 10.1016/j.tins.2010.01.001. [DOI] [PubMed] [Google Scholar]
- Kaufman SB. Sex differences in mental rotation and spatial visualization ability: Can they be accounted for by differences in working memory capacity? Intelligence. 2007;35:211–223. [Google Scholar]
- Keding TJ, Herringa RJ. Abnormal structure of fear circuitry in pediatric posttraumatic stress disorder. Neuropsychopharmacology. 2015;40:537–545. doi: 10.1038/npp.2014.239. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kee N, Teixeira CM, Wang AH, Frankland PW. Preferential incorporation of adult-generated granule cells into spatial memory networks in the dentate gyrus. Nat Neurosci. 2007;10:355–362. doi: 10.1038/nn1847. [DOI] [PubMed] [Google Scholar]
- Keller J, Gomez R, Williams G, Lembke A, Lazzeroni L, Murphy GM, Jr, Schatzberg AF. HPA axis in major depression: cortisol, clinical symptomatology and genetic variation predict cognition. Mol Psychiatry. 2017;22:527–536. doi: 10.1038/mp.2016.120. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kesler SR, Garrett A, Bender B, Yankowitz J, Zeng SM, Reiss AL. Amygdala and hippocampal volumes in Turner syndrome: a high-resolution MRI study of X-monosomy. Neuropsychologia. 2004;42:1971–1978. doi: 10.1016/j.neuropsychologia.2004.04.021. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kessler RC. Epidemiology of women and depression. J Affect Disord. 2003;74:5–13. doi: 10.1016/s0165-0327(02)00426-3. [DOI] [PubMed] [Google Scholar]
- Kessler RC, McGonagle KA, Zhao S, Nelson CB, Hughes M, Eshleman S, Wittchen HU, Kendler KS. Results from the National Comorbidity Survey. Vol. 51. Arch Gen Psychiatry; 1994. Lifetime and 12-month prevalence of DSM-III-R psychiatric disorders in the United States; pp. 8–19. [DOI] [PubMed] [Google Scholar]
- Khaksari M, Mahmoodi M, Rezvani ME, Sajjadi MA, Karam GA, Hajizadeh S. Differences between male and female students in cardiovascular and endocrine responses to examination stress. J Ayub Med Coll Abbottabad. 2005;17:15–19. [PubMed] [Google Scholar]
- Khalili-Mahani N, Dedovic K, Engert V, Pruessner M, Pruessner JC. Hippocampal activation during a cognitive task is associated with subsequent neuroendocrine and cognitive responses to psychological stress. Hippocampus. 2010;20:323–334. doi: 10.1002/hipo.20623. [DOI] [PubMed] [Google Scholar]
- Kheirbek MA, Klemenhagen KC, Sahay A, Hen R. Neurogenesis and generalization: a new approach to stratify and treat anxiety disorders. Nat Neurosci. 2012;15:1613–1620. doi: 10.1038/nn.3262. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kikusui T, Ichikawa S, Mori Y. Maternal deprivation by early weaning increases corticosterone and decreases hippocampal BDNF and neurogenesis in mice. Psychoneuroendocrinology. 2009;34:762–772. doi: 10.1016/j.psyneuen.2008.12.009. [DOI] [PubMed] [Google Scholar]
- Kim GW, Jeong GW. Neural mechanisms underlying sexual arousal in connection with sexual hormone levels: a comparative study of the postoperative male-to-female transsexuals and premenopausal and menopausal women. NeuroReport. 2014;25:693–700. doi: 10.1097/WNR.0000000000000159. [DOI] [PubMed] [Google Scholar]
- Kimura D, Seal BN. Sex differences in recall of real or nonsense words. Psychol Rep. 2003;93:263–264. doi: 10.2466/pr0.2003.93.1.263. [DOI] [PubMed] [Google Scholar]
- Kindler J, Weickert CS, Skilleter AJ, Catts SV, Lenroot R, Weickert TW. Selective estrogen receptor modulation increases hippocampal activity during probabilistic association learning in schizophrenia. Neuropsychopharmacology. 2015;40:2388–2397. doi: 10.1038/npp.2015.88. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kirschbaum C, Wust S, Hellhammer D. Consistent sex differences in cortisol responses to psychological stress. Psychosom Med. 1992;54:648–657. doi: 10.1097/00006842-199211000-00004. [DOI] [PubMed] [Google Scholar]
- Kirschbaum C, Kudielka BM, Gaab J, Schommer NC, Hellhammer DH. Impact of gender, menstrual cycle phase, and oral contraceptives on the activity of the hypothalamus-pituitary-adrenal axis. Psychosom Med. 1999;61:154–162. doi: 10.1097/00006842-199903000-00006. [DOI] [PubMed] [Google Scholar]
- Klaassen EB, de Groot RH, Evers EA, Nicolson NA, Veltman DJ, Jolles J. Cortisol and induced cognitive fatigue: effects on memory activation in healthy males. Biol Psychol. 2013;94:167–174. doi: 10.1016/j.biopsycho.2013.05.015. [DOI] [PubMed] [Google Scholar]
- Knoops AJ, Gerritsen L, van der Graaf Y, Mali WP, Geerlings MI. Basal hypothalamic pituitary adrenal axis activity and hippocampal volumes: the SMART-Medea study. Biol Psychiatry. 2010;67:1191–1198. doi: 10.1016/j.biopsych.2010.01.025. [DOI] [PubMed] [Google Scholar]
- Kogler L, Gur RC, Derntl B. Sex differences in cognitive regulation of psychosocial achievement stress: brain and behavior. Hum Brain Mapp. 2015;36:1028–1042. doi: 10.1002/hbm.22683. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kogler L, Muller VI, Seidel EM, Boubela R, Kalcher K, Moser E, Habel U, Gur RC, Eickhoff SB, Derntl B. Sex differences in the functional connectivity of the amygdalae in association with cortisol. Neuroimage. 2016;134:410–423. doi: 10.1016/j.neuroimage.2016.03.064. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kogler L, Seidel EM, Metzler H, Thaler H, Boubela RN, Pruessner JC, Kryspin-Exner I, Gur RC, Windischberger C, Moser E, Habel U, Derntl B. Impact of self-esteem and sex on stress reactions. Sci Rep. 2017;7 doi: 10.1038/s41598-017-17485-w. 17210. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kole MH, Costoli T, Koolhaas JM, Fuchs E. Bidirectional shift in the cornu ammonis 3 pyramidal dendritic organization following brief stress. Neuroscience. 2004;125:337–347. doi: 10.1016/j.neuroscience.2004.02.014. [DOI] [PubMed] [Google Scholar]
- Komatsuzaki Y, Murakami G, Tsurugizawa T, Mukai H, Tanabe N, Mitsuhashi K, Kawata M, Kimoto T, Ooishi Y, Kawato S. Rapid spinogenesis of pyramidal neurons induced by activation of glucocorticoid receptors in adult male rat hippocampus. Biochem Biophys Res Commun. 2005;335:1002–1007. doi: 10.1016/j.bbrc.2005.07.173. [DOI] [PubMed] [Google Scholar]
- Kong L, Chen K, Womer F, Jiang W, Luo X, Driesen N, Liu J, Blumberg H, Tang Y, Xu K, Wang F. Sex differences of gray matter morphology in cortico-limbic-striatal neural system in major depressive disorder. J Psychiatr Res. 2013;47:733–739. doi: 10.1016/j.jpsychires.2013.02.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kong F, Zhen Z, Li J, Huang L, Wang X, Song Y, Liu J. Sex-related neu-roanatomical basis of emotion regulation ability. PLoS ONE. 2014;9:e97071. doi: 10.1371/journal.pone.0097071. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Koss WA, Frick KM. Sex differences in hippocampal function. J Neurosci Res. 2017;95:539–562. doi: 10.1002/jnr.23864. [DOI] [PubMed] [Google Scholar]
- Kretz O, Fester L, Wehrenberg U, Zhou L, Brauckmann S, Zhao S, Prange-Kiel J, Naumann T, Jarry H, Frotscher M, Rune GM. Hippocampal synapses depend on hippocampal estrogen synthesis. J Neurosci. 2004;24:5913–5921. doi: 10.1523/JNEUROSCI.5186-03.2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kuipers SD, Trentani A, Westenbroek C, Bramham CR, Korf J, Kema IP, Ter Horst GJ, Den Boer JA. Unique patterns of FOS, phospho-CREB and BrdU immunoreactivity in the female rat brain following chronic stress and citalopram treatment. Neuropharmacology. 2006;50:428–440. doi: 10.1016/j.neuropharm.2005.10.006. [DOI] [PubMed] [Google Scholar]
- Kukolja J, Thiel CM, Wolf OT, Fink GR. Increased cortisol levels in cogni-tively challenging situations are beneficial in young but not older subjects. Psychopharmacology. 2008;201:293–304. doi: 10.1007/s00213-008-1275-8. [DOI] [PubMed] [Google Scholar]
- Kukolja J, Klingmuller D, Maier W, Fink GR, Hurlemann R. Noradrenergic-glucocorticoid modulation of emotional memory encoding in the human hippocampus. Psychol Med. 2011;41:2167–2176. doi: 10.1017/S0033291711000183. [DOI] [PubMed] [Google Scholar]
- Lagace DC, Fischer SJ, Eisch AJ. Gender and endogenous levels of estradiol do not influence adult hippocampal neurogenesis in mice. Hippocampus. 2007;17:175–180. doi: 10.1002/hipo.20265. [DOI] [PubMed] [Google Scholar]
- Lajud N, Roque A, Cajero M, Gutierrez-Ospina G, Torner L. Periodic maternal separation decreases hippocampal neurogenesis without affecting basal corticosterone during the stress hyporesponsive period, but alters HPA axis and coping behavior in adulthood. Psychoneuroendocrinology. 2012;37:410–420. doi: 10.1016/j.psyneuen.2011.07.011. [DOI] [PubMed] [Google Scholar]
- Landre L, Destrieux C, Baudry M, Barantin L, Cottier JP, Martineau J, Hommet C, Isingrini M, Belzung C, Gaillard P, Camus V, El Hage W. Preserved subcortical volumes and cortical thickness in women with sexual abuse-related PTSD. Psychiatry Res. 2010;183:181–186. doi: 10.1016/j.pscychresns.2010.01.015. [DOI] [PubMed] [Google Scholar]
- Langelaan S, Bakker AB, Schaufeli WB, van Rhenen W, van Doornen LJ. Do burned-out and work-engaged employees differ in the functioning of the hypotha-lamic-pituitary-adrenal axis? Scand. J Work Environ Health. 2006;32:339–348. doi: 10.5271/sjweh.1029. [DOI] [PubMed] [Google Scholar]
- Lassalle JM, Bataille T, Halley H. Reversible inactivation of the hippocampal mossy fiber synapses in mice impairs spatial learning, but neither consolidation nor memory retrieval, in the Morris navigation task. Neurobiol Learn Mem. 2000;73:243–257. doi: 10.1006/nlme.1999.3931. [DOI] [PubMed] [Google Scholar]
- Lawton CA. Gender differences in way-finding strategies: relationship to spatial ability and spatial anxiety. Sex Roles. 1994;30:765–779. [Google Scholar]
- Lawton CA. Gender and regional differences in spatial referents used in direction giving. Sex Roles. 2001;44:321–337. [Google Scholar]
- Lawton C, Charleston S, Zieles A. Individual- and gender-related differences in indoor wayfinding. Environ Behav. 1996;28:204–219. [Google Scholar]
- Le Melledo JM, Baker G. Role of progesterone and other neuroactive steroids in anxiety disorders. Expert Rev Neurother. 2004;4:851–860. doi: 10.1586/14737175.4.5.851. [DOI] [PubMed] [Google Scholar]
- Lebron-Milad K, Abbs B, Milad MR, Linnman C, Rougemount-Bucking A, Zeidan MA, Holt DJ, Goldstein JM. Sex differences in the neurobiology of fear conditioning and extinction: a preliminary fMRI study of shared sex differences with stress-arousal circuitry. Biol Mood Anxiety Disord. 2012;2:7. doi: 10.1186/2045-5380-2-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lee JH, Byun MS, Yi D, Choe YM, Choi HJ, Baek H, Sohn BK, Lee JY, Kim HJ, Kim JW, Lee Y, et al. Sex-specific association of sex hormones and gonadotropins, with brain amyloid and hippocampal neurodegeneration. Neurobiol Aging. 2017;58:34–40. doi: 10.1016/j.neurobiolaging.2017.06.005. [DOI] [PubMed] [Google Scholar]
- Lee SJ, Campomanes CR, Sikat PT, Greenfield AT, Allen PB, McEwen BS. Estrogen induces phosphorylation of cyclic AMP response element binding (pCREB) in primary hippocampal cells in a time-dependent manner. Neuroscience. 2004;124:549–560. doi: 10.1016/j.neuroscience.2003.11.035. [DOI] [PubMed] [Google Scholar]
- Lee KJ, Kim SJ, Kim SW, Choi SH, Shin YC, Park SH, Moon BH, Cho E, Lee MS, Choi SH, Chun BG, et al. Chronic mild stress decreases survival, but not proliferation, of new-born cells in adult rat hippocampus. Exp Mol Med. 2006;38:44–54. doi: 10.1038/emm.2006.6. [DOI] [PubMed] [Google Scholar]
- Lemaire V, Koehl M, Le Moal M, Abrous DN. Prenatal stress produces learning deficits associated with an inhibition of neurogenesis in the hippocampus. Proc Natl Acad Sci U S A. 2000;97:11032–11037. doi: 10.1073/pnas.97.20.11032. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Leslie AT, Akers KG, Krakowski AD, Stone SS, Sakaguchi M, Arruda-Carvalho M, Frankland PW. Impact of early adverse experience on complexity of adult-generated neurons. Transl Psychiatry. 2011;1:e35. doi: 10.1038/tp.2011.38. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lete I, Lapuente O. Contraceptive options for women with premenstrual dysphoric disorder: current insights and a narrative review. Open Access J Contracept. 2016;7:117–125. doi: 10.2147/OAJC.S97013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Leuner B, Gould E, Shors TJ. Is there a link between adult neurogenesis and learning? Hippocampus. 2006;16:216–224. doi: 10.1002/hipo.20153. [DOI] [PubMed] [Google Scholar]
- Lewin C, Herlitz A. Sex differences in face recognition-women's faces make the difference. Brain Cogn. 2002;50:121–128. doi: 10.1016/s0278-2626(02)00016-7. [DOI] [PubMed] [Google Scholar]
- Li W, van Tol MJ, Li M, Miao W, Jiao Y, Heinze HJ, Bogerts B, He H, Walter M. Regional specificity of sex effects on subcortical volumes across the lifespan in healthy aging. Hum Brain Mapp. 2014;35:238–247. doi: 10.1002/hbm.22168. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lisofsky N, Martensson J, Eckert A, Lindenberger U, Gallinat J, Kuhn S. Hippocampal volume and functional connectivity changes during the female menstrual cycle. Neuroimage. 2015;118:154–162. doi: 10.1016/j.neuroimage.2015.06.012. [DOI] [PubMed] [Google Scholar]
- Lisofsky N, Lindenberger U, Kuhn S. Amygdala/hippocampal activation during the menstrual cycle: evidence for lateralization of effects across different tasks. Neuropsychologia. 2015;67:55–62. doi: 10.1016/j.neuropsychologia.2014.12.005. [DOI] [PubMed] [Google Scholar]
- Liu J, Chaplin TM, Wang F, Sinha R, Mayes LC, Blumberg HP. Stress reactivity and corticolimbic response to emotional faces in adolescents. J Am Acad Child Adolesc Psychiatry. 2012;51:304–312. doi: 10.1016/j.jaac.2011.12.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Liu L, Zhao L, She H, Chen S, Wang JM, Wong C, McClure K, Sitruk-Ware R, Brinton RD. Clinically relevant progestins regulate neurogenic and neuro-protective responses in vitro and in vivo. Endocrinology. 2010;151:5782–5794. doi: 10.1210/en.2010-0005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Logue MW, van Rooij SJH, Dennis EL, Davis SL, Hayes JP, Stevens JS, Densmore M, Haswell CC, Ipser J, Koch SBJ, Korgaonkar M, et al. Smaller hippocampal volume in posttraumatic stress disorder: a multisite ENIGMA-PGC study: subcortical volumetry results from posttraumatic stress disorder consortia. Biol Psychiatry. 2018;83:244–253. doi: 10.1016/j.biopsych.2017.09.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lord C, Buss C, Lupien SJ, Pruessner JC. Hippocampal volumes are larger in postmenopausal women using estrogen therapy compared to past users, never users and men: a possible window of opportunity effect. Neurobiol Aging. 2008;29:95–101. doi: 10.1016/j.neurobiolaging.2006.09.001. [DOI] [PubMed] [Google Scholar]
- Lord C, Engert V, Lupien SJ, Pruessner JC. Effect of sex and estrogen therapy on the aging brain: a voxel-based morphometry study. Menopause. 2010;17:846–851. doi: 10.1097/gme.0b013e3181e06b83. [DOI] [PubMed] [Google Scholar]
- Lorenzetti V, Allen NB, Fornito A, Yucel M. Structural brain abnormalities in major depressive disorder: a selective review of recent MRI studies. J Affect Disord. 2009;117:1–17. doi: 10.1016/j.jad.2008.11.021. [DOI] [PubMed] [Google Scholar]
- Lovallo WR, Robinson JL, Glahn DC, Fox PT. Acute effects of hydrocortisone on the human brain: an fMRI study. Psychoneuroendocrinology. 2010;35:15–20. doi: 10.1016/j.psyneuen.2009.09.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Luo J, Wang T, Liang S, Hu X, Li W, Jin F. Experimental gastritis leads to anxiety- and depression-like behaviors in female but not male rats. Behav Brain Funct. 2013;9:46. doi: 10.1186/1744-9081-9-46. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lupien SJ, de Leon M, de Santi S, Convit A, Tarshish C, Nair NP, Thakur M, McEwen BS, Hauger RL, Meaney MJ. Cortisol levels during human aging predict hippocampal atrophy and memory deficits. Nat Neurosci. 1998;1:69–73. doi: 10.1038/271. [DOI] [PubMed] [Google Scholar]
- Lymer J, Robinson A, Winters BD, Choleris E. Rapid effects of dorsal hippo-campal G-protein coupled estrogen receptor on learning in female mice. Psychoneuroendocrinology. 2017;77:131–140. doi: 10.1016/j.psyneuen.2016.11.019. [DOI] [PubMed] [Google Scholar]
- Mackiewicz KL, Sarinopoulos I, Cleven KL, Nitschke JB. The effect of anticipation and the specificity of sex differences for amygdala and hippocampus function in emotional memory. Proc Natl Acad Sci USA. 2006;103:14200–14205. doi: 10.1073/pnas.0601648103. [DOI] [PMC free article] [PubMed] [Google Scholar]
- MacLusky NJ, Luine VN, Hajszan T, Leranth C. The 17alpha and 17beta isomers of estradiol both induce rapid spine synapse formation in the CA1 hippo-campal subfield of ovariectomized female rats. Endocrinology. 2005;146:287–293. doi: 10.1210/en.2004-0730. [DOI] [PubMed] [Google Scholar]
- MacMaster FP, Carrey N, Langevin LM, Jaworska N, Crawford S. Disorder-specific volumetric brain difference in adolescent major depressive disorder and bipolar depression. Brain Imaging Behav. 2014;8:119–127. doi: 10.1007/s11682-013-9264-x. [DOI] [PubMed] [Google Scholar]
- Magarinos AM, McEwen BS. Stress-induced atrophy of apical dendrites of hippocampal CA3c neurons: involvement of glucocorticoid secretion and excitatory amino acid receptors. Neuroscience. 1995;69:89–98. doi: 10.1016/0306-4522(95)00259-l. [DOI] [PubMed] [Google Scholar]
- Mahmoud R, Wainwright SR, Galea LA. Sex hormones and adult hippocampal neurogenesis: Regulation, implications, and potential mechanisms. Front Neuroendocrinol. 2016;41:129–152. doi: 10.1016/j.yfrne.2016.03.002. [DOI] [PubMed] [Google Scholar]
- Maki PM. Critical window hypothesis of hormone therapy and cognition: a scientific update on clinical studies. Menopause. 2013;20:695–709. doi: 10.1097/GME.0b013e3182960cf8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Maki PM, Dumas J. Mechanisms of action ofestrogen in the brain: insights from human neuroimaging and psychopharmacologic studies. Semin Reprod Med. 2009;27:250–259. doi: 10.1055/s-0029-1216278. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Maki PM, Dennerstein L, Clark M, Guthrie J, LaMontagne P, Fornelli D, Little D, Henderson VW, Resnick SM. Perimenopausal use of hormone therapy is associated with enhanced memory and hippocampal function later in life. Brain Res. 2011;1379:232–243. doi: 10.1016/j.brainres.2010.11.030. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Maki PM, Resnick SM. Longitudinal effects of estrogen replacement therapy on PET cerebral blood flow and cognition. Neurobiol Aging. 2000;21:373–383. doi: 10.1016/s0197-4580(00)00123-8. [DOI] [PubMed] [Google Scholar]
- Maki PM, Mordecai KL, Rubin LH, Sundermann E, Savarese A, Eatough E, Drogos L. Menstrual cycle effects on cortisol responsivity and emotional retrieval following a psychosocial stressor. Horm Behav. 2015;74:201–208. doi: 10.1016/j.yhbeh.2015.06.023. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Malberg JE, Duman RS. Cell proliferation in adult hippocampus is decreased by inescapable stress: reversal by fluoxetine treatment. Neuropsychopharmacology. 2003;28:1562–1571. doi: 10.1038/sj.npp.1300234. [DOI] [PubMed] [Google Scholar]
- Maller JJ, Reglade-Meslin C, Anstey KJ, Sachdev P. Sex and symmetry differences in hippocampal volumetrics: before and beyond the opening of the crus of the fornix. Hippocampus. 2006;16:80–90. doi: 10.1002/hipo.20133. [DOI] [PubMed] [Google Scholar]
- Malykhin NV, Huang Y, Hrybouski S, Olsen F. Differential vulnerability of hippocampal subfields and anteroposterior hippocampal subregions in healthy cognitive aging. Neurobiol Aging. 2017;59:121–134. doi: 10.1016/j.neurobiolaging.2017.08.001. [DOI] [PubMed] [Google Scholar]
- Mandyam CD, Crawford EF, Eisch AJ, Rivier CL, Richardson HN. Stress experienced in utero reduces sexual dichotomies in neurogenesis, microenvironment, and cell death in the adult rat hippocampus. Dev Neurobiol. 2008;68:575–589. doi: 10.1002/dneu.20600. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Mareckova K, Holsen LM, Admon R, Makris N, Seidman L, Buka S, Whitfield-Gabrieli S, Goldstein JM. Brain activity and connectivity in response to negative affective stimuli: impact of dysphoric mood and sex across diagnoses. Hum Brain Mapp. 2016;37(11):3733–3744. doi: 10.1002/hbm.23271. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Mareckova K, Holsen L, Admon R, Whitfield-Gabrieli S, Seidman LJ, Buka SL, Klibanski A, Goldstein JM. Neural - hormonal responses to negative affective stimuli: impact of dysphoric mood and sex. J Affect Disord. 2017;222:88–97. doi: 10.1016/j.jad.2017.06.050. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Marinari KT, Leshner AI, Doyle MP. Menstrual cycle status and adrenocortical reactivity to psychological stress. Psychoneuroendocrinology. 1976;1:213–218. doi: 10.1016/0306-4530(76)90011-1. [DOI] [PubMed] [Google Scholar]
- Markham JA, McKian KP, Stroup TS, Juraska JM. Sexually dimorphic aging of dendritic morphology in CA1 of hippocampus. Hippocampus. 2005;15:97–103. doi: 10.1002/hipo.20034. [DOI] [PubMed] [Google Scholar]
- Markowska AL. Sex dimorphisms in the rate of age-related decline in spatial memory: relevance to alterations in the estrous cycle. J Neurosci: Off J Soc Neurosci. 1999;19:8122–8133. doi: 10.1523/JNEUROSCI.19-18-08122.1999. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Mazzucco CA, Lieblich SE, Bingham BI, Williamson MA, Viau V, Galea LA. Both estrogen receptor alpha and estrogen receptor beta agonists enhance cell proliferation in the dentate gyrus of adult female rats. Neuroscience. 2006;141:1793–1800. doi: 10.1016/j.neuroscience.2006.05.032. [DOI] [PubMed] [Google Scholar]
- McClure RE, Barha CK, Galea LA. 17beta-Estradiol, but not estrone, increases the survival and activation of new neurons in the hippocampus in response to spatial memory in adult female rats. Horm Behav. 2013;63:144–157. doi: 10.1016/j.yhbeh.2012.09.011. [DOI] [PubMed] [Google Scholar]
- McCormick CM, Nixon F, Thomas C, Lowie B, Dyck J. Hippocampal cell proliferation and spatial memory performance after social instability stress in adolescence in female rats. Behav Brain Res. 2010;208:23–29. doi: 10.1016/j.bbr.2009.11.003. [DOI] [PubMed] [Google Scholar]
- McCormick CM, Thomas CM, Sheridan CS, Nixon F, Flynn JA, Mathews IZ. Social instability stress in adolescent male rats alters hippocampal neurogen-esis and produces deficits in spatial location memory in adulthood. Hippocampus. 2012;22:1300–1312. doi: 10.1002/hipo.20966. [DOI] [PubMed] [Google Scholar]
- McEwen BS, Woolley CS. Estradiol and progesterone regulate neuronal structure and synaptic connectivity in adult as well as developing brain. Exp Gerontol. 1994;29:431–436. doi: 10.1016/0531-5565(94)90022-1. [DOI] [PubMed] [Google Scholar]
- McHugh TL, Saykin AJ, Wishart HA, Flashman LA, Cleavinger HB, Rabin LA, Mamourian AC, Shen L. Hippocampal volume and shape analysis in an older adult population. Clin Neuropsychol. 2007;21:130–145. doi: 10.1080/13854040601064534. [DOI] [PMC free article] [PubMed] [Google Scholar]
- McIntyre RS, Mancini D, Eisfeld BS, Soczynska JK, Grupp L, Konarski JZ, Kennedy SH. Calculated bioavailable testosterone levels and depression in middle-aged men. Psychoneuroendocrinology. 2006;31:1029–1035. doi: 10.1016/j.psyneuen.2006.06.005. [DOI] [PubMed] [Google Scholar]
- McKinnon MC, Yucel K, Nazarov A, MacQueen GM. A meta-analysis examining clinical predictors of hippocampal volume in patients with major depressive disorder. J Psychiatry Neurosci. 2009;34:41–54. [PMC free article] [PubMed] [Google Scholar]
- McKittrick CR, Magarinos AM, Blanchard DC, Blanchard RJ, McEwen BS, Sakai RR. Chronic social stress reduces dendritic arbors in CA3 of hippocampus and decreases binding to serotonin transporter sites. Synapse. 2000;36:85–94. doi: 10.1002/(SICI)1098-2396(200005)36:2<85::AID-SYN1>3.0.CO;2-Y. [DOI] [PubMed] [Google Scholar]
- McLean CP, Asnaani A, Litz BT, Hofmann SG. Gender differences in anxiety disorders: prevalence, course of illness, comorbidity and burden of illness. J Psychiatr Res. 2011;45:1027–1035. doi: 10.1016/j.jpsychires.2011.03.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Merz CJ, Tabbert K, Schweckendiek J, Klucken T, Vaitl D, Stark R, Wolf OT. Investigating the impact of sex and cortisol on implicit fear conditioning with fMRI. Psychoneuroendocrinology. 2010;35:33–46. doi: 10.1016/j.psyneuen.2009.07.009. [DOI] [PubMed] [Google Scholar]
- Merz CJ, Wolf OT, Schweckendiek J, Klucken T, Vaitl D, Stark R. Stress differentially affects fear conditioning in men and women. Psychoneuroendocrinology. 2013;38:2529–2541. doi: 10.1016/j.psyneuen.2013.05.015. [DOI] [PubMed] [Google Scholar]
- Merz CJ, Hamacher-Dang TC, Stark R, Wolf OT, Hermann A. Neural underpinnings of cortisol effects on fear extinction. Neuropsychopharmacology. 2018;43:384–392. doi: 10.1038/npp.2017.227. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Merz CJ, Wolf OT. Sex differences in stress effects on emotional learning. J Neurosci Res. 2017;95:93–105. doi: 10.1002/jnr.23811. [DOI] [PubMed] [Google Scholar]
- Meyer CE, Kurth F, Lepore S, Gao JL, Johnsonbaugh H, Oberoi MR, Sawiak SJ, MacKenzie-Graham A. In vivo magnetic resonance images reveal neuroana-tomical sex differences through the application of voxel-based morphometry in C57BL/6 mice. Neuroimage. 2017;163:197–205. doi: 10.1016/j.neuroimage.2017.09.027. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Milad MR, Igoe SA, Lebron-Milad K, Novales JE. Estrous cycle phase and gonadal hormones influence conditioned fear extinction. Neuroscience. 2009;164:887–895. doi: 10.1016/j.neuroscience.2009.09.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Miller KK, Perlis RH, Papakostas GI, Mischoulon D, Losifescu DV, Brick DJ, Fava M. Low-dose transdermal testosterone augmentation therapy improves depression severity in women. CNS Spectr. 2009;14:688–694. doi: 10.1017/s1092852900023944. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Milne AM, MacQueen GM, Hall GB. Abnormal hippocampal activation in patients with extensive history of major depression: an fMRI study. J Psychiatry Neurosci. 2012;37:28–36. doi: 10.1503/jpn.110004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Mirescu C, Peters JD, Gould E. Early life experience alters response of adult neurogenesis to stress. Nat Neurosci. 2004;7:841–846. doi: 10.1038/nn1290. [DOI] [PubMed] [Google Scholar]
- Moffat SD, Resnick SM. Long-term measures of free testosterone predict regional cerebral blood flow patterns in elderly men. Neurobiol Aging. 2007;28:914–920. doi: 10.1016/j.neurobiolaging.2006.04.001. [DOI] [PubMed] [Google Scholar]
- Moffat SD, Zonderman AB, Metter EJ, Kawas C, Blackman MR, Harman SM, Resnick SM. Free testosterone and risk for Alzheimer disease in older men. Neurology. 2004;62:188–193. doi: 10.1212/wnl.62.2.188. [DOI] [PubMed] [Google Scholar]
- Montero-Lopez E, Santos-Ruiz A, Garcia-Rios MC, Rodriguez-Blazquez M, Rogers HL, Peralta-Ramirez MI. The relationship between the menstrual cycle and cortisol secretion: daily and stress-invoked cortisol patterns. Int J Psychophysiol. 2018;131:67–72. doi: 10.1016/j.ijpsycho.2018.03.021. [DOI] [PubMed] [Google Scholar]
- Moon CM, Kim GW, Jeong GW. Whole-brain gray matter volume abnormalities in patients with generalized anxiety disorder: voxel-based morphometry. NeuroReport. 2014;25:184–189. doi: 10.1097/WNR.0000000000000100. [DOI] [PubMed] [Google Scholar]
- Morris RG, Garrud P, Rawlins JN, O'Keefe J. Place navigation impaired in rats with hippocampal lesions. Nature. 1982;297:681–683. doi: 10.1038/297681a0. [DOI] [PubMed] [Google Scholar]
- Moses-Kolko EL, Berga SL, Kalro B, Sit DK, Wisner KL. Transdermal estradiol for postpartum depression: a promising treatment option. Clin Obstet Gynecol. 2009;52:516–529. doi: 10.1097/GRF.0b013e3181b5a395. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Mueller SC. Magnetic resonance imaging in paediatric psychoneuroendocrinology: a new frontier for understanding the impact of hormones on emotion and cognition. J Neuroendocrinol. 2013;25:762–770. doi: 10.1111/jne.12048. [DOI] [PubMed] [Google Scholar]
- Mueller SC, Mandell D, Leschek EW, Pine DS, Merke DP, Ernst M. Early hyperandrogenism affects the development of hippocampal function: preliminary evidence from a functional magnetic resonance imaging study of boys with familial male precocious puberty. J Child Adolesc Psychopharmacol. 2009;19:41–50. doi: 10.1089/cap.2008.031. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Mukai H, Tsurugizawa T, Murakami G, Kominami S, Ishii H, Ogiue-Ikeda M, Takata N, Tanabe N, Furukawa A, Hojo Y, Ooishi Y, et al. Rapid modulation of long-term depression and spinogenesis via synaptic estrogen receptors in hippocampal principal neurons. J Neurochem. 2007;100:950–967. doi: 10.1111/j.1471-4159.2006.04264.x. [DOI] [PubMed] [Google Scholar]
- Murphy DG, DeCarli C, McIntosh AR, Daly E, Mentis MJ, Pietrini P, Szczepanik J, Schapiro MB, Grady CL, Horwitz B, Rapoport SI. Sex differences in human brain morphometry and metabolism: an in vivo quantitative magnetic resonance imaging and positron emission tomography study on the effect of aging. Arch Gen Psychiatry. 1996;53:585–594. doi: 10.1001/archpsyc.1996.01830070031007. [DOI] [PubMed] [Google Scholar]
- Murray F, Smith DW, Hutson PH. Chronic low dose corticosterone exposure decreased hippocampal cell proliferation, volume and induced anxiety and depression like behaviours in mice. Eur J Pharmacol. 2008;583:115–127. doi: 10.1016/j.ejphar.2008.01.014. [DOI] [PubMed] [Google Scholar]
- Naghdi N, Nafisy N, Majlessi N. The effects of intrahippocampal testosterone and flutamide on spatial localization in the Morris water maze. Brain Res. 2001;897:44–51. doi: 10.1016/s0006-8993(00)03261-3. [DOI] [PubMed] [Google Scholar]
- Nagy AI, Ormerod BK, Mazzucco CA, Galea LA. Estradiol-induced enhancement in cell proliferation is mediated through estrogen receptors in the dentate gyrus of adult female rats. Drug Develop Res. 2006;66:142–149. [Google Scholar]
- Nakajima M, Allen S, Al'Absi M. Influences of the Menstrual phase on cortisol response to stress in nicotine dependent women: a preliminary examination. Nicotine Tob Res. 2019;21:617–622. doi: 10.1093/ntr/nty071. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Naninck EF, Hoeijmakers L, Kakava-Georgiadou N, Meesters A, Lazic SE, Lucassen PJ, Korosi A. Chronic early life stress alters developmental and adult neurogenesis and impairs cognitive function in mice. Hippocampus. 2014;25:309–328. doi: 10.1002/hipo.22374. [DOI] [PubMed] [Google Scholar]
- Naninck EF, Hoeijmakers L, Kakava-Georgiadou N, Meesters A, Lazic SE, Lucassen PJ, Korosi A. Chronic early life stress alters developmental and adult neurogenesis and impairs cognitive function in mice. Hippocampus. 2015;25:309–328. doi: 10.1002/hipo.22374. [DOI] [PubMed] [Google Scholar]
- Neufang S, Specht K, Hausmann M, Gunturkun O, Herpertz-Dahlmann B, Fink GR, Konrad K. Sex differences and the impact of steroid hormones on the developing human brain. Cereb Cortex. 2009;19:464–473. doi: 10.1093/cercor/bhn100. [DOI] [PubMed] [Google Scholar]
- Nguyen TV, Lew J, Albaugh MD, Botteron KN, Hudziak JJ, Fonov VS, Collins DL, Ducharme S, McCracken JT. Sex-specific associations of testosterone with prefrontal-hippocampal development and executive function. Psychoneuroendocrinology. 2017;76:206–217. doi: 10.1016/j.psyneuen.2016.12.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Nini M, Hongna Z, Zhiying L, Li Y, Xia W. Gender differences in dynamic functional connectivity based on resting-state fMRI. Conf Proc IEEE Eng Med Biol Soc; 2017. pp. 2940–2943. [DOI] [PubMed] [Google Scholar]
- Noack H, Nolte L, Nieratschker V, Habel U, Derntl B. Imaging stress: an overview of stress induction methods in the MR scanner. J Neural Transm (Vienna) 2019 doi: 10.1007/s00702-018-01965-y. [DOI] [PubMed] [Google Scholar]
- Nordin K, Herlitz A, Larsson EM, Soderlund H. Overlapping effects of age on associative memory and the anterior hippocampus from middle to older age. Behav Brain Res. 2017;317:350–359. doi: 10.1016/j.bbr.2016.10.002. [DOI] [PubMed] [Google Scholar]
- Nowakowski RS, Hayes NL, Galea LA. Cold Spring Harbour Laboratory Press; Cold Spring Harbour: Numerology of Neurogenesis: Characterizing the Cell Type of Neurostem Cells. [Google Scholar]
- Oberg C, Larsson M, Backman L. Differential sex effects in olfactory functioning: the role of verbal processing. J Int Neuropsychol Soc. 2002;8:691–698. doi: 10.1017/s1355617702801424. [DOI] [PubMed] [Google Scholar]
- Oberlander JG, Woolley CS. 17beta-Estradiol acutely potentiates glutamatergic synaptic transmission in the hippocampus through distinct mechanisms in males and females. J Neurosci. 2016;36:2677–2690. doi: 10.1523/JNEUROSCI.4437-15.2016. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Oei NY, Elzinga BM, Wolf OT, de Ruiter MB, Damoiseaux JS, Kuijer JP, Veltman DJ, Scheltens P, Rombouts SA. Glucocorticoids decrease hippocampal and prefrontal activation during declarative memory retrieval in young men. Brain Imaging Behav. 2007;1:31–41. doi: 10.1007/s11682-007-9003-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ohnishi T, Matsuda H, Hirakata M, Ugawa Y. Navigation ability dependent neural activation in the human brain: an fMRI study. Neurosci Res. 2006;55:361–369. doi: 10.1016/j.neures.2006.04.009. [DOI] [PubMed] [Google Scholar]
- Olderbak S, Wilhelm O, Hildebrandt A, Quoidbach J. Sex differences in facial emotion perception ability across the lifespan. Cogn Emot. 2019;33:579–588. doi: 10.1080/02699931.2018.1454403. [DOI] [PubMed] [Google Scholar]
- Olff M. Sex and gender differences in post-traumatic stress disorder: an update. Eur J Psychotraumatol. 2017:8. [Google Scholar]
- Ooishi Y, Kawato S, Hojo Y, Hatanaka Y, Higo S, Murakami G, Komatsuzaki Y, Ogiue-Ikeda M, Kimoto T, Mukai H. Modulation of synaptic plasticity in the hippocampus by hippocampus-derived estrogen and androgen. J Steroid Biochem Mol Biol. 2012;131:37–51. doi: 10.1016/j.jsbmb.2011.10.004. [DOI] [PubMed] [Google Scholar]
- Oomen CA, Girardi CE, Cahyadi R, Verbeek EC, Krugers H, Joels M, Lucassen PJ. Opposite effects of early maternal deprivation on neurogenesis in male versus female rats. PLoS ONE. 2009;4:e3675. doi: 10.1371/journal.pone.0003675. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Oomen CA, Soeters H, Audureau N, Vermunt L, van Hasselt FN, Manders EM, Joels M, Lucassen PJ, Krugers H. Severe early life stress hampers spatial learning and neurogenesis, but improves hippocampal synaptic plasticity and emotional learning under high-stress conditions in adulthood. J Neurosci. 2010;30:6635–6645. doi: 10.1523/JNEUROSCI.0247-10.2010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Oomen CA, Soeters H, Audureau N, Vermunt L, van Hasselt FN, Manders EM, Joels M, Krugers H, Lucassen PJ. Early maternal deprivation affects dentate gyrus structure and emotional learning in adult female rats. Psychopharmacology. 2011;214:249–260. doi: 10.1007/s00213-010-1922-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Opel N, Redlich R, Zwanzger P, Grotegerd D, Arolt V, Heindel W, Konrad C, Kugel H, Dannlowski U. Hippocampal atrophy in major depression: a function of childhood maltreatment rather than diagnosis? Neuropsychopharmacology. 2014;39:2723–2731. doi: 10.1038/npp.2014.145. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Oreland S, Nylander I, Pickering C. Prolonged maternal separation decreases granule cell number in the dentate gyrus of 3-week-old male rats. Int J Dev Neurosci. 2010;28:139–144. [Google Scholar]
- Ormerod BK, Galea LA. Reproductive status influences cell proliferation and cell survival in the dentate gyrus of adult female meadow voles: a possible regulatory role for estradiol. Neuroscience. 2001;102:369–379. doi: 10.1016/s0306-4522(00)00474-7. [DOI] [PubMed] [Google Scholar]
- Ormerod BK, Galea LA. Reproductive status influences the survival of new cells in the dentate gyrus of adult male meadow voles. Neurosci Lett. 2003;346:25–28. doi: 10.1016/s0304-3940(03)00546-9. [DOI] [PubMed] [Google Scholar]
- Ormerod BK, Lee TT, Galea LA. Estradiol initially enhances but subsequently suppresses (via adrenal steroids) granule cell proliferation in the dentate gyrus of adult female rats. J Neurobiol. 2003;55:247–260. doi: 10.1002/neu.10181. [DOI] [PubMed] [Google Scholar]
- Ormerod BK, Lee TT, Galea LA. Estradiol enhances neurogenesis in the dentate gyri of adult male meadow voles by increasing the survival of young granule neurons. Neuroscience. 2004;128:645–654. doi: 10.1016/j.neuroscience.2004.06.039. [DOI] [PubMed] [Google Scholar]
- Orsini CA, Moorman DE, Young JW, Setlow B, Floresco SB. Neural mechanisms regulating different forms of risk-related decision-making: Insights from animal models. Neurosci Biobehav Rev. 2015;58:147–167. doi: 10.1016/j.neubiorev.2015.04.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Palanza P. Animal models of anxiety and depression: how are females different? Neurosci Biobehav Rev. 2001;25:219–233. doi: 10.1016/s0149-7634(01)00010-0. [DOI] [PubMed] [Google Scholar]
- Panizzon MS, Hauger RL, Eaves LJ, Chen CH, Dale AM, Eyler LT, Fischl B, Fennema-Notestine C, Franz CE, Grant MD, Jacobson KC, et al. Genetic influences on hippocampal volume differ as a function of testosterone level in middle-aged men. Neuroimage. 2012;59:1123–1131. doi: 10.1016/j.neuroimage.2011.09.044. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Pawluski JL, Brummelte S, Barha CK, Crozier TM, Galea LA. Effects of steroid hormones on neurogenesis in the hippocampus of the adult female rodent during the estrous cycle, pregnancy, lactation and aging. Front Neuroendocrinol. 2009;30:343–357. doi: 10.1016/j.yfrne.2009.03.007. [DOI] [PubMed] [Google Scholar]
- Pegues MP, Rogers LJ, Amend D, Vinogradov S, Deicken RF. Anterior hippocampal volume reduction in male patients with schizophrenia. Schizophr Res. 2003;60:105–115. doi: 10.1016/s0920-9964(02)00288-8. [DOI] [PubMed] [Google Scholar]
- Perani CV, Slattery DA. Using animal models to study post-partum psychiatric disorders. Br J Pharmacol. 2014;171:4539–4555. doi: 10.1111/bph.12640. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Perez-Martin M, Azcoitia I, Trejo JL, Sierra A, Garcia-Segura LM. An antagonist of estrogen receptors blocks the induction of adult neurogenesis by insulinlike growth factor-I in the dentate gyrus of adult female rat. Eur J Neurosci. 2003;18:923–930. doi: 10.1046/j.1460-9568.2003.02830.x. [DOI] [PubMed] [Google Scholar]
- Perez-Martin M, Salazar V, Castillo C, Ariznavarreta C, Azcoitia I, Garcia-Segura LM, Tresguerres JA. Estradiol and soy extract increase the production of new cells in the dentate gyrus of old rats. Exp Gerontol. 2005;40:450–453. doi: 10.1016/j.exger.2005.03.003. [DOI] [PubMed] [Google Scholar]
- Perl DP. The Mount Sinai J Med. Vol. 77. New York; 2010. Neuropathology of Alzheimer's disease; pp. 32–42. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Perlaki G, Orsi G, Plozer E, Altbacker A, Darnai G, Nagy SA, Horvath R, Toth A, Doczi T, Kovacs N, Bogner P, et al. Are there any gender differences in the hippocampus volume after head-size correction? A volumetric and voxel-based morphometric study. Neurosci Lett. 2014;570:119–123. doi: 10.1016/j.neulet.2014.04.013. [DOI] [PubMed] [Google Scholar]
- Perrot-Sinal TS, Kostenuik MA, Ossenkopp KP, Kavaliers M. Sex differences in performance in the Morris water maze and the effects of initial nonstationary hidden platform training. Behav Neurosci. 1996;110:1309–1320. doi: 10.1037//0735-7044.110.6.1309. [DOI] [PubMed] [Google Scholar]
- Persson J, Herlitz A, Engman J, Morell A, Sjolie D, Wikstrom J, Soderlund H. Remembering our origin: gender differences in spatial memory are reflected in gender differences in hippocampal lateralization. Behav Brain Res. 2013;256:219–228. doi: 10.1016/j.bbr.2013.07.050. [DOI] [PubMed] [Google Scholar]
- Persson J, Spreng RN, Turner G, Herlitz A, Morell A, Stening E, Wahlund LO, Wikstrom J, Soderlund H. Sex differences in volume and structural covar-iance of the anterior and posterior hippocampus. Neuroimage. 2014;99:215–225. doi: 10.1016/j.neuroimage.2014.05.038. [DOI] [PubMed] [Google Scholar]
- Pham K, Nacher J, Hof PR, McEwen BS. Repeated restraint stress suppresses neurogenesis and induces biphasic PSA-NCAM expression in the adult rat dentate gyrus. Eur J Neurosci. 2003;17:879–886. doi: 10.1046/j.1460-9568.2003.02513.x. [DOI] [PubMed] [Google Scholar]
- Phan A, Lancaster KE, Armstrong JN, MacLusky NJ, Choleris E. Rapid effects of estrogen receptor alpha and beta selective agonists on learning and dendritic spines in female mice. Endocrinology. 2011;152:1492–1502. doi: 10.1210/en.2010-1273. [DOI] [PubMed] [Google Scholar]
- Phan A, Gabor CS, Favaro KJ, Kaschack S, Armstrong JN, MacLusky NJ, Choleris E. Low doses of 17beta-estradiol rapidly improve learning and increase hippocampal dendritic spines. Neuropsychopharmacology. 2012;37:2299–2309. doi: 10.1038/npp.2012.82. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Phan A, Suschkov S, Molinaro L, Reynolds K, Lymer JM, Bailey CD, Kow LM, MacLusky NJ, Pfaff DW, Choleris E. Rapid increases in immature synapses parallel estrogen-induced hippocampal learning enhancements. Proc Natl Acad Sci USA. 2015;112:16018–16023. doi: 10.1073/pnas.1522150112. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Pibiri F, Nelson M, Guidotti A, Costa E, Pinna G. Decreased corticolimbic allopregnanolone expression during social isolation enhances contextual fear: a model relevant for posttraumatic stress disorder. Proc Natl Acad Sci USA. 2008;105:5567–5572. doi: 10.1073/pnas.0801853105. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Pillemer DB, Wink P, DiDonato TE, Sanborn RL. Gender differences in autobiographical memory styles of older adults. Memory. 2003;11:525–532. doi: 10.1080/09658210244000117. [DOI] [PubMed] [Google Scholar]
- Pintzka CW, Evensmoen HR, Lehn H, Haberg AK. Changes in spatial cognition and brain activity after a single dose of testosterone in healthy women. Behav Brain Res. 2016;298:78–90. doi: 10.1016/j.bbr.2015.10.056. [DOI] [PubMed] [Google Scholar]
- Pintzka CW, Haberg AK. Perimenopausal hormone therapy is associated with regional sparing of the CA1 subfield: a HUNT MRI study. Neurobiol Aging. 2015;36:2555–2562. doi: 10.1016/j.neurobiolaging.2015.05.023. [DOI] [PubMed] [Google Scholar]
- Pletzer B. Sex-specific strategy use and global-local processing: a perspective toward integrating sex differences in cognition. Front Neurosci. 2014;8:425. doi: 10.3389/fnins.2014.00425. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Pletzer B. Sex hormones and gender role relate to greymatter volumes in sexually dimorphic brain areas. Front Neurosci. 2019 doi: 10.3389/fnins.2019.00592. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Pletzer B, Kronbichler M, Aichhorn M, Bergmann J, Ladurner G, Kerschbaum H. Menstrual cycle and hormonal contraceptive use modulate human brain structure. Brain Res. 2010;1348:55–62. doi: 10.1016/j.brainres.2010.06.019. [DOI] [PubMed] [Google Scholar]
- Pletzer B, Kronbichler M, Kerschbaum H. Differential effects of androgenic and anti-androgenic progestins on fusiform and frontal gray matter volume and face recognition performance. Brain Res. 2015;1596:108–115. doi: 10.1016/j.brainres.2014.11.025. [DOI] [PubMed] [Google Scholar]
- Pletzer B, Harris T, Hidalgo-Lopez E. Subcortical structural changes along the menstrual cycle: beyond the hippocampus. Sci Rep. 2018;8 doi: 10.1038/s41598-018-34247-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Pletzer B, Harris TA, Scheuringer A, Hidalgo-Lopez E. The cycling brain: menstrual cycle related fluctuations in hippocampal and fronto-striatal activation and connectivity during cognitive tasks. Neuropsychopharmacology. 2019 doi: 10.1038/s41386-019-0435-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Pletzer B, Harris T, Hidalgo-Lopez E. Previous contraceptive treatment relates to grey matter volumes in the hippocampus and basal ganglia. Sci Rep. 2019;9 doi: 10.1038/s41598-019-47446-4. 11003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Powers SI, Laurent HK, Gunlicks-Stoessel M, Balaban S, Bent E. Depression and anxiety predict sex-specific cortisol responses to interpersonal stress. Psychoneuroendocrinology. 2016;69:172–179. doi: 10.1016/j.psyneuen.2016.04.007. [DOI] [PubMed] [Google Scholar]
- Protopopescu X, Butler T, Pan H, Root J, Altemus M, Polanecsky M, McEwen B, Silbersweig D, Stern E. Hippocampal structural changes across the menstrual cycle. Hippocampus. 2008;18:985–988. doi: 10.1002/hipo.20468. [DOI] [PubMed] [Google Scholar]
- Pruessner JC, Dedovic K, Khalili-Mahani N, Engert V, Pruessner M, Buss C, Renwick R, Dagher A, Meaney MJ, Lupien S. Deactivation of the limbic system during acute psychosocial stress: evidence from positron emission tomography and functional magnetic resonance imaging studies. Biol Psychiatry. 2008;63:234–240. doi: 10.1016/j.biopsych.2007.04.041. [DOI] [PubMed] [Google Scholar]
- Pruessner JC, Dedovic K, Pruessner M, Lord C, Buss C, Collins L, Dagher A, Lupien SJ. Stress regulation in the central nervous system: evidence from structural and functional neuroimaging studies in human populations -2008 Curt Richter Award Winner. Psychoneuroendocrinology. 2008;35(2010):179–191. doi: 10.1016/j.psyneuen.2009.02.016. [DOI] [PubMed] [Google Scholar]
- Pruessner M, Pruessner JC, Hellhammer DH, Bruce Pike G, Lupien SJ. The associations among hippocampal volume, cortisol reactivity, and memory performance in healthy young men. Psychiatry Res. 2007;155:1–10. doi: 10.1016/j.pscychresns.2006.12.007. [DOI] [PubMed] [Google Scholar]
- Pruessner M, Lepage M, Collins DL, Pruessner JC, Joober R, Malla AK. Reduced hippocampal volume and hypothalamus-pituitary-adrenal axis function in first episode psychosis: evidence for sex differences. Neuroimage Clin. 2015;7:195–202. doi: 10.1016/j.nicl.2014.12.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Pulopulos MM, Hidalgo V, Puig-Perez S, Salvador A. Psychophysiological response to social stressors: relevance of sex and age. Psicothema. 2018;30:171–176. doi: 10.7334/psicothema2017.200. [DOI] [PubMed] [Google Scholar]
- Quas JA, Castro A, Bryce CI, Granger DA. Stress physiology and memory for emotional information: moderation by individual differences in pubertal hormones. Dev Psychol. 2018;54:1606–1620. doi: 10.1037/dev0000532. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Rayen I, Gemmel M, Pauley G, Steinbusch HW, Pawluski JL. Developmental exposure to SSRIs, in addition to maternal stress, has long-term sex-dependent effects on hippocampal plasticity. Psychopharmacology. 2015;232:1231–1244. doi: 10.1007/s00213-014-3758-0. [DOI] [PubMed] [Google Scholar]
- Raz N, Gunning-Dixon F, Head D, Rodrigue KM, Williamson A, Acker JD. Aging, sexual dimorphism, and hemispheric asymmetry of the cerebral cortex: re-plicability of regional differences in volume. Neurobiol Aging. 2004;25:377–396. doi: 10.1016/S0197-4580(03)00118-0. [DOI] [PubMed] [Google Scholar]
- Redlich R, Almeida JJ, Grotegerd D, Opel N, Kugel H, Heindel W, Arolt V, Phillips ML, Dannlowski U. Brain morphometric biomarkers distinguishing unipolar and bipolar depression. A voxel-based morphometry-pattern classification approach JAMA Psychiatry. 2014;71:1222–1230. doi: 10.1001/jamapsychiatry.2014.1100. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Reschke-Hernandez AE, Okerstrom KL, Bowles Edwards A, Tranel D. Sex and stress: Men and women show different cortisol responses to psychological stress induced by the Trier social stress test and the Iowa singing social stress test. J Neurosci Res. 2017;95:106–114. doi: 10.1002/jnr.23851. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Resnick SM, Espeland MA, Jaramillo SA, Hirsch C, Stefanick ML, Murray AM, Ockene J, Davatzikos C. Postmenopausal hormone therapy and regional brain volumes: the WHIMS-MRI study. Neurology. 2009;72:135–142. doi: 10.1212/01.wnl.0000339037.76336.cf. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Revest JM, Dupret D, Koehl M, Funk-Reiter C, Grosjean N, Piazza PV, Abrous DN. Adult hippocampal neurogenesis is involved in anxiety-related behaviors. Mol Psychiatry. 2009;14:959–967. doi: 10.1038/mp.2009.15. [DOI] [PubMed] [Google Scholar]
- Riggins T, Geng F, Botdorf M, Canada K, Cox L, Hancock GR. Protracted hippocampal development is associated with age-related improvements in memory during early childhood. Neuroimage. 2018;174:127–137. doi: 10.1016/j.neuroimage.2018.03.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Roberts AD, Wessely S, Chalder T, Papadopoulos A, Cleare AJ. Salivary cortisol response to awakening in chronic fatigue syndrome. Br J Psychiatry. 2004;184:136–141. doi: 10.1192/bjp.184.2.136. [DOI] [PubMed] [Google Scholar]
- Roca CA, Schmidt PJ, Altemus M, Deuster P, Danaceau MA, Putnam K, Rubinow DR. Differential menstrual cycle regulation of hypothalamic-pituitary-adrenal axis in women with premenstrual syndrome and controls. J Clin Endocrinol Metab. 2003;88:3057–3063. doi: 10.1210/jc.2002-021570. [DOI] [PubMed] [Google Scholar]
- Romeo RD, Lee SJ, McEwen BS. Differential stress reactivity in intact and ovariectomized prepubertal and adult female rats. Neuroendocrinology. 2004;80:387–393. doi: 10.1159/000084203. [DOI] [PubMed] [Google Scholar]
- Romer B, Pfeiffer N, Lewicka S, Ben-Abdallah N, Vogt MA, Deuschle M, Vollmayr B, Gass P. Finasteride treatment inhibits adult hippocampal neurogenesis in male mice. Pharmacopsychiatry. 2010;43:174–178. doi: 10.1055/s-0030-1249095. [DOI] [PubMed] [Google Scholar]
- Rosario ER, Chang L, Head EH, Stanczyk FZ, Pike CJ. Brain levels of sex steroid hormones in men and women during normal aging and in Alzheimer's disease. Neurobiol Aging. 2011;32:604–613. doi: 10.1016/j.neurobiolaging.2009.04.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ruigrok AN, Salimi-Khorshidi G, Lai MC, Baron-Cohen S, Lombardo MV, Tait RJ, Suckling J. A meta-analysis of sex differences in human brain structure. Neurosci Biobehav Rev. 2014;39:34–50. doi: 10.1016/j.neubiorev.2013.12.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Rummel J, Epp JR, Galea LA. Estradiol does not influence strategy choice but place strategy choice is associated with increased cell proliferation in the hippocampus of female rats. Horm Behav. 2010;58:582–590. doi: 10.1016/j.yhbeh.2010.07.009. [DOI] [PubMed] [Google Scholar]
- Rutkowsky JM, Wallace BK, Wise PM, O'Donnell ME. Effects of estradiol on ischemic factor-induced astrocyte swelling and AQP4 protein abundance. Am J Physiol Cell Physiol. 2011;301:C204–C212. doi: 10.1152/ajpcell.00399.2010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sah A, Schmuckermair C, Sartori SB, Gaburro S, Kandasamy M, Irschick R, Klimaschewski L, Landgraf R, Aigner L, Singewald N. Anxiety- rather than depression-like behavior is associated with adult neurogenesis in a female mouse model of higher trait anxiety- and comorbid depression-like behavior. Transl Psychiatry. 2012;2:e171. doi: 10.1038/tp.2012.94. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sahay A, Scobie KN, Hill AS, O'Carroll CM, Kheirbek MA, Burghardt NS, Fenton AA, Dranovsky A, Hen R. Increasing adult hippocampal neuro-genesis is sufficient to improve pattern separation. Nature. 2011;472:466–470. doi: 10.1038/nature09817. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Samplin E, Ikuta T, Malhotra AK, Szeszko PR, Derosse P. Sex differences in resilience to childhood maltreatment: effects of trauma history on hippocampal volume, general cognition and subclinical psychosis in healthy adults. J Psychiatr Res. 2013;47:1174–1179. doi: 10.1016/j.jpsychires.2013.05.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Santarelli L, Saxe M, Gross C, Surget A, Battaglia F, Dulawa S, Weisstaub N, Lee J, Duman R, Arancio O, Belzung C, et al. Requirement of hippocampal neurogenesis for the behavioral effects of antidepressants. Science. 2003;301:805–809. doi: 10.1126/science.1083328. [DOI] [PubMed] [Google Scholar]
- Sapolsky RM, Meaney MJ. Maturation of the adrenocortical stress response: neuroendocrine control mechanisms and the stress hyporesponsive period. Brain Res. 1986;396:64–76. doi: 10.1016/s0006-8993(86)80190-1. [DOI] [PubMed] [Google Scholar]
- Saravia F, Beauquis J, Pietranera L, De Nicola AF. Neuroprotective effects of estradiol in hippocampal neurons and glia of middle age mice. Psychoneuroendocrinology. 2007;32:480–492. doi: 10.1016/j.psyneuen.2007.02.012. [DOI] [PubMed] [Google Scholar]
- Satterthwaite TD, Vandekar S, Wolf DH, Ruparel K, Roalf DR, Jackson C, Elliott MA, Bilker WB, Calkins ME, Prabhakaran K, Davatzikos C, et al. Sex differences in the effect of puberty on hippocampal morphology. J Am Acad Child Adolesc Psychiatry. 2014;53:341–50 e1. doi: 10.1016/j.jaac.2013.12.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Saucier DM, Green SM, Leason J, MacFadden A, Bell S, Elias LJ. Are sex differences in navigation caused by sexually dimorphic strategies or by differences in the ability to use the strategies? Behav Neurosci. 2002;116:403–410. doi: 10.1037//0735-7044.116.3.403. [DOI] [PubMed] [Google Scholar]
- Scharfman HE, MacLusky NJ. Sex differences in hippocampal area CA3 pyramidal cells. J Neurosci Res. 2017;95:563–575. doi: 10.1002/jnr.23927. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Scharfman HE, Myers CE. Corruption of the dentate gyrus by “dominant” granule cells: Implications for dentate gyrus function in health and disease. Neurobiol Learn Mem. 2016;129:69–82. doi: 10.1016/j.nlm.2015.09.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Scheff SW, Price DA, Schmitt FA, Mufson EJ. Hippocampal synaptic loss in early Alzheimer's disease and mild cognitive impairment. Neurobiol Aging. 2006;27:1372–1384. doi: 10.1016/j.neurobiolaging.2005.09.012. [DOI] [PubMed] [Google Scholar]
- Schlotz W, Hellhammer J, Schulz P, Stone AA. Perceived work overload and chronic worrying predict weekend-weekday differences in the cortisol awakening response. Psychosom Med. 2004;66:207–214. doi: 10.1097/01.psy.0000116715.78238.56. [DOI] [PubMed] [Google Scholar]
- Schmaal L, Yucel M, Ellis R, Vijayakumar N, Simmons JG, Allen NB, Whittle S. Brain structural signatures of adolescent depressive symptom trajectories: a longitudinal magnetic resonance imaging study. J Am Acad Child Adolesc Psychiatry. 2017;56:593–601 e9. doi: 10.1016/j.jaac.2017.05.008. [DOI] [PubMed] [Google Scholar]
- Schoning S, Engelien A, Kugel H, Schafer S, Schiffbauer H, Zwitserlood P, Pletziger E, Beizai P, Kersting A, Ohrmann P, Greb RR, et al. Functional anatomy ofvisuo-spatial working memory during mental rotation is influenced by, sex menstrual cycle, and sex steroid hormones. Neuropsychologia. 2007;45:3203–3214. doi: 10.1016/j.neuropsychologia.2007.06.011. [DOI] [PubMed] [Google Scholar]
- Schuck RK, Flores RE, Fung LK. Brief report: sex/gender differences in symptomology and camouflaging in adults with autism spectrum disorder. J Autism Dev Disord. 2019;49:2597–2604. doi: 10.1007/s10803-019-03998-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Schuhmacher A, Mossner R, Jessen F, Scheef L, Block W, Belloche AC, Lennertz L, Welper H, Hofels S, Pfeiffer U, Wagner M, et al. Association of amygdala volumes with cortisol secretion in unipolar depressed patients. Psychiatry Res. 2012;202:96–103. doi: 10.1016/j.pscychresns.2011.09.007. [DOI] [PubMed] [Google Scholar]
- Schwabe L, Tegenthoff M, Hoffken O, Wolf OT. Mineralocorticoid receptor blockade prevents stress-induced modulation of multiple memory systems in the human brain. Biol Psychiatry. 2013;74:801–808. doi: 10.1016/j.biopsych.2013.06.001. [DOI] [PubMed] [Google Scholar]
- Schwabe L, Wolf OT. Stress modulates the engagement of multiple memory systems in classification learning. J Neurosci. 2012;32:11042–11049. doi: 10.1523/JNEUROSCI.1484-12.2012. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sebastian V, Estil JB, Chen D, Schrott LM, Serrano PA. Acute physiological stress promotes clustering of synaptic markers and alters spine morphology in the hippocampus. PLoS ONE. 2013;8:e79077. doi: 10.1371/journal.pone.0079077. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Seeman MV. Psychopathology in women and men: focus on female hormones. Am J Psychiatry. 1997;154:1641–1647. doi: 10.1176/ajp.154.12.1641. [DOI] [PubMed] [Google Scholar]
- Seeman TE, McEwen BS, Singer BH, Albert MS, Rowe JW. Increase in urinary cortisol excretion and memory declines: MacArthur studies of successful aging. J Clin Endocrinol Metab. 1997;82:2458–2465. doi: 10.1210/jcem.82.8.4173. [DOI] [PubMed] [Google Scholar]
- Seiger R, Hahn A, Hummer A, Kranz GS, Ganger S, Woletz M, Kraus C, Sladky R, Kautzky A, Kasper S, Windischberger C, et al. Subcortical gray matter changes in transgender subjects after long-term cross-sex hormone administration. Psychoneuroendocrinology. 2016;74:371–379. doi: 10.1016/j.psyneuen.2016.09.028. [DOI] [PubMed] [Google Scholar]
- Seo D, Jia Z, Lacadie CM, Tsou KA, Bergquist K, Sinha R. Sex differences in neural responses to stress and alcohol context cues. Hum Brain Mapp. 2011;32:1998–2013. doi: 10.1002/hbm.21165. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Seo D, Rabinowitz AG, Douglas RJ, Sinha R. Limbic response to stress linking life trauma and hypothalamus-pituitary-adrenal axis function. Psychoneuroendocrinology. 2019;99:38–46. doi: 10.1016/j.psyneuen.2018.08.023. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Shafir T, Love T, Berent-Spillson A, Persad CC, Wang H, Reame NK, Frey KA, Zubieta JK, Smith YR. Postmenopausal hormone use impact on emotion processing circuitry. Behav Brain Res. 2012;226:147–153. doi: 10.1016/j.bbr.2011.09.012. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sheline YI, Sanghavi M, Mintun MA, Gado MH. Depression duration but not age predicts hippocampal volume loss in medically healthy women with recurrent major depression. J Neurosci. 1999;19:5034–5043. doi: 10.1523/JNEUROSCI.19-12-05034.1999. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sheppard PAS, Choleris E, Galea LAM. Structural plasticity of the hippocampus in response to estrogens in female rodents. Mol Brain. 2019;12:22. doi: 10.1186/s13041-019-0442-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sheridan MA, How J, Araujo M, Schamberg MA, Nelson CA. What are the links between maternal social status, hippocampal function, and HPA axis function in children? Dev Sci. 2013;16:665–675. doi: 10.1111/desc.12087. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Shima N, Yamaguchi Y, Yuri K. Distribution of estrogen receptor beta mRNA-containing cells in ovariectomized and estrogen-treated female rat brain. Anat Sci Int. 2003;78:85–97. doi: 10.1046/j.0022-7722.2003.00042.x. [DOI] [PubMed] [Google Scholar]
- Shin NY, Bak Y, Nah Y, Han S, Kim DJ, Kim SJ, Lee JE, Lee SG, Lee SK. Author correction: disturbed retrieval network and prospective memory decline in postpartum women. Sci Rep. 2018;8 doi: 10.1038/s41598-018-23875-5. 14437. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Shin NY, Bak Y, Nah Y, Han S, Kim DJ, Kim SJ, Lee JE, Lee SG, Lee SK. Disturbed retrieval network and prospective memory decline in postpartum women. Sci Rep. 2018;8 doi: 10.1038/s41598-018-23875-5. 5476. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Shinohara Y, Hirase H, Watanabe M, Itakura M, Takahashi M, Shigemoto R. Left-right asymmetry of the hippocampal synapses with differential subunit allocation of glutamate receptors. Proc Natl Acad Sci USA. 2008;105:19498–19503. doi: 10.1073/pnas.0807461105. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Shores MM, Sloan KL, Matsumoto AM, Moceri VM, Felker B, Kivlahan DR. Increased incidence of diagnosed depressive illness in hypogonadal older men. Arch Gen Psychiatry. 2004;61:162–167. doi: 10.1001/archpsyc.61.2.162. [DOI] [PubMed] [Google Scholar]
- Shores MM, Moceri VM, Sloan KL, Matsumoto AM, Kivlahan DR. Low testosterone levels predict incident depressive illness in older men: effects of age and medical morbidity. J Clin Psychiatry. 2005;66:7–14. doi: 10.4088/jcp.v66n0102. [DOI] [PubMed] [Google Scholar]
- Shores MM, Kivlahan DR, Sadak TI, Li EJ, Matsumoto AM. A randomized, double-blind, placebo-controlled study of testosterone treatment in hypogonadal older men with subthreshold depression (dysthymia or minor depression) J Clin Psychiatry. 2009;70:1009–1016. doi: 10.4088/jcp.08m04478. [DOI] [PubMed] [Google Scholar]
- Shors TJ, Chua C, Falduto J. Sex differences and opposite effects of stress on dendritic spine density in the male versus female hippocampus. J Neurosci. 2001;21:6292–6297. doi: 10.1523/JNEUROSCI.21-16-06292.2001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Shors TJ, Miesegaes G, Beylin A, Zhao M, Rydel T, Gould E. Neurogenesis in the adult is involved in the formation of trace memories. Nature. 2001;410:372–376. doi: 10.1038/35066584. [DOI] [PubMed] [Google Scholar]
- Shors TJ, Mathew J, Sisti HM, Edgecomb C, Beckoff S, Dalla C. Neurogenesis and helplessness are mediated by controllability in males but not in females. Biol Psychiatry. 2007;62:487–495. doi: 10.1016/j.biopsych.2006.10.033. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Shughrue PJ, Lane MV, Merchenthaler I. Comparative distribution of estrogen receptor-alpha and -beta mRNA in the rat central nervous system. J Comp Neurol. 1997;388:507–525. doi: 10.1002/(sici)1096-9861(19971201)388:4<507::aid-cne1>3.0.co;2-6. [DOI] [PubMed] [Google Scholar]
- Siddiqui A, Romeo RD. Sex differences and similarities in hippocampal cellular proliferation and the number of immature neurons during adolescence in rats. Dev Neurosci. 2019;41:132–138. doi: 10.1159/000502056. [DOI] [PubMed] [Google Scholar]
- Silva-Gomez AB, Aguilar-Salgado Y, Reyes-Hernandez DO, Flores G. Dexamethasone induces different morphological changes in the dorsal and ventral hippocampus of rats. J Chem Neuroanat. 2013;47:71–78. doi: 10.1016/j.jchemneu.2012.12.004. [DOI] [PubMed] [Google Scholar]
- Silverman II, Choi J, Mackewn A, Fisher M, Moro J, Olshansky E. Evolved mechanisms underlying wayfinding. Further studies on the hunter-gatherer theory of spatial sex differences Evol Hum Behav. 2000;21:201–213. doi: 10.1016/s1090-5138(00)00036-2. [DOI] [PubMed] [Google Scholar]
- Sivakumar PT, Kalmady SV, Venkatasubramanian G, Bharath S, Reddy NN, Rao NP, Kovoor JM, Jain S, Varghese M. Volumetric analysis of hippocampal sub-regions in late onset depression: a 3 tesla magnetic resonance imaging study. Asian J Psychiatr. 2015;13:38–43. doi: 10.1016/j.ajp.2014.11.005. [DOI] [PubMed] [Google Scholar]
- Skakkebaek A, Gravholt CH, Rasmussen PM, Bojesen A, Jensen JS, Fedder J, Laurberg P, Hertz JM, Ostergaard JR, Pedersen AD, Wallentin M. Neuroanatomical correlates of Klinefelter syndrome studied in relation to the neuropsychological profile. Neuroimage Clin. 2014;4:1–9. doi: 10.1016/j.nicl.2013.10.013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Slavich GM, Sacher J. Stress, sex hormones, inflammation, and major depressive disorder: Extending Social Signal Transduction Theory of Depression to account for sex differences in mood disorders. Psychopharmacology. 2019 doi: 10.1007/s00213-019-05326-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Smith CC, McMahon LL. Estradiol-induced increase in the magnitude of long-term potentiation is prevented by blocking NR2B-containing receptors. J Neurosci: Off J Soc Neurosci. 2006;26:8517–8522. doi: 10.1523/JNEUROSCI.5279-05.2006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sneider JT, Sava S, Rogowska J, Yurgelun-Todd DA. A preliminary study of sex differences in brain activation during a spatial navigation task in healthy adults. Percept Mot Skills. 2011;113:461–480. doi: 10.2466/04.22.24.27.PMS.113.5.461-480. [DOI] [PubMed] [Google Scholar]
- Sneider JT, Cohen-Gilbert JE, Hamilton DA, Stein ER, Golan N, Oot EN, Seraikas AM, Rohan ML, Harris SK, Nickerson LD, Silveri MM. Adolescent hippocampal and prefrontal brain activation during performance of the virtual morris water task. Front Hum Neurosci. 2018;12:238. doi: 10.3389/fnhum.2018.00238. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Soleman RS, Schagen SE, Veltman DJ, Kreukels BP, Cohen-Kettenis PT, Lambalk CB, Wouters F, Delemarre-van de Waal HA. Sex differences in verbal fluency during adolescence: a functional magnetic resonance imaging study in gender dysphoric and control boys and girls. J Sex Med. 2013;10:1969–1977. doi: 10.1111/jsm.12083. [DOI] [PubMed] [Google Scholar]
- Sousa N, Lukoyanov NV, Madeira MD, Almeida OF, Paula-Barbosa MM. Reorganization of the morphology of hippocampal neurites and synapses after stress-induced damage correlates with behavioral improvement. Neuroscience. 2000;97:253–266. doi: 10.1016/s0306-4522(00)00050-6. [DOI] [PubMed] [Google Scholar]
- Sovijit WN, Sovijit WE, Pu S, Usuda K, Inoue R, Watanabe G, Yamaguchi H, Nagaoka K. Ovarian progesterone suppresses depression and anxiety-like behaviors by increasing the Lactobacillus population of gut microbiota in ovar-iectomized mice. Neurosci Res. 2019 doi: 10.1016/j.neures.2019.04.005. [DOI] [PubMed] [Google Scholar]
- Spalletta G, Piras F, Caltagirone C, Fagioli S. Hippocampal multimodal structural changes and subclinical depression in healthy individuals. J Affect Disord. 2014;152-154:105–112. doi: 10.1016/j.jad.2013.05.068. [DOI] [PubMed] [Google Scholar]
- Spritzer MD, Galea LA. Testosterone and dihydrotestosterone, but not estradiol, enhance survival of new hippocampal neurons in adult male rats. Dev Neurobiol. 2007;67:1321–1333. doi: 10.1002/dneu.20457. [DOI] [PubMed] [Google Scholar]
- Spritzer MD, Gill M, Weinberg A, Galea LA. Castration differentially affects spatial working and reference memory in male rats. Arch Sex Behav. 2008;37:19–29. doi: 10.1007/s10508-007-9264-2. [DOI] [PubMed] [Google Scholar]
- Spritzer MD, Ibler E, Inglis W, Curtis MG. Testosterone and social isolation influence adult neurogenesis in the dentate gyrus of male rats. Neuroscience. 2011;195:180–190. doi: 10.1016/j.neuroscience.2011.08.034. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Starkman MN, Giordani B, Gebarski SS, Schteingart DE. Improvement in learning associated with increase in hippocampal formation volume. Biol Psychiatry. 2003;53:233–238. doi: 10.1016/s0006-3223(02)01750-x. [DOI] [PubMed] [Google Scholar]
- Steinman K, Ross J, Lai S, Reiss A, Hoeft F. Structural and functional neu-roimaging in Klinefelter (47, XXY) syndrome: a review of the literature and preliminary results from a functional magnetic resonance imaging study of language. Dev Disabil Res Rev. 2009;15:295–308. doi: 10.1002/ddrr.84. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Stephens MA, Mahon PB, McCaul ME, Wand GS. Hypothalamic-pituitary-adrenal axis response to acute psychosocial stress: Effects of biological sex and circulating sex hormones. Psychoneuroendocrinology. 2016;66:47–55. doi: 10.1016/j.psyneuen.2015.12.021. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Stomby A, Boraxbekk CJ, Lundquist A, Nordin A, Nilsson LG, Adolfsson R, Nyberg L, Olsson T. Higher diurnal salivary cortisol levels are related to smaller prefrontal cortex surface area in elderly men and women. Eur J Endocrinol. 2016;175:117–126. doi: 10.1530/EJE-16-0352. [DOI] [PubMed] [Google Scholar]
- Stroud LR, Papandonatos GD, D'Angelo CM, Brush B, Lloyd-Richardson EE. Sex differences in biological response to peer rejection and performance challenge across development: a pilot study. Physiol Behav. 2017;169:224–233. doi: 10.1016/j.physbeh.2016.12.005. [DOI] [PubMed] [Google Scholar]
- Suda S, Segi-Nishida E, Newton SS, Duman RS. A postpartum model in rat: behavioral and gene expression changes induced by ovarian steroid deprivation. Biol Psychiatry. 2008;64:311–319. doi: 10.1016/j.biopsych.2008.03.029. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sudheimer KD, O'Hara R, Spiegel D, Powers B, Kraemer HC, Neri E, Weiner M, Hardan A, Hallmayer J, Dhabhar FS. Cortisol, cytokines, and hippocampal volume interactions in the elderly. Front Aging Neurosci. 2014;6:153. doi: 10.3389/fnagi.2014.00153. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sundstrom Poromaa I, Gingnell M. Menstrual cycle influence on cognitive function and emotion processing-from a reproductive perspective. Front Neurosci. 2014;8:380. doi: 10.3389/fnins.2014.00380. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Suri D, Veenit V, Sarkar A, Thiagarajan D, Kumar A, Nestler EJ, Galande S, Vaidya VA. Early stress evokes age-dependent biphasic changes in hippocampal neurogenesis, BDNF expression, and cognition. Biol Psychiatry. 2013;73:658–666. doi: 10.1016/j.biopsych.2012.10.023. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Suzuki M, Hagino H, Nohara S, Zhou SY, Kawasaki Y, Takahashi T, Matsui M, Seto H, Ono T, Kurachi M. Male-specific volume expansion of the human hippocampus during adolescence. Cereb Cortex. 2005;15:187–193. doi: 10.1093/cercor/bhh121. [DOI] [PubMed] [Google Scholar]
- Szeszko PR, Lehrner A, Yehuda R. Glucocorticoids and hippocampal structure and function in PTSD. Harv Rev Psychiatry. 2018;26:142–157. doi: 10.1097/HRP.0000000000000188. [DOI] [PubMed] [Google Scholar]
- Tan A, Ma W, Vira A, Marwha D, Eliot L. The human hippocampus is not sexually-dimorphic: meta-analysis of structural MRI volumes. Neuroimage. 2016;124:350–366. doi: 10.1016/j.neuroimage.2015.08.050. [DOI] [PubMed] [Google Scholar]
- Tanapat P, Hastings NB, Reeves AJ, Gould E. Estrogen stimulates a transient increase in the number of new neurons in the dentate gyrus of the adult female rat. J Neurosci: Off J Soc Neurosci. 1999;19:5792–5801. doi: 10.1523/JNEUROSCI.19-14-05792.1999. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Tanapat P, Hastings NB, Rydel TA, Galea LA, Gould E. Exposure to foxodor inhibits cell proliferation in the hippocampus of adult rats via an adrenal hormone-dependent mechanism. J Comp Neurol. 2001;437:496–504. doi: 10.1002/cne.1297. [DOI] [PubMed] [Google Scholar]
- Tanapat P, Hastings NB, Gould E. Ovarian steroids influence cell proliferation in the dentate gyrus of the adult female rat in a dose- and time-dependent manner. J Comp Neurol. 2005;481:252–265. doi: 10.1002/cne.20385. [DOI] [PubMed] [Google Scholar]
- Tang SS, Ren Y, Ren XQ, Cao JR, Hong H, Ji H, Hu QH. ERalpha and/or ERbeta activation ameliorates cognitive impairment, neurogenesis and apoptosis in type 2 diabetes mellitus mice. Exp Neurol. 2019;311:33–43. doi: 10.1016/j.expneurol.2018.09.002. [DOI] [PubMed] [Google Scholar]
- Taylor WD, Steffens DC, Payne ME, MacFall JR, Marchuk DA, Svenson IK, Krishnan KR. Influence of serotonin transporter promoter region polymorphisms on hippocampal volumes in late-life depression. Arch Gen Psychiatry. 2005;62:537–544. doi: 10.1001/archpsyc.62.5.537. [DOI] [PubMed] [Google Scholar]
- Tessner KD, Walker EF, Dhruv SH, Hochman K, Hamann S. The relation of cortisol levels with hippocampus volumes under baseline and challenge conditions. Brain Res. 2007;1179:70–78. doi: 10.1016/j.brainres.2007.05.027. [DOI] [PubMed] [Google Scholar]
- Thomas RM, Hotsenpiller G, Peterson DA. Acute psychosocial stress reduces cell survival in adult hippocampal neurogenesis without altering proliferation. J Neurosci. 2007;27:2734–2743. doi: 10.1523/JNEUROSCI.3849-06.2007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Thomason ME, Tocco MA, Quednau KA, Bedway AR, Carre JM. Idle behaviors of the hippocampus reflect endogenous cortisol levels in youth. J Am Acad Child Adolesc Psychiatry. 2013;52:642–52 e1. doi: 10.1016/j.jaac.2013.04.004. [DOI] [PubMed] [Google Scholar]
- Toffol E, Heikinheimo O, Partonen T. Hormone therapy and mood in perime-nopausal and postmenopausal women: a narrative review. Menopause. 2015;22:564–578. doi: 10.1097/GME.0000000000000323. [DOI] [PubMed] [Google Scholar]
- Torner L, Karg S, Blume A, Kandasamy M, Kuhn HG, Winkler J, Aigner L, Neumann ID. Prolactin prevents chronic stress-induced decrease of adult hippocampal neurogenesis and promotes neuronal fate. J Neurosci. 2009;29:1826–1833. doi: 10.1523/JNEUROSCI.3178-08.2009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Totterman TH, Carlsson M, Simonsson B, Bengtsson M, Nilsson K. T-cell activation and subset patterns are altered in B-CLL and correlate with the stage of the disease. Blood. 1989;74:786–792. [PubMed] [Google Scholar]
- Toufexis DJ, Davis C, Hammond A, Davis M. Progesterone attenuates corti-cotropin-releasing factor-enhanced but not fear-potentiated startle via the activity of its neuroactive metabolite, allopregnanolone. J Neurosci. 2004;24:10280–10287. doi: 10.1523/JNEUROSCI.1386-04.2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Toufexis D, Rivarola MA, Lara H, Viau V. Stress and the reproductive axis. J Neuroendocrinol. 2014;26:573–586. doi: 10.1111/jne.12179. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Travis SG, Coupland NJ, Hegadoren K, Silverstone PH, Huang Y, Carter R, Fujiwara E, Seres P, Malykhin NV. Effects of cortisol on hippocampal subfields volumes and memory performance in healthy control subjects and patients with major depressive disorder. J Affect Disord. 2016;201:34–41. doi: 10.1016/j.jad.2016.04.049. [DOI] [PubMed] [Google Scholar]
- Tsurugizawa T, Mukai H, Tanabe N, Murakami G, Hojo Y, Kominami S, Mitsuhashi K, Komatsuzaki Y, Morrison JH, Janssen WG, Kimoto T, Kawato S. Estrogen induces rapid decrease in dendritic thorns of CA3 pyramidal neurons in adult male rat hippocampus. Biochem Biophys Res Commun. 2005;337:1345–1352. doi: 10.1016/j.bbrc.2005.09.188. [DOI] [PubMed] [Google Scholar]
- Tuscher JJ, Luine V, Frankfurt M, Frick KM. Estradiol-mediated spine changes in the dorsal hippocampus and medial prefrontal cortex of ovariectomized female mice depend on ERK and mTOR activation in the dorsal hippocampus. J Neurosci. 2016;36:1483–1489. doi: 10.1523/JNEUROSCI.3135-15.2016. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Uno H, Tarara R, Else JG, Suleman MA, Sapolsky RM. Hippocampal damage associated with prolonged and fatal stress in primates. J Neurosci. 1989;9:1705–1711. doi: 10.1523/JNEUROSCI.09-05-01705.1989. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Vachon-Presseau E, Roy M, Martel MO, Caron E, Marin MF, Chen J, Albouy G, Plante I, Sullivan MJ, Lupien SJ, Rainville P. The stress model of chronic pain: evidence from basal cortisol and hippocampal structure and function in humans. Brain. 2013;136:815–827. doi: 10.1093/brain/aws371. [DOI] [PubMed] [Google Scholar]
- Valli I, Crossley NA, Day F, Stone J, Tognin S, Mondelli V, Howes O, Valmaggia L, Pariante C, McGuire P. HPA-axis function and grey matter volume reductions: imaging the diathesis-stress model in individuals at ultra-high risk of psychosis. Transl Psychiatry. 2016;6:e797. doi: 10.1038/tp.2016.68. [DOI] [PMC free article] [PubMed] [Google Scholar]
- van Stegeren AH. Imaging stress effects on memory: a review of neuroimaging studies. Can J Psychiatry. 2009;54:16–27. doi: 10.1177/070674370905400105. [DOI] [PubMed] [Google Scholar]
- van Stegeren AH, Roozendaal B, Kindt M, Wolf OT, Joels M. Interacting noradrenergic and corticosteroid systems shift human brain activation patterns during encoding. Neurobiol Learn Mem. 2010;93:56–65. doi: 10.1016/j.nlm.2009.08.004. [DOI] [PubMed] [Google Scholar]
- van Wingen G, van Broekhoven F, Verkes RJ, Petersson KM, Backstrom T, Buitelaar J, Fernandez G. How progesterone impairs memory for biologically salient stimuli in healthy young women. J Neurosci. 2007;27:11416–11423. doi: 10.1523/JNEUROSCI.1715-07.2007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- van Wingen G, Mattern C, Verkes RJ, Buitelaar J, Fernandez G. Testosterone biases automatic memory processes in women towards potential mates. Neuroimage. 2008;43:114–120. doi: 10.1016/j.neuroimage.2008.07.002. [DOI] [PubMed] [Google Scholar]
- Viau V, Meaney MJ. Variations in the hypothalamic-pituitary-adrenal response to stress during the estrous cycle in the rat. Endocrinology. 1991;129:2503–2511. doi: 10.1210/endo-129-5-2503. [DOI] [PubMed] [Google Scholar]
- Victor TA, Drevets WC, Misaki M, Bodurka J, Savitz J. Sex differences in neural responses to subliminal sad and happy faces in healthy individuals: implications for depression. J Neurosci Res. 2017;95:703–710. doi: 10.1002/jnr.23870. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Vierk R, Glassmeier G, Zhou L, Brandt N, Fester L, Dudzinski D, Wilkars W, Bender RA, Lewerenz M, Gloger S, Graser L, et al. Aromatase inhibition abolishes LTP generation in female but not in male mice. J Neurosci. 2012;32:8116–8126. doi: 10.1523/JNEUROSCI.5319-11.2012. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Vogel S, Klumpers F, Kroes MC, Oplaat KT, Krugers HJ, Oitzl MS, Joels M, Fernandez G. A stress-induced shift from trace to delay conditioning depends on the mineralocorticoid receptor. Biol Psychiatry. 2015;78:830–839. doi: 10.1016/j.biopsych.2015.02.014. [DOI] [PubMed] [Google Scholar]
- Vogel S, Kluen LM, Fernandez G, Schwabe L. Stress leads to aberrant hip-pocampal involvement when processing schema-related information. Learn Mem. 2018;25:21–30. doi: 10.1101/lm.046003.117. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Voyer D, Bryden MP. Gender, level ofspatial ability, and lateralization ofmental rotation. Brain Cogn. 1990;13:18–29. doi: 10.1016/0278-2626(90)90037-o. [DOI] [PubMed] [Google Scholar]
- Voyer D, Voyer S, Bryden MP. Magnitude of sex differences in spatial abilities: a meta-analysis and consideration of critical variables. Psychol Bull. 1995;117:250–270. doi: 10.1037/0033-2909.117.2.250. [DOI] [PubMed] [Google Scholar]
- Voyer D, Postma A, Brake B, Imperato-McGinley J. Gender differences in object location memory: a meta-analysis. Psychon Bull Rev. 2007;14:23–38. doi: 10.3758/bf03194024. [DOI] [PubMed] [Google Scholar]
- Vythilingam M, Vermetten E, Anderson GM, Luckenbaugh D, Anderson ER, Snow J, Staib LH, Charney DS, Bremner JD. Hippocampal volume, memory, and cortisol status in major depressive disorder: effects of treatment. Biol Psychiatry. 2004;56:101–112. doi: 10.1016/j.biopsych.2004.04.002. [DOI] [PubMed] [Google Scholar]
- Waddell J, Bowers JM, Edwards NS, Jordan CL, McCarthy MM. Dysregulation of neonatal hippocampal cell genesis in the androgen insensitive Tfm rat. Horm Behav. 2013;64:144–152. doi: 10.1016/j.yhbeh.2013.05.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wainwright SR, Workman JL, Tehrani A, Hamson DK, Chow C, Lieblich SE, Galea LA. Testosterone has antidepressant-like efficacy and facilitates imi-pramine-induced neuroplasticity in male rats exposed to chronic unpredictable stress. Horm Behav. 2016;79:58–69. doi: 10.1016/j.yhbeh.2016.01.001. [DOI] [PubMed] [Google Scholar]
- Walf AA, Frye CA. A review and update of mechanisms of estrogen in the hippocampus and amygdala for anxiety and depression behavior. Neuropsychopharm: Off Publ Am Coll Neuropsychopharm. 2006;31:1097–1111. doi: 10.1038/sj.npp.1301067. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Walf AA, Frye CA. Administration of estrogen receptor beta-specific selective estrogen receptor modulators to the hippocampus decrease anxiety and depressive behavior of ovariectomized rats. Pharmacol Biochem Behav. 2007;86:407–414. doi: 10.1016/j.pbb.2006.07.003. [DOI] [PubMed] [Google Scholar]
- Walf AA, Frye CA. Estradiol decreases anxiety behavior and enhances inhibitory avoidance and gestational stress produces opposite effects. Stress. 2007;10:251–260. doi: 10.1080/00958970701220416. [DOI] [PubMed] [Google Scholar]
- Walf AA, Frye CA. Effects of two estradiol regimens on anxiety and depressive behaviors and trophic effects in peripheral tissues in a rodent model. Gend Med. 2009;6:300–311. doi: 10.1016/j.genm.2009.04.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Walf AA, Sumida K, Frye CA. Inhibiting 5alpha-reductase in the amygdala attenuates antianxiety and antidepressive behavior of naturally receptive and hormone-primed ovariectomized rats. Psychopharmacology. 2006;186:302–311. doi: 10.1007/s00213-005-0100-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wang Y, Xu Q, Li S, Li G, Zuo C, Liao S, Long Y, Li S, Joshi RM. Gender differences in anomalous subcortical morphology for children with ADHD. Neurosci Lett. 2018;665:176–181. doi: 10.1016/j.neulet.2017.12.006. [DOI] [PubMed] [Google Scholar]
- Warren SG, Juraska JM. Spatial and nonspatial learning across the rat estrous cycle. Behav Neurosci. 1997;111:259–266. doi: 10.1037//0735-7044.111.2.259. [DOI] [PubMed] [Google Scholar]
- Warren SG, Humphreys AG, Juraska JM, Greenough WT. LTP varies across the estrous cycle: enhanced synaptic plasticity in proestrus rats. Brain Res. 1995;703:26–30. doi: 10.1016/0006-8993(95)01059-9. [DOI] [PubMed] [Google Scholar]
- Watanabe R, Kakeda S, Watanabe K, Liu X, Katsuki A, Umeno-Nakano W, Hori H, Abe O, Yoshimura R, Korogi Y. Relationship between the hippocampal shape abnormality and serum cortisol levels in first-episode and drug-naive major depressive disorder patients. Depress Anxiety. 2017;34:401–409. doi: 10.1002/da.22604. [DOI] [PubMed] [Google Scholar]
- Weerda R, Muehlhan M, Wolf OT, Thiel CM. Effects of acute psychosocial stress on working memory related brain activity in men. Hum Brain Mapp. 2010;31:1418–1429. doi: 10.1002/hbm.20945. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wegman J, Fonteijn HM, van Ekert J, Tyborowska A, Jansen C, Janzen G. Gray and white matter correlates of navigational ability in humans. Hum Brain Mapp. 2014;35:2561–2572. doi: 10.1002/hbm.22349. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wei SM, Baller EB, Kohn PD, Kippenhan JS, Kolachana B, Soldin SJ, Rubinow DR, Schmidt PJ, Berman KF. Brain-derived neurotrophic factor Val(66) Met genotype and ovarian steroids interactively modulate working memory-related hippocampal function in women: a multimodal neuroimaging study. Mol Psychiatry. 2018;23:1066–1075. doi: 10.1038/mp.2017.72. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Weinstock M. Prenatal stressors in rodents: Effects on behavior. Neurobiol Stress. 2016;6:3–13. doi: 10.1016/j.ynstr.2016.08.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wessa M, Rohleder N, Kirschbaum C, Flor H. Altered cortisol awakening response in posttraumatic stress disorder. Psychoneuroendocrinology. 2006;31:209–215. doi: 10.1016/j.psyneuen.2005.06.010. [DOI] [PubMed] [Google Scholar]
- Westenbroek C, Den Boer JA, Veenhuis M, Ter Horst GJ. Chronic stress and social housing differentially affect neurogenesis in male and female rats. Brain Res Bull. 2004;64:303–308. doi: 10.1016/j.brainresbull.2004.08.006. [DOI] [PubMed] [Google Scholar]
- Whittle S, Yap MB, Sheeber L, Dudgeon P, Yucel M, Pantelis C, Simmons JG, Allen NB. Hippocampal volume and sensitivity to maternal aggressive behavior: a prospective study of adolescent depressive symptoms. Dev Psychopathol. 2011;23:115–129. doi: 10.1017/S0954579410000684. [DOI] [PubMed] [Google Scholar]
- Wieck A. Oestradiol and psychosis: clinical findings and biological mechanisms. Curr Top Behav Neurosci. 2011;8:173–187. doi: 10.1007/7854_2011_127. [DOI] [PubMed] [Google Scholar]
- Wierenga LM, Bos MGN, Schreuders E, Vd Kamp F, Peper JS, Tamnes CK, Crone EA. Unraveling age, puberty and testosterone effects on subcortical brain development across adolescence. Psychoneuroendocrinology. 2018;91:105–114. doi: 10.1016/j.psyneuen.2018.02.034. [DOI] [PubMed] [Google Scholar]
- Williams CL, Barnett AM, Meck WH. Organizational effects of early gonadal secretions on sexual differentiation in spatial memory. Behav Neurosci. 1990;104:84–97. doi: 10.1037//0735-7044.104.1.84. [DOI] [PubMed] [Google Scholar]
- Wisse LE, Biessels GJ, Stegenga BT, Kooistra M, van der Veen PH, Zwanenburg JJ, van der Graaf Y, Geerlings MI. Major depressive episodes over the course of 7 years and hippocampal subfield volumes at 7 tesla MRI: the PREDICT-MR study. J Affect Disord. 2015;175:1–7. doi: 10.1016/j.jad.2014.12.052. [DOI] [PubMed] [Google Scholar]
- Wolf OT. Stress and memory in humans: twelve years of progress? Brain Res. 2009;1293:142–154. doi: 10.1016/j.brainres.2009.04.013. [DOI] [PubMed] [Google Scholar]
- Wolf OT, Convit A, de Leon MJ, Caraos C, Qadri SF. Basal hypothalamo-pituitary-adrenal axis activity and corticotropin feedback in young and older men: relationships to magnetic resonance imaging-derived hippocampus and cingulate gyrus volumes. Neuroendocrinology. 2002;75:241–249. doi: 10.1159/000054715. [DOI] [PubMed] [Google Scholar]
- Wolfram M, Bellingrath S, Kudielka BM. The cortisol awakening response (CAR) across the female menstrual cycle. Psychoneuroendocrinology. 2011;36:905–912. doi: 10.1016/j.psyneuen.2010.12.006. [DOI] [PubMed] [Google Scholar]
- Woolley CS, McEwen BS. Roles of estradiol and progesterone in regulation of hippocampal dendritic spine density during the estrous cycle in the rat. J Comp Neurol. 1993;336:293–306. doi: 10.1002/cne.903360210. [DOI] [PubMed] [Google Scholar]
- Woolley CS, Gould E, McEwen BS. Exposure to excess glucocorticoids alters dendritic morphology of adult hippocampal pyramidal neurons. Brain Res. 1990;531:225–231. doi: 10.1016/0006-8993(90)90778-a. [DOI] [PubMed] [Google Scholar]
- Woolley CS, Gould E, Frankfurt M, McEwen BS. Naturally occurring fluctuation in dendritic spine density on adult hippocampal pyramidal neurons. J Neurosci. 1990;10:4035–4039. doi: 10.1523/JNEUROSCI.10-12-04035.1990. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Woolley DG, Vermaercke B, Op de Beeck H, Wagemans J, Gantois I, D'Hooge R, Swinnen SP, Wenderoth N. Sex differences in human virtual water maze performance: novel measures reveal the relative contribution of directional responding and spatial knowledge. Behav Brain Res. 2010;208:408–414. doi: 10.1016/j.bbr.2009.12.019. [DOI] [PubMed] [Google Scholar]
- Woon F, Hedges DW. Gender does notmoderate hippocampal volume deficits in adults with posttraumatic stress disorder: a meta-analysis. Hippocampus. 2011;21:243–252. doi: 10.1002/hipo.20746. [DOI] [PubMed] [Google Scholar]
- Wust S, Federenko I, Hellhammer DH, Kirschbaum C. Genetic factors, perceived chronic stress, and the free cortisol response to awakening. Psychoneuroendocrinology. 2000;25:707–720. doi: 10.1016/s0306-4530(00)00021-4. [DOI] [PubMed] [Google Scholar]
- Wust S, Wolf J, Hellhammer DH, Federenko I, Schommer N, Kirschbaum C. The cortisol awakening response - normal values and confounds. Noise Health. 2000;2:79–88. [PubMed] [Google Scholar]
- Xiao L, Jordan CL. Sex differences, laterality, and hormonal regulation of androgen receptor immunoreactivity in rat hippocampus. Horm Behav. 2002;42:327–336. doi: 10.1006/hbeh.2002.1822. [DOI] [PubMed] [Google Scholar]
- Yagi S, Galea LAM. Sex differences in hippocampal cognition and neurogenesis. Neuropsychopharmacology. 2019;44:200–213. doi: 10.1038/s41386-018-0208-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Yagi S, Drewczynski D, Wainwright SR, Barha CK, Hershorn O, Galea LAM. Sex and estrous cycle differences in immediate early gene activation in the hippocampus and the dorsal striatum after the cue competition task. Horm Behav. 2017;87:69–79. doi: 10.1016/j.yhbeh.2016.10.019. [DOI] [PubMed] [Google Scholar]
- Yamasue H, Abe O, Suga M, Yamada H, Inoue H, Tochigi M, Rogers M, Aoki S, Kato N, Kasai K. Gender-common and -specific neuroanatomical basis of human anxiety-related personality traits. Cereb Cortex. 2008;18:46–52. doi: 10.1093/cercor/bhm030. [DOI] [PubMed] [Google Scholar]
- Yao S, Song J, Gao J, Lin P, Yang M, Zahid KR, Yan Y, Cao C, Ma P, Zhang H, Li Z, et al. Cognitive function and serum hormone levels are associated with gray matter volume decline in female patients with pro-lactinomas. Front Neurol. 2017;8:742. doi: 10.3389/fneur.2017.00742. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Yap JJ, Takase LF, Kochman LJ, Fornal CA, Miczek KA, Jacobs BL. Repeated brief social defeat episodes in mice: effects on cell proliferation in the dentate gyrus. Behav Brain Res. 2006;172:344–350. doi: 10.1016/j.bbr.2006.05.027. [DOI] [PubMed] [Google Scholar]
- Ycaza Herrera A, Mather M. Actions and interactions of estradiol and glucocorticoids in cognition and the brain: Implications for aging women. Neurosci Biobehav Rev. 2015;55:36–52. doi: 10.1016/j.neubiorev.2015.04.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Yonker JE, Eriksson E, Nilsson LG, Herlitz A. Sex differences in episodic memory: minimal influence of estradiol. Brain Cogn. 2003;52:231–238. doi: 10.1016/s0278-2626(03)00074-5. [DOI] [PubMed] [Google Scholar]
- Yoshiya M, Komatsuzaki Y, Hojo Y, Ikeda M, Mukai H, Hatanaka Y, Murakami G, Kawata M, Kimoto T, Kawato S. Corticosterone rapidly increases thorns of CA3 neurons via synaptic/extranuclear glucocorticoid receptor in rat hippocampus. Front Neural Circuits. 2013;7:191. doi: 10.3389/fncir.2013.00191. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Young KD, Bellgowan PS, Bodurka J, Drevets WC. Functional neuroimaging of sex differences in autobiographical memory recall. Hum Brain Mapp. 2013;34:3320–3332. doi: 10.1002/hbm.22144. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Yue Y, Hu L, Tian QJ, Jiang JM, Dong YL, Jin ZY, Cheng YH, Hong X, Ge QS, Zuo PP. Effects of long-term, low-dose sex hormone replacement therapy on hippocampus and cognition of postmenopausal women of different apoE genotypes. Acta Pharmacol Sin. 2007;28:1129–1135. doi: 10.1111/j.1745-7254.2007.00618.x. [DOI] [PubMed] [Google Scholar]
- Yuste R, Bonhoeffer T. Morphological changes in dendritic spines associated with long-term synaptic plasticity. Annu Rev Neurosci. 2001;24:1071–1089. doi: 10.1146/annurev.neuro.24.1.1071. [DOI] [PubMed] [Google Scholar]
- Yuste R, Bonhoeffer T. Genesis of dendritic spines: insights from ultrastructural and imaging studies. Nat Rev Neurosci. 2004;5:24–34. doi: 10.1038/nrn1300. [DOI] [PubMed] [Google Scholar]
- Zarrouf FA, Artz S, Griffith J, Sirbu C, Kommor M. Testosterone and depression: systematic review and meta-analysis. J Psychiatric Pract. 2009;15:289–305. doi: 10.1097/01.pra.0000358315.88931.fc. [DOI] [PubMed] [Google Scholar]
- Zhang JM, Konkle AT, Zup SL, McCarthy MM. Impact of sex and hormones on new cells in the developing rat hippocampus: a novel source of sex dimorphism? Eur. J Neurosci. 2008;27:791–800. doi: 10.1111/j.1460-9568.2008.06073.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zhang Z, Yang R, Zhou R, Li L, Sokabe M, Chen L. Progesterone promotes the survival of newborn neurons in the dentate gyrus of adult male mice. Hippocampus. 2010;20:402–412. doi: 10.1002/hipo.20642. [DOI] [PubMed] [Google Scholar]
- Zhao Z, Taylor WD, Styner M, Steffens DC, Krishnan KR, MacFall JR. Hippocampus shape analysis and late-life depression. PLoS ONE. 2008;3:e1837. doi: 10.1371/journal.pone.0001837. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zhao H, Tian Z, Hao J, Chen B. Extragonadal aromatization increases with time after ovariectomy in rats. Reprod Biol Endocrinol. 2005;3:6. doi: 10.1186/1477-7827-3-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zhou L, Fester L, von Blittersdorff B, Hassu B, Nogens H, Prange-Kiel J, Jarry H, Wegscheider K, Rune GM. Aromatase inhibitors induce spine synapse loss in the hippocampus of ovariectomized mice. Endocrinology. 2010;151:1153–1160. doi: 10.1210/en.2009-0254. [DOI] [PubMed] [Google Scholar]
- Zorn JV, Schur RR, Boks MP, Kahn RS, Joels M, Vinkers CH. Cortisol stress reactivity across psychiatric disorders: A systematic review and meta-analysis. Psychoneuroendocrinology. 2017;77:25–36. doi: 10.1016/j.psyneuen.2016.11.036. [DOI] [PubMed] [Google Scholar]
- Zuena AR, Mairesse J, Casolini P, Cinque C, Alema GS, Morley-Fletcher S, Chiodi V, Spagnoli LG, Gradini R, Catalani A, Nicoletti F, et al. Prenatal restraint stress generates two distinct behavioral and neurochemical profiles in male and female rats. PLoS ONE. 2008;3:e2170. doi: 10.1371/journal.pone.0002170. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zuo Y, Lin A, Chang P, Gan WB. Development of long-term dendritic spine stability in diverse regions of cerebral cortex. Neuron. 2005;46:181–189. doi: 10.1016/j.neuron.2005.04.001. [DOI] [PubMed] [Google Scholar]