Abstract
Neuropsychiatric disorders, which are associated with stress hormone dysregulation, occur at different rates in men and women. Moreover, nowadays, preclinical and clinical evidence demonstrates that sex and gender can lead to differences in stress responses that predispose males and females to different expressions of similar pathologies. In this curated review, we focus on what is known about sex differences in classic mechanisms of stress response, such as glucocorticoid hormones and corticotrophin-releasing factor (CRF), which are components of the hypothalamic-pituitary-adrenal (HPA) axis. Then, we present sex differences in neurotransmitter levels, such as serotonin, dopamine, glutamate and GABA, as well as indices of neurodegeneration, such as amyloid β and Tau. Gonadal hormone effects, such as estrogens and testosterone, are also discussed throughout the review. We also review in detail preclinical data investigating sex differences caused by recently-recognized regulators of stress and disease, such as the immune system, genetic and epigenetic mechanisms, as well neurosteroids. Finally, we discuss how understanding sex differences in stress responses, as well as in pharmacology, can be leveraged into novel, more efficacious therapeutics for all. Based on the supporting evidence, it is obvious that incorporating sex as a biological variable into preclinical research is imperative for the understanding and treatment of stress-related neuropsychiatric disorders, such as depression, anxiety and Alzheimer’s disease.
Keywords: Stress, sex differences, immune, neurodegeneration, antidepressants, HPA axis, glucocorticoids
1. INTRODUCTION
Stressful experiences are a part of life, but the way people respond to stress varies. In general, the stress response itself is meant to be adaptive, particularly to acute stressors, as it allows for mobilizing energy stores while suppressing growth reproduction and drives the immune system to prepare the body to deal with a threat environment [1, 2]. In some cases, prior stress can promote resilience to future stressors, a phenomenon called stress inoculation [3, 4]. However, the effects of a maladaptive response to stress can be multidimensional and may include different neurobiological and psychological outcomes, like alexithymia [5]. For example, maladaptive responses have recently been observed in stressed healthcare professionals during the recent COVID-19 pandemic [6]. Moreover, chronic stress or an unchecked stress response is a risk factor for a range of psychiatric and neurodegenerative disorders, including major depression and Alzheimer’s disease (AD) [7-9].
There are many factors that can contribute to variability in how people respond to stress, ranging from genetic factors to environmental (e.g., social support). Here, we focus on how sex/gender can lead to differences in stress responses that predispose males and females to different pathology.
Evidence that sex/gender can influence stress reactions comes, in part, from epidemiological data that reveal that disorders linked to stress hormone dysregulation occur at different rates in men and women [10]. For example, rates of major depression are nearly twice as high in women as in men [11, 12]. Women are also more likely to suffer from anxiety disorders than men, with a lifetime female-to-male prevalence ratio of 1.7:1 [13]. This sex/gender disparity is observed in various disorders linked to high levels of perceived stress and trauma and stress hormones, including generalized anxiety disorder, panic disorder, social anxiety disorder, specific phobias, and post-traumatic stress disorder (PTSD) [14, 15]. Other neurological and medical conditions - such as AD, migraines, insomnia, and irritable bowel syndrome - that are more common in women than men are often comorbid with depression and anxiety, perhaps suggesting some common underlying pathology [16-25]. However, it is an oversimplification to assume that women are simply more vulnerable to stress. There are disorders exacerbated by stress, such as schizophrenia, that have a male bias in rates and age of onset [26-28]. Additionally, psychological and sociocultural factors play a role. For example, diagnostic criteria, which for psychiatric disorders are completely symptom-based can influence outcomes. One study using a broader diagnosis for depression that includes additional symptoms, such as anger attacks/aggression, substance abuse, and risk-taking, found that this expanded criterion eliminates sex disparities in disease prevalence [29].
Based on epidemiological data alone, it is difficult to obtain a clear picture of whether sex-related variables, such as gonadal hormones, are important to consider in the understanding of the etiology of stress-related disorders and treatment development. To address these limitations, the field has turned to non-human animal models of stressor exposure to determine whether there are sex differences in stress responses relevant to human health. These models are crucial in driving drug development [30], so it may be surprising that, historically, they excluded female animals [31]. However, in response to pressure from funding agencies in the United States of America and Canada [32], in the past five years or so, more basic and preclinical studies have included both sexes in their designs, although there are still major gaps in proper sex comparisons [33, 34], as well as targeted funding [35].
Despite the limited data including female subjects, here we focus on what is known about sex differences in classic mechanisms of responding to stress. We also talk about more recently recognized regulators of stress and disease, such as the immune system, epigenetic mechanisms, neurodegeneration and sex differences therein. Finally, we discuss how understanding sex differences in stress responses can be leveraged into novel therapeutics that better treat psychiatric and neurological disorders in everyone.
2. CLASSICAL MECHANISMS AND STRESS RESPONSES
The hypothalamus-pituitary-adrenal (HPA) axis is activated, in part, to provide energy to the body in response to stress, and its dysregulation has been implicated in stress-related disorders. Sex differences in the HPA axis have been described in detail [36], with female rodents having higher plasma corticosterone, the most abundant glucocorticoid found in rats, in comparison to males [37-39]. However, this sex difference seems to depend on several factors, such as strain, age, time of sampling, housing conditions, diet and estrous cycle phase, reproductive status, etc. [39-42]. Interestingly, globulin corticosteroid binding, which determines the amount of the unbound, active corticosterone that reaches the brain, is also sex-differentiated [43] and is influenced by stress [44].
Stress-induced activation of the HPA axis is more robust in female rats than in males, but this activation does not seem to correlate with the female behavioral response to stress [36, 45, 46]. For example, stress enhances corticotropin-releasing factor (CRF), ACTH, and corticosterone more in female than male rats [41, 47, 48], but when surgical adrenalectomy is performed in female rats, and corticosterone is substituted, resulting in stable corticosterone levels, females continue to demonstrate stress associated behavior in the FST. This finding suggests that generally, the female behavioral response to stress, as evidenced in this case from the immobility, swimming and climbing FST behaviors, is less influenced by the HPA axis, whereas in males, their behavioral response to stress more accurately tracks the HPA axis (dys)function [45].
Similarly, in another study, adrenalectomy also did not alter a well-described stress effect in female rats. Specifically, associative learning in trace eyeblink condition is decreased in female rats as a consequence of acute stress exposure (30 min of tail shock in a restrainer tube) that has been applied 24 hours in advance. However, in male rats, the same surgery, which prevents HPA axis activation, abolished the effects of acute stress in enhancing male eyeblink conditioning [49]. Notably, these acute stress effects require the hippocampus in both sexes [50] and cause respective sex-differentiated stress responses in the density of spines in the CA1 area of the hippocampus, i.e., decrease in females and increase in males. The phase of the estrous cycle is important in determining the female stress effect on learning and spine density on the hippocampus, which is a measure of synaptic plasticity. Specifically, this is evident when females are stressed in the proestrous phase of the cycle and are sacrificed or begin testing in the diestrous phase when estrogens are low [51]. Interestingly, this female stress effect is also dependent on the organizational effects of gonadal hormones, as the female response can be masculinized with one injection of testosterone on the first day of birth [52]. Both associative learning and spine density, in response to acute stress in adult masculinized female rats (which do not have a cycle) are enhanced in a similar fashion to male rats [53, 54].
Sex differences in neurotransmitter levels are also present in response to stress [40, 55, 56]. In particular, rats exposed to the FST, which consists of two sessions of swim stress on two consecutive days, had enhanced serotonergic activity. This was indicated by an increased 5-HIAA/5-HT turnover ratio in the hippocampus and levels of serotonin’s metabolite 5-HIAA in the prefrontal cortex on the second day [39]. Interestingly, serotonergic activity in the prefrontal cortex correlates with behavior in the FST, whereas hippocampal serotonin does not [57]. In response to 6 weeks of chronic mild stress, only female rats exhibit decreased serotonergic activity in the hippocampus and the hypothalamus, which might be linked with a higher rate of female depression and stronger response to SSRIs [40].
Sex differences in the response of the dopaminergic system to short-term stress also occur. Specifically, dopaminergic activity is enhanced in females following the forced swim and this has been considered as an adaptive mechanism [39, 58]. Previous studies have also shown that stress exposure influences amino acid levels in the prefrontal cortex and the hippocampus of rats, two brain regions involved in stress-related and affective disorders [59-61]. In particular, exposure to two sessions of swim stress enhanced glutamate, glutamine, GABA and taurine levels in female rats. In males only GABA and taurine levels were enhanced [39]. It has been suggested that stress-induced glutamatergic and GABAergic enhancements are linked with changes in serotonergic and dopaminergic activity in the PFC [62, 63].
Overall, enhancements in neurotransmitter levels in response to short-term or acute stress can be considered as a typical adaptive response, whereas chronic stressful experience results in decreased neurotransmission that contributes to the neurobiology of stress-related and affective disorders.
3. SEX DIFFERENCES IN THE CORTICOTROPIN RELEASING FACTOR - CRF SYSTEM
CRF dysregulation is implicated in psychiatric disorders, such as major depressive disorder (MDD), and neurodegenerative disorders, such as AD. In stress-related psychiatric disorders, high CRF levels are found in the cerebrospinal fluid (CSF) of humans with depression and PTSD [64, 65]. In MDD, the elevated CRF in CSF normalizes with successful treatment, correlating CRF levels with symptomatology [66]. In postmortem brain tissue from people with depression, high levels of CRF are found in the paraventricular: nucleus (PVN) and in neuromodulatory regions, including the raphe and locus coeruleus (LC) [67, 68]. CRF is also linked to neurodegenerative disorders. In AD, chronic stress, which leads to high CRF levels, increases the risk for the disorder [69, 70]. However, CRF immunoreactivity is reduced in postmortem tissue from people with AD, but this is accompanied by CRF receptor upregulation, perhaps as a compensatory response to counter the lower CRF levels [71, 72]. The impact of these changes is unclear, but it has led to the theory that initial CRF hypersecretion due to stress increases AD risk, but the lasting dysregulation of central CRF may decrease its tone on important regions for memory and cognition, contributing to cognitive deficits [69]. As noted, MDD and AD occur more often in women, but unfortunately, sex/gender differences in CRF in patients with these conditions have not been investigated. In neurotypical populations, peripheral administration of CRF causes an increased ACTH response in women compared to men [73]. This could indicate that women have a greater HPA axis response to CRF release, which could bias them towards these disorders linked to CRF.
Despite the paucity of data on sex differences in CRF in humans, there are many rodent studies showing that, in regions relevant to affect and cognition, there are sex differences in CRF that range from the inputs that regulate CRF neurons to CRF’s postsynaptic efficacy [74]. There is some evidence that female rats have greater CRF expression than males in the PVN [48, 75]. In target regions of the PVN, including the pituitary and the medial septum, CRF binding protein (CRF-BP), which binds free CRF to reduce its bioavailability, is higher in female than male rodents [76, 77]. This increase in female CRF-BP may help compensate for higher levels of CRF released into these regions from the PVN.
There are two types of CRF receptors: CRF1 and CRF2. Global knockout studies have found largely opposing effects of these receptors where CRF1 initiates the HPA axis and increases anxiety, while CRF2 mediates the duration of the HPA axis response, promoting stress recovery and reducing anxiety [78-80]. There are sex differences in CRF receptor function. For example, CRF1 binding typically reflects receptor number, and it is increased in the cortex, nucleus accumbens, and amygdala of adult female rats [81]. In the rostral portion of the anteroventral periventricular nucleus of the hypothalamus, a region that regulates maternal behavior, female mice have more CRF1-positive neurons than males, and these sex differences in exacerbated by chronic variable stress [82, 83]. There are also sex differences in CRF2 binding, which tends to be male-biased in that higher levels of CRF2 binding in the bed nucleus of the stria terminalis, amygdala, and hypothalamus are found in male compared to female rats [81]. Given the differential roles of CRF1 and CRF2, these sex differences in binding may bias females toward stress reactivity and anxiety and males towards stress recovery. In addition to differences in receptor amount, sex differences in the distribution of CRF receptors on different cell types have also been reported. In the dorsal raphe, CRF1 receptors have a higher colocalization with parvalbumin-containing GABA neurons in male mice, while in the hippocampal CA1 region, there is great CRF receptor colocalization with delta opioid receptor-containing dendrites in female rats [84, 85]. Sex differences in receptor distribution can influence how regions respond to stress and their downstream effects on efferent targets.
CRF receptors are G-protein coupled receptors, and while they preferentially bind the Gs protein and signal through the cyclic AMP (cAMP) protein kinase A (PKA) signaling pathway, they can also bind other G proteins and β-arrestin [86, 87]. Thus, the downstream effects of CRF receptors are not only regulated by their receptor number and localization but also by their signaling. Sex differences in CRF1 signaling are found in the LC, a noradrenergic-containing nucleus that projects to many regions, including the cortex, to increase levels of arousal [88-90]. Specifically, CRF1 receptors in the LC signal more through the cAMP-PKA pathway in females compared to male rats, which increases the sensitivity of female LC neurons to CRF [91]. This sex difference in sensitivity is linked to sex differences in cortical network activity, as CRF in the LC increases theta oscillations in the medial prefrontal cortex and its coherence with the orbitofrontal cortex in females but not in males [92]. Under acute or moderate stress, this increased sensitivity of LC neurons to CRF in females may help promote alertness and cognitive processing. However, under conditions of CRF hypersecretion, it could lead to hyperarousal, a negative state of being on edge that contributes to some symptoms of depression and PTSD and may be more prominent in women [93-96].
Similar sex differences in CRF1 are also found in the cortex, where CRF1 receptors are more highly coupled to Gs in females but to β-arrestin in males [91]. β-arrestin can activate its own suite of signaling cascades that are often distinct from those activated by G-proteins [97, 98]. Using a phosphoproteomic approach in CRF overexpressing (CRF-OE) mice, it was found that CRF hypersecretion increased activation of phosphopeptides in cortical Gs signaling pathways in females and β-arrestin signaling pathways in males [99]. This indicates that the signaling of the CRF1 is sex-biased [100]. An additional discovery was that CRF-OE female mice had an overrepresentation of phosphopeptides in the AD pathway and increased tau phosphorylation in the cortex [99]. In a mouse model of AD pathology, female mice that express human APP and also have an overexpression of CRF in the forebrain have an increased formation of amyloid β plaques and cognitive impairments relative to males [99]. Together, these studies suggest that CRF hypersecretion can bias females toward AD pathology (Fig. 1).
Fig. (1).

CRF and glucocorticoid signaling interplay in male and female AD brain. A schematic representation of the complex interplay between sex hormones, corticotrophin-releasing factor (CRF), and glucocorticoid (GC) signaling in male and female Altzheimer disease (AD) brain. Under chronic stress conditions, both CRF and GC receptor (GR) signaling seems to participate in the stress-driven Aβ overproduction as well as Tau hyperphosphorylation and accumulation in the AD brain with female brain exhibiting a CRF-Gs-PKA cascade activation that contributes to both AD pathomechanisms. Note the counteracting role of sex steroids in stress-induced GR activation and downstream induction of AD-related pathomechanisms, suggesting that reduction of male or female sex steroids (e.g., by aging) and the concomitant exposure to prolonged stress and high GC levels increase Aβ overproduction and/or Tau hyperphosphorylation and accumulation thus, endangering neuronal function and triggering AD neuropathology.
Given the link between high levels of CRF and brain disorders, there has been an effort to develop CRF1 receptor antagonists to treat psychiatric disorders [101]. These antagonists were initially tested in preclinical studies using male rodents [102-106]. At the time, there was no evidence that the CRF1 receptor could signal differently in males and females because researchers were not using female rodents in their studies. However, this different signaling likely reflects a sex difference in the conformation of the CRF1 receptor, which could alter the efficacy of antagonists. The one clinical trial that showed some efficacy of CRF1 antagonists in depression included only men [107]. The other unsuccessful CRF1 antagonist clinical trials for depression tested CRF1 antagonists in mixed-sex/gender groups or only in women [36]. Unfortunately, the data from the mixed-sex/gender trials was not disambiguated by sex, so we are unaware whether these drugs were actually effective in men in these other trials. These findings highlight the problems with excluding females from basic and preclinical research and not disambiguating data by sex/gender in clinical trials. Moreover, they underscore that sex differences in pharmacodynamics, such as receptor function, should be assessed and considered in developing therapeutics.
4. THE PRECIPITATING ROLE OF CHRONIC STRESS ON NEURODEGENERATION: THE ROLE OF SEX
As noted, clinical studies have suggested that lifetime stress is associated with the early onset of AD pathology [108, 109]. In addition to CRF, other stress hormones, such as glucocorticoids (GCs), are associated with the initiation and progression of AD. For instance, chronic stress may advance the age of onset of the familial form of AD, while cortisol levels in AD patients correlate with their memory deficits [110-112]. In addition, high cortisol levels, the abundant GC in humans, are commonly found in AD patients’ plasma, saliva, and CSF [113-115], while AD patients also show higher total daily secretion of cortisol [116]. It is noteworthy that female AD patients show higher cortisol levels than male patients [117], suggesting that a sex difference in the stress response may contribute to the increased risk of women for AD with a potential role for both GC and centrally active CRF.
Focusing on the accumulation of amyloid β (Aβ), a molecular hallmark of AD brain, a recent clinical study that used Pittsburgh compound B positron emission tomography (PiB - PET) technology correlated high cortisol levels with elevated Αβ levels in the AD brain [118]. In line with clinical evidence, animal studies showed that elevated GC levels or exposure to chronic stressful conditions increased the levels and accumulation of Aβ in the brain, resulting in impaired cognitive function [119, 120]. In addition, neuronal mechanisms involved in the degradation or excretion of Aβ may be inactivated by stress; e.g., chronic stress affects the brain's excretion properties by reducing the expression of aquaporin 4 (AQP4) protein, exacerbating the accumulation of Aβ [121]. As mentioned above, CRF may also play a critical role in the stress-driven precipitation of AD, with females being more vulnerable to males [99, 122]. However, the role of GC and their signaling in the interplay of stress and sex in AD precipitation remains mainly unclear.
Different studies suggest that chronic stress also triggers different parameters of Tau pathology, the other hallmark of the AD brain. Exposure of animals to chronic stressful conditions resulted in the hyperphosphorylation of Tau and its accumulation in both neuronal dendrites and synapses, leading to neuronal malfunction and impairments of synaptic signaling [123-125]. This stress- or GC-driven accumulation of abnormal forms of Tau protein may occur via the inhibition of different degradation mechanisms of Tau. For instance, the autophagy mechanism and the endolysosomal degradation pathway are inhibited under stressful conditions, and this leads to pathological accumulation of Tau protein and neuronal malfunction in the brain of experimental AD rodent models [126, 127]. In addition, molecular chaperones, e.g., heat shock proteins (Hsp90 and Hsp70) involved in Tau degradation, are also shown to be dysregulated by chronic stress [124]. Hsp90 and Hsp70 maintain GC receptors in a high-affinity state, thus suggesting a point at which GC/GC receptor signaling and Tau degradation machinery can intersect. Interestingly, the above mechanism related to Tau accumulation is suggested to be involved in the increased vulnerability of the female hippocampus to the detrimental effect of chronic stress. Compared to males, females exhibited higher levels of Tau pathology and neuronal malfunction, as well as cognitive impairment in response to prolonged stress with particular role for molecular chaperones [124]. Note that prolonged exogenous administration of GCs presents similar effects, demonstrating their central role in the pathological process triggered by chronic stress [125, 128]. It is now clear that the HPA axis, GCs, and CRF are involved in the regulation of AD pathological mechanisms under exposure to prolonged chronic stress, resulting in the accumulation of Aβ and Tau protein in the brain [129]. For example, animal studies suggest the involvement of CRF receptors in stress-induced hyperphosphorylation of Tau. As noted, CRF overexpression increases the hyperphosphorylation of Tau with a greater effect in females than males [130, 131].
Notably, it is important to mention here a potential interaction of sex hormones with different components of stress (e.g., GC and GC signaling). For instance, it is suggested that loss of the neuroprotective effect of estrogens could contribute to the increased vulnerability of females to stress-driven AD brain pathology [132], as de-masculinization of neonatal male AD Tg mice narrows the gender gap in terms of Aβ pathology [133]. Moreover, there is strong evidence for an interplay between GC and sex steroids, in particular with respect to the regulation of neuroendocrine function and behavior. Previous studies demonstrate that the depletion of male gonadal steroids exacerbates the GC-driven Tau hyperphosphorylation [134], while clinical evidence recently showed that testosterone can counteract GC-induced hippocampal atrophy and memory deficits in middle-aged men [135]. Given that age is a risk for AD and that GC and sex steroid levels are inversely regulated (increased and decreased, respectively) during aging [136], future studies should further clarify the molecular underpinning of the complex interplay between sex, aging and stress in the precipitation of AD, as well as further dissect the GC and CRF contribution to the stress-driven AD brain pathology.
5. BEYOND CLASSICAL STRESS RESPONSES: SEX DIFFERENCES IN GENETICS, EPIGENETICS, AND IMMUNE RESPONSE TO STRESS
Translational studies have identified genetic, epigenetic, and immune mechanisms that contribute to stress susceptibility and are relevant to human mood disorders. Preclinical studies often use stress to induce behavior that overlaps with symptoms/domains of depression. These stress paradigms vary with different laboratories using forms of social, variable, or unpredictable stress applied chronically to induce changes in behavior [137]. In some of these paradigms, not only do males and females engage in different behaviors in response to stress, but the underlying transcriptional response is different or even opposite [138-144]. Even when depression-like behaviors are similarly induced in male and female mice, there is less than 30% overlap in stress-induced gene expression in the nucleus accumbens (NAc) and pPFC [145]. In humans’ different transcriptional signatures have been identified in these brain regions of men and women with depression [146, 147]. A variety of epigenetic mechanisms contribute to transcriptional sex differences in both humans with depression and rodents exposed to stress. These include sex-specific regulation by DNA methyltransferase (DNMTs), histone modifications, microRNAs (miRNA) and long non-coding RNA (lncRNA).
DNMTs are enzymes that covalently link a methyl group to the 5’ position of cytosine nucleotides of DNA, resulting in the suppression of gene expression [148]. There are several classes of DNMTs, including DNMT1, which maintains methylation between progenitor and daughter cells [149]. DNMTs 3a & b are involved with de novo methylation, for which they methylate sites that were previously unmethylated and/or recruit methylation binding domain proteins to produce a variety of histone modifications [149]. DNA methylation is a component of typical development, and constitutive knockout is lethal. Rodent studies suggest males and females have different baseline patterns of methylation, which is important to feminizing the brain and its response to a variety of stimuli [140, 150]. The NAc, a key region involved in reward, DNMT3a over-expression shifts both sexes to be more sensitive to stress [140]. In males, DNMT3a over-expression, in the absence of stress, increases spine density in the NAc, similar to the effects of either cocaine administration or social stress [151, 152]. Blocking DNMT3a activation by 6-day variable stress in the NAc of female mice shifts their behavior to a male-like response and promotes behavioral resilience [140]. Bulk sequencing of the NAc demonstrated that this manipulation removed many of the pre-existing transcriptional differences induced by stress between males and females, resulting in greater overlap of transcription. When intracerebral DNMT3a was repressed in female mice during early prenatal development, they engaged in male-like sex behaviors in adulthood after priming with testosterone and exposure to a receptive female [150].
While DNMT3a knockdown promoted a male-like response to stress in females, stress can be blocked in males by decreasing DNMT1 expression and increasing histone modifications [153]. Male mice given the phytochemicals dihydrocaffeic acid (DHCA) and malvidin-3′-O-glucoside (Mal-gluc) are resilient to social defeat stress and have reduced interleukin-6 (IL-6) expression in the periphery and decreased spine density in the NAc. In both male and female mice exposed to social defeat stress or variable stress, DHCA/Mal-gluc blocks the effects of stress on behavior, but through different mechanisms resulting in different transcriptional changes and alterations of different peripheral cytokines [153, 154].
Histone modifications result from additions or removal of marks on the N-terminal tails of the histone core in nucleosomes [155-158]. These modifications can open or close chromatin structures, resulting in the ability to express or suppress gene transcription [157, 159, 160]. Histone modifications during development have long-lasting effects by sex-specifically shaping brain regions and their subsequent stress response. The bed nucleus of the stria terminalis (BNST) is a sexually dimorphic structure involved in emotional responses to stress. It is masculinized through histone acylation in combination with testosterone during an early postnatal critical window [161]. Neurons in the female BNST undergo apoptosis during this critical window, resulting in a smaller volume in females compared to males [161]. If male mice are treated during this time point with a histone deacetylase inhibitor (HDAC), they have a feminized BNST [161]. Injection of testosterone into females during this same time will produce a male-like BNST [162]. In adulthood, masculinized females respond to acute stress like males, resulting in enhanced learning ability [162, 163], suggesting that epigenetic modification of the BNST may be involved in stress resilience. Histone modifications in adulthood can alter the behavioral response of male rodents to social stress through hyperacetylation of the BDNF promoter in the hippocampus, which also occurs following chronic antidepressant treatment [164]. Sex differences in long-term retrieval of fear memory are also dependent on histone modifications, specifically acetylation of the Cyclin-dependent kinase 5 promoter [165].
In addition to epigenetic mechanisms that act on promoter regions, noncoding RNAs also contribute to sex differences in the stress response. MicroRNAs (miRs) are short (~20 nucleotides) non-coding RNAs that act upon mRNAs to suppress protein translation [166]. Male and female mice exposed to variable stress have no overlap in the downregulation of NAc miRs and only 3 overlapping upregulated miRs. Network analysis indicated that many of the genes upregulated by female miRs are immune-related pathways, whereas in males’ upregulation of miRs is associated with neuronal signaling pathways [167]. In male mice exposed to social defeat stress, an miR in peripheral immune cells regulates the behavioral and immune response to stress [168]. Social defeat stress increased the population of immature, pro-inflammatory monocytes (Ly6chigh) in both susceptible and resilient mice. However, differences in expression of the miR106b~25 cluster in bone marrow-derived bone marrow leukocytes regulated the behavioral response to stress. Leukocyte-specific knockout of this miR cluster promoted resilience. MiR-144-3p in red blood cells has also been identified as a biomarker of depression in humans and stress susceptibility in mice [169]. Furthermore, this miR has the potential to identify who will respond to ketamine treatment or not [169].
Non-coding RNA in sperm can also contribute to epigenetic and transgenerational effects of stress. Paternal stress transmits to the next generation, producing a stress-susceptible phenotype in offspring [170-172]. Both miRs and long non-coding (lncRNA) have been identified as driving these transgenerational effects [170, 172]. LncRNA is implicated in female depression and stress susceptibility. Women with depression have altered primate-specific lncRNAs LINC00473 and Rp11-298d21 (FEDORA) in the PFC [143, 144]. Viral-mediated upregulation of these lncRNAs in the PFC of mice promotes stress resilience or stress susceptibility, respectively, in females but not in male mice. Interestingly, FEDORA expression in the blood was also a potential biomarker of depression for women [143].
Epigenetic regulation of stress/depression also occurs from the ability of immune-associated genes to escape X inactivation (Fig. 2). The X chromosome contains more immune-related genes than any other chromosome [173], and at least 9 of these genes escape X inactivation, resulting in a larger dose for females [174]. These include the toll-like 7 receptor which is activated by single-strand RNA viruses like COVID-19, and CXCR3, a chemokine receptor downstream of interferon signaling and CD40LG, which modulates T cell communication to B cells. Females have a stronger immune system than males in that they have a more effective response to vaccines, greater production of antibodies, greater release of cytokines during infection, and stronger rejection of tumors and/or transplanted tissue [175]. The tradeoff for this enhanced protection is a greater risk of auto-immune disorders. Women account for 70-80% of the population experiencing autoimmune disorders [174, 175].
Fig. (2).

Novel mechanisms of stress susceptibility beyond the HPA axis. Recent rodent studies suggest that multiple epigenetic mechanisms contribute to sex differences in stress susceptibility. These include the ability of alleles on the X chromosome to escape X inactivation in females, DNA methylation and histone modifications that impact the likelihood that a gene will be expressed and noncoding RNAs, including lncRNA and miRs. These mechanisms strongly impact the immune system and gonadal hormones, which also engage in bidirectional communication. Cytokines released by peripheral immune cells and hormones can act in an endocrine fashion to alter immune cells in the brain that, in turn, impact synaptic plasticity. Made with Biorender bly, brexanolone, which is a pharm disorders and depression are intertwined, as experiencing one disease results in an increased risk of developing the other.
Recent research has identified additional peripheral influences that contribute to depression in humans and the stress response in rodents. One theory that is currently being explored is the leaky gut hypothesis [176]. The concept is that stress loosens the intestinal barrier, allowing endotoxins that escape and increase inflammatory signaling in the body and brain. Striking evidence that the gut microbiome contributes to depression symptoms comes from a study that transplanted gut microbes from donors with depression into adult male rats [177]. Rats that got recolonized with microbiota from depressed but not control donors expressed anhedonia and exploratory anxiety-associated behaviors. Ongoing research explores the mechanisms involved and how the gut microbiome can be reshaped to treat mood disorders [178-180]. The gut microbiome can differ by sex [181]. Opposite-sex microbiome transplants confer some of the immune properties of the host [182]; however, more research is needed to understand how sex interacts with gut microbes to shape stress behavioral responses to stress and its relevance to depression.
Young males and females have differences in blood-brain-barrier (BBB) permeability, which can be further altered by stress [183]. Females have greater permeability of the PFC and a more inflamed immune profile at baseline than males [184]. Stress increases the permeability of PFC in females, whereas stress increases the permeability of the striatum in males [185, 186]. These sex differences likely explain why different areas of the brain are more vulnerable to peripheral inflammation in males and females. Increased permeability in response to stress occurs via downregulation of the tight junction protein claudin 5. In males, this occurs in the NAc, allowing increased amounts of peripheral cytokines to enter that brain region [185]. In female mice, stress also caused the downregulation of claudin 5 to increase BBB permeability. However, the impact was on the PFC rather than the NAc [186, 187]. The authors also found corresponding changes in genes associated with BBB permeability in post-mortem PFC tissue of women with MDD, suggesting that the PFC may be more vulnerable to neurovascular damage than the NAc in females across species.
Moreover, females have a higher number of reactive microglia, the innate immune cells of the brain within the PFC [188]. Following stress, males express a more reactive immune profile, and females express a greater number of homeostatic markers. Increased microglia activation in the PFC of humans with depression has been identified using post-mortem tissue, and more recently, it was suggested by PET imaging studies that use Translocator protein 4 (TSPO4), which is expressed by microglia, astrocytes and endothelial cells [189]. This initial downregulation of microglia activation in the PFC by females may be a protective compensatory response given the greater permeability of this region. Inversely, microglia from females are activated by stress in the NAc, whereas males seem to be able to suppress activation, maintaining homeostasis [190].
Sex differences in depression and stress responsivity are also influenced by gonadal hormones. Estrogens, progesterone, and androgen receptors are present in most immune cells [191-193]. Because estrogens and progesterone fluctuate across the cycle, they have a dose-dependent impact on immune cell function. Low levels of estrogens stimulate activation of these cells, whereas high levels of estrogens suppress immune function [194]. Studies on stress suggest that estrogen is a modulator of inflammation, as well as a modulator of behavioral responses [151, 195]. Estrogens can increase the secretion of pro-inflammatory interleukins (IL-6 and IL-8) in the innate immune system and increase the secretion of antibodies and regulatory T cells by B cells, increasing the number of regulatory T cells [194]. In general, testosterone suppresses immune activation, particularly by the adaptive immune system [196]. Testosterone induces apoptosis of T cells, resulting in a reduced number in males [197, 198]. Suppressive effects of testosterone on the immune system may, in part, protect males from the immune-mediated effects of stress. For example, testosterone replacement in male or female gonadectomized mice blocked stimulation of the pro-inflammatory cytokine Tumor necrosis factor alpha (TNF-α) with the endotoxin lipopolysaccharide (LPS) [199]. LPS is often used to induce “sickness behavior,” which overlaps with symptoms of depression [200]. In humans, men and women have different immune responses to LPS injection, including higher levels of circulating pro-inflammatory cytokines TNF-α and interleukin-6 along with greater activation of cortisol, whereas men have an increase in the anti-inflammatory cytokine interleukin 10 [201]. Testosterone has a diurnal rhythm, and little is known about how it may differently regulate the immune system during the light vs. dark cycle [202]. Most studies that have examined the effect of testosterone on the immune system have examined it in the context of removal or addition. Men who have naturally low levels of testosterone do not exhibit a diurnal rhythm. Further complicating the matter is that cortisol is a regulator of both testosterone secretion and the immune system [202]. In both sexes, the HPA axis traditionally acts to suppress immune responses. As such, sex differences in HPA axis activity also contribute to sex differences in the immune response to stress.
6. SEX DIFFERENCES IN DRUG RESPONSE
Depression, anxiety, AD and other stress-related disorders require a multifaced treatment plan, which may include psychotherapy, psychosocial interventions and neuropsychopharmacological treatment, according to the severity of the disorder and the individual patient needs [203]. Several medication classes are available, prominently including selective serotonin reuptake inhibitors (SSRI) and serotonin-noradrenaline reuptake inhibitors (SNRI). Such medications are misleadingly referred to as “antidepressants” [204] but are considered effective treatments for anxiety, obsessive-compulsive disorder and other brain diseases where the monoaminergic neurotransmission may be altered. They are also often prescribed to treat psychiatric symptoms, which are present in neurodegenerative disorders.
To date, there is evidence of sex differences in the pharmacokinetic and pharmacodynamic properties of SSRIs [205-207]. Moreover, it is postulated that many of the pharmacodynamic sex differences may be based on the underlying pharmacokinetic differences between sexes, i.e., sex differences in absorption, distribution, metabolism, and excretion. For example, in women, gastric acid secretion is less pronounced, and the gastrointestinal tract transit time is elongated, and as a result, the maximum drug concentration may be reduced [206, 208]. On the other hand, bioavailability is often found to be enhanced in women [209, 210]. Regarding the distribution of drugs, protein binding is less in women, thus increasing the fraction of unbound active drugs [211]. Another important aspect of sex differences in distribution is that women have a larger fat/muscle ratio than men. As CNS-acting drugs must pass the BBB and thus are designed as highly lipophilic, their initial distribution in women is broader, and then they display a lower redistribution rate and clearance. Moreover, hormonal fluctuations during women's menstrual cycle may further affect the absorption and distribution of psychotropics [210, 212]. To date, the most robust evidence regarding pharmacokinetic sex differences is for the metabolism of antidepressants [206, 213]. However, there are no proper guidelines regarding different dosing of most psychotropics in men and women, although generally, women are probably exposed to higher drug levels. In relevance to that, recently, regulatory agencies issued warnings about using lower doses of some hypnotic medications, such as zolpidem, in women [214].
Similar to pharmacokinetics, important sex differences are thought to exist in the pharmacodynamics of many licensed psychotropics [206, 215]. Regarding antidepressants, data is inconclusive and, at times, conflicting. Some studies support the existence of sex differences [205, 216-218], whereas others fail to identify clinically significant differences [219, 220]. As suggested above regarding CRF-antagonists that failed in clinical trials, a possible explanation for these discrepancies is the lack of proper stratification in clinical studies according to sex, as well as to women’s hormonal status [221]. Indeed, when age and hormonal status are considered, premenopausal women respond better to SSRI, whereas older postmenopausal women do not respond as well [217, 222]. The role of estrogens in facilitating drug response was further highlighted by the finding that in post-menopausal women, hormonal replacement therapy co-administered with SSRIs increased favorable outcomes [221, 223]. Several studies produced similar findings, supporting the beneficial interplay between estrogens and antidepressants [224-228]. However, it is worth mentioning that there have been negative studies as well [229, 230], suggesting that the mediating effect of estrogens may be more complicated and context-dependent in various patient populations, according to the underlying neurobiology and especially that of the serotonin transporter (SERT) binding [231].
Pharmacokinetic and pharmacodynamic sex differences are also found in several preclinical studies of psychotropics. Behavioral tests such as the Tail Suspension Test (TST) and the Forced Swim Test (FST) are often used to study the effects of antidepressants. When properly validated, such tests can also highlight important sex differences [232, 233]. For example, compared to males, female rats display higher levels of immobility and lower head shake counts during the FST [45, 58, 234]. Most antidepressants typically reduce immobility and increase swimming duration [223, 235, 236], and female rodents respond more favorably to lower doses of several SSRIs [234, 237-239].
Sex differences may also exist for several other psychotropics, not directly acting on the monoaminergic neurotransmission. Esketamine, a stereoisomer of the racemic drug ketamine, is a newly licensed medication for severe depression and suicidality [240, 241]. Although clinical studies have yet to show important pharmacodynamic sex differences, few preclinical studies suggest sex differences, as females present higher sensitivity to ketamine’s actions than male animals [242]. Moreover, in social isolation stress models, females recovered from depressive-like behaviors with lower doses of ketamine than males [243].
Regarding cholinesterase inhibitors that are mainly used for treating AD, limited data suggest that women respond better to treatment than men [244], but overall, there is an almost complete lack of sex-specific data reported in clinical trials for AD drugs. Also, there is no sex-specific reporting of adverse events related to these treatments [245]. Therefore, more sex-specific designed studies are needed in AD research, as well.
As mentioned, research on therapy for stress-related disorders has focused lately on other targets beyond classical ones. Apart from those already discussed above, these include NMDA receptors and glutamatergic pathways, serotonergic receptors (e.g., 5-HT2A as targets of psychedelics), the GABAergic system, neuropeptides, endocannabinoids and many more [246-248]. Another very interesting line of research includes neurosteroids, which are produced de novo locally in the brain, as nowadays, it is known that the brain possesses all the enzymes required for the de novo synthesis of steroids from cholesterol and not just from steroid precursors synthesized in the gonads or adrenals, which subsequently enter the brain through the bloodstream [249]. Also, there is accumulating evidence that these neurosteroids play a significant role in neuropsychiatric disorders. For example, allopregnanolone, which is the most investigated, is modulated by stress and is involved in PTSD and depression [250]. Notably, brexanolone, which is a pharmaceutical preparation of allopregnanolone, has been licensed as a treatment for post-partum depression [251].
Estrogens can also act as neurosteroids synthesized locally in the brain from steroid precursors, such as testosterone. This conversion is catalyzed by the rate-limiting enzyme aromatase, encoded by the CYP19 gene, and is happening locally in the brain of both males and females [249]. These neuroestrogens are known to be involved in several brain functions, including neuroprotection, cognition and mood [252-254]. As mentioned, testosterone can also derive from de novo synthesis locally in the brain or from circulating sources that enter the brain. As known, testosterone is converted to estrogens by aromatase or to non-aromatizable androgens (such as DHT) that have also been found to have an important impact on brain functions, especially cognition and neurodegeneration [249]. As mentioned above, sex steroids, especially testosterone, are known to interact with the HPA axis [255, 256].
Regarding stress studies, there is evidence that the enzyme aromatase is modulated by stress in the hypothalamus of male and female adult quails 257], as well as in the male, but not in the female adult rat hypothalamus 39]. In preclinical models of antidepressant activity, short-term subacute administration of letrozole produced a clear antidepressant effect, which was comparable in effect size to that of fluoxetine, an established antidepressant treatment 258]. However, in other studies of repeated letrozole administration over several days (7-21 days), there were no clear antidepressant behavioral effects despite a persisting modulation of the monoaminergic neurotransmission systems [258, 259]. Sustained aromatase inhibition decreased noradrenaline and dopaminergic activity, as demonstrated by the dopaminergic turnover rates in the hippocampus and PFC of male and female adult rats [39]. Moreover, aromatase inhibition enhanced serotonergic activity, as demonstrated by the serotonin turnover rate in the hippocampus of males and females [39]. These effects were not influenced by adult gonadectomy of rats, which suggests that inhibition of estrogen locally in the brain may play a role [39]. These findings are further supported by the fact that adult ovariectomized aromatase knockout female mice also exhibit enhanced serotonergic activity in the hippocampus, suggesting a modulatory role of neuro-estrogens on hippocampal function [260].
These and other studies suggest that neuro estrogens, as well as their receptors, could be interesting, druggable targets for the treatment of stress-related disorders. Importantly, estrogen’s receptors include classical intracellular ERα and ERβ receptors, as well as the membrane G protein-coupled estrogen receptor 1 (GPER1) receptor, which is involved in rapid, non-genomic actions of estrogens in the brain [261, 262]. ERα and ERβ receptors, as well as their modulators, such as tamoxifen and raloxifene, have long been studied for their role in anxiety, cognition and depression, especially during menopause [249, 263, 264]. ERβ seems to be more involved in mood regulation [264]. Also, tamoxifen has been suggested as a potential treatment for episodes of mania, but more studies are needed [265, 266]. The GPER1 has also been recently involved in stress response and anxiety in male and female mice 267, 268], as well as in depression in men and women [269]. More studies are needed to elucidate the role of pharmacological treatments targeting estrogen receptors in anxiety, affective disorders and AD.
CONCLUSION
As discussed in detail above, several animal and human studies confirm the existence of various sex differences in the neurobiological mechanisms of stress response that are linked with the pathophysiology of depression, anxiety and AD [138]. However, only recently, preclinical studies started to include both sexes in stress animal models. A significant emphasis has been given by the National Institutes of Health (NIH) policy to consider sex as a biological variable (SABV) in basic research [270]. For practical recommendations on SABV experimental design, the role of gonadal hormones, as well as relevant statistics, the readers are referred to an NIH-funded 18-part video series made by Cohen Veterans Bioscience. These videos are open-access and available online for researchers [271].
Moreover, funding agencies in the USA, Canada, Australia and the European Union vigorously request the inclusion of sex and gender in research, aiming to facilitate better disease understanding [272]. In particular, the inclusion of SABV in neuropsychopharmacology and stress research may significantly increase the translatability of preclinical findings to clinical setups, which in turn can lead to the development of more efficacious treatment for stress-related disorders [273-275]. In fact, the often-observed sex mismatch between preclinical and clinical trials may account for other confounders, for the problem of limited reproducibility in research [276, 277]. Therefore, more sex-aware preclinical research may facilitate the generation of leads for further clinical testing and expedite the early recognition of adverse events that may appear more frequently or at lower doses in one sex or the other [278]. Finally, training a new generation of physicians and health care professionals to account for sex and gender in their practice will pave the way for more personalized care, especially regarding stress-related disorders that, as presented in this review, are heavily characterized by sex-dependent neurobiology.
ACKNOWLEDGEMENTS
Declared none.
LIST OF ABBREVIATIONS
- AD
Alzheimer’s Disease
- BBB
Blood-brain-barrier
- CSF
Cerebrospinal Fluid
- FST
Forced Swim Test
- GCs
Glucocorticoids
- HPA
Hypothalamus-pituitary-adrenal
- LC
Locus Coeruleus
- LPS
Lipopolysaccharide
- MDD
Major Depressive Disorder
- miRNA
microRNAs
- NAc
Nucleus Accumbens
- PTSD
Post-traumatic Stress Disorder
- SNRI
Serotonin-noradrenaline Reuptake Inhibitors
- TNF-α
Tumor Necrosis Factor Alpha
- TST
Tail Suspension Test
CONSENT FOR PUBLICATION
Not applicable.
FUNDING
This work has been supported by NIH grants R01 DA049837, R21 MH129020, R01 DA056534, and NSF grant IOS-1929829 to D.A.B., an NIMH R56MH124930-01A1 to G.E.H. and a Hellenic Foundation for Research and Innovation (H.F.R.I.) Grant with Number: HFRI‐FM17‐1676 to C.D.
CONFLICT OF INTEREST
The authors declare no conflict of interest, financial or otherwise.
REFERENCES
- 1.Munck A., Guyre P.M., Holbrook N.J. Physiological functions of glucocorticoids in stress and their relation to pharmacological actions. Endocr. Rev. 1984;5(1):25–44. doi: 10.1210/edrv-5-1-25. [DOI] [PubMed] [Google Scholar]
- 2.McEwen B.S., Gianaros P.J. Stress- and allostasis-induced brain plasticity. Annu. Rev. Med. 2011;62(1):431–445. doi: 10.1146/annurev-med-052209-100430. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Lyons D.M., Parker K.J., Schatzberg A.F. Animal models of early life stress: Implications for understanding resilience. Dev. Psychobiol. 2010;52(7):616–624. doi: 10.1002/dev.20500. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Masten A.S. Ordinary magic: Resilience processes in development. Am. Psychol. 2001;56(3):227–238. doi: 10.1037/0003-066X.56.3.227. [DOI] [PubMed] [Google Scholar]
- 5.De Berardis D., Fornaro M., Orsolini L. Editorial: “No Words for Feelings, Yet!” exploring alexithymia, disorder of affect regulation, and the “Mind-Body” connection. Front. Psychiatry. 2020;11:593462. doi: 10.3389/fpsyt.2020.593462. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Grandinetti P., Gooney M., Scheibein F., Testa R., Ruggieri G., Tondo P., Corona A., Boi G., Floris L., Profeta V.F., G. Wells J.S., De Berardis D. Stress and maladaptive coping of italians health care professionals during the first wave of the pandemic. Brain Sci. 2021;11(12):1586. doi: 10.3390/brainsci11121586. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Wilson R.S., Arnold S.E., Schneider J.A., Kelly J.F., Tang Y., Bennett D.A. Chronic psychological distress and risk of Alzheimer’s disease in old age. Neuroepidemiology. 2006;27(3):143–153. doi: 10.1159/000095761. [DOI] [PubMed] [Google Scholar]
- 8.Riboni F.V., Belzung C. Stress and psychiatric disorders: From categorical to dimensional approaches. Curr. Opin. Behav. Sci. 2017;14:72–77. doi: 10.1016/j.cobeha.2016.12.011. [DOI] [Google Scholar]
- 9.Newman S.C., Bland R.C. Life events and the 1-year prevalence of major depressive episode, generalized anxiety disorder, and panic disorder in a community sample. Compr. Psychiatry. 1994;35(1):76–82. doi: 10.1016/0010-440X(94)90173-2. [DOI] [PubMed] [Google Scholar]
- 10.Altemus M., Sarvaiya N., Epperson N.C. Sex differences in anxiety and depression clinical perspectives. Front. Neuroendocrinol. 2014;35(3):320–330. doi: 10.1016/j.yfrne.2014.05.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Kessler R.C., Petukhova M., Sampson N.A., Zaslavsky A.M., Wittchen H.U. Twelve-month and lifetime prevalence and lifetime morbid risk of anxiety and mood disorders in the United States. Int. J. Methods Psychiatr. Res. 2012;21(3):169–184. doi: 10.1002/mpr.1359. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Marcus S.M., Young E.A., Kerber K.B., Kornstein S., Farabaugh A.H., Mitchell J., Wisniewski S.R., Balasubramani G.K., Trivedi M.H., Rush A.J. Gender differences in depression: Findings from the STAR*D study. J. Affect. Disord. 2005;87(2-3):141–150. doi: 10.1016/j.jad.2004.09.008. [DOI] [PubMed] [Google Scholar]
- 13.McLean C.P., Asnaani A., Litz B.T., Hofmann S.G. Gender differences in anxiety disorders: Prevalence, course of illness, comorbidity and burden of illness. J. Psychiatr. Res. 2011;45(8):1027–1035. doi: 10.1016/j.jpsychires.2011.03.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Kessler R.C., Aguilar-Gaxiola S., Alonso J., Chatterji S., Lee S., Ormel J., Üstün T.B., Wang P.S. The global burden of mental disorders: An update from the WHO World Mental Health (WMH) Surveys. Epidemiol. Psichiatr. Soc. 2009;18(1):23–33. doi: 10.1017/S1121189X00001421. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Tolin D.F., Foa E.B. Sex differences in trauma and posttraumatic stress disorder: A quantitative review of 25 years of research. Psychol. Bull. 2006;132(6):959–992. doi: 10.1037/0033-2909.132.6.959. [DOI] [PubMed] [Google Scholar]
- 16.Lydiard R.B. Irritable bowel syndrome, anxiety, and depression: what are the links? J. Clin. Psychiatry. 2001;62(S8):38–45. [PubMed] [Google Scholar]
- 17.Beghi E., Allais G., Cortelli P., D’Amico D., De Simone R., d’Onofrio F., Genco S., Manzoni G.C., Moschiano F., Tonini M.C., Torelli P., Quartaroli M., Roncolato M., Salvi S., Bussone G. Headache and anxiety-depressive disorder comorbidity: The HADAS study. Neurol. Sci. 2007;28(S2):S217–S219. doi: 10.1007/s10072-007-0780-6. [DOI] [PubMed] [Google Scholar]
- 18.van Mill J.G., Hoogendijk W.J.G., Vogelzangs N., van Dyck R., Penninx B.W.J.H. Insomnia and sleep duration in a large cohort of patients with major depressive disorder and anxiety disorders. J. Clin. Psychiatry. 2010;71(3):239–246. doi: 10.4088/JCP.09m05218gry. [DOI] [PubMed] [Google Scholar]
- 19.Lipton R.B., Stewart W.F., Diamond S., Diamond M.L., Reed M. Prevalence and burden of migraine in the United States: Data from the American Migraine Study II. Headache. 2001;41(7):646–657. doi: 10.1046/j.1526-4610.2001.041007646.x. [DOI] [PubMed] [Google Scholar]
- 20.Singareddy R., Vgontzas A.N., Fernandez-Mendoza J., Liao D., Calhoun S., Shaffer M.L., Bixler E.O. Risk factors for incident chronic insomnia: A general population prospective study. Sleep Med. 2012;13(4):346–353. doi: 10.1016/j.sleep.2011.10.033. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Drossman D.A., Thompson W.G., Talley N.J., Funch-Jensen P., Janssens J., Whitehead W.E. Identification of sub-groups of functional gastrointestinal disorders. Gastroenterol. Intl. 1990;3(4):159–172. [Google Scholar]
- 22.Gao S., Hendrie H.C., Hall K.S., Hui S. The relationships between age, sex, and the incidence of dementia and Alzheimer disease: A meta-analysis. Arch. Gen. Psychiatry. 1998;55(9):809–815. doi: 10.1001/archpsyc.55.9.809. [DOI] [PubMed] [Google Scholar]
- 23.Medeiros A.M., Silva R.H. Sex differences in Alzheimer’s Disease: Where do we stand? J. Alzheimers Dis. 2019;67(1):35–60. doi: 10.3233/JAD-180213. [DOI] [PubMed] [Google Scholar]
- 24.Novais F., Starkstein S. Phenomenology of depression in Alzheimer’s Disease. J. Alzheimers Dis. 2015;47(4):845–855. doi: 10.3233/JAD-148004. [DOI] [PubMed] [Google Scholar]
- 25.Kouzoupis A.V., Lyrakos D., Kokras N., Panagiotarakou M., Syrigos K.N., Papadimitriou G.N. Dysfunctional remembered parenting in oncology outpatients affects psychological distress symptoms in a gender‐specific manner. Stress Health. 2012;28(5):381–388. doi: 10.1002/smi.2460. [DOI] [PubMed] [Google Scholar]
- 26.Riecher-Rössler A., Butler S., Kulkarni J. Sex and gender differences in schizophrenic psychoses-a critical review. Arch. Women Ment. Health. 2018;21(6):627–648. doi: 10.1007/s00737-018-0847-9. [DOI] [PubMed] [Google Scholar]
- 27.McGrath J., Saha S., Chant D., Welham J. Schizophrenia: A concise overview of incidence, prevalence, and mortality. Epidemiol. Rev. 2008;30(1):67–76. doi: 10.1093/epirev/mxn001. [DOI] [PubMed] [Google Scholar]
- 28.Green M.J., Girshkin L., Teroganova N., Quidé Y. Stress,Schizophrenia and Bipolar Disorder; In: Behavioral Neurobiology of Stress-related Disorders, SpringerLink. 2014. pp. 217–235. [DOI] [PubMed] [Google Scholar]
- 29.Martin L.A., Neighbors H.W., Griffith D.M. The experience of symptoms of depression in men vs. women: Analysis of the national comorbidity survey replication. JAMA Psychiatry. 2013;70(10):1100–1106. doi: 10.1001/jamapsychiatry.2013.1985. [DOI] [PubMed] [Google Scholar]
- 30.Gururajan A., Reif A., Cryan J.F., Slattery D.A. The future of rodent models in depression research. Nat. Rev. Neurosci. 2019;20(11):686–701. doi: 10.1038/s41583-019-0221-6. [DOI] [PubMed] [Google Scholar]
- 31.Beery A.K., Zucker I. Sex bias in neuroscience and biomedical research. Neurosci. Biobehav. Rev. 2011;35(3):565–572. doi: 10.1016/j.neubiorev.2010.07.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Tannenbaum C., Schwarz J.M., Clayton J.A., de Vries G.J., Sullivan C. Evaluating sex as a biological variable in preclinical research: The devil in the details. Biol. Sex Differ. 2016;7(1):13. doi: 10.1186/s13293-016-0066-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Mamlouk G.M., Dorris D.M., Barrett L.R., Meitzen J. Sex bias and omission in neuroscience research is influenced by research model and journal, but not reported NIH funding. Front. Neuroendocrinol. 2020;57:100835. doi: 10.1016/j.yfrne.2020.100835. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Rechlin R.K., Splinter T.F.L., Hodges T.E., Albert A.Y., Galea L.A.M. An analysis of neuroscience and psychiatry papers published from 2009 and 2019 outlines opportunities for increasing discovery of sex differences. Nat. Commun. 2022;13(1):2137. doi: 10.1038/s41467-022-29903-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Dalla C. Integrating sex and gender in mental health research: Enhanced funding for better treatments. Nat. Mental Health. 2023;1(6):383–384. doi: 10.1038/s44220-023-00076-2. [DOI] [Google Scholar]
- 36.Kokras N., Hodes G.E., Bangasser D.A., Dalla C. Sex differences in the hypothalamic-pituitary-adrenal axis: An obstacle to antidepressant drug development? Br. J. Pharmacol. 2019;176(21):4090–4106. doi: 10.1111/bph.14710. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Atkinson H.C., Waddell B.J. Circadian variation in basal plasma corticosterone and adrenocorticotropin in the rat: sexual dimorphism and changes across the estrous cycle. Endocrinology. 1997;138(9):3842–3848. doi: 10.1210/endo.138.9.5395. [DOI] [PubMed] [Google Scholar]
- 38.Weinstock M., Razin M., Schorer-apelbaum D., Men D., McCarty R. Gender differences in sympathoadrenal activity in rats at rest and in response to footshock stress. Int. J. Dev. Neurosci. 1998;16(3-4):289–295. doi: 10.1016/S0736-5748(98)00021-5. [DOI] [PubMed] [Google Scholar]
- 39.Kokras N., Pastromas N., Papasava D., de Bournonville C., Cornil C.A., Dalla C. Sex differences in behavioral and neurochemical effects of gonadectomy and aromatase inhibition in rats. Psychoneuroendocrinology. 2018;87:93–107. doi: 10.1016/j.psyneuen.2017.10.007. [DOI] [PubMed] [Google Scholar]
- 40.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(3):703–714. doi: 10.1016/j.neuroscience.2005.06.068. [DOI] [PubMed] [Google Scholar]
- 41.Bangasser D.A., Valentino R.J. Sex differences in stress-related psychiatric disorders: Neurobiological perspectives. Front. Neuroendocrinol. 2014;35(3):303–319. doi: 10.1016/j.yfrne.2014.03.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Kokras N., Sotiropoulos I., Pitychoutis P.M., Almeida O.F.X., Papadopoulou-Daifoti Z. Citalopram-mediated anxiolysis and differing neurobiological responses in both sexes of a genetic model of depression. Neuroscience. 2011;194:62–71. doi: 10.1016/j.neuroscience.2011.07.077. [DOI] [PubMed] [Google Scholar]
- 43.Gala R.R., Westphal U. Further studies on the corticosteroid-binding globulin in the rat: Proposed endocrine control. Endocrinology. 1966;79(1):67–76. doi: 10.1210/endo-79-1-67. [DOI] [PubMed] [Google Scholar]
- 44.Oyola M.G., Handa R.J. Hypothalamic-pituitary-adrenal and hypothalamic-pituitary-gonadal axes: Sex differences in regulation of stress responsivity. Stress. 2017;20(5):476–494. doi: 10.1080/10253890.2017.1369523. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Kokras N., Dalla C., Sideris A.C., Dendi A., Mikail H.G., Antoniou K., Papadopoulou-Daifoti Z. Behavioral sexual dimorphism in models of anxiety and depression due to changes in HPA axis activity. Neuropharmacology. 2012;62(1):436–445. doi: 10.1016/j.neuropharm.2011.08.025. [DOI] [PubMed] [Google Scholar]
- 46.Kokras N., Krokida S., Varoudaki T.Z., Dalla C. Do corticosterone levels predict female depressive‐like behavior in rodents? J. Neurosci. Res. 2021;99(1):324–331. doi: 10.1002/jnr.24686. [DOI] [PubMed] [Google Scholar]
- 47.Rivier C. Gender, sex steroids, corticotropin-releasing factor, nitric oxide, and the HPA response to stress. Pharmacol. Biochem. Behav. 1999;64(4):737–751. doi: 10.1016/S0091-3057(99)00148-3. [DOI] [PubMed] [Google Scholar]
- 48.Viau V., Bingham B., Davis J., Lee P., Wong M. Gender and puberty interact on the stress-induced activation of parvocellular neurosecretory neurons and corticotropin-releasing hormone messenger ribonucleic acid expression in the rat. Endocrinology. 2005;146(1):137–146. doi: 10.1210/en.2004-0846. [DOI] [PubMed] [Google Scholar]
- 49.Wood G.E., Beylin A.V., Shors T.J. The contribution of adrenal and reproductive hormones to the opposing effects of stress on trace conditioning males versus females. Behav. Neurosci. 2001;115(1):175–187. doi: 10.1037/0735-7044.115.1.175. [DOI] [PubMed] [Google Scholar]
- 50.Bangasser D.A., Shors T.J. The hippocampus is necessary for enhancements and impairments of learning following stress. Nat. Neurosci. 2007;10(11):1401–1403. doi: 10.1038/nn1973. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Dalla C., Shors T.J. Sex differences in learning processes of classical and operant conditioning. Physiol. Behav. 2009;97(2):229–238. doi: 10.1016/j.physbeh.2009.02.035. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Dalla C., Whetstone A.S., Hodes G.E., Shors T.J. Stressful experience has opposite effects on dendritic spines in the hippocampus of cycling versus masculinized females. Neurosci. Lett. 2009;449(1):52–56. doi: 10.1016/j.neulet.2008.10.051. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Shors T.J., 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(16):6292–6297. doi: 10.1523/JNEUROSCI.21-16-06292.2001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Leuner B., Shors T.J. New spines, new memories. Mol. Neurobiol. 2004;29(2):117–130. doi: 10.1385/MN:29:2:117. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Dalla C., Pitychoutis P.M., Kokras N., Papadopoulou-Daifoti Z. Sex differences in response to stress and expression of depressive-like behaviours in the rat. Curr. Top. Behav. Neurosci. 2011;8:97–118. doi: 10.1007/7854_2010_94. [DOI] [PubMed] [Google Scholar]
- 56.Kokras N., Antoniou K., Dalla C., Bekris S., Xagoraris M., Ovestreet D.H., Papadopoulou-Daifoti Z. Sex-related differential response to clomipramine treatment in a rat model of depression. J. Psychopharmacol. 2009;23(8):945–956. doi: 10.1177/0269881108095914. [DOI] [PubMed] [Google Scholar]
- 57.Mikail H.G., Dalla C., Kokras N., Kafetzopoulos V., Papadopoulou-Daifoti Z. Sertraline behavioral response associates closer and dose-dependently with cortical rather than hippocampal serotonergic activity in the rat forced swim stress. Physiol. Behav. 2012;107(2):201–206. doi: 10.1016/j.physbeh.2012.06.016. [DOI] [PubMed] [Google Scholar]
- 58.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(3):595–605. doi: 10.1016/j.physbeh.2007.10.020. [DOI] [PubMed] [Google Scholar]
- 59.Kokras N., Antoniou K., Polissidis A., Papadopoulou-Daifoti Z. Antidepressants induce regionally discrete, sex-dependent changes in brain’s glutamate content. Neurosci. Lett. 2009;464(2):98–102. doi: 10.1016/j.neulet.2009.08.011. [DOI] [PubMed] [Google Scholar]
- 60.Shors T.J., Falduto J., Leuner B. The opposite effects of stress on dendritic spines in male vs. female rats are NMDA receptor-dependent. Eur. J. Neurosci. 2004;19(1):145–150. doi: 10.1046/j.1460-9568.2003.03065.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Kokras N., Sotiropoulos I., Besinis D., Tzouveka E.L., Almeida O.F.X., Sousa N., Dalla C. Neuroplasticity-related correlates of environmental enrichment combined with physical activity differ between the sexes. Eur. Neuropsychopharmacol. 2019;29(1):1–15. doi: 10.1016/j.euroneuro.2018.11.1107. [DOI] [PubMed] [Google Scholar]
- 62.Andolina D., Maran D., Viscomi M.T., Puglisi-Allegra S. Strain-dependent variations in stress coping behavior are mediated by a 5-HT/GABA interaction within the prefrontal corticolimbic system. Int. J. Neuropsychopharmacol. 2015;18(3):pyu074. doi: 10.1093/ijnp/pyu074. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Treccani G., Musazzi L., Perego C., Milanese M., Nava N., Bonifacino T., Lamanna J., Malgaroli A., Drago F., Racagni G., Nyengaard J.R., Wegener G., Bonanno G., Popoli M. Acute stress rapidly increases the readily releasable pool of glutamate vesicles in prefrontal and frontal cortex through non-genomic action of corticosterone. Mol. Psychiatry. 2014;19(4):401. doi: 10.1038/mp.2014.20. [DOI] [PubMed] [Google Scholar]
- 64.Bremner J.D., Licinio J., Darnell A., Krystal J.H., Owens M.J., Southwick S.M., Nemeroff C.B., Charney D.S. Elevated CSF corticotropin-releasing factor concentrations in posttraumatic stress disorder. Am. J. Psychiatry. 1997;154(5):624–629. doi: 10.1176/ajp.154.5.624. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Banki C.M., Karmacsi L., Bissette G., Nemeroff C.B. CSF corticotropin-releasing hormone and somatostatin in major depression: Response to antidepressant treatment and relapse. Eur. Neuropsychopharmacol. 1992;2(2):107–113. doi: 10.1016/0924-977X(92)90019-5. [DOI] [PubMed] [Google Scholar]
- 66.Heuser I., Bissette G., Dettling M., Schweiger U., Gotthardt U., Schmider J., Lammers C.H., Nemeroff C.B., Holsboer F. Cerebrospinal fluid concentrations of corticotropin-releasing hormone, vasopressin, and somatostatin in depressed patients and healthy controls: Response to amitriptyline treatment. Depress. Anxiety. 1998;8(2):71–79. doi: 10.1002/(SICI)1520-6394(1998)8:2<71:AID-DA5>3.0.CO;2-N. [DOI] [PubMed] [Google Scholar]
- 67.Austin M.C., Janosky J.E., Murphy H.A. Increased corticotropin-releasing hormone immunoreactivity in monoamine-containing pontine nuclei of depressed suicide men. Mol. Psychiatry. 2003;8(3):324–332. doi: 10.1038/sj.mp.4001250. [DOI] [PubMed] [Google Scholar]
- 68.Bissette G., Klimek V., Pan J., Stockmeier C., Ordway G. Elevated concentrations of CRF in the locus coeruleus of depressed subjects. Neuropsychopharmacology. 2003;28(7):1328–1335. doi: 10.1038/sj.npp.1300191. [DOI] [PubMed] [Google Scholar]
- 69.Vandael D., Gounko N.V. Corticotropin releasing factor-binding protein (CRF-BP) as a potential new therapeutic target in Alzheimer’s disease and stress disorders. Transl. Psychiatry. 2019;9(1):272. doi: 10.1038/s41398-019-0581-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Pomara N., Greenberg W.M., Branford M.D., Doraiswamy P.M. Therapeutic implications of HPA axis abnormalities in Alzheimer’s disease: Review and update. Psychopharmacol. Bull. 2003;37(2):120–134. [PubMed] [Google Scholar]
- 71.Whitehouse P.J., Vale W.W., Zweig R.M., Singer H.S., Mayeux R., Kuhar M.J., Price D.L., De Souza E.B. Reductions in corticotropin releasing factor-like immunoreactivity in cerebral cortex in Alzheimer’s disease, Parkinson’s disease, and progressive supranuclear palsy. Neurology. 1987;37(6):905–909. doi: 10.1212/WNL.37.6.905. [DOI] [PubMed] [Google Scholar]
- 72.Souza E.B.D. CRH defects in Alzheimer’s and other neurologic diseases. Hosp. Pract. 1988;23(9):59–71. doi: 10.1080/21548331.1988.11703535. [DOI] [PubMed] [Google Scholar]
- 73.Gallucci W.T., Baum A., Laue L., Rabin D.S., Chrousos G.P., Gold P.W., Kling M.A. Sex differences in sensitivity of the hypothalamic-pituitary-adrenal axis. Health Psychol. 1993;12(5):420–425. doi: 10.1037/0278-6133.12.5.420. [DOI] [PubMed] [Google Scholar]
- 74.Bangasser D.A., Wiersielis K.R. Sex differences in stress responses: A critical role for corticotropin-releasing factor. Hormones. 2018;17(1):5–13. doi: 10.1007/s42000-018-0002-z. [DOI] [PubMed] [Google Scholar]
- 75.Dunčko, R.; Kiss, A.; Škultétyová, I.; Rusnák, M.; Ježová, D. Corticotropin-releasing hormone mRNA levels in response to chronic mild stress rise in male but not in female rats while tyrosine hydroxylase mRNA levels decrease in both sexes. Psychoneuroendocrinology. 2001;26(1):77–89. doi: 10.1016/S0306-4530(00)00040-8. [DOI] [PubMed] [Google Scholar]
- 76.Speert D.B., McClennen S.J., Seasholtz A.F. Sexually dimorphic expression of corticotropin-releasing hormone-binding protein in the mouse pituitary. Endocrinology. 2002;143(12):4730–4741. doi: 10.1210/en.2002-220556. [DOI] [PubMed] [Google Scholar]
- 77.Wiersielis K.R., Ceretti A., Hall A., Famularo S.T., Salvatore M., Ellis A.S., Jang H., Wimmer M.E., Bangasser D.A. Sex differences in corticotropin releasing factor regulation of medial septum-mediated memory formation. Neurobiol. Stress. 2019;10:100150. doi: 10.1016/j.ynstr.2019.100150. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Bale T.L., Vale W.W. Increased depression-like behaviors in corticotropin-releasing factor receptor-2-deficient mice: sexually dichotomous responses. J. Neurosci. 2003;23(12):5295–5301. doi: 10.1523/JNEUROSCI.23-12-05295.2003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Bale T.L., Picetti R., Contarino A., Koob G.F., Vale W.W., Lee K.F. Mice deficient for both corticotropin-releasing factor receptor 1 (CRFR1) and CRFR2 have an impaired stress response and display sexually dichotomous anxiety-like behavior. J. Neurosci. 2002;22(1):193–199. doi: 10.1523/JNEUROSCI.22-01-00193.2002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Bale T.L. Sensitivity to stress: Dysregulation of CRF pathways and disease development. Horm. Behav. 2005;48(1):1–10. doi: 10.1016/j.yhbeh.2005.01.009. [DOI] [PubMed] [Google Scholar]
- 81.Weathington J.M., Hamki A., Cooke B.M. Sex- and region-specific pubertal maturation of the corticotropin-releasing factor receptor system in the rat. J. Comp. Neurol. 2014;522(6):1284–1298. doi: 10.1002/cne.23475. [DOI] [PubMed] [Google Scholar]
- 82.Rosinger Z.J., Jacobskind J.S., Park S.G., Justice N.J., Zuloaga D.G. Distribution of corticotropin-releasing factor receptor 1 in the developing mouse forebrain: A novel sex difference revealed in the rostral periventricular hypothalamus. Neuroscience. 2017;361:167–178. doi: 10.1016/j.neuroscience.2017.08.016. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Rosinger Z.J., De Guzman R.M., Jacobskind J.S., Saglimbeni B., Malone M., Fico D., Justice N.J., Forni P.E., Zuloaga D.G. Sex-dependent effects of chronic variable stress on discrete corticotropin-releasing factor receptor 1 cell populations. Physiol. Behav. 2020;219:112847. doi: 10.1016/j.physbeh.2020.112847. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84.Howerton A.R., Roland A.V., Fluharty J.M., Marshall A., Chen A., Daniels D., Beck S.G., Bale T.L. Sex differences in corticotropin-releasing factor receptor-1 action within the dorsal raphe nucleus in stress responsivity. Biol. Psychiatry. 2014;75(11):873–883. doi: 10.1016/j.biopsych.2013.10.013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85.Williams T.J., Akama K.T., Knudsen M.G., McEwen B.S., Milner T.A. Ovarian hormones influence corticotropin releasing factor receptor colocalization with delta opioid receptors in CA1 pyramidal cell dendrites. Exp. Neurol. 2011;230(2):186–196. doi: 10.1016/j.expneurol.2011.04.012. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86.Hauger R.L., Risbrough V., Oakley R.H., Olivares-Reyes J.A., Dautzenberg F.M. Role of CRF receptor signaling in stress vulnerability, anxiety, and depression. Ann. N. Y. Acad. Sci. 2009;1179(1):120–143. doi: 10.1111/j.1749-6632.2009.05011.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87.Hillhouse E.W., Grammatopoulos D.K. The molecular mechanisms underlying the regulation of the biological activity of corticotropin-releasing hormone receptors: implications for physiology and pathophysiology. Endocr. Rev. 2006;27(3):260–286. doi: 10.1210/er.2005-0034. [DOI] [PubMed] [Google Scholar]
- 88.Berridge C.W., Foote S.L. Effects of locus coeruleus activation on electroencephalographic activity in neocortex and hippocampus. J. Neurosci. 1991;11(10):3135–3145. doi: 10.1523/JNEUROSCI.11-10-03135.1991. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89.Berridge C.W., Waterhouse B.D. The locus coeruleus-noradrenergic system: Modulation of behavioral state and state-dependent cognitive processes. Brain Res. Brain Res. Rev. 2003;42(1):33–84. doi: 10.1016/S0165-0173(03)00143-7. [DOI] [PubMed] [Google Scholar]
- 90.Gary Aston-Jones. In: M.G. Role of the locus coeruleusnorepinephrine system in arousal and circadian regulation of the sleep-wake cycle. In: Brain norepinephrine: Neurobiology and therapeutics. Ordway G.A, Frazer A, editors. Cambridge University Press,; 2007. pp. 157–195. [Google Scholar]
- 91.Bangasser D.A., Curtis A., Reyes B.A., Bethea T.T., Parastatidis I., Ischiropoulos H., Van Bockstaele E.J., Valentino R.J. Sex differences in corticotropin-releasing factor receptor signaling and trafficking: Potential role in female vulnerability to stress-related psychopathology. Mol. Psychiatry. 2010;15(9):877-896–904. doi: 10.1038/mp.2010.89. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92.Bates M.L.S., Arner J.R., Curtis A.L., Valentino R., Bhatnagar S. Sex-specific alterations in corticotropin-releasing factor regulation of coerulear-cortical network activity. Neuropharmacology. 2023;223:109317. doi: 10.1016/j.neuropharm.2022.109317. [DOI] [PubMed] [Google Scholar]
- 93.Coker A.L., Weston R., Creson D.L., Justice B., Blakeney P. PTSD symptoms among men and women survivors of intimate partner violence: the role of risk and protective factors. Violence Vict. 2005;20(6):625–643. doi: 10.1891/0886-6708.20.6.625. [DOI] [PubMed] [Google Scholar]
- 94.Breslau N., Chilcoat H.D., Kessler R.C., Peterson E.L., Lucia V.C. Vulnerability to assaultive violence: further specification of the sex difference in post-traumatic stress disorder. Psychol. Med. 1999;29(4):813–821. doi: 10.1017/S0033291799008612. [DOI] [PubMed] [Google Scholar]
- 95.Plante D.T., Landsness E.C., Peterson M.J., Goldstein M.R., Riedner B.A., Wanger T., Guokas J.J., Tononi G., Benca R.M. Sex-related differences in sleep slow wave activity in major depressive disorder: A high-density EEG investigation. BMC Psychiatry. 2012;12(1):146. doi: 10.1186/1471-244X-12-146. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96.Nolen-Hoeksema S., Larson J., Grayson C. Explaining the gender difference in depressive symptoms. J. Pers. Soc. Psychol. 1999;77(5):1061–1072. doi: 10.1037/0022-3514.77.5.1061. [DOI] [PubMed] [Google Scholar]
- 97.Lefkowitz R.J., Shenoy S.K. Transduction of receptor signals by beta-arrestins. Science. 2005;308(5721):512–517. doi: 10.1126/science.1109237. [DOI] [PubMed] [Google Scholar]
- 98.Violin J.D. Lefkowitz, R.J. β-Arrestin-biased ligands at seven-transmembrane receptors. Trends Pharmacol. Sci. 2007;28(8):416–422. doi: 10.1016/j.tips.2007.06.006. [DOI] [PubMed] [Google Scholar]
- 99.Bangasser D.A., Dong H., Carroll J., Plona Z., Ding H., Rodriguez L., McKennan C., Csernansky J.G., Seeholzer S.H., Valentino R.J. Corticotropin-releasing factor overexpression gives rise to sex differences in Alzheimer’s disease-related signaling. Mol. Psychiatry. 2017;22(8):1126–1133. doi: 10.1038/mp.2016.185. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100.Valentino R.J., Van Bockstaele E., Bangasser D. Sex-specific cell signaling: The corticotropin-releasing factor receptor model. Trends Pharmacol. Sci. 2013;34(8):437–444. doi: 10.1016/j.tips.2013.06.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101.Murrough J.W., Charney D.S. Corticotropin-releasing factor type 1 receptor antagonists for stress-related disorders: Time to call it quits? Biol. Psychiatry. 2017;82(12):858–860. doi: 10.1016/j.biopsych.2017.10.012. [DOI] [PubMed] [Google Scholar]
- 102.Mansbach R.S., Brooks E.N., Chen Y.L. Antidepressant-like effects of CP-154,526, a selective CRF1 receptor antagonist. Eur. J. Pharmacol. 1997;323(1):21–26. doi: 10.1016/S0014-2999(97)00025-3. [DOI] [PubMed] [Google Scholar]
- 103.Schulz D.W., Mansbach R.S., Sprouse J., Braselton J.P., Collins J., Corman M., Dunaiskis A., Faraci S., Schmidt A.W., Seeger T., Seymour P., Tingley F.D., III, Winston E.N., Chen Y.L., Heym J. CP-154,526: A potent and selective nonpeptide antagonist of corticotropin releasing factor receptors. Proc. Natl. Acad. Sci. . 1996;93(19):10477–10482. doi: 10.1073/pnas.93.19.10477. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104.Deak T., Nguyen K.T., Ehrlich A.L., Watkins L.R., Spencer R.L., Maier S.F., Licinio J., Wong M.L., Chrousos G.P., Webster E., Gold P.W. The impact of the nonpeptide corticotropin-releasing hormone antagonist antalarmin on behavioral and endocrine responses to stress. Endocrinology. 1999;140(1):79–86. doi: 10.1210/endo.140.1.6415. [DOI] [PubMed] [Google Scholar]
- 105.Zorrilla E.P., Valdez G.R., Nozulak J., Koob G.F., Markou A. Effects of antalarmin, a CRF type 1 receptor antagonist, on anxiety-like behavior and motor activation in the rat. Brain Res. 2002;952(2):188–199. doi: 10.1016/S0006-8993(02)03189-X. [DOI] [PubMed] [Google Scholar]
- 106.Chaki S., Nakazato A., Kennis L., Nakamura M., Mackie C., Sugiura M., Vinken P., Ashton D., Langlois X., Steckler T. Anxiolytic- and antidepressant-like profile of a new CRF1 receptor antagonist, R278995/CRA0450. Eur. J. Pharmacol. 2004;485(1-3):145–158. doi: 10.1016/j.ejphar.2003.11.032. [DOI] [PubMed] [Google Scholar]
- 107.Ising M., Zimmermann U.S., Künzel H.E., Uhr M., Foster A.C., Learned-Coughlin S.M., Holsboer F., Grigoriadis D.E. High-affinity CRF1 receptor antagonist NBI-34041: preclinical and clinical data suggest safety and efficacy in attenuating elevated stress response. Neuropsychopharmacology. 2007;32(9):1941–1949. doi: 10.1038/sj.npp.1301328. [DOI] [PubMed] [Google Scholar]
- 108.Caruso A., Nicoletti F., Gaetano A., Scaccianoce S. Risk factors for Alzheimer’s disease: Focus on stress. Front. Pharmacol. 2019;10:976. doi: 10.3389/fphar.2019.00976. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 109.Ouanes S., Popp J. High cortisol and the risk of dementia and alzheimer’s disease: A review of the literature. Front. Aging Neurosci. 2019;11:43. doi: 10.3389/fnagi.2019.00043. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110.Csernansky J.G., Dong H., Fagan A.M., Wang L., Xiong C., Holtzman D.M., Morris J.C. Plasma cortisol and progression of dementia in subjects with Alzheimer-type dementia. Am. J. Psychiatry. 2006;163(12):2164–2169. doi: 10.1176/ajp.2006.163.12.2164. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 111.Elgh E., Lindqvist Åstot A., Fagerlund M., Eriksson S., Olsson T., Näsman B. Cognitive dysfunction, hippocampal atrophy and glucocorticoid feedback in Alzheimer’s disease. Biol. Psychiatry. 2006;59(2):155–161. doi: 10.1016/j.biopsych.2005.06.017. [DOI] [PubMed] [Google Scholar]
- 112.Vyas S., Rodrigues A.J., Silva J.M., Tronche F., Almeida O.F.X., Sousa N., Sotiropoulos I. Chronic stress and glucocorticoids: From neuronal plasticity to neurodegeneration. Neural Plast. 2016;2016:1–15. doi: 10.1155/2016/6391686. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 113.Hatzinger M. Z’Brun, A.; Hemmeter, U.; Seifritz, E.; Baumann, F.; Holsboer-Trachsler, E.; Heuser, I.J. Hypothalamic-pituitary-adrenal system function in patients with alzheimer’s disease. Neurobiol. Aging. 1995;16(2):205–209. doi: 10.1016/0197-4580(94)00159-6. [DOI] [PubMed] [Google Scholar]
- 114.Peskind E.R., Wilkinson C.W., Petrie E.C., Schellenberg G.D., Raskind M.A. Increased CSF cortisol in AD is a function of APOE genotype. Neurology. 2001;56(8):1094–1098. doi: 10.1212/WNL.56.8.1094. [DOI] [PubMed] [Google Scholar]
- 115.Greenwald B.S., Mathé A.A., Mohs R.C., Levy M.I., Johns C.A., Davis K.L. Cortisol and Alzheimer’s disease, II: Dexamethasone suppression, dementia severity, and affective symptoms. Am. J. Psychiatry. 1986;143(4):442–446. doi: 10.1176/ajp.143.4.442. [DOI] [PubMed] [Google Scholar]
- 116.Hartmann A., Veldhuis J.D., Deuschle M., Standhardt H., Heuser I. Twenty-four hour cortisol release profiles in patients with Alzheimer’s and Parkinson’s disease compared to normal controls: Ultradian secretory pulsatility and diurnal variation. Neurobiol. Aging. 1997;18(3):285–289. doi: 10.1016/S0197-4580(97)80309-0. [DOI] [PubMed] [Google Scholar]
- 117.Rasmuson S., Näsman B., Olsson T. Increased serum levels of dehydroepiandrosterone (DHEA) and interleukin-6 (IL-6) in women with mild to moderate Alzheimer’s disease. Int. Psychogeriatr. 2011;23(9):1386–1392. doi: 10.1017/S1041610211000810. [DOI] [PubMed] [Google Scholar]
- 118.Toledo J.B., Toledo E., Weiner M.W., Jack C.R., Jr, Jagust W., Lee V.M.Y., Shaw L.M., Trojanowski J.Q. Cardiovascular risk factors, cortisol, and amyloid‐β deposition in Alzheimer’s Disease Neuroimaging Initiative. Alzheimers Dement. 2012;8(6):483–489. doi: 10.1016/j.jalz.2011.08.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 119.Catania C., Sotiropoulos I., Silva R., Onofri C., Breen K.C., Sousa N., Almeida O F X. The amyloidogenic potential and behavioral correlates of stress. Mol. Psychiatry. 2009;14(1):95–105. doi: 10.1038/sj.mp.4002101. [DOI] [PubMed] [Google Scholar]
- 120.Green K.N., Billings L.M., Roozendaal B., McGaugh J.L., LaFerla F.M. Glucocorticoids increase amyloid-beta and tau pathology in a mouse model of Alzheimer’s disease. J. Neurosci. 2006;26(35):9047–9056. doi: 10.1523/JNEUROSCI.2797-06.2006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 121.Xia M., Yang L., Sun G., Qi S., Li B. Mechanism of depression as a risk factor in the development of Alzheimer’s disease: The function of AQP4 and the glymphatic system. Psychopharmacology. 2017;234(3):365–379. doi: 10.1007/s00213-016-4473-9. [DOI] [PubMed] [Google Scholar]
- 122.Devi L., Alldred M.J., Ginsberg S.D., Ohno M. Sex- and brain region-specific acceleration of β-amyloidogenesis following behavioral stress in a mouse model of Alzheimer’s disease. Mol. Brain. 2010;3(1):34. doi: 10.1186/1756-6606-3-34. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 123.Sotiropoulos I., Catania C., Pinto L.G., Silva R., Pollerberg G.E., Takashima A., Sousa N., Almeida O.F.X. Stress acts cumulatively to precipitate Alzheimer’s disease-like tau pathology and cognitive deficits. J. Neurosci. 2011;31(21):7840–7847. doi: 10.1523/JNEUROSCI.0730-11.2011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 124.Sotiropoulos I., Silva J., Kimura T., Rodrigues A.J., Costa P., Almeida O.F.X., Sousa N., Takashima A. Female hippocampus vulnerability to environmental stress, a precipitating factor in Tau aggregation pathology. J. Alzheimers Dis. 2014;43(3):763–774. doi: 10.3233/JAD-140693. [DOI] [PubMed] [Google Scholar]
- 125.Lopes S., Vaz-Silva J., Pinto V., Dalla C., Kokras N., Bedenk B., Mack N., Czisch M., Almeida O.F.X., Sousa N., Sotiropoulos I. Tau protein is essential for stress-induced brain pathology. Proc. Natl. Acad. Sci. 2016;113(26):E3755–E3763. doi: 10.1073/pnas.1600953113. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 126.Silva J.M., Rodrigues S., Sampaio-Marques B., Gomes P., Neves-Carvalho A., Dioli C., Soares-Cunha C., Mazuik B.F., Takashima A., Ludovico P., Wolozin B., Sousa N., Sotiropoulos I. Dysregulation of autophagy and stress granule-related proteins in stress-driven Tau pathology. Cell Death Differ. 2019;26(8):1411–1427. doi: 10.1038/s41418-018-0217-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 127.Vaz-Silva J., Gomes P., Jin Q., Zhu M., Zhuravleva V., Quintremil S., Meira T., Silva J., Dioli C., Soares-Cunha C., Daskalakis N.P., Sousa N., Sotiropoulos I., Waites C.L. Endolysosomal degradation of Tau and its role in glucocorticoid‐driven hippocampal malfunction. EMBO J. 2018;37(20):e99084. doi: 10.15252/embj.201899084. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 128.Pinheiro S., Silva J., Mota C., Vaz-Silva J., Veloso A., Pinto V., Sousa N., Cerqueira J., Sotiropoulos I. Tau mislocation in glucocorticoid-triggered hippocampal pathology. Mol. Neurobiol. 2016;53(7):4745–4753. doi: 10.1007/s12035-015-9356-2. [DOI] [PubMed] [Google Scholar]
- 129.Sotiropoulos I., Silva J.M., Gomes P., Sousa N., Almeida O.F.X. Stress and the etiopathogenesis of alzheimer’s disease and depression. Adv. Exp. Med. Biol. 2019;1184:241–257. doi: 10.1007/978-981-32-9358-8_20. [DOI] [PubMed] [Google Scholar]
- 130.Rissman R.A., Lee K.F., Vale W., Sawchenko P.E. Corticotropin-releasing factor receptors differentially regulate stress-induced tau phosphorylation. J. Neurosci. 2007;27(24):6552–6562. doi: 10.1523/JNEUROSCI.5173-06.2007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 131.Rissman R.A., Staup M.A., Lee A.R., Justice N.J., Rice K.C., Vale W., Sawchenko P.E. Corticotropin-releasing factor receptor-dependent effects of repeated stress on tau phosphorylation, solubility, and aggregation. Proc. Natl. Acad. Sci. USA. 2012;109(16):6277–6282. doi: 10.1073/pnas.1203140109. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 132.Gandy S., Duff K. Post-menopausal estrogen deprivation and Alzheimer’s disease. Exp. Gerontol. 2000;35(4):503–511. doi: 10.1016/S0531-5565(00)00116-9. [DOI] [PubMed] [Google Scholar]
- 133.Carroll J.C., Rosario E.R., Kreimer S., Villamagna A., Gentzschein E., Stanczyk F.Z., Pike C.J. Sex differences in β-amyloid accumulation in 3xTg-AD mice: Role of neonatal sex steroid hormone exposure. Brain Res. 2010;1366:233–245. doi: 10.1016/j.brainres.2010.10.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 134.Monteiro-Fernandes D., Sousa N., Almeida O.F.X., Sotiropoulos I. Sex hormone depletion augments glucocorticoid induction of tau hyperphosphorylation in male rat brain. Neuroscience. 2021;454:140–150. doi: 10.1016/j.neuroscience.2020.05.049. [DOI] [PubMed] [Google Scholar]
- 135.Panizzon M.S., Hauger R.L., Xian H., Jacobson K., Lyons M.J., Franz C.E., Kremen W.S. Interactive effects of testosterone and cortisol on hippocampal volume and episodic memory in middle-aged men. Psychoneuroendocrinology. 2018;91:115–122. doi: 10.1016/j.psyneuen.2018.03.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 136.Fiacco S., Walther A., Ehlert U. Steroid secretion in healthy aging. Psychoneuroendocrinology. 2019;105:64–78. doi: 10.1016/j.psyneuen.2018.09.035. [DOI] [PubMed] [Google Scholar]
- 137.Italia M., Forastieri C., Longaretti A., Battaglioli E., Rusconi F. Rationale, relevance, and limits of stress-induced psychopathology in rodents as models for psychiatry research: An introductory overview. Int. J. Mol. Sci. 2020;21(20):7455. doi: 10.3390/ijms21207455. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 138.Kokras N., Dalla C. Sex differences in animal models of psychiatric disorders. Br. J. Pharmacol. 2014;171(20):4595–4619. doi: 10.1111/bph.12710. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 139.Hodes G.E. A primer on sex differences in the behavioral response to stress. Curr. Opin. Behav. Sci. 2018;23:75–83. doi: 10.1016/j.cobeha.2018.03.012. [DOI] [Google Scholar]
- 140.Hodes G.E., Pfau M.L., Purushothaman I., Ahn H.F., Golden S.A., Christoffel D.J., Magida J., Brancato A., Takahashi A., Flanigan M.E., Ménard C., Aleyasin H., Koo J.W., Lorsch Z.S., Feng J., Heshmati M., Wang M., Turecki G., Neve R., Zhang B., Shen L., Nestler E.J., Russo S.J. Sex differences in nucleus accumbens transcriptome profiles associated with susceptibility versus resilience to subchronic variable stress. J. Neurosci. 2015;35(50):16362–16376. doi: 10.1523/JNEUROSCI.1392-15.2015. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 141.van der Zee Y.Y., Lardner C.K., Parise E.M., Mews P., Ramakrishnan A., Patel V., Teague C.D., Salery M., Walker D.M., Browne C.J., Labonté B., Parise L.F., Kronman H., Penã C.J., Torres-Berrío A., Duffy J.E., de Nijs L., Eijssen L.M.T., Shen L., Rutten B., Issler O., Nestler E.J. Sex-specific role for SLIT1 in regulating stress susceptibility. Biol. Psychiatry. 2022;91(1):81–91. doi: 10.1016/j.biopsych.2021.01.019. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 142.Lorsch Z.S., Loh Y.H.E., Purushothaman I., Walker D.M., Parise E.M., Salery M., Cahill M.E., Hodes G.E., Pfau M.L., Kronman H., Hamilton P.J., Issler O., Labonté B., Symonds A.E., Zucker M., Zhang T.Y., Meaney M.J., Russo S.J., Shen L., Bagot R.C., Nestler E.J. Estrogen receptor α drives pro-resilient transcription in mouse models of depression. Nat. Commun. 2018;9(1):1116. doi: 10.1038/s41467-018-03567-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 143.Issler O., van der Zee Y.Y., Ramakrishnan A., Xia S., Zinsmaier A.K., Tan C., Li W., Browne C.J., Walker D.M., Salery M., Torres-Berrío A., Futamura R., Duffy J.E., Labonte B., Girgenti M.J., Tamminga C.A., Dupree J.L., Dong Y., Murrough J.W., Shen L., Nestler E.J. The long noncoding RNA FEDORA is a cell type- and sex-specific regulator of depression. Sci. Adv. 2022;8(48):eabn9494. doi: 10.1126/sciadv.abn9494. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 144.Issler O., van der Zee Y.Y., Ramakrishnan A., Wang J., Tan C., Loh Y.H.E., Purushothaman I., Walker D.M., Lorsch Z.S., Hamilton P.J., Peña C.J., Flaherty E., Hartley B.J., Torres-Berrío A., Parise E.M., Kronman H., Duffy J.E., Estill M.S., Calipari E.S., Labonté B., Neve R.L., Tamminga C.A., Brennand K.J., Dong Y., Shen L., Nestler E.J. Sex-specific role for the long non-coding RNA LINC00473 in depression. Neuron. 2020;106(6):912–926.e5. doi: 10.1016/j.neuron.2020.03.023. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 145.Labonté B., Engmann O., Purushothaman I., Menard C., Wang J., Tan C., Scarpa J.R., Moy G., Loh Y.H.E., Cahill M., Lorsch Z.S., Hamilton P.J., Calipari E.S., Hodes G.E., Issler O., Kronman H., Pfau M., Obradovic A.L.J., Dong Y., Neve R.L., Russo S., Kasarskis A., Tamminga C., Mechawar N., Turecki G., Zhang B., Shen L., Nestler E.J. Sex-specific transcriptional signatures in human depression. Nat. Med. 2017;23(9):1102–1111. doi: 10.1038/nm.4386. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 146.Seney M.L., Chang L.C., Oh H., Wang X., Tseng G.C., Lewis D.A., Sibille E. The role of genetic sex in affect regulation and expression of gaba-related genes across species. Front. Psychiatry. 2013;4:104. doi: 10.3389/fpsyt.2013.00104. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 147.Seney M.L., Huo Z., Cahill K., French L., Puralewski R., Zhang J., Logan R.W., Tseng G., Lewis D.A., Sibille E. Opposite molecular signatures of depression in men and women. Biol. Psychiatry. 2018;84(1):18–27. doi: 10.1016/j.biopsych.2018.01.017. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 148.Bestor T.H. The DNA methyltransferases of mammals. Hum. Mol. Genet. 2000;9(16):2395–2402. doi: 10.1093/hmg/9.16.2395. [DOI] [PubMed] [Google Scholar]
- 149.Feng J., Fan G. The role of DNA methylation in the central nervous system and neuropsychiatric disorders. Int. Rev. Neurobiol. 2009;89:67–84. doi: 10.1016/S0074-7742(09)89004-1. [DOI] [PubMed] [Google Scholar]
- 150.Nugent B.M., Wright C.L., Shetty A.C., Hodes G.E., Lenz K.M., Mahurkar A., Russo S.J., Devine S.E., McCarthy M.M. Brain feminization requires active repression of masculinization via DNA methylation. Nat. Neurosci. 2015;18(5):690–697. doi: 10.1038/nn.3988. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 151.LaPlant Q., Vialou V., Covington H.E., III, Dumitriu D., Feng J., Warren B.L., Maze I., Dietz D.M., Watts E.L., Iñiguez S.D., Koo J.W., Mouzon E., Renthal W., Hollis F., Wang H., Noonan M.A., Ren Y., Eisch A.J., Bolaños C.A., Kabbaj M., Xiao G., Neve R.L., Hurd Y.L., Oosting R.S., Fan G., Morrison J.H., Nestler E.J. Dnmt3a regulates emotional behavior and spine plasticity in the nucleus accumbens. Nat. Neurosci. 2010;13(9):1137–1143. doi: 10.1038/nn.2619. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 152.Christoffel D.J. Golden, S.A.; Dumitriu, D.; Robison, A.J.; Janssen, W.G.; Ahn, H.F.; Krishnan, V.; Reyes, C.M.; Han, M.H.; Ables, J.L.; Eisch, A.J.; Dietz, D.M.; Ferguson, D.; Neve, R.L.; Greengard, P.; Kim, Y.; Morrison, J.H.; Russo, S.J. IκB kinase regulates social defeat stress-induced synaptic and behavioral plasticity. J. Neurosci. 2011;31(1):314–321. doi: 10.1523/JNEUROSCI.4763-10.2011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 153.Wang J., Hodes G.E., Zhang H., Zhang S., Zhao W., Golden S.A., Bi W., Menard C., Kana V., Leboeuf M., Xie M., Bregman D., Pfau M.L., Flanigan M.E., Esteban-Fernández A., Yemul S., Sharma A., Ho L., Dixon R., Merad M., Han M.H., Russo S.J., Pasinetti G.M. Epigenetic modulation of inflammation and synaptic plasticity promotes resilience against stress in mice. Nat. Commun. 2018;9(1):477. doi: 10.1038/s41467-017-02794-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 154.Deonaraine K.K., Wang Q., Cheng H., Chan K.L., Lin H.Y., Liu K., Parise L.F., Cathomas F., Leclair K.B., Flanigan M.E., Li L., Aleyasin H., Guevara C., Hao K., Zhang B., Russo S.J., Wang J. Sex‐specific peripheral and central responses to stres induced depression and treatment in a mouse model. J. Neurosci. Res. 2020;98(12):2541–2553. doi: 10.1002/jnr.24724. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 155.Peña C.J., Bagot R.C., Labonté B., Nestler E.J. Epigenetic signaling in psychiatric disorders. J. Mol. Biol. 2014;426(20):3389–3412. doi: 10.1016/j.jmb.2014.03.016. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 156.Jenuwein T., Allis C.D. Translating the histone code. Science. 2001;293(5532):1074–1080. doi: 10.1126/science.1063127. [DOI] [PubMed] [Google Scholar]
- 157.Sun H., Kennedy P.J., Nestler E.J. Epigenetics of the depressed brain: Role of histone acetylation and methylation. Neuropsychopharmacology. 2013;38(1):124–137. doi: 10.1038/npp.2012.73. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 158.Fischle W., Wang Y., Allis D.C. Binary switches and modification cassettes in histone biology and beyond. Nature. 2003;425(6957):475–479. doi: 10.1038/nature02017. [DOI] [PubMed] [Google Scholar]
- 159.Vialou V., Feng J., Robison A.J., Nestler E.J. Epigenetic mechanisms of depression and antidepressant action. Annu. Rev. Pharmacol. Toxicol. 2013;53(1):59–87. doi: 10.1146/annurev-pharmtox-010611-134540. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 160.Iizuka M., Smith M.M. Functional consequences of histone modifications. Curr. Opin. Genet. Dev. 2003;13(2):154–160. doi: 10.1016/S0959-437X(03)00020-0. [DOI] [PubMed] [Google Scholar]
- 161.Murray E.K., Hien A., de Vries G.J., Forger N.G. Epigenetic control of sexual differentiation of the bed nucleus of the stria terminalis. Endocrinology. 2009;150(9):4241–4247. doi: 10.1210/en.2009-0458. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 162.Bangasser D.A., Shors T.J. The bed nucleus of the stria terminalis modulates learning after stress in masculinized but not cycling females. J. Neurosci. 2008;28(25):6383–6387. doi: 10.1523/JNEUROSCI.0831-08.2008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 163.Bangasser D.A., Santollo J., Shors T.J. The bed nucleus of the stria terminalis is critically involved in enhancing associative learning after stressful experience. Behav. Neurosci. 2005;119(6):1459–1466. doi: 10.1037/0735-7044.119.6.1459. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 164.Tsankova N.M., Berton O., Renthal W., Kumar A., Neve R.L., Nestler E.J. Sustained hippocampal chromatin regulation in a mouse model of depression and antidepressant action. Nat. Neurosci. 2006;9(4):519–525. doi: 10.1038/nn1659. [DOI] [PubMed] [Google Scholar]
- 165.Sase A.S., Lombroso S.I., Santhumayor B.A., Wood R.R., Lim C.J., Neve R.L., Heller E.A. Sex-specific regulation of fear memory by targeted epigenetic editing of Cdk5. Biol. Psychiatry. 2019;85(8):623–634. doi: 10.1016/j.biopsych.2018.11.022. [DOI] [PubMed] [Google Scholar]
- 166.O’Carroll D., Schaefer A. General principals of miRNA biogenesis and regulation in the brain. Neuropsychopharmacology. 2013;38(1):39–54. doi: 10.1038/npp.2012.87. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 167.Pfau M.L., Purushothaman I., Feng J., Golden S.A., Aleyasin H., Lorsch Z.S., Cates H.M., Flanigan M.E., Menard C., Heshmati M., Wang Z., Ma’ayan A., Shen L., Hodes G.E., Russo S.J. Integrative analysis of sex-specific microRNA networks following stress in mouse nucleus accumbens. Front. Mol. Neurosci. 2016;9:144. doi: 10.3389/fnmol.2016.00144. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 168.Pfau M.L., Menard C., Cathomas F., Desland F., Kana V., Chan K.L., Shimo Y., LeClair K., Flanigan M.E., Aleyasin H., Walker D.M., Bouchard S., Mack M., Hodes G.E., Merad M.M., Russo S.J. Role of monocyte-derived microRNA106b∼25 in resilience to social stress. Biol. Psychiatry. 2019;86(6):474–482. doi: 10.1016/j.biopsych.2019.02.023. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 169.van der Zee Y.Y., Eijssen L.M.T., Mews P., Ramakrishnan A., Alvarez K., Lardner C.K., Cates H.M., Walker D.M., Torres-Berrío A., Browne C.J., Cunningham A., Cathomas F., Kronman H., Parise E.M., de Nijs L., Shen L., Murrough J.W., Rutten B.P.F., Nestler E.J., Issler O. Blood miR-144-3p: A novel diagnostic and therapeutic tool for depression. Mol. Psychiatry. 2022;27(11):4536–4549. doi: 10.1038/s41380-022-01712-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 170.Rodgers A.B., Morgan C.P., Leu N.A., Bale T.L. Transgenerational epigenetic programming via sperm microRNA recapitulates effects of paternal stress. Proc. Natl. Acad. Sci. . 2015;112(44):13699–13704. doi: 10.1073/pnas.1508347112. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 171.Dietz D.M., LaPlant Q., Watts E.L., Hodes G.E., Russo S.J., Feng J., Oosting R.S., Vialou V., Nestler E.J. Paternal transmission of stress-induced pathologies. Biol. Psychiatry. 2011;70(5):408–414. doi: 10.1016/j.biopsych.2011.05.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 172.Cunningham A.M., Walker D.M., Ramakrishnan A., Doyle M.A., Bagot R.C., Cates H.M., Peña C.J., Issler O., Lardner C.K., Browne C., Russo S.J., Shen L., Nestler E.J. Sperm transcriptional state associated with paternal transmission of stress phenotypes. J. Neurosci. 2021;41(29):6202–6216. doi: 10.1523/JNEUROSCI.3192-20.2021. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 173.Bianchi I., Lleo A., Gershwin M.E., Invernizzi P. The X chromosome and immune associated genes. J. Autoimmun. 2012;38(2-3):J187–J192. doi: 10.1016/j.jaut.2011.11.012. [DOI] [PubMed] [Google Scholar]
- 174.Youness A., Miquel C.H., Guéry J.C. Escape from X chromosome inactivation and the female predominance in autoimmune diseases. Int. J. Mol. Sci. 2021;22(3):1114. doi: 10.3390/ijms22031114. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 175.Klein S.L., Flanagan K.L. Sex differences in immune responses. Nat. Rev. Immunol. 2016;16(10):626–638. doi: 10.1038/nri.2016.90. [DOI] [PubMed] [Google Scholar]
- 176.Maes M., Kubera M., Leunis J.C. The gut-brain barrier in major depression: intestinal mucosal dysfunction with an increased translocation of LPS from gram negative enterobacteria (leaky gut) plays a role in the inflammatory pathophysiology of depression. Neuroendocrinol. Lett. 2008;29(1):117–124. [PubMed] [Google Scholar]
- 177.Kelly J.R., Borre Y., O’ Brien C., Patterson E., El Aidy S., Deane J., Kennedy P.J., Beers S., Scott K., Moloney G., Hoban A.E., Scott L., Fitzgerald P., Ross P., Stanton C., Clarke G., Cryan J.F., Dinan T.G. Transferring the blues: Depression-associated gut microbiota induces neurobehavioural changes in the rat. J. Psychiatr. Res. 2016;82:109–118. doi: 10.1016/j.jpsychires.2016.07.019. [DOI] [PubMed] [Google Scholar]
- 178.Lyte M. Microbial endocrinology and the microbiota-gut-brain axis. Adv. Exp. Med. Biol. 2014;817:3–24. doi: 10.1007/978-1-4939-0897-4_1. [DOI] [PubMed] [Google Scholar]
- 179.Cruz-Pereira J.S., Rea K., Nolan Y.M., O’Leary O.F., Dinan T.G., Cryan J.F. Depression’s unholy trinity: Dysregulated stress, immunity, and the microbiome. Annu. Rev. Psychol. 2020;71(1):49–78. doi: 10.1146/annurev-psych-122216-011613. [DOI] [PubMed] [Google Scholar]
- 180.Foster J.A., Rinaman L., Cryan J.F. Stress & the gut-brain axis: Regulation by the microbiome. Neurobiol. Stress. 2017;7:124–136. doi: 10.1016/j.ynstr.2017.03.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 181.Kim Y.S., Unno T., Kim B.Y., Park M.S. Sex differences in gut microbiota. World J. Mens Health. 2020;38(1):48–60. doi: 10.5534/wjmh.190009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 182.Markle J.G.M., Frank D.N., Mortin-Toth S., Robertson C.E., Feazel L.M., Rolle-Kampczyk U., von Bergen M., McCoy K.D., Macpherson A.J., Danska J.S. Sex differences in the gut microbiome drive hormone-dependent regulation of autoimmunity. Science. 2013;339(6123):1084–1088. doi: 10.1126/science.1233521. [DOI] [PubMed] [Google Scholar]
- 183.Dalla C., Pavlidi P., Sakelliadou D.G., Grammatikopoulou T., Kokras N. Sex differences in blood-brain barrier transport of psychotropic drugs. Front. Behav. Neurosci. 2022;16:844916. doi: 10.3389/fnbeh.2022.844916. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 184.Kumar M., Rainville J.R., Williams K., Lile J.A., Hodes G.E., Vassoler F.M., Turner J.R. Sexually dimorphic neuroimmune response to chronic opioid treatment and withdrawal. Neuropharmacology. 2021;186:108469. doi: 10.1016/j.neuropharm.2021.108469. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 185.Menard C., Pfau M.L., Hodes G.E., Kana V., Wang V.X., Bouchard S., Takahashi A., Flanigan M.E., Aleyasin H., LeClair K.B., Janssen W.G., Labonté B., Parise E.M., Lorsch Z.S., Golden S.A., Heshmati M., Tamminga C., Turecki G., Campbell M., Fayad Z.A., Tang C.Y., Merad M., Russo S.J. Social stress induces neurovascular pathology promoting depression. Nat. Neurosci. 2017;20(12):1752–1760. doi: 10.1038/s41593-017-0010-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 186.Dion-Albert L., Bandeira Binder L., Daigle B., Hong-Minh A., Lebel M., Menard C. Sex differences in the blood-brain barrier: Implications for mental health. Front. Neuroendocrinol. 2022;65:100989. doi: 10.1016/j.yfrne.2022.100989. [DOI] [PubMed] [Google Scholar]
- 187.Dion-Albert L., Cadoret A., Doney E., Kaufmann F.N., Dudek K.A., Daigle B., Parise L.F., Cathomas F., Samba N., Hudson N., Lebel M., Aardema F., Ait Bentaleb L., Beauchamp J., Bendahmane H., Benoit E., Bergeron L., Bertone A., Bertrand N., Berube F-A., Blanchet P., Boissonneault J., Bolduc C.J., Bonin J-P., Borgeat F., Boyer R., Breault C., Breton J-J., Briand C., Brodeur J., Brule K., Brunet L., Carriere S., Chartrand C., Chenard-Soucy R., Chevrette T., Cloutier E., Cloutier R., Cormier H., Cote G., Cyr J., David P., De Benedictis L., Delisle M-C., Deschenes P., Desjardins C.D., Desmarais G., Dubreucq J-L., Dumont M., Dumais A., Ethier G., Feltrin C., Felx A., Findlay H., Fortier L., Fortin D., Fortin L., Francois N., Gagne V., Gagnon M-P., Gignac-Hens M-C., Giguere C-E., Godbout R., Grou C., Guay S., Guillem F., Hachimi-Idrissi N., Herry C., Hodgins S., Homayoun S., Jemel B., Joyal C., Kouassi E., Labelle R., Lafortune D., Lahaie M., Lahlafi S., Lalonde P., Landry P., Lapaige V., Larocque G., Larue C., Lavoie M., Leclerc J-J., Lecomte T., Lecours C., Leduc L., Lelan M-F., Lemieux A., Lesage A., Letarte A., Lepage J., Levesque A., Lipp O., Luck D., Lupien S., Lusignan F-A., Lusignan R., Luyet A.J., Lynhiavu A., Melun J-P., Morin C., Nicole L., Noel F., Normandeau L., O’Connor K., Ouellette C., Parent V., Parizeau M-H., Pelletier J-F., Pelletier J., Pelletier M., Plusquellec P., Poirier D., Potvin S., Prevost G., Prevost M-J., Racicot P., Racine-Gagne M-F., Renaud P., Ricard N., Rivet S., Rolland M., Sasseville M., Safadi G., Smith S., Smolla N., Stip E., Teitelbaum J., Thibault A., Thibault L., Thibault S., Thomas F., Todorov C., Tourjman V., Tranulis C., Trudeau S., Trudel G., Vacri N., Valiquette L., Vanier C., Villeneuve K., Villeneuve M., Vincent P., Wolfe M., Xiong L., Zizzi A., Campbell M., Turecki G., Mechawar N., Menard C. Vascular and blood-brain barrier-related changes underlie stress responses and resilience in female mice and depression in human tissue. Nat. Commun. 2022;13(1):164. doi: 10.1038/s41467-021-27604-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 188.Bollinger J.L., Salinas I., Fender E., Sengelaub D.R., Wellman C.L. Gonadal hormones differentially regulate sex‐specific stress effects on glia in the medial prefrontal cortex. J. Neuroendocrinol. 2019;31(8):e12762. doi: 10.1111/jne.12762. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 189.Van Camp N., Lavisse S., Roost P., Gubinelli F., Hillmer A., Boutin H. TSPO imaging in animal models of brain diseases. Eur. J. Nucl. Med. Mol. Imaging. 2021;49(1):77–109. doi: 10.1007/s00259-021-05379-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 190.Tsyglakova M., Huskey A.M., Hurst E.H., Telep N.M., Wilding M.C., Babington M.E., Rainville J.R., Hodes G.E. Sex and region-specific effects of variable stress on microglia morphology. Brain, Behav. Immun. Health. 2021;18:100378. doi: 10.1016/j.bbih.2021.100378. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 191.Keselman A., Heller N. Estrogen signaling modulates allergic inflammation and contributes to sex differences in asthma. Front. Immunol. 2015;6:568. doi: 10.3389/fimmu.2015.00568. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 192.Molero L., García-Durán M., Diaz-Recasens J., Rico L., Casado S., López-Farré A. Expression of estrogen receptor subtypes and neuronal nitric oxide synthase in neutrophils from women and men Regulation by estrogen. Cardiovasc. Res. 2002;56(1):43–51. doi: 10.1016/S0008-6363(02)00505-9. [DOI] [PubMed] [Google Scholar]
- 193.Zierau O., Zenclussen A.C., Jensen F. Role of female sex hormones, estradiol and progesterone, in mast cell behavior. Front. Immunol. 2012;3:169. doi: 10.3389/fimmu.2012.00169. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 194.Rainville J.R., Tsyglakova M., Hodes G.E. Deciphering sex differences in the immune system and depression. Front. Neuroendocrinol. 2018;50:67–90. doi: 10.1016/j.yfrne.2017.12.004. [DOI] [PubMed] [Google Scholar]
- 195.Finnell J.E., Muniz B.L., Padi A.R., Lombard C.M., Moffitt C.M., Wood C.S., Wilson L.B., Reagan L.P., Wilson M.A., Wood S.K. Essential role of ovarian hormones in susceptibility to the consequences of witnessing social defeat in female rats. Biol. Psychiatry. 2018;84(5):372–382. doi: 10.1016/j.biopsych.2018.01.013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 196.Furman D., Hejblum B.P., Simon N., Jojic V., Dekker C.L., Thiébaut R., Tibshirani R.J., Davis M.M. Systems analysis of sex differences reveals an immunosuppressive role for testosterone in the response to influenza vaccination. Proc. Natl. Acad. Sci. 2014;111(2):869–874. doi: 10.1073/pnas.1321060111. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 197.McMurray R.W., Suwannaroj S., Ndebele K., Jenkins J.K. Differential effects of sex steroids on T and B cells: modulation of cell cycle phase distribution, apoptosis and bcl-2 protein levels. Pathobiology. 2001;69(1):44–58. doi: 10.1159/000048757. [DOI] [PubMed] [Google Scholar]
- 198.Trigunaite A., Dimo J., Jørgensen T.N. Suppressive effects of androgens on the immune system. Cell. Immunol. 2015;294(2):87–94. doi: 10.1016/j.cellimm.2015.02.004. [DOI] [PubMed] [Google Scholar]
- 199.Gaillard R.C., Spinedi E. Sex- and stress-steroids interactions and the immune system: Evidence for a neuroendocrine-immunological sexual dimorphism. Domest. Anim. Endocrinol. 1998;15(5):345–352. doi: 10.1016/S0739-7240(98)00028-9. [DOI] [PubMed] [Google Scholar]
- 200.Dantzer R., Kelley K.W. Stress and immunity: An integrated view of relationships between the brain and the immune system. Life Sci. 1989;44(26):1995–2008. doi: 10.1016/0024-3205(89)90345-7. [DOI] [PubMed] [Google Scholar]
- 201.Engler H., Benson S., Wegner A., Spreitzer I., Schedlowski M., Elsenbruch S. Men and women differ in inflammatory and neuroendocrine responses to endotoxin but not in the severity of sickness symptoms. Brain Behav. Immun. 2016;52:18–26. doi: 10.1016/j.bbi.2015.08.013. [DOI] [PubMed] [Google Scholar]
- 202.Harden K.P., Wrzus C., Luong G., Grotzinger A., Bajbouj M., Rauers A., Wagner G.G., Riediger M. Diurnal coupling between testosterone and cortisol from adolescence to older adulthood. Psychoneuroendocrinology. 2016;73:79–90. doi: 10.1016/j.psyneuen.2016.07.216. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 203.Andrews G., Bell C., Boyce P., Gale C., Lampe L., Marwat O., Rapee R., Wilkins G. Royal australian and new zealand college of psychiatrists clinical practice guidelines for the treatment of panic disorder, social anxiety disorder and generalised anxiety disorder. Aust. N. Z. J. Psychiatry. 2018;52(12):1109–1172. doi: 10.1177/0004867418799453. [DOI] [Google Scholar]
- 204.Zohar J., Stahl S., Moller H.J., Blier P., Kupfer D., Yamawaki S., Uchida H., Spedding M., Goodwin G.M., Nutt D. A review of the current nomenclature for psychotropic agents and an introduction to the Neuroscience-based Nomenclature. Eur. Neuropsychopharmacol. 2015;25(12):2318–2325. doi: 10.1016/j.euroneuro.2015.08.019. [DOI] [PubMed] [Google Scholar]
- 205.Khan A., Brodhead A.E., Schwartz K.A., Kolts R.L., Brown W.A. Sex differences in antidepressant response in recent antidepressant clinical trials. J. Clin. Psychopharmacol. 2005;25(4):318–324. doi: 10.1097/01.jcp.0000168879.03169.ce. [DOI] [PubMed] [Google Scholar]
- 206.Kokras N., Dalla C., Papadopoulou-Daifoti Z. Sex differences in pharmacokinetics of antidepressants. Expert Opin. Drug Metab. Toxicol. 2011;7(2):213–226. doi: 10.1517/17425255.2011.544250. [DOI] [PubMed] [Google Scholar]
- 207.Sramek J.J., Murphy M.F., Cutler N.R. Sex differences in the psychopharmacological treatment of depression. Dialogues Clin. Neurosci. 2016;18(4):447–457. doi: 10.31887/DCNS.2016.18.4/ncutler. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 208.Hutson W.R., Roehrkasse R.L., Wald A. Influence of gender and menopause on gastric emptying and motility. Gastroenterology. 1989;96(1):11–17. doi: 10.1016/0016-5085(89)90758-0. [DOI] [PubMed] [Google Scholar]
- 209.Marazziti D., Baroni S., Picchetti M., Piccinni A., Carlini M., Vatteroni E., Falaschi V., Lombardi A., Dell’Osso L. Pharmacokinetics and pharmacodinamics of psychotropic drugs: Effect of sex. CNS Spectr. 2013;18(3):118–127. doi: 10.1017/S1092852912001010. [DOI] [PubMed] [Google Scholar]
- 210.Nicolas J.M., Espie P., Molimard M. Gender and interindividual variability in pharmacokinetics. Drug Metab. Rev. 2009;41(3):408–421. doi: 10.1080/10837450902891485. [DOI] [PubMed] [Google Scholar]
- 211.Kristensen C.B. Imipramine serum protein binding in healthy subjects. Clin. Pharmacol. Ther. 1983;34(5):689–694. doi: 10.1038/clpt.1983.233. [DOI] [PubMed] [Google Scholar]
- 212.Anderson G.D. Sex and racial differences in pharmacological response: where is the evidence? Pharmacogenetics, pharmacokinetics, and pharmacodynamics. J. Womens Health. 2005;14(1):19–29. doi: 10.1089/jwh.2005.14.19. [DOI] [PubMed] [Google Scholar]
- 213.Schwartz J.B. The current state of knowledge on age, sex, and their interactions on clinical pharmacology. Clin. Pharmacol. Ther. 2007;82(1):87–96. doi: 10.1038/sj.clpt.6100226. [DOI] [PubMed] [Google Scholar]
- 214.Farkas R.H., Unger E.F., Temple R. Zolpidem and driving impairment-identifying persons at risk. N. Engl. J. Med. 2013;369(8):689–691. doi: 10.1056/NEJMp1307972. [DOI] [PubMed] [Google Scholar]
- 215.Bigos K.L., Pollock B.G., Stankevich B.A., Bies R.R. Sex differences in the pharmacokinetics and pharmacodynamics of antidepressants: An updated review. Gend. Med. 2009;6(4):522–543. doi: 10.1016/j.genm.2009.12.004. [DOI] [PubMed] [Google Scholar]
- 216.Berlanga C., Flores-Ramos M. Different gender response to serotonergic and noradrenergic antidepressants. A comparative study of the efficacy of citalopram and reboxetine. J. Affect. Disord. 2006;95(1-3):119–123. doi: 10.1016/j.jad.2006.04.029. [DOI] [PubMed] [Google Scholar]
- 217.Kornstein S.G., Schatzberg A.F., Thase M.E., Yonkers K.A., McCullough J.P., Keitner G.I., Gelenberg A.J., Davis S.M., Harrison W.M., Keller M.B. Gender differences in treatment response to sertraline versus imipramine in chronic depression. Am. J. Psychiatry. 2000;157(9):1445–1452. doi: 10.1176/appi.ajp.157.9.1445. [DOI] [PubMed] [Google Scholar]
- 218.Young E.A., Kornstein S.G., Marcus S.M., Harvey A.T., Warden D., Wisniewski S.R., Balasubramani G.K., Fava M., Trivedi M.H., John Rush A. Sex differences in response to citalopram: A STAR∗D report. J. Psychiatr. Res. 2009;43(5):503–511. doi: 10.1016/j.jpsychires.2008.07.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 219.Parker G., Parker K., Austin M.P., Mitchell P., Brotchie H. Gender differences in response to differing antidepressant drug classes: Two negative studies. Psychol. Med. 2003;33(8):1473–1477. doi: 10.1017/S0033291703007918. [DOI] [PubMed] [Google Scholar]
- 220.Kornstein S.G., Pedersen R.D., Holland P.J., Nemeroff C.B., Rothschild A.J., Thase M.E., Trivedi M.H., Ninan P.T., Keller M.B. Influence of sex and menopausal status on response, remission, and recurrence in patients with recurrent major depressive disorder treated with venlafaxine extended release or fluoxetine: Analysis of data from the PREVENT study. J. Clin. Psychiatry. 2014;75(1):62–68. doi: 10.4088/JCP.12m07841. [DOI] [PubMed] [Google Scholar]
- 221.Thase M.E., Entsuah R., Cantillon M., Kornstein S.G. Relative antidepressant efficacy of venlafaxine and SSRIs: sex-age interactions. J. Womens Health. 2005;14(7):609–616. doi: 10.1089/jwh.2005.14.609. [DOI] [PubMed] [Google Scholar]
- 222.Naito S., Sato K., Yoshida K., Higuchi H., Takahashi H., Kamata M., Ito K., Ohkubo T., Shimizu T. Gender differences in the clinical effects of fluvoxamine and milnacipran in Japanese major depressive patients. Psychiatry Clin. Neurosci. 2007;61(4):421–427. doi: 10.1111/j.1440-1819.2007.01679.x. [DOI] [PubMed] [Google Scholar]
- 223.Williams A.V., Trainor B.C. The impact of sex as a biological variable in the search for novel antidepressants. Front. Neuroendocrinol. 2018;50:107–117. doi: 10.1016/j.yfrne.2018.05.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 224.Keating C., Tilbrook A., Kulkarni J. Oestrogen: an overlooked mediator in the neuropsychopharmacology of treatment response? Int. J. Neuropsychopharmacol. 2011;14(4):553–566. doi: 10.1017/S1461145710000982. [DOI] [PubMed] [Google Scholar]
- 225.Schneider L.S., Small G.W., Clary C.M. Estrogen replacement therapy and antidepressant response to sertraline in older depressed women. Am. J. Geriatr. Psychiatry. 2001;9(4):393–399. doi: 10.1097/00019442-200111000-00007. [DOI] [PubMed] [Google Scholar]
- 226.Schneider L.S., Small G.W., Hamilton S.H., Bystritsky A., Nemeroff C.B., Meyers B.S. Estrogen replacement and response to fluoxetine in a multicenter geriatric depression trial. Am. J. Geriatr. Psychiatry. 1997;5(2):97–106. doi: 10.1097/00019442-199721520-00002. [DOI] [PubMed] [Google Scholar]
- 227.Stahl S.M. Basic psychopharmacology of antidepressants, part 2: Estrogen as an adjunct to antidepressant treatment. J. Clin. Psychiatry. 1998;59(S4):15–24. [PubMed] [Google Scholar]
- 228.Richardson T.A., Robinson R.D. Menopause and depression: A review of psychologic function and sex steroid neurobiology during the menopause. Prim. Care Update Ob Gyns. 2000;7(6):215–223. doi: 10.1016/S1068-607X(00)00049-4. [DOI] [PubMed] [Google Scholar]
- 229.Shapira B., Oppenheim G., Zohar J., Segal M., Malach D., Belmaker R.H. Lack of efficacy of estrogen supplementation to imipramine in resistant female depressives. Biol. Psychiatry. 1985;20(5):576–579. doi: 10.1016/0006-3223(85)90031-9. [DOI] [PubMed] [Google Scholar]
- 230.Amsterdam J., Garcia-España F., Fawcett J., Quitkin F., Reimherr F., Rosenbaum J., Beasley C. Fluoxetine efficacy in menopausal women with and without estrogen replacement. J. Affect. Disord. 1999;55(1):11–17. doi: 10.1016/S0165-0327(98)00203-1. [DOI] [PubMed] [Google Scholar]
- 231.Frokjaer V.G., Pinborg A., Holst K.K., Overgaard A., Henningsson S., Heede M., Larsen E.C., Jensen P.S., Agn M., Nielsen A.P., Stenbæk D.S., da Cunha-Bang S., Lehel S., Siebner H.R., Mikkelsen J.D., Svarer C., Knudsen G.M. Role of serotonin transporter changes in depressive responses to sex-steroid hormone manipulation: A positron emission tomography study. Biol. Psychiatry. 2015;78(8):534–543. doi: 10.1016/j.biopsych.2015.04.015. [DOI] [PubMed] [Google Scholar]
- 232.Kokras N., Dalla C. Preclinical sex differences in depression and antidepressant response: Implications for clinical research. J. Neurosci. Res. 2017;95(1-2):731–736. doi: 10.1002/jnr.23861. [DOI] [PubMed] [Google Scholar]
- 233.Eid R.S., Gobinath A.R., Galea L.A.M. Sex differences in depression: Insights from clinical and preclinical studies. Prog. Neurobiol. 2019;176:86–102. doi: 10.1016/j.pneurobio.2019.01.006. [DOI] [PubMed] [Google Scholar]
- 234.Kokras N., Antoniou K., Mikail H.G., Kafetzopoulos V., Papadopoulou-Daifoti Z., Dalla C. Forced swim test: What about females? Neuropharmacology. 2015;99:408–421. doi: 10.1016/j.neuropharm.2015.03.016. [DOI] [PubMed] [Google Scholar]
- 235.Dalla C., Pitychoutis P.M., Kokras N., Papadopoulou-Daifoti Z. Sex differences in animal models of depression and antidepressant response. Basic Clin. Pharmacol. Toxicol. 2010;106(3):226–233. doi: 10.1111/j.1742-7843.2009.00516.x. [DOI] [PubMed] [Google Scholar]
- 236.Saland S.K., Duclot F., Kabbaj M. Integrative analysis of sex differences in the rapid antidepressant effects of ketamine in preclinical models for individualized clinical outcomes. Curr. Opin. Behav. Sci. 2017;14:19–26. doi: 10.1016/j.cobeha.2016.11.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 237.Fernández-Guasti A., Olivares-Nazario M., Reyes R., Martínez-Mota L. Sex and age differences in the antidepressant-like effect of fluoxetine in the forced swim test. Pharmacol. Biochem. Behav. 2017;152:81–89. doi: 10.1016/j.pbb.2016.01.011. [DOI] [PubMed] [Google Scholar]
- 238.Gómez M.L., Martínez-Mota L., Estrada-Camarena E., Fernández-Guasti A. Influence of the brain sexual differentiation process on despair and antidepressant-like effect of fluoxetine in the rat forced swim test. Neuroscience. 2014;261:11–22. doi: 10.1016/j.neuroscience.2013.12.035. [DOI] [PubMed] [Google Scholar]
- 239.David D.J.P., Nic Dhonnchadha B.Á., Jolliet P., Hascoët M., Bourin M. Are there gender differences in the temperature profile of mice after acute antidepressant administration and exposure to two animal models of depression? Behav. Brain Res. 2001;119(2):203–211. doi: 10.1016/S0166-4328(00)00351-X. [DOI] [PubMed] [Google Scholar]
- 240.Melo A., Kokras N., Dalla C., Ferreira C., Ventura-Silva A.P., Sousa N., Pêgo J.M. The positive effect on ketamine as a priming adjuvant in antidepressant treatment. Transl. Psychiatry. 2015;5(5):e573–e573. doi: 10.1038/tp.2015.66. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 241.Pavlidi P., Megalokonomou A., Sofron A., Kokras N., Dalla C. Pharmacology of ketamine and esketamine as rapid-acting antidepressants. Psychiatriki. 2021;32(S1):55–63. doi: 10.22365/jpsych.2021.050. [DOI] [PubMed] [Google Scholar]
- 242.Carrier N., Kabbaj M. Sex differences in the antidepressant-like effects of ketamine. Neuropharmacology. 2013;70:27–34. doi: 10.1016/j.neuropharm.2012.12.009. [DOI] [PubMed] [Google Scholar]
- 243.Sarkar A., Kabbaj M. Sex differences in effects of ketamine on behavior, spine density, and synaptic proteins in socially isolated rats. Biol. Psychiatry. 2016;80(6):448–456. doi: 10.1016/j.biopsych.2015.12.025. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 244.Scacchi R., Gambina G., Broggio E., Corbo R.M. Sex and ESR1 genotype may influence the response to treatment with donepezil and rivastigmine in patients with Alzheimer’s disease. Int. J. Geriatr. Psychiatry. 2014;29(6):610–615. doi: 10.1002/gps.4043. [DOI] [PubMed] [Google Scholar]
- 245.Mehta N., Rodrigues C., Lamba M., Wu W., Bronskill S.E., Herrmann N., Gill S.S., Chan A.W., Mason R., Day S., Gurwitz J.H., Rochon P.A. Systematic review of sex‐specific reporting of data: Cholinesterase inhibitor example. J. Am. Geriatr. Soc. 2017;65(10):2213–2219. doi: 10.1111/jgs.15020. [DOI] [PubMed] [Google Scholar]
- 246.Marwaha S., Palmer E., Suppes T., Cons E., Young A.H., Upthegrove R. Novel and emerging treatments for major depression. Lancet. 2023;401(10371):141–153. doi: 10.1016/S0140-6736(22)02080-3. [DOI] [PubMed] [Google Scholar]
- 247.Garakani A., Murrough J.W., Freire R.C., Thom R.P., Larkin K., Buono F.D., Iosifescu D.V. Pharmacotherapy of anxiety disorders: Current and emerging treatment options. Front. Psychiatry. 2020;11:595584. doi: 10.3389/fpsyt.2020.595584. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 248.Sartori S.B., Singewald N. Novel pharmacological targets in drug development for the treatment of anxiety and anxiety-related disorders. Pharmacol. Ther. 2019;204:107402. doi: 10.1016/j.pharmthera.2019.107402. [DOI] [PubMed] [Google Scholar]
- 249.Gillies G.E., McArthur S. Estrogen actions in the brain and the basis for differential action in men and women: A case for sex-specific medicines. Pharmacol. Rev. 2010;62(2):155–198. doi: 10.1124/pr.109.002071. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 250.Almeida F.B., Pinna G., Barros H.M.T. The Role of HPA Axis and Allopregnanolone on the Neurobiology of Major Depressive Disorders and PTSD. Int. J. Mol. Sci. 2021;22(11):5495. doi: 10.3390/ijms22115495. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 251.Meltzer-Brody S., Colquhoun H., Riesenberg R., Epperson C.N., Deligiannidis K.M., Rubinow D.R., Li H., Sankoh A.J., Clemson C., Schacterle A., Jonas J., Kanes S. Brexanolone injection in post-partum depression: Two multicentre, double-blind, randomised, placebo-controlled, phase 3 trials. Lancet. 2018;392(10152):1058–1070. doi: 10.1016/S0140-6736(18)31551-4. [DOI] [PubMed] [Google Scholar]
- 252.Arevalo M.A., Azcoitia I., Garcia-Segura L.M. The neuroprotective actions of oestradiol and oestrogen receptors. Nat. Rev. Neurosci. 2015;16(1):17–29. doi: 10.1038/nrn3856. [DOI] [PubMed] [Google Scholar]
- 253.Srivastava D.P., Woolfrey K.M., Penzes P. Insights into rapid modulation of neuroplasticity by brain estrogens. Pharmacol. Rev. 2013;65(4):1318–1350. doi: 10.1124/pr.111.005272. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 254.Pavlidi P., Kokras N., Dalla C. Sex differences in depression and anxiety. Curr. Top. Behav. Neurosci. 2022;62:103–132. doi: 10.1007/7854_2022_375. [DOI] [PubMed] [Google Scholar]
- 255.Handa R.J., Weiser M.J. Gonadal steroid hormones and the hypothalamo-pituitary-adrenal axis. Front. Neuroendocrinol. 2014;35(2):197–220. doi: 10.1016/j.yfrne.2013.11.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 256.Juster R.P., Raymond C., Desrochers A.B., Bourdon O., Durand N., Wan N., Pruessner J.C., Lupien S.J. Sex hormones adjust “sex-specific” reactive and diurnal cortisol profiles. Psychoneuroendocrinology. 2016;63:282–290. doi: 10.1016/j.psyneuen.2015.10.012. [DOI] [PubMed] [Google Scholar]
- 257.Balthazart J., Charlier T.D., Cornil C.A., Dickens M.J., Harada N., Konkle A.T.M., Voigt C., Ball G.F. Sex differences in brain aromatase activity: genomic and non-genomic controls. Front. Endocrinol. 2011;2:34. doi: 10.3389/fendo.2011.00034. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 258.Kokras N., Pastromas N., Porto T.H., Kafetzopoulos V., Mavridis T., Dalla C. Acute but not sustained aromatase inhibition displays antidepressant properties. Int. J. Neuropsychopharmacol. 2014;17(8):1307–1313. doi: 10.1017/S1461145714000212. [DOI] [PubMed] [Google Scholar]
- 259.Chaiton J.A., Wong S.J., Galea L.A.M. 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]
- 260.Dalla C., Antoniou K., Papadopoulou-Daifoti Z., Balthazart J., Bakker J. Oestrogen-deficient female aromatase knockout (ArKO) mice exhibit ‘depressive-like’ symptomatology. Eur. J. Neurosci. 2004;20(1):217–228. doi: 10.1111/j.1460-9568.2004.03443.x. [DOI] [PubMed] [Google Scholar]
- 261.Alexander A., Irving A.J., Harvey J. Emerging roles for the novel estrogen-sensing receptor GPER1 in the CNS. Neuropharmacology. 2017;113((Pt B)):652–660. doi: 10.1016/j.neuropharm.2016.07.003. [DOI] [PubMed] [Google Scholar]
- 262.Tang H., Zhang Q., Yang L., Dong Y., Khan M., Yang F., Brann D.W., Wang R. GPR30 mediates estrogen rapid signaling and neuroprotection. Mol. Cell. Endocrinol. 2014;387(1-2):52–58. doi: 10.1016/j.mce.2014.01.024. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 263.Yang Z.D., Yu J., Zhang Q. Effects of raloxifene on cognition, mental health, sleep and sexual function in menopausal women: A systematic review of randomized controlled trials. Maturitas. 2013;75(4):341–348. doi: 10.1016/j.maturitas.2013.05.010. [DOI] [PubMed] [Google Scholar]
- 264.Solomon M.B., Herman J.P. Sex differences in psychopathology: Of gonads, adrenals and mental illness. Physiol. Behav. 2009;97(2):250–258. doi: 10.1016/j.physbeh.2009.02.033. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 265.Carmassi C., Cordone A., Dell’Oste V., Pedrinelli V., Pardini F., Simoncini M., Dell’Osso L. Prescribing tamoxifen in patients with mood disorders. J. Clin. Psychopharmacol. 2021;41(4):450–460. doi: 10.1097/JCP.0000000000001412. [DOI] [PubMed] [Google Scholar]
- 266.Palacios J., Yildiz A., Young A.H., Taylor M.J. Tamoxifen for bipolar disorder: Systematic review and meta-analysis. J. Psychopharmacol. 2019;33(2):177–184. doi: 10.1177/0269881118822167. [DOI] [PubMed] [Google Scholar]
- 267.Kastenberger I., Schwarzer C. GPER1 (GPR30) knockout mice display reduced anxiety and altered stress response in a sex and paradigm dependent manner. Horm. Behav. 2014;66(4):628–636. doi: 10.1016/j.yhbeh.2014.09.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 268.Kastenberger I., Lutsch C., Schwarzer C. Activation of the G-protein-coupled receptor GPR30 induces anxiogenic effects in mice, similar to oestradiol. Psychopharmacology (Berl.) 2012;221(3):527–535. doi: 10.1007/s00213-011-2599-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 269.Findikli E., Kurutas E.B., Camkurt M.A., Karaaslan M.F., Izci F. Fındıklı, H.A.; Kardaş S.; Dag, B.; Altun, H. Increased serum g protein-coupled estrogen receptor 1 levels and its diagnostic value in drug naïve patients with major depressive disorder. Clin. Psychopharmacol. Neurosci. 2017;15(4):337–342. doi: 10.9758/cpn.2017.15.4.337. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 270.Miller L.R., Marks C., Becker J.B., Hurn P.D., Chen W.J., Woodruff T., McCarthy M.M., Sohrabji F., Schiebinger L., Wetherington C.L., Makris S., Arnold A.P., Einstein G., Miller V.M., Sandberg K., Maier S., Cornelison T.L., Clayton J.A. Considering sex as a biological variable in preclinical research. FASEB J. 2017;31(1):29–34. doi: 10.1096/fj.201600781r. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 271.Accounting for Neglected Factors and Applying Practical Solutions to Enhance Rigor and Reproducibility. 2023. Available from: https://www.preclinicaldataforum.org/addressing-sex-as-a-biological-variable-training/
- 272.Clayton J.A., Collins F.S. Policy: NIH to balance sex in cell and animal studies. Nature. 2014;509(7500):282–283. doi: 10.1038/509282a. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 273.Pawluski J.L., Kokras N., Charlier T.D., Dalla C. Sex matters in neuroscience and neuropsychopharmacology. Eur. J. Neurosci. 2020;52(1):2423–2428. doi: 10.1111/ejn.14880. [DOI] [PubMed] [Google Scholar]
- 274.Shansky R.M. Are hormones a “female problem” for animal research? Science. 2019;364(6443):825–826. doi: 10.1126/science.aaw7570. [DOI] [PubMed] [Google Scholar]
- 275.Butlen-Ducuing F., Balkowiec-Iskra E., Dalla C., Slattery D.A., Ferretti M.T., Kokras N., Balabanov P., De Vries C., Mellino S., Chadha S.A. Implications of sex-related differences in central nervous system disorders for drug research and development. Nat. Rev. Drug Discov. 2021;20(12):881–882. doi: 10.1038/d41573-021-00115-6. [DOI] [PubMed] [Google Scholar]
- 276.Bespalov A., Steckler T. Lacking quality in research: Is behavioral neuroscience affected more than other areas of biomedical science? J. Neurosci. Methods. 2018;300:4–9. doi: 10.1016/j.jneumeth.2017.10.018. [DOI] [PubMed] [Google Scholar]
- 277.Bespalov A., Steckler T., Altevogt B., Koustova E., Skolnick P., Deaver D., Millan M.J., Bastlund J.F., Doller D., Witkin J., Moser P., O’Donnell P., Ebert U., Geyer M.A., Prinssen E., Ballard T., Macleod M. Failed trials for central nervous system disorders do not necessarily invalidate preclinical models and drug targets. Nat. Rev. Drug Discov. 2016;15(7):516. doi: 10.1038/nrd.2016.88. [DOI] [PubMed] [Google Scholar]
- 278.Hodes G.E., Kropp D.R. Sex as a biological variable in stress and mood disorder research. Nat. Mental Health. 2023;1(7):453–461. doi: 10.1038/s44220-023-00083-3. [DOI] [Google Scholar]
