Next Article in Journal
Structural Analysis of Janus Tyrosine Kinase Variants in Hematological Malignancies: Implications for Drug Development and Opportunities for Novel Therapeutic Strategies
Previous Article in Journal
Nicotine: From Discovery to Biological Effects
Previous Article in Special Issue
Real-Time PCR Quantification of 87 miRNAs from Cerebrospinal Fluid: miRNA Dynamics and Association with Extracellular Vesicles after Severe Traumatic Brain Injury
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Sex Differences in Brain Disorders

by
Malgorzata Ziemka-Nalecz
,
Paulina Pawelec
,
Karolina Ziabska
and
Teresa Zalewska
*
NeuroRepair Department, Mossakowski Medical Research Institute, Polish Academy of Sciences, 5, A. Pawinskiego Str., 02-106 Warsaw, Poland
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2023, 24(19), 14571; https://doi.org/10.3390/ijms241914571
Submission received: 6 September 2023 / Revised: 20 September 2023 / Accepted: 22 September 2023 / Published: 26 September 2023
(This article belongs to the Special Issue New Mechanisms and Therapeutics in Neurological Diseases 2.0)

Abstract

:
A remarkable feature of the brain is its sexual dimorphism. Sexual dimorphism in brain structure and function is associated with clinical implications documented previously in healthy individuals but also in those who suffer from various brain disorders. Sex-based differences concerning some features such as the risk, prevalence, age of onset, and symptomatology have been confirmed in a range of neurological and neuropsychiatric diseases. The mechanisms responsible for the establishment of sex-based differences between men and women are not fully understood. The present paper provides up-to-date data on sex-related dissimilarities observed in brain disorders and highlights the most relevant features that differ between males and females. The topic is very important as the recognition of disparities between the sexes might allow for the identification of therapeutic targets and pharmacological approaches for intractable neurological and neuropsychiatric disorders.

1. Introduction

A remarkable feature of the brain is its sexual dimorphism. Brain dimorphism is a complex process, with multiple contributing mechanisms and pathways resulting in differences. Sex-based differences in terms of prevalence, age of onset, pathophysiology, and symptomatology have been identified in a range of neuropsychiatric illnesses (Figure 1). Mechanisms determining sexual dimorphism in pathological states may vary according to location and species. The majority of the literature indicates the key role of a complex interaction between genetic factors and sex hormones, which are probably responsible for the different susceptibility of men and women and may change the severity of symptoms. However, other sex-specific factors, e.g., environmental factors and gene–environment interactions, are also considered. It should be noted that many of the participating factors suggested by researchers remain elusive and require further attention. Despite recent advances, the precise link between the proposed agents and brain impairments is not completely understood. The present paper aims to provide an overview providing up-to-date information on the more recent findings on sex-related differences in neuropsychiatric diseases (Table 1). A better understanding of the mechanisms underlying sex-based answers to pathological conditions may be important in the identification of therapeutic targets and pharmacological treatments for these intractable disorders.

2. Autism Spectrum Disorder (ASD)

Autism spectrum disorder (ASD) comprises a heterogeneous group of neurodevelopmental disorders defined by the early developmental onset of continuous life-long symptoms characterized by social communication deficits and a pattern of restricted behaviors of interest [50,51]. One of the most widely accepted theories is that autism stems from some neurodevelopmental anomaly (genetic, environmental, and hormonal). Since the etiopathogenesis of autism has not been elucidated so far, the diagnosis of individuals is mainly based on the presentation of three symptoms: profound alteration in social interaction, communication deficits, and stereotyped behaviors. A comparative clinical study indicated a significant predominance of ASD pathology in males. According to available information, males are about 3–4 times more likely to be diagnosed than females [6,7,8,9]. So far, some contrasting data postulate that females have less restricted interests and activities and more significant social communication difficulties than males with ASD [3,4,5]. In contrast, one study reported that females and males with ASD did not differ in terms of the severity of autism [52]. Thus, till now, the results concerning differential sex effects in ASD are highly variable. The inconsistency of subtle yet potentially meaningful ASD phenotypic differences between males and females results from methodological issues [53]. It should be mentioned that while there is no evidence of sex differences in early childhood cognitive functioning, an executive functioning difference exists in adulthood. Therefore, new research will probably be needed for the elucidation of the differences between sexes, covering both the onset and severity of the symptoms.
In contrast to the clinical presentation, only a few studies have systematically explored the effect of sex in mouse models of autism. A summary of the knowledge about sex-dependent behavioral differences is provided by Romano et al. [54]. However, because of the prevalence of males in the analyzed group, it was rather impossible to draw any conclusion. Recent data in models suggest that females with autism when compared to affected males, present less severe repetitive/restricted behaviors but comparable deficits in social and communication domains [55].
The precise mechanism responsible for the differences between men and women remains unclear. Many researchers considering the developmental effects of steroid hormones in the brain during premature organization, as well as the maintenance of sexually dimorphic structures, have focused attention towards their engagement in setting sex-specific liability [7,56,57]. The action of hormones was further strongly supported by the finding that abnormal exposure to testosterone during pregnancy correlated positively with several autistic features and inversely with social skills in children and had a profound effect on sexual dimorphism in human behavior and ASD [1,2]. Testosterone may interact with downstream molecules like neurotransmitters, neuropeptides, and immune pathways and, by these, contribute to male vulnerability. In an interesting report, Baruch et al. (2018) state that altered androgen and estrogen levels could both lead, via different pathways, to alterations of cognitive, linguistic, social and emotional circuits [57].
Male and female hormones control the expression of autistic candidate genes differently. For example, the candidate gene for the increased susceptibility to autism is the RORA gene, the expression of which is low in the frontal cortex of autistic individuals and oppositely regulated by the male and female hormones. Males seem to be more susceptible to disruptions in RORA and an increase in testosterone [58]. Therefore, genes associated with ASD may be modulated by sex hormones.
Various studies have attributed the male preponderance of ASD to sex-specific single-nucleotide polymorphism, single-nucleotide variants, copy number variants, and proteins (some of them involved in synapse formation and cell adhesion [8,59,60,61,62]. However, the above findings have not been consistently replicated [63].
Despite several advances, the precise link between the considered factors and sex-based differences in ASD is not completely understood. The summary of existing hypotheses and phenomena that contribute to the differences between males and females in ASD are presented in an elegant, detailed review prepared by Ferri et al. [64]. This work may provoke researchers to perform further studies for the recognition of sex-based mechanisms in ASD, which are necessary for the introduction of more specific therapies.

3. Down Syndrome

Down syndrome (DS) is the most common genetic cause of intellectual disability, with a prevalence of 1 in every 700 births. Down syndrome (DS) is caused by partial or full triplication of human chromosome 21 (Hsa21) [65]. Data taken from the literature indicate a prevalence of males in children with all trisomy 21 variants with a sex ratio of 1.24. The most prominent feature of DS is the learning and memory deficiency that affects 100% of patients. The disease also demonstrates other behavioral phenotypes, such as the incomplete and delayed acquisition of motor, linguistic, and visual–spatial abilities and neurobehavioral disorders. It was detected that DS males, as compared to females, have major behavioral problems, including attention deficit, hyperactivity-like behaviors in childhood, and depression in adulthood [12]. Other occurring phenotypes diagnosed in DS patients are related to cardiovascular disturbances, breathing, and skeletal alterations. It was noted that males were affected twice as much as females, suffering a higher prevalence of breathing disorders during sleep [13]. In contrast, a lower bone mineral density was detected in DS females [66].
Research examining sex differences postulated a general agreement that women with DS are at higher risk of developing Alzheimer-like dementia [10,11]. The early age of menopause onset in relation to DS and the loss, as a consequence, of endogenous bioavailable estradiol levels cause women to be four times more likely to develop Alzheimer disease (AD) [67]. Thus, levels of estradiol were found to correlate with the onset of cognitive decline in female DS patients [68]. It is known that the hormonal status of DS patients appears to be highly affected, but to date, no clinical trials concerning estrogen, as well as other hormone replacement therapies, have been published for women with DS. In contrast to the lack of clinical trials in humans, a prolonged estrogen treatment improved cognitive deficits in Ts65Dn mice and restored deficits in the brain’s cholinergic system and function [68,69,70,71]. The results might be coupled to the normalization of the serum levels of estrogen, which was reduced in DS female mutant mice. This leads us to the conclusion that estrogen probably plays a key role in delaying the onset and severity of dementia in women with AD [72,73]. Thus, the results of the preclinical study forced us to assess this speculation in human patients.

4. Rett Syndrome

Rett Syndrome (RTT) is a progressive X-linked neurodevelopmental disorder. RTT affects almost exclusively girls, with a prevalence of 1 in 10,000 births [15,16,17,18]. Diagnosis of RTT in males is rare as patients do not often survive past infancy [19], except for a few cases recognized beyond early childhood [74,75,76]. Typical symptoms are onset at months 6–18 after normal neurological and physical development and progress over several stages with a regression period characterized by a profound loss of acquired developmental communication skills [77]. Further on, disease development reaches a plateau, joined by deterioration of motor abilities, breathing and feeding disturbances, EEG impairment, sleep problems, and autistic-like behaviors [14,77,78,79]. Rett Syndrome is the most abundant cause of intellectual disability, neurological regression, loss of speech, and learning deficits. One of the most common symptoms of RTT is epilepsy, which occurs in 60–80% of RTT cases. Neuropathological studies showed that RTT patients exhibit reduced brain volume and neuronal hypotrophy [14]. The studies have reported that RTT patients also demonstrate other symptoms caused by metabolic complications in organs other than the brain [80,81]. These include gastric problems, cardiorespiratory problems, decreased bone density, dyslipidemia, inflammation of the gallbladder, urological dysfunction, and sleep disturbances [79,82].
RTT results primarily from mutations in the X-linked gene (MECP2), encoding methyl-CpG binding protein 2 (MeCP2) [83]. The protein MeCP2 binds to methylated genomic DNA and functions as a transcriptional regulator through interactions with other important genes [84]. Evidence indicates that MeCP2 expression correlates with the postnatal maturation of the CNS and neuronal differentiation [85]. Importantly, MeCP2 plays a particularly significant role in the development and maintenance of functional mature neurons [86,87]. Mutation in MeCP2 accounts for more than 90% of classical RTT causes [14]. Moreover, mutation status combined with a degree of X chromosome inactivation and the presence of modifier genes might be a predictor of disease severity [79]. Furthermore, selectively re-expressing MeCP2 in the postmitotic neurons of adult mice results in partially reversing the symptoms of RTT [88,89,90].
Clinical presentation in Rett Syndrome differs significantly among individuals based on the degree of phenotypic variation resulting from specific mutations.
The monogenic nature of the disease has led to the development of Mecp21-mutant mouse models, some of which recapitulate certain phenotypes of human RTT. A comparison of behavioral analyses showed sex-dependent differences in the major mouse model of RTT [54]. The effects could be linked to differential expression of MeCP2 in male and female brains during development [91]. It seems logical to hypothesize that in the early programming of sex-based differences, a complex interaction between sex hormones and MeCP2 takes place. Unfortunately, in contrast to the reported data indicating the involvement of hormones in neurodevelopmental diseases, there is no information on the levels of sex hormones at different ages in RTT mouse models. Further knowledge on this topic may be necessary for a better understanding of the different roles played by MeCP2 in the development of sex-based differences [54].
The studies in the RTT field have recently focused on finding an effective treatment. One of the considered strategies is to deliver a functional version of the gene to increase MeCP2 expression and could compensate for the loss of function [92]. The other one is to target downstream factors of MeCP2 by improving neurotransmitter signaling or upregulation genes under MeCP2 control.
Introducing the first strategy may have functional implications, as MeCP2 overexpression may reactivate X-linked genes to pathogenic levels. Thus, it is necessary to use a molecule that can reactivate only MeCP2 expression [93,94].
The second strategy requires the identification of the precise functions of MeCP2, which are currently still limited [79]. Therefore, future research should consider improving the quality of life of RTT patients.

5. Fragile X Syndrome

Fragile X Syndrome (FXS) is a monogenetic neurodevelopmental disorder caused by an expansion of unstable trinucleotide repeats (CGG) within the promoter of mental retardation gene 1 (FMR1), encoding for the Fragile X Mental Retardation Protein (FMRP) [21,95,96]. FMRP is an RNA-binding protein involved in several functions through the modulation of the translation of several proteins engaged in brain activity. Additionally, FMRP, as a component of the ribonucleoprotein complex, participates in shuttling between the nucleus and cytoplasm, in transport along dendrites, and in association with polyribosomes [22,97,98,99]. Genetic aberration reduces or even eliminates FMRP, leading to neurodevelopmental impairments. Studies have shown that the lowered level of FMRP expression correlates with the degree of disorders [100,101]. Unexpectedly, the assessment of function suggests that despite the higher level of FMRP in females vs. males, females’ FXS do not necessarily achieve better outcomes than their male counterparts [102].
Clinical features of individuals with FXS include intellectual impairment and stereotyped behaviors such as locomotor hyperactivity, sensory hypersensitivity, and unusual physical marks such as an elongated face, flat feet, and extended finger joints. Fragile X Syndrome is usually diagnosed around 2–3 years of age and predicts life-long disability. Till now, disease-modifying drugs are still not available [103,104]. The data sets present a lower incidence of FXS in females, at a ratio of 1:7000, while in males, this FXS syndrome affects 1 in every 4000 of the population [105,106]. In general, female human patients typically display less severe symptoms than males, probably due to compensation by the second not-affected X chromosome [20]. In contrast, others have thought that such compensation alone may not be sufficient to explain all sex-dependent differences.
Recent data postulated the regulation of Fmr1 gene expression by sex hormones, particularly 17 beta-estradiol and testosterone [54]. The precise analysis performed by Yang et al. [107] found that the alteration in estradiol level in Fmr1 KO neurons is associated with changes in LTP, synaptic architecture, and fear memory. This finding supports a complicated interaction between FMRP and sex hormones, although the research has addressed that other factors, such as different development or different neural circuitry, also participate in these processes [108].
Sex-dependent differences have been evidenced in cognitive and clinical profiles [109]. Investigations have challenged the notion that FXS males present a greater likelihood of displaying autistic-like behaviors, while females may have normal intelligence or mild intellectual disability [21,22,23].
The topic of sex-dependent differences in symptoms in FXS patients was developed further in preclinical rodent models in rats [110] and mice [21,111,112]. The data obtained via an analysis of Fmr1 KO of both sexes are consistent with the above-described milder phenotype in females compared to affected males [113,114,115,116]. The detailed study of Wong’s research group found differences in EEG and in repetitive behavior tests, wherein males displayed increased activity while females did not. In addition, females but not males showed abnormal fear memory. In the opinion of Nolan et al. [117], the effect is probably associated with the loss of FMRP.
However, there is a lack of information concerning the interaction between the investigated genotypes and hormones in Fmr1 KO animals. So far, there are also no data presenting hormone profiles in these models. Therefore, the discussion about the potential therapeutic value of the pharmacological modulation of hormone levels is, at present, impossible and requires more studies.

6. CDKL-5 Associated Disorders (CDD)

Cyclin-dependent kinase 5 (CDKL-5 known as STk9) encodes a serine/threonine kinase, with the highest levels of expression in the brain [118]. CDKL-5 protein is vital for normal brain development and function. It participates in basic processes such as neuronal morphogenesis and plasticity through interaction or association with other proteins or via phosphorylation modification [119,120,121,122]. A lack of CDKL-5 expression results in decreased dendritic branching, impaired neuronal circuit connection, impaired synaptic vesicle stability, and neurite outgrowth, as demonstrated in vitro [123,124,125] and in vivo [120,122,126].
Mutations in the X-linked CDKL5 gene and consequential ion deficiency in cyclin-dependent kinase-like 5 caused severe neurodevelopment disorders (CDD). Individuals suffer infantile-onset drug-resistant seizures and lifelong disability, including motor, visual and social impairments. CDKL-5 disorders primarily affect females despite CDD being rather rare, with a prevalence of 1:40,000 or 1:60,000 births [127,128,129]. Pathogenic variants in CDKL-5 are among the most common genetic causes of severe epilepsy in childhood. No effective treatment is currently available, and medical management is only symptomatic and supportive.
The generation of knock-out mice carrying inactivating mutations in the catalytic domain allowed for the characterization of the in vivo consequences of the loss of function of this protein [130,131,132]. Behavioral research shows that mutant mice display impulsivity together with defective cognitive performance and autistic-like abnormalities. However, there are no complete data comparing the phenotype of males and females in this KO model. Only one study found a sex-dependent subtle difference in locomotor activity [130]. However, data are very scarce and do not allow for precise conclusions.

7. Bipolar Disorder

Bipolar disorder (BD) is a serious and insistent complex psychiatric disorder resulting in substantial morbidity and mortality. BD is characterized by extreme fluctuations in mood, demonstrated by manic, hypomanic and depressive episodes. A substantial proportion of patients with bipolar disorder experience neurocognitive impairments [133,134,135]. Lifetime prevalence reaches 1.3–1.6% in all human populations. BD begins in late adolescence or early adulthood with a mean onset age of before 21 years [136]. The majority of studies indicate an equal age of onset across the sexes, although some data have shown that women are slightly older than men when the disease manifests [28,137]. The ratio of BD incidence in men and women is approximately 1:1. Important but still limited and sometimes contrasting findings indicate the existence of sex differences concerning the epidemiological, clinical and psychopathological characteristics of the disease [138]. Usually, women show a higher rate of rapid cycles [27], mixed features [139], a higher number of depressive episodes [28,29] and suicide attempts [30,31]. Conversely, manic episodes and unipolar mania are more common among men [28].
A substantial proportion of patients with BD Ie neurocognitive impairments [135], but this subject has not been explored in the literature relating to BD. Lastly, the field of BD research has focused on the relative contribution of sex to cognitive profiles in BD [140,141].
It was found that men with BD perform worse than women in some aspects, like visuospatial construction [142,143], verbal memory performance [144], and reduced sensorimotor gating [145]. By contrast, males performed better in spatial memory and sustained attention tasks [146]. Recently, Xu observed that scores regarding attention and delayed memory were lower in males than in females [140]. In addition, one study showed that women with BD experience greater cognitive benefits from vigorous physical activity than men with BD [147].
Previous studies stated that the sex-specific degree of neurocognitive impairments is closely linked to sex dimorphism in brain structures. In truth, no full anatomical analysis was performed on a brain with BD. Only two reports have indicated significant evidence of anatomical differences. Shi [148] observed that right hippocampal volume loss was more evident in females than males. The second publication by Mitchell et al. [149] found that two structures (the left supramarginal gyrus and right inferior parietal lobule) are smaller in males with BD than in females and were correlated with worse cognitive performances. Due to the limited findings, it is still not clear if such differences influence the described cognitive reactions.
Further systematic studies showed that sex-mediated differences in symptoms and cognitive dysfunction in BD are attributed to sex hormones (gonadal and neurosteroids) [150,151,152]. Worsening BD symptoms in women during periods of low levels of sex hormones and in clinical trials indicate that steroid hormones can improve specific symptoms of BD, which is consistent with the above suggestion. Importantly, two other studies showed the benefits of using synthetic progesterone for the treatment of acute mania [153,154].
Despite these important findings, speculation that hormones determine cognitive sex differences in BD would be rather premature. The existing literature containing research data showing hormones profile in the BD disorder is still sparse and inconsistent [141]. It seems clear that sex-mediated differences need to be determined in future investigations.
Apart from hormones, it is thought that genes are likely to explain sex-mediated differences in neurocognitive impairments. However, no distinct gene has been unequivocally associated with BD [24]. Detailed analysis of gene expression in post-mortem tissue has shown dysregulation of several genes and pathways, including those associated with the ubiquitin pathway, synaptic functions [24], intracellular protein trafficking, and protein metabolic processes [25] in BD patients. The reported results are not precise, and future studies will need to address this subject to achieve an understanding of the complex mechanism.
Furthermore, others have focused on the potential effect of inflammation. A body of evidence implies that the difference in exacerbated inflammation occurring in BD may be a causative factor in different levels of cognitive decline in females and males. Dysregulation of cytokine levels in the peripheral blood and central nervous system (CNS) of patients and the relapsing–remitting nature of the disease might point out the significance of immune responses in the pathogenesis of BD [155]. It has also been detected that an increase in proinflammatory markers TNFalfa and IL8 is associated with both depressive and maniac episodes [26,156,157]. In general, little is known about the potential role of inflammation in sex-based differences. Importantly, only one study reported the upregulated expression of SOCS genes (suppressors of cytokine signaling), and only in male subjects [158]. These results provide support to the hypothesis that sex-dependent differences in patients with BD might be, at least partially, explained by inflammation. A significant effect of sex was later confirmed when considering altered inflammation markers in BD phases [159]. More work is needed to prove a direct causal link between inflammation and sex differences.

8. Epilepsy

Epilepsy is a chronic neurological disorder that affects more than 70 million people worldwide [160]. The disease is characterized by an enduring predisposition to recurrent spontaneous and unpredictable seizures, which can result in brain damage and substantial body injury. The clinical manifestation of epilepsy includes sudden and abnormal episodes of motor, sensory, autonomic, or psychological origin. The disorder can arise as a result of a dysfunction of the normal connectivity of neural networks or/and imbalance of inhibitory and excitatory neurotransmitters. Comparative clinical data have shown that the overall incidence of epilepsy, the susceptibility to excitability episodes, and the occurrence of epileptic seizures are generally higher in males [32,33,34,35]. Nevertheless, exacerbation of cyclic series seizures in women during the specific phases of the menstrual cycle was also documented. Such occurring episodes are named catamenial epilepsy. Studies indicate that the menstrual-related fluctuations of seizures may be linked to alterations in the concentration of neurosteroids in the brain [161,162,163].
Despite such inconsistency, sex-dependent differences are observed across the different forms of epilepsy and seizure conditions [164]. The mechanisms that cause the establishment of dimorphism in epilepsy still remain unclear. Two groups of researchers reported that sex-linked disparities in regional brain morphology, cerebral asymmetry, and variations in functional connectivity in the brain may determine sex-dependent vulnerability to seizures and epileptogenic cascades [165,166]. However, the precise molecular events that lead from the alteration of brain morphological structures to sexual differences are not completely understood.
A few but convincing studies showed a strong connection between sex hormones and epileptic conditions. It was found that steroids derived from the peripheral system, as well as neurosteroids, likely exert a changeable and complex effect on seizure sensitivity. Since then, these agents have received considerable attention, spurring justified hope that many hormones may represent new therapeutic strategies against seizures [167,168,169,170]. Detailed analysis of neurosteroid (allopregnanolone and androstanediol) function revealed that they act as positive modulators of inhibitory GABAA receptors [171]. Consistent with this, chloride influx into the cell increased, producing neuronal inhibition [167].
Despite the fact that, to date, there have been no studies to demonstrate that reduced levels of neurosteroids can influence epileptogenesis, its effect on neuronal inhibition may serve as a basis for sex differences in epilepsy. One particularly illuminating experiment conducted by Reddy et al. [172] showed that extrasynaptic receptor GABAA contributes to sex differences in seizure susceptibility and neurosteroid protection. In addition, his results confirmed greater sensitivity in females than males. This corresponds partially with the earlier statement that steroid hormones play a key role in the neuroendocrine control of neuronal excitability and seizure susceptibility [173].
It has been shown that the action of another intensively analyzed hormone, progesterone, is associated with anticonvulsant activity due to the negative impacts of glutamatergic transmission [174,175]. It is worth noting that only a few studies performed clinical experiments with progesterone administration. This leads to reduced seizures in women with epilepsy [162,176]. In contrast, estrogen presents a highly variable effect on seizure susceptibility, while, in general, it exhibits pro-convulsant and epileptogenic properties in animals and humans via the potentiation of glutamatergic transmission [174,175,177]; on the other hand, some studies demonstrate protective anticonvulsant activity. It is apparent that the final action is likely due to treatment duration, dosage, hormonal status, and the seizure model [178,179].
Interestingly, several studies have also shown that androgens and testosterone serve as bimodal modulators of seizure susceptibility, demonstrating, under specific conditions, anticonvulsant as well as pro-convulsant properties. The data produced by Reddy [180] postulated that the metabolization of testosterone to estrogen increases seizure susceptibility in both animal and human models. In contrast, testosterone, through conversion to androstanediol, a positive allosteric modulator of GABAA [181,182], activated GABAA receptors, inducing chloride influx into the cell and leading to the hyperpolarization of the neuron, which can prevent neuronal hyperexcitability and diminish epileptic seizures.
Together, the above results emphasize the similarities and differences among hormone functions in epileptic conditions. Whether the action is beneficial or detrimental is still a matter of debate. The most important finding is that the antiseizure action of neurosteroids presents sex differences [172]. Future development of this research is needed for the further recognition of sex differences in epilepsies to design personalized therapies.
Despite the postulated key role of hormones, we cannot rule out the possibility of other potential factors that may determine the sex-mediated differences in epilepsy. Among the factors participating in this phenomenon are neurogenesis, glial response, chloride homeostasis, and neurotrophic factors [164]. Despite the fact that they likely play a role in determining sex differences, till now, there has been limited direct evidence of the precise functional role of these agents in models of epilepsy.
Importantly, future work is needed to understand sex differences for the design of effective therapies for epilepsy and disease modification of epileptogenesis.

9. Schizophrenia

Schizophrenia (SZ) is one of the most complex and least understood psychiatric disorders, showing a wide range of inter-individual variation in many aspects of the disease. It is typically referred to as a chronic and debilitating condition because it may lead to a progressive functional decline impacting cognitive, affective, and social domains [183]. The heterogenic symptoms of the disease range from hallucinations and delusions, disorganized speech and behavior, flat affect, lack of motivation, and cognitive deficits [184,185]. Cognitive impairments exist in almost all patients with schizophrenia and may determine the functional outcome of the disorder [186]. To date, the exact mechanism underlying schizophrenia is not clear.
Schizophrenia affects up to 1% of the world’s population. SZ commonly develops during adolescence or early adulthood [187]. The incidence of schizophrenia is generally higher in men than in women in ratio estimates ranging from approximately 1.2 to 1.5 [188,189]. The reverse of this ratio and the higher prevalence in women is seen in the second half of life (after 40 years).
Clinical observations and evidence in epidemiological surveys revealed significant sex differences in the incidence, clinical course, expression, and premorbid functions, as well as response to clinical treatment in schizophrenia patients [190,191,192]. In addition to the earlier age of onset and greater premorbidity dysfunction, men with schizophrenia experience more severe negative and cognitive symptoms (social withdrawal, lack of motivation, poverty of speech) relative to women [42,43,44]. Women, on the other hand, display more affective symptoms, including depression, impulsivity, self-harm, and suicide attempts [41]. The diversity most probably arises from the interplay of sex hormones and neurodevelopmental and psychosocial sex differences, but the identification of origin continues to be controversial.
The early studies tried to link sex-mediated differences to diverse structural and functional brain abnormalities in schizophrenia patients. Finally, the performed analysis yielded inconsistent results. It was found that the disparities occur in areas that normally show sexual dimorphism, implying that the same factors are engaged in both normal neurodevelopmental processes and those associated with schizophrenia. Thus, the gathering of data sets allowed us to state that men’s brains present more severe morphological abnormalities than women, including reduced frontal and temporal volumes [41,188,193,194,195].
Further systematic studies have revealed a strong connection between the levels of sex hormones and the pathophysiology of schizophrenia [36,196,197,198,199]. Importantly, it has been convincingly shown that patients with schizophrenia benefit from hormone treatments such as estrogen, progesterone, and testosterone [200,201]. The protective effect of reproductive hormones was also observed in the clinical symptoms characterized by later onset and fewer symptoms in women [200,201,202]. A significant piece of evidence concerning the action of estrogen came from the observation that during low estrogenic phases in women, greater psychosis onset and more severe symptoms were noted. The administration of hormones to postmenopausal women reduced the risk of disease, as was already noted in other impairments [198,203]. In addition, it was detected that high levels of estrogen during pregnancy may prevent psychotic relapse. Interestingly, the negative correlation between plasma estrogen levels and schizophrenia symptoms was also reported in male patients [204]. Thus, the action of estragon has received considerable attention as a probable therapeutic agent.
It has also been reported that estrogen plays a neuroprotective role by influencing major neurotransmitter systems relevant to schizophrenia as a dopaminergic system. It is known that action on the dopaminergic system is mediated by both non-genomic activity expressed stimulation of specific signaling pathways and genomic effects involving gene transcription [205]. It is hypothesized that the effect of estrogen on dopamine signaling pathways is linked to reduced impulsivity and risk of substance abuse in women [37,198]. Moreover, estragon may modify the effects of serotonergic [38,206,207] or glutamatergic systems [38,39,40] in animal studies. Estrogen increased the density of 5HT2a receptors in specific areas of the brain.
In contrast to several pieces of data concerning estragon, there is only a small amount of information available describing the effect of progesterone in the pathophysiology of schizophrenia. Halari et al. reported an association between higher circulating progesterone levels and poorer performance, specifically in spatial memory [208]. Some of the data showed that this reaction may be implicated in cognitive and emotional processing. Summarizing this discussion, it became clear that existing observations on estrogen and progesterone in schizophrenia are inconsistent, and some studies have also mentioned that these hormones have no significant effect on schizophrenic patients [209].
Another set of informative data came from analyzing the effect of testosterone. It showed that a low level of testosterone (the main male hormone) is probably associated with more severe symptoms, although results are still not completely consistent [210,211,212]. Testosterone receptors, similarly to estrogen receptors, have been implicated in the modulation of mood states and cognitive performance [213,214]. More recent reports have demonstrated that the plasma level of testosterone was inversely correlated with the severity of negative symptoms in male patients. The authors of [196,211,215] found diminished levels of testosterone in male patients and enhanced levels in female patients. These elevated levels in females correlated with brain activation during mental rotation.
Another important hormone in terms of reproductive function, oxytocin, was considered a potential therapeutic target for schizophrenia. In agreement with this suggestion, patients presenting with high levels of plasma oxytocin developed fewer psychotic symptoms [216,217] and improved cognition [218]. The inhibition of the antipsychotic effect may be related to its ability to act as a dopamine regulator [219].
A limited number of studies have been performed to identify the genetic variant that differentiates males and females [220]. So, a gender-specific association of certain dopaminergic genes, such as catechol-o-methyltransferase (COMT) and mono-amino-oxidase (MAO), with schizophrenia was suggested. Further detailed research indicated that sex-specific DNA methylation of the MAO-promoter has been associated with schizophrenia [221]. Furthermore, sex differences have also been found in GABA-ergic genes [222]. The data obtained by using GWAS found that a gene variant that encodes the zinc finger protein responsible for the regulation of dendritic spine maintenance and implicated in cognitive performance [223,224,225] was related to schizophrenia, specifically in women [226]. Similarly, the SLC30A3 gene (which encodes protein zinc transporter 3) increases the risk of schizophrenia, again, specifically in women [227].
It is concluded that sex may affect the overall clinical picture of schizophrenia. Based on the data taken from the literature, it is suggested that the interplay of chromosomal factors and gonadal hormones may contribute to sex differences. However, attention should be paid to several additional factors that may play a role in sexual differentiation without the involvement of hormones [228]. These include differences in epigenetic mechanisms, different severity, and perinatal complications as well as exposure to viruses and environmental toxins, which could affect men and women presenting with different predispositions to schizophrenia.

10. Stress-Related Depressive Disorders

Stress-related disorders such as major depressive disorder (MDD) and post-traumatic stress disorder (PTSD) constitute some of the most common disabling diseases worldwide, with an approximately two-fold greater prevalence in women than in men [49,151,229,230,231]. The disorders are characterized by depressed mood, diminished interest in all daily activities, a decline in cognitive ability, and vegetative symptoms, such as disturbed sleep and appetite. They may also cause secondary disability, as patients with depression are more likely to develop chronic medical illnesses. Conventionally, depressive symptoms have been attributed to an imbalance in the hypothalamic–pituitary–adrenal axis following extreme stress created during trauma [232,233], as well as to dysregulation in the neuromodulation of neurotransmitters. Depressive disorders are also associated with inflammation [45,46].
Comprehensive studies have shown that MDD is ranked at the top of neuropsychiatric impairments as a leading cause of mortality [234]. MDD is associated with alterations in regional brain volumes, particularly the hippocampus, and with functional changes in brain circuits, such as cognitive control networks. The etiology of MDD is multifactorial, and its heritability is probably about 35% [49]. The evidence has pointed to environmental factors, such as sexual, physical or emotional abuse during childhood, which are strongly associated with the risk of developing MDD.
Epidemiological studies of MDD and PTSD found sex-based differences in the rate of disorder presentation, with an increased risk of depressive episodes in women. They may be related to the lower threshold for the manifestation of disorders. For example, it was observed that women present different initial reactivity to trauma with increased peri-traumatic dissociation and a greater negative recall, which suggests greater negative memory consolidation following stress [47]. It is worth pointing out that the progression rate of depression in females compared to males is directly proportional to the development of female reproductive function during puberty [235,236]. It indicates the interconnection of sex hormone levels and their receptors with sex differences.
In recent decades, considerable attention has been received by gonadal hormones as important mediators of sex differences between men and women [233,237]. This hypothesis is in agreement with the results of a meta-analysis that evidenced the influence of hormones on stress reactivity. Moreover, this group of researchers identified estradiol as a possible related risk factor in the prevalence and severity of PTSD between sexes. They discovered that estrogen-response elements on the pituitary adenylate cyclase-activating peptide receptor gene, PACIRl, determined severity in women but not men [48].
According to the previous data set, estradiol is an important regulator of the recall of extinct memories [238], which is the essential process underlying recovery from a traumatic experience. Women with higher levels of endogenous estradiol more successfully extinguish traumatic memories compared to women using contraceptives [239,240].
Summarizing the available data, PTSD and MDD disorders display clear differences between males and females. Management primarily comprises pharmacological treatment. However, there is still very little evidence of sex-related differences in terms of the efficacy of antidepressant drug treatments despite all the research performed in relation to the topic. Since both pharmacokinetics and pharmacodynamics may differ between males and females, it may become necessary to consider sex in clinical trials and provide adequately stratified guidelines. To improve therapeutic approaches, it is essential to understand the underlying mechanisms that cause males and females to respond differently to clinical interventions. Therefore, the major efforts of researchers should be directed at fully identifying the factors related to these sex-based phenomena.
An important set of supporting information came from studies on animal models including males and females. The results of several studies have revealed sex differences and the sexually dimorphic effect of some factors’ actions [241,242]. Valentino et al. have evidenced that there are sex differences in trafficking and signaling of the main modulator of the stress response-the corticotropin-releasing factor (CRF1) receptor. Sex-biased association of the CRF1 with the Gs-protein and beta-arrestin 2 would render females more sensitive to acute stress and less capable of adapting to chronic stress [241]. Moreover, Torrisi et al. discovered that the same stress-induced memory impairment in both sexes can be triggered by different sex-dependent molecular mechanisms [243]. The results of this work suggest a possible elevated NMDA and AMPA receptor sensitivity to the acute stress-induced aberrant and prolonged glutamate release in the dorsal hippocampus of female mice. Some, but not all data could be extrapolated to humans.

11. Conclusions

The clinical evidence reviewed here clearly illustrates pronounced sex differences in some aspects of neuropsychiatric disturbances. What emerges from this review is that much needs to be done. In particular, detailed investigations should be directed at identifying the mechanisms responsible for the sex-based difference in impairments. For now, it is still too early to translate the preliminary results obtained to routine clinical use. Nevertheless, although many questions remain to be answered, this topic may lead to avenues for novel sex-specific strategies and therapeutic options in men and women suffering from neuropsychiatric disturbances.

Author Contributions

T.Z. was the major contributor in collecting the data and writing the manuscript. P.P. and K.Z. participated in writing the manuscript, M.Z.-N. participated in writing the manuscript and preparing the figures. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by the Mossakowski Medical Research Institute Statutory Fund no 6/2023.

Conflicts of Interest

The authors declare no conflict of interest. The funders had no role in the design of the study; in the collection, analyses, or interpretation of data; in the writing of the manuscript; or in the decision to publish the results.

References

  1. Knickmeyer, R.C.; Baron-Cohen, S. Topical Review: Fetal Testosterone and Sex Differences in Typical Social Development and in Autism. J. Child. Neurol. 2006, 21, 825–845. [Google Scholar] [CrossRef]
  2. Baron-Cohen, S.; Auyeung, B.; Nørgaard-Pedersen, B.; Hougaard, D.M.; Abdallah, M.W.; Melgaard, L.; Cohen, A.S.; Chakrabarti, B.; Ruta, L.; Lombardo, M.V. Elevated Fetal Steroidogenic Activity in Autism. Mol. Psychiatry 2015, 20, 369–376. [Google Scholar] [CrossRef]
  3. Carter, C.S. Sex Differences in Oxytocin and Vasopressin: Implications for Autism Spectrum Disorders? Behav. Brain Res. 2007, 176, 170–186. [Google Scholar] [CrossRef]
  4. Hartley, S.L.; Sikora, D.M. Sex Differences in Autism Spectrum Disorder: An Examination of Developmental Functioning, Autistic Symptoms, and Coexisting Behavior Problems in Toddlers. J. Autism Dev. Disord. 2009, 39, 1715–1722. [Google Scholar] [CrossRef]
  5. Frazier, T.W.; Georgiades, S.; Bishop, S.L.; Hardan, A.Y. Behavioral and Cognitive Characteristics of Females and Males with Autism in the Simons Simplex Collection. J. Am. Acad. Child Adolesc. Psychiatry 2014, 53, 329–340.e3. [Google Scholar] [CrossRef]
  6. Baron-Cohen, S.; Lombardo, M.V.; Auyeung, B.; Ashwin, E.; Chakrabarti, B.; Knickmeyer, R. Why Are Autism Spectrum Conditions More Prevalent in Males? PLoS Biol. 2011, 9, e1001081. [Google Scholar] [CrossRef]
  7. Lai, M.-C.; Lombardo, M.V.; Suckling, J.; Ruigrok, A.N.V.; Chakrabarti, B.; Ecker, C.; Deoni, S.C.L.; Craig, M.C.; Murphy, D.G.M.; Bullmore, E.T.; et al. Biological Sex Affects the Neurobiology of Autism. Brain 2013, 136, 2799–2815. [Google Scholar] [CrossRef]
  8. Werling, D.M.; Geschwind, D.H. Sex Differences in Autism Spectrum Disorders. Curr. Opin. Neurol. 2013, 26, 146–153. [Google Scholar] [CrossRef]
  9. Werling, D.M. The Role of Sex-Differential Biology in Risk for Autism Spectrum Disorder. Biol. Sex Differ. 2016, 7, 58. [Google Scholar] [CrossRef]
  10. Matthews, D.C.; Lukic, A.S.; Andrews, R.D.; Marendic, B.; Brewer, J.; Rissman, R.A.; Mosconi, L.; Strother, S.C.; Wernick, M.N.; Mobley, W.C.; et al. Dissociation of Down Syndrome and Alzheimer’s Disease Effects with Imaging. Alzheimer’s Dement. Transl. Res. Clin. Interv. 2016, 2, 69–81. [Google Scholar] [CrossRef]
  11. Fiest, K.M.; Roberts, J.I.; Maxwell, C.J.; Hogan, D.B.; Smith, E.E.; Frolkis, A.; Cohen, A.; Kirk, A.; Pearson, D.; Pringsheim, T.; et al. The Prevalence and Incidence of Dementia Due to Alzheimer’s Disease: A Systematic Review and Meta-Analysis. Can. J. Neurol. Sci. 2016, 43 (Suppl. S1), S51–S82. [Google Scholar] [CrossRef] [PubMed]
  12. Määttä, T.; Tervo-Määttä, T.; Taanila, A.; Kaski, M.; Iivanainen, M. Mental Health, Behaviour and Intellectual Abilities of People with Down Syndrome. Down Syndr. Res. Pract. 2006, 11, 37–43. [Google Scholar] [CrossRef] [PubMed]
  13. de Miguel-Díez, J.; Villa-Asensi, J.R.; Alvarez-Sala, J.L. Prevalence of Sleep-Disordered Breathing in Children with Down Syndrome: Polygraphic Findings in 108 Children. Sleep 2003, 26, 1006–1009. [Google Scholar] [CrossRef]
  14. Chahrour, M.; Zoghbi, H.Y. The Story of Rett Syndrome: From Clinic to Neurobiology. Neuron 2007, 56, 422–437. [Google Scholar] [CrossRef]
  15. Rett, A. On a unusual brain atrophy syndrome in hyperammonemia in childhood. Wien Med. Wochenschr. 1966, 116, 723–726. [Google Scholar]
  16. Haas, R.H. The History and Challenge of Rett Syndrome. J. Child. Neurol. 1988, 3 (Suppl. S1), S3–S5. [Google Scholar] [CrossRef]
  17. Renieri, A.; Meloni, I.; Longo, I.; Ariani, F.; Mari, F.; Pescucci, C.; Cambi, F. Rett Syndrome: The Complex Nature of a Monogenic Disease. J. Mol. Med. (Berl.) 2003, 81, 346–354. [Google Scholar] [CrossRef]
  18. Segatto, M.; Trapani, L.; Di Tunno, I.; Sticozzi, C.; Valacchi, G.; Hayek, J.; Pallottini, V. Cholesterol Metabolism Is Altered in Rett Syndrome: A Study on Plasma and Primary Cultured Fibroblasts Derived from Patients. PLoS ONE 2014, 9, e104834. [Google Scholar] [CrossRef]
  19. Reichow, B.; George-Puskar, A.; Lutz, T.; Smith, I.C.; Volkmar, F.R. Brief Report: Systematic Review of Rett Syndrome in Males. J. Autism Dev. Disord. 2015, 45, 3377–3383. [Google Scholar] [CrossRef]
  20. Kazdoba, T.M.; Leach, P.T.; Silverman, J.L.; Crawley, J.N. Modeling Fragile X Syndrome in the Fmr1 Knockout Mouse. Intractable Rare Dis. Res. 2014, 3, 118–133. [Google Scholar] [CrossRef]
  21. Dahlhaus, R. Of Men and Mice: Modeling the Fragile X Syndrome. Front. Mol. Neurosci. 2018, 11, 41. [Google Scholar] [CrossRef] [PubMed]
  22. Maurin, T.; Zongaro, S.; Bardoni, B. Fragile X Syndrome: From Molecular Pathology to Therapy. Neurosci. Biobehav. Rev. 2014, 46, 242–255. [Google Scholar] [CrossRef] [PubMed]
  23. Reiss, A.L.; Freund, L. Fragile X Syndrome, DSM-HI-R, and Autism. J. Am. Acad. Child Adolesc. Psychiatry 1990, 29, 885–891. [Google Scholar] [CrossRef] [PubMed]
  24. Ryan, M.M.; Lockstone, H.E.; Huffaker, S.J.; Wayland, M.T.; Webster, M.J.; Bahn, S. Gene Expression Analysis of Bipolar Disorder Reveals Downregulation of the Ubiquitin Cycle and Alterations in Synaptic Genes. Mol. Psychiatry 2006, 11, 965–978. [Google Scholar] [CrossRef] [PubMed]
  25. Chen, D.T.; Jiang, X.; Akula, N.; Shugart, Y.Y.; Wendland, J.R.; Steele, C.J.M.; Kassem, L.; Park, J.-H.; Chatterjee, N.; Jamain, S.; et al. Genome-Wide Association Study Meta-Analysis of European and Asian-Ancestry Samples Identifies Three Novel Loci Associated with Bipolar Disorder. Mol. Psychiatry 2013, 18, 195–205. [Google Scholar] [CrossRef] [PubMed]
  26. Liu, H.-C.; Yang, Y.-Y.; Chou, Y.-M.; Chen, K.-P.; Shen, W.W.; Leu, S.-J. Immunologic Variables in Acute Mania of Bipolar Disorder. J. Neuroimmunol. 2004, 150, 116–122. [Google Scholar] [CrossRef]
  27. Slyepchenko, A.; Frey, B.N.; Lafer, B.; Nierenberg, A.A.; Sachs, G.S.; Dias, R.S. Increased Illness Burden in Women with Comorbid Bipolar and Premenstrual Dysphoric Disorder: Data from 1 099 Women from STEP-BD Study. Acta Psychiatr. Scand. 2017, 136, 473–482. [Google Scholar] [CrossRef]
  28. Diflorio, A.; Jones, I. Is Sex Important? Gender Differences in Bipolar Disorder. Int. Rev. Psychiatry 2010, 22, 437–452. [Google Scholar] [CrossRef]
  29. Saunders, E.F.H.; Nazir, R.; Kamali, M.; Ryan, K.A.; Evans, S.; Langenecker, S.; Gelenberg, A.J.; McInnis, M.G. Gender Differences, Clinical Correlates, and Longitudinal Outcome of Bipolar Disorder with Comorbid Migraine. J. Clin. Psychiatry 2014, 75, 7630. [Google Scholar] [CrossRef]
  30. Nivoli, A.M.A.; Pacchiarotti, I.; Rosa, A.R.; Popovic, D.; Murru, A.; Valenti, M.; Bonnin, C.M.; Grande, I.; Sanchez-Moreno, J.; Vieta, E.; et al. Gender Differences in a Cohort Study of 604 Bipolar Patients: The Role of Predominant Polarity. J. Affect. Disord. 2011, 133, 443–449. [Google Scholar] [CrossRef]
  31. Clements, C.; Morriss, R.; Jones, S.; Peters, S.; Roberts, C.; Kapur, N. Suicide in Bipolar Disorder in a National English Sample, 1996–2009: Frequency, Trends and Characteristics. Psychol. Med. 2013, 43, 2593–2602. [Google Scholar] [CrossRef]
  32. Christensen, J.; Kjeldsen, M.J.; Andersen, H.; Friis, M.L.; Sidenius, P. Gender Differences in Epilepsy. Epilepsia 2005, 46, 956–960. [Google Scholar] [CrossRef] [PubMed]
  33. Christensen, J.; Vestergaard, M.; Pedersen, M.G.; Pedersen, C.B.; Olsen, J.; Sidenius, P. Incidence and Prevalence of Epilepsy in Denmark. Epilepsy Res. 2007, 76, 60–65. [Google Scholar] [CrossRef] [PubMed]
  34. Christensen, J.; Pedersen, M.G.; Pedersen, C.B.; Sidenius, P.; Olsen, J.; Vestergaard, M. Long-Term Risk of Epilepsy after Traumatic Brain Injury in Children and Young Adults: A Population-Based Cohort Study. Lancet 2009, 373, 1105–1110. [Google Scholar] [CrossRef] [PubMed]
  35. Benn, E.K.T.; Hauser, W.A.; Shih, T.; Leary, L.; Bagiella, E.; Dayan, P.; Green, R.; Andrews, H.; Thurman, D.J.; Hesdorffer, D.C. Estimating the Incidence of First Unprovoked Seizure and Newly Diagnosed Epilepsy in the Low-Income Urban Community of Northern Manhattan, New York City. Epilepsia 2008, 49, 1431–1439. [Google Scholar] [CrossRef]
  36. Seeman, M.V. Menstrual Exacerbation of Schizophrenia Symptoms. Acta Psychiatr. Scand. 2012, 125, 363–371. [Google Scholar] [CrossRef]
  37. Riecher-Rössler, A.; Butler, S.; Kulkarni, J. Sex and Gender Differences in Schizophrenic Psychoses—A Critical Review. Arch. Womens Ment. Health 2018, 21, 627–648. [Google Scholar] [CrossRef] [PubMed]
  38. Vollenweider, F.X. Advances and Pathophysiological Models of Hallucinogenic Drug Actions in Humans: A Preamble to Schizophrenia Research. Pharmacopsychiatry 1998, 31, 92–103. [Google Scholar] [CrossRef]
  39. Javitt, D.C.; Zukin, S.R. Recent Advances in the Phencyclidine Model of Schizophrenia. Am. J. Psychiatry 1991, 148, 1301–1308. [Google Scholar] [CrossRef]
  40. Tamminga CA Gender and Schizophrenia. J. Clin. Psychiatry 1997, 58 (Suppl. S15), 33–37.
  41. Mendrek, A.; Mancini-Marïe, A. Sex/Gender Differences in the Brain and Cognition in Schizophrenia. Neurosci. Biobehav. Rev. 2016, 67, 57–78. [Google Scholar] [CrossRef]
  42. Morgan, C.; Fisher, H. Environment and Schizophrenia: Environmental Factors in Schizophrenia: Childhood Trauma—A Critical Review. Schizophr. Bull. 2007, 33, 3–10. [Google Scholar] [CrossRef]
  43. Chang, Y.-T.; Chen, P.-C.; Tsai, I.-J.; Sung, F.-C.; Chin, Z.-N.; Kuo, H.-T.; Tsai, C.-H.; Chou, I.-C. Bidirectional Relation between Schizophrenia and Epilepsy: A Population-Based Retrospective Cohort Study. Epilepsia 2011, 52, 2036–2042. [Google Scholar] [CrossRef] [PubMed]
  44. Sommer, I.E.; Tiihonen, J.; van Mourik, A.; Tanskanen, A.; Taipale, H. The Clinical Course of Schizophrenia in Women and Men—A Nation-Wide Cohort Study. NPJ Schizophr. 2020, 6, 12. [Google Scholar] [CrossRef] [PubMed]
  45. Slavich, G.M.; Sacher, J. Stress, Sex Hormones, Inflammation, and Major Depressive Disorder: Extending Social Signal Transduction Theory of Depression to Account for Sex Differences in Mood Disorders. Psychopharmacology 2019, 236, 3063–3079. [Google Scholar] [CrossRef] [PubMed]
  46. Fries, G.R.; Saldana, V.A.; Finnstein, J.; Rein, T. Molecular Pathways of Major Depressive Disorder Converge on the Synapse. Mol. Psychiatry 2023, 28, 284–297. [Google Scholar] [CrossRef]
  47. Felmingham, K.L.; Bryant, R.A. Gender Differences in the Maintenance of Response to Cognitive Behavior Therapy for Posttraumatic Stress Disorder. J. Consult. Clin. Psychol. 2012, 80, 196–200. [Google Scholar] [CrossRef]
  48. Lind, M.J.; Marraccini, M.E.; Sheerin, C.M.; Bountress, K.; Bacanu, S.-A.; Amstadter, A.B.; Nugent, N.R. Association of Posttraumatic Stress Disorder with Rs2267735 in the ADCYAP1R1 Gene: A Meta-Analysis. J. Trauma. Stress 2017, 30, 389–398. [Google Scholar] [CrossRef]
  49. Otte, C.; Gold, S.M.; Penninx, B.W.; Pariante, C.M.; Etkin, A.; Fava, M.; Mohr, D.C.; Schatzberg, A.F. Major Depressive Disorder. Nat. Rev. Dis. Primers 2016, 2, 16065. [Google Scholar] [CrossRef]
  50. Lai, W.W.; Oei, T.P.S. Coping in Parents and Caregivers of Children with Autism Spectrum Disorders (ASD): A Review. Rev. J. Autism Dev. Disord. 2014, 1, 207–224. [Google Scholar] [CrossRef]
  51. Christensen, J.; Overgaard, M.; Parner, E.T.; Vestergaard, M.; Schendel, D. Risk of Epilepsy and Autism in Full and Half Siblings—A Population-Based Cohort Study. Epilepsia 2016, 57, 2011–2018. [Google Scholar] [CrossRef] [PubMed]
  52. Prosperi, M.; Turi, M.; Guerrera, S.; Napoli, E.; Tancredi, R.; Igliozzi, R.; Apicella, F.; Valeri, G.; Lattarulo, C.; Gemma, A.; et al. Sex Differences in Autism Spectrum Disorder: An Investigation on Core Symptoms and Psychiatric Comorbidity in Preschoolers. Front. Integr. Neurosci. 2021, 14, 594082. [Google Scholar] [CrossRef] [PubMed]
  53. de Giambattista, C.; Ventura, P.; Trerotoli, P.; Margari, F.; Margari, L. Sex Differences in Autism Spectrum Disorder: Focus on High Functioning Children and Adolescents. Front. Psychiatry 2021, 12, 539835. [Google Scholar] [CrossRef] [PubMed]
  54. Romano, E.; Cosentino, L.; Laviola, G.; De Filippis, B. Genes and Sex Hormones Interaction in Neurodevelopmental Disorders. Neurosci. Biobehav. Rev. 2016, 67, 9–24. [Google Scholar] [CrossRef] [PubMed]
  55. Supekar, K.; Menon, V. Sex Differences in Structural Organization of Motor Systems and Their Dissociable Links with Repetitive/Restricted Behaviors in Children with Autism. Mol. Autism 2015, 6, 50. [Google Scholar] [CrossRef]
  56. Mottron, L.; Duret, P.; Mueller, S.; Moore, R.D.; Forgeot d’Arc, B.; Jacquemont, S.; Xiong, L. Sex Differences in Brain Plasticity: A New Hypothesis for Sex Ratio Bias in Autism. Mol. Autism 2015, 6, 33. [Google Scholar] [CrossRef]
  57. Baruah, A.; Singla, K.; Narwat, P.; Das, N.; Chapadgaonkar, S.S. Gender Bias in Autism Spectrum Disorders—A Review. J. Clin. Diagn. Res. 2018, 12, 1–7. [Google Scholar] [CrossRef]
  58. Hu, V.W.; Sarachana, T.; Sherrard, R.M.; Kocher, K.M. Investigation of Sex Differences in the Expression of RORA and Its Transcriptional Targets in the Brain as a Potential Contributor to the Sex Bias in Autism. Mol. Autism 2015, 6, 7. [Google Scholar] [CrossRef]
  59. Carayol, J.; Sacco, R.; Tores, F.; Rousseau, F.; Lewin, P.; Hager, J.; Persico, A.M. Converging Evidence for an Association of ATP2B2 Allelic Variants with Autism in Male Subjects. Biol. Psychiatry 2011, 70, 880–887. [Google Scholar] [CrossRef]
  60. Tropeano, M.; Ahn, J.W.; Dobson, R.J.B.; Breen, G.; Rucker, J.; Dixit, A.; Pal, D.K.; McGuffin, P.; Farmer, A.; White, P.S.; et al. Male-Biased Autosomal Effect of 16p13.11 Copy Number Variation in Neurodevelopmental Disorders. PLoS ONE 2013, 8, e61365. [Google Scholar] [CrossRef]
  61. Steeb, H.; Ramsey, J.M.; Guest, P.C.; Stocki, P.; Cooper, J.D.; Rahmoune, H.; Ingudomnukul, E.; Auyeung, B.; Ruta, L.; Baron-Cohen, S.; et al. Serum Proteomic Analysis Identifies Sex-Specific Differences in Lipid Metabolism and Inflammation Profiles in Adults Diagnosed with Asperger Syndrome. Mol. Autism 2014, 5, 4. [Google Scholar] [CrossRef] [PubMed]
  62. Tropeano, M.; Howley, D.; Gazzellone, M.J.; Wilson, C.E.; Ahn, J.W.; Stavropoulos, D.J.; Murphy, C.M.; Eis, P.S.; Hatchwell, E.; Dobson, R.J.B.; et al. Microduplications at the Pseudoautosomal SHOX Locus in Autism Spectrum Disorders and Related Neurodevelopmental Conditions. J. Med. Genet. 2016, 53, 536–547. [Google Scholar] [CrossRef] [PubMed]
  63. Mitra, I.; Tsang, K.; Ladd-Acosta, C.; Croen, L.A.; Aldinger, K.A.; Hendren, R.L.; Traglia, M.; Lavillaureix, A.; Zaitlen, N.; Oldham, M.C.; et al. Pleiotropic Mechanisms Indicated for Sex Differences in Autism. PLoS Genet. 2016, 12, e1006425. [Google Scholar] [CrossRef] [PubMed]
  64. Ferri, S.L.; Abel, T.; Brodkin, E.S. Sex Differences in Autism Spectrum Disorder: A Review. Curr. Psychiatry Rep. 2018, 20, 9. [Google Scholar] [CrossRef]
  65. Antonarakis, S.E.; Skotko, B.G.; Rafii, M.S.; Strydom, A.; Pape, S.E.; Bianchi, D.W.; Sherman, S.L.; Reeves, R.H. Down Syndrome. Nat. Rev. Dis. Primers 2020, 6, 9. [Google Scholar] [CrossRef] [PubMed]
  66. González-Agüero, A.; Vicente-Rodríguez, G.; Moreno, L.A.; Casajús, J.A. Bone Mass in Male and Female Children and Adolescents with Down Syndrome. Osteoporos Int. 2011, 22, 2151–2157. [Google Scholar] [CrossRef]
  67. Schupf, N.; Winsten, S.; Patel, B.; Pang, D.; Ferin, M.; Zigman, W.B.; Silverman, W.; Mayeux, R. Bioavailable Estradiol and Age at Onset of Alzheimer’s Disease in Postmenopausal Women with Down Syndrome. Neurosci. Lett. 2006, 406, 298–302. [Google Scholar] [CrossRef]
  68. Granholm, A.-C.; Sanders, L.; Seo, H.; Lin, L.; Ford, K.; Isacson, O. Estrogen Alters Amyloid Precursor Protein as Well as Dendritic and Cholinergic Markers in a Mouse Model of Down Syndrome. Hippocampus 2003, 13, 905–914. [Google Scholar] [CrossRef]
  69. Granholm, A.-C.E.; Ford, K.A.; Hyde, L.A.; Bimonte, H.A.; Hunter, C.L.; Nelson, M.; Albeck, D.; Sanders, L.A.; Mufson, E.J.; Crnic, L.S. Estrogen Restores Cognition and Cholinergic Phenotype in an Animal Model of Down Syndrome. Physiol. Behav. 2002, 77, 371–385. [Google Scholar] [CrossRef]
  70. Faizi, M.; Bader, P.L.; Tun, C.; Encarnacion, A.; Kleschevnikov, A.; Belichenko, P.; Saw, N.; Priestley, M.; Tsien, R.W.; Mobley, W.C.; et al. Comprehensive Behavioral Phenotyping of Ts65Dn Mouse Model of Down Syndrome: Activation of Β1-Adrenergic Receptor by Xamoterol as a Potential Cognitive Enhancer. Neurobiol. Dis. 2011, 43, 397–413. [Google Scholar] [CrossRef]
  71. Zampieri, B.L.; Fernandez, F.; Pearson, J.N.; Stasko, M.R.; Costa, A.C.S. Ultrasonic Vocalizations during Male–Female Interaction in the Mouse Model of Down Syndrome Ts65Dn. Physiol. Behav. 2014, 128, 119–125. [Google Scholar] [CrossRef] [PubMed]
  72. Simpkins, J.W.; Perez, E.; Wang, X.; Yang, S.; Wen, Y.; Singh, M. The Potential for Estrogens in Preventing Alzheimer’s Disease and Vascular Dementia. Ther. Adv. Neurol. Disord. 2009, 2, 31–49. [Google Scholar] [CrossRef] [PubMed]
  73. Cosgrave, M.P.; Tyrrell, J.; McCarron, M.; Gill, M.; Lawlor, B.A. Age at Onset of Dementia and Age of Menopause in Women with Down’s Syndrome. J. Intellect. Disabil. Res. 1999, 43, 461–465. [Google Scholar] [CrossRef] [PubMed]
  74. Pitzianti, M.B.; Santamaria Palombo, A.; Esposito, S.; Pasini, A. Rett Syndrome in Males: The Different Clinical Course in Two Brothers with the Same Microduplication MECP2 Xq28. Int. J. Environ. Res. Public Health 2019, 16, 3075. [Google Scholar] [CrossRef]
  75. Takeguchi, R.; Takahashi, S.; Kuroda, M.; Tanaka, R.; Suzuki, N.; Tomonoh, Y.; Ihara, Y.; Sugiyama, N.; Itoh, M. MeCP2_e2 Partially Compensates for Lack of MeCP2_e1: A Male Case of Rett Syndrome. Mol. Genet Genom. Med. 2020, 8, e1088. [Google Scholar] [CrossRef]
  76. Neul, J.L.; Benke, T.A.; Marsh, E.D.; Skinner, S.A.; Merritt, J.; Lieberman, D.N.; Standridge, S.; Feyma, T.; Heydemann, P.; Peters, S.; et al. The Array of Clinical Phenotypes of Males with Mutations in Methyl-CpG Binding Protein 2. Am. J. Med. Genet B Neuropsychiatr. Genet 2019, 180, 55–67. [Google Scholar] [CrossRef]
  77. Hagberg, B.; Hanefeld, F.; Percy, A.; Skjeldal, O. An Update on Clinically Applicable Diagnostic Criteria in Rett Syndrome. Comments to Rett Syndrome Clinical Criteria Consensus Panel Satellite to European Paediatric Neurology Society Meeting, Baden Baden, Germany, 11 September 2001. Eur. J. Paediatr. Neurol. 2002, 6, 293–297. [Google Scholar] [CrossRef]
  78. Mount, R.H.; Hastings, R.P.; Reilly, S.; Cass, H.; Charman, T. Behavioural and Emotional Features in Rett Syndrome. Disabil. Rehabil. 2001, 23, 129–138. [Google Scholar] [CrossRef]
  79. Vashi, N.; Justice, M.J. Treating Rett Syndrome: From Mouse Models to Human Therapies. Mamm. Genome 2019, 30, 90–110. [Google Scholar] [CrossRef]
  80. Kyle, S.M.; Vashi, N.; Justice, M.J. Rett Syndrome: A Neurological Disorder with Metabolic Components. Open Biol. 2018, 8, 170216. [Google Scholar] [CrossRef]
  81. Mouro, F.M.; Miranda-Lourenço, C.; Sebastião, A.M.; Diógenes, M.J. From Cannabinoids and Neurosteroids to Statins and the Ketogenic Diet: New Therapeutic Avenues in Rett Syndrome? Front. Neurosci. 2019, 13, 680. [Google Scholar] [CrossRef] [PubMed]
  82. Good, K.V.; Vincent, J.B.; Ausió, J. MeCP2: The Genetic Driver of Rett Syndrome Epigenetics. Front. Genet 2021, 12, 620859. [Google Scholar] [CrossRef] [PubMed]
  83. Amir, R.E.; Van den Veyver, I.B.; Wan, M.; Tran, C.Q.; Francke, U.; Zoghbi, H.Y. Rett Syndrome Is Caused by Mutations in X-Linked MECP2, Encoding Methyl-CpG-Binding Protein 2. Nat. Genet 1999, 23, 185–188. [Google Scholar] [CrossRef] [PubMed]
  84. Chahrour, M.; Jung, S.Y.; Shaw, C.; Zhou, X.; Wong, S.T.C.; Qin, J.; Zoghbi, H.Y. MeCP2, a Key Contributor to Neurological Disease, Activates and Represses Transcription. Science 2008, 320, 1224–1229. [Google Scholar] [CrossRef]
  85. Kishi, N.; Macklis, J.D. MECP2 Is Progressively Expressed in Post-Migratory Neurons and Is Involved in Neuronal Maturation Rather than Cell Fate Decisions. Mol. Cell. Neurosci. 2004, 27, 306–321. [Google Scholar] [CrossRef]
  86. McGraw, C.M.; Samaco, R.C.; Zoghbi, H.Y. Adult Neural Function Requires MeCP2. Science 2011, 333, 186. [Google Scholar] [CrossRef]
  87. Nguyen, M.V.C.; Du, F.; Felice, C.A.; Shan, X.; Nigam, A.; Mandel, G.; Robinson, J.K.; Ballas, N. MeCP2 Is Critical for Maintaining Mature Neuronal Networks and Global Brain Anatomy during Late Stages of Postnatal Brain Development and in the Mature Adult Brain. J. Neurosci. 2012, 32, 10021–10034. [Google Scholar] [CrossRef]
  88. Giacometti, E.; Luikenhuis, S.; Beard, C.; Jaenisch, R. Partial Rescue of MeCP2 Deficiency by Postnatal Activation of MeCP2. Proc. Natl. Acad. Sci. USA 2007, 104, 1931–1936. [Google Scholar] [CrossRef]
  89. Guy, J.; Gan, J.; Selfridge, J.; Cobb, S.; Bird, A. Reversal of Neurological Defects in a Mouse Model of Rett Syndrome. Science 2007, 315, 1143–1147. [Google Scholar] [CrossRef]
  90. Luikenhuis, S.; Giacometti, E.; Beard, C.F.; Jaenisch, R. Expression of MeCP2 in Postmitotic Neurons Rescues Rett Syndrome in Mice. Proc. Natl. Acad. Sci. USA 2004, 101, 6033–6038. [Google Scholar] [CrossRef]
  91. Kurian, J.R.; Forbes-Lorman, R.M.; Auger, A.P. Sex Difference in Mecp2 Expression during a Critical Period of Rat Brain Development. Epigenetics 2007, 2, 173–178. [Google Scholar] [CrossRef] [PubMed]
  92. Tillotson, R.; Cholewa-Waclaw, J.; Chhatbar, K.; Connelly, J.C.; Kirschner, S.A.; Webb, S.; Koerner, M.V.; Selfridge, J.; Kelly, D.A.; De Sousa, D.; et al. Neuronal Non-CG Methylation Is an Essential Target for MeCP2 Function. Mol. Cell 2021, 81, 1260–1275.e12. [Google Scholar] [CrossRef] [PubMed]
  93. Carrette, L.L.G.; Blum, R.; Ma, W.; Kelleher, R.J.; Lee, J.T. Tsix–Mecp2 Female Mouse Model for Rett Syndrome Reveals That Low-Level MECP2 Expression Extends Life and Improves Neuromotor Function. Proc. Natl. Acad. Sci. USA 2018, 115, 8185–8190. [Google Scholar] [CrossRef] [PubMed]
  94. Minkovsky, A.; Sahakyan, A.; Bonora, G.; Damoiseaux, R.; Dimitrova, E.; Rubbi, L.; Pellegrini, M.; Radu, C.G.; Plath, K. A High-Throughput Screen of Inactive X Chromosome Reactivation Identifies the Enhancement of DNA Demethylation by 5-Aza-2′-DC upon Inhibition of Ribonucleotide Reductase. Epigenetics Chromatin 2015, 8, 42. [Google Scholar] [CrossRef]
  95. Pieretti, M.; Zhang, F.; Fu, Y.-H.; Warren, S.T.; Oostra, B.A.; Caskey, C.T.; Nelson, D.L. Absence of Expression of the FMR-1 Gene in Fragile X Syndrome. Cell 1991, 66, 817–822. [Google Scholar] [CrossRef]
  96. Verkerk, A.J.M.H.; Pieretti, M.; Sutcliffe, J.S.; Fu, Y.-H.; Kuhl, D.P.A.; Pizzuti, A.; Reiner, O.; Richards, S.; Victoria, M.F.; Zhang, F.; et al. Identification of a Gene (FMR-1) Containing a CGG Repeat Coincident with a Breakpoint Cluster Region Exhibiting Length Variation in Fragile X Syndrome. Cell 1991, 65, 905–914. [Google Scholar] [CrossRef]
  97. Maurin, T.; Bardoni, B. Fragile X Mental Retardation Protein: To Be or Not to Be a Translational Enhancer. Front. Mol. Biosci. 2018, 5, 113. [Google Scholar] [CrossRef]
  98. Richter, J.D.; Zhao, X. The Molecular Biology of FMRP: New Insights into Fragile X Syndrome. Nat. Rev. Neurosci. 2021, 22, 209–222. [Google Scholar] [CrossRef]
  99. Bach, S.; Shovlin, S.; Moriarty, M.; Bardoni, B.; Tropea, D. Rett Syndrome and Fragile X Syndrome: Different Etiology with Common Molecular Dysfunctions. Front. Cell. Neurosci. 2021, 15, 764761. [Google Scholar] [CrossRef]
  100. Budimirovic, D.B.; Schlageter, A.; Filipovic-Sadic, S.; Protic, D.D.; Bram, E.; Mahone, E.M.; Nicholson, K.; Culp, K.; Javanmardi, K.; Kemppainen, J.; et al. A Genotype-Phenotype Study of High-Resolution FMR1 Nucleic Acid and Protein Analyses in Fragile X Patients with Neurobehavioral Assessments. Brain Sci. 2020, 10, 694. [Google Scholar] [CrossRef]
  101. Tassone, F.; Hagerman, R.J.; Iklé, D.N.; Dyer, P.N.; Lampe, M.; Willemsen, R.; Oostra, B.A.; Taylor, A.K. FMRP Expression as a Potential Prognostic Indicator in Fragile X Syndrome. Am. J. Med. Genet. 1999, 84, 250–261. [Google Scholar] [CrossRef]
  102. Bartholomay, K.L.; Lee, C.H.; Bruno, J.L.; Lightbody, A.A.; Reiss, A.L. Closing the Gender Gap in Fragile X Syndrome: Review of Females with Fragile X Syndrome and Preliminary Research Findings. Brain Sci. 2019, 9, 11. [Google Scholar] [CrossRef] [PubMed]
  103. Erickson, C.A.; Kaufmann, W.E.; Budimirovic, D.B.; Lachiewicz, A.; Haas-Givler, B.; Miller, R.M.; Weber, J.D.; Abbeduto, L.; Hessl, D.; Hagerman, R.J.; et al. Best Practices in Fragile X Syndrome Treatment Development. Brain Sci. 2018, 8, 224. [Google Scholar] [CrossRef] [PubMed]
  104. Gallagher, A.; Hallahan, B. Fragile X-Associated Disorders: A Clinical Overview. J. Neurol. 2012, 259, 401–413. [Google Scholar] [CrossRef] [PubMed]
  105. Loesch, D.Z.; Huggins, R.M.; Hagerman, R.J. Phenotypic Variation and FMRP Levels in Fragile X. Ment. Retard. Dev. Disabil. Res. Rev. 2004, 10, 31–41. [Google Scholar] [CrossRef] [PubMed]
  106. Schaefer, T.L.; Davenport, M.H.; Erickson, C.A. Emerging Pharmacologic Treatment Options for Fragile X Syndrome. Appl. Clin. Genet. 2015, 8, 75–93. [Google Scholar] [CrossRef] [PubMed]
  107. Yang, Q.; Yang, L.; Zhang, K.; Guo, Y.-Y.; Liu, S.-B.; Wu, Y.-M.; Li, X.-Q.; Song, Q.; Zhuo, M.; Zhao, M.-G. Increased Coupling of Caveolin-1 and Estrogen Receptor α Contributes to the Fragile X Syndrome. Ann. Neurol. 2015, 77, 618–636. [Google Scholar] [CrossRef]
  108. Mailick, M.R.; Movaghar, A.; Hong, J.; Greenberg, J.S.; DaWalt, L.S.; Zhou, L.; Jackson, J.; Rathouz, P.J.; Baker, M.W.; Brilliant, M.; et al. Health Profiles of Mosaic Versus Non-Mosaic FMR1 Premutation Carrier Mothers of Children with Fragile X Syndrome. Front. Genet. 2018, 9, 173. [Google Scholar] [CrossRef]
  109. Rinehart, N.J.; Cornish, K.M.; Tonge, B.J. Gender Differences in Neurodevelopmental Disorders: Autism and Fragile X Syndrome. In Biological Basis of Sex Differences in Psychopharmacology; Neill, J.C., Kulkarni, J., Eds.; Current Topics in Behavioral Neurosciences; Springer: Berlin/Heidelberg, Germany, 2011; pp. 209–229. ISBN 978-3-642-20006-9. [Google Scholar]
  110. Kulkarni, P.; Sevilimedu, A. The Known Unknowns: Missing Pieces in in Vivo Models of Fragile X Syndrome. J. Rare Dis. Res. Treat. 2020, 5, 1–9. [Google Scholar] [CrossRef]
  111. The Dutch-Belgian Fragile X Consorthium; Bakker, C.E.; Verheij, C.; Willemsen, R.; van der Helm, R.; Oerlemans, F.; Vermey, M.; Bygrave, A.; Hoogeveen, A.; Oostra, B.A.; et al. Fmr1 Knockout Mice: A Model to Study Fragile X Mental Retardation. Cell 1994, 78, 23–33. [Google Scholar] [CrossRef]
  112. Mientjes, E.J.; Nieuwenhuizen, I.; Kirkpatrick, L.; Zu, T.; Hoogeveen-Westerveld, M.; Severijnen, L.; Rifé, M.; Willemsen, R.; Nelson, D.L.; Oostra, B.A. The Generation of a Conditional Fmr1 Knock out Mouse Model to Study Fmrp Function in Vivo. Neurobiol. Dis. 2006, 21, 549–555. [Google Scholar] [CrossRef] [PubMed]
  113. Baker, S.; Hooper, S.; Skinner, M.; Hatton, D.; Schaaf, J.; Ornstein, P.; Bailey, D. Working Memory Subsystems and Task Complexity in Young Boys with Fragile X Syndrome. J. Intellect. Disabil. Res. 2011, 55, 19–29. [Google Scholar] [CrossRef] [PubMed]
  114. Ding, Q.; Sethna, F.; Wang, H. Behavioral Analysis of Male and Female Fmr1 Knockout Mice on C57BL/6 Background. Behav. Brain Res. 2014, 271, 72–78. [Google Scholar] [CrossRef] [PubMed]
  115. Qin, M.; Entezam, A.; Usdin, K.; Huang, T.; Liu, Z.-H.; Hoffman, G.E.; Smith, C.B. A Mouse Model of the Fragile X Premutation: Effects on Behavior, Dendrite Morphology, and Regional Rates of Cerebral Protein Synthesis. Neurobiol. Dis. 2011, 42, 85–98. [Google Scholar] [CrossRef] [PubMed]
  116. Wong, H.; Hooper, A.W.M.; Niibori, Y.; Lee, S.J.; Hategan, L.A.; Zhang, L.; Karumuthil-Melethil, S.; Till, S.M.; Kind, P.C.; Danos, O.; et al. Sexually Dimorphic Patterns in Electroencephalography Power Spectrum and Autism-Related Behaviors in a Rat Model of Fragile X Syndrome. Neurobiol. Dis. 2020, 146, 105118. [Google Scholar] [CrossRef]
  117. Nolan, S.O.; Reynolds, C.D.; Smith, G.D.; Holley, A.J.; Escobar, B.; Chandler, M.A.; Volquardsen, M.; Jefferson, T.; Pandian, A.; Smith, T.; et al. Deletion of Fmr1 Results in Sex-Specific Changes in Behavior. Brain Behav. 2017, 7, e00800. [Google Scholar] [CrossRef] [PubMed]
  118. Rusconi, L.; Salvatoni, L.; Giudici, L.; Bertani, I.; Kilstrup-Nielsen, C.; Broccoli, V.; Landsberger, N. CDKL5 Expression Is Modulated during Neuronal Development and Its Subcellular Distribution Is Tightly Regulated by the C-Terminal Tail. J. Biol. Chem. 2008, 283, 30101–30111. [Google Scholar] [CrossRef]
  119. Kilstrup-Nielsen, C.; Rusconi, L.; La Montanara, P.; Ciceri, D.; Bergo, A.; Bedogni, F.; Landsberger, N. What We Know and Would Like to Know about CDKL5 and Its Involvement in Epileptic Encephalopathy. Neural Plast. 2012, 2012, e728267. [Google Scholar] [CrossRef]
  120. Ricciardi, S.; Ungaro, F.; Hambrock, M.; Rademacher, N.; Stefanelli, G.; Brambilla, D.; Sessa, A.; Magagnotti, C.; Bachi, A.; Giarda, E.; et al. CDKL5 Ensures Excitatory Synapse Stability by Reinforcing NGL-1–PSD95 Interaction in the Postsynaptic Compartment and Is Impaired in Patient IPSC-Derived Neurons. Nat. Cell Biol. 2012, 14, 911–923. [Google Scholar] [CrossRef]
  121. Valli, E.; Trazzi, S.; Fuchs, C.; Erriquez, D.; Bartesaghi, R.; Perini, G.; Ciani, E. CDKL5, a Novel MYCN-Repressed Gene, Blocks Cell Cycle and Promotes Differentiation of Neuronal Cells. Biochim. Et Biophys. Acta (BBA)—Gene Regul. Mech. 2012, 1819, 1173–1185. [Google Scholar] [CrossRef]
  122. Van Bergen, N.J.; Massey, S.; Quigley, A.; Rollo, B.; Harris, A.R.; Kapsa, R.M.I.; Christodoulou, J. CDKL5 Deficiency Disorder: Molecular Insights and Mechanisms of Pathogenicity to Fast-Track Therapeutic Development. Biochem. Soc. Trans. 2022, 50, 1207–1224. [Google Scholar] [CrossRef]
  123. Chen, Q.; Zhu, Y.-C.; Yu, J.; Miao, S.; Zheng, J.; Xu, L.; Zhou, Y.; Li, D.; Zhang, C.; Tao, J.; et al. CDKL5, a Protein Associated with Rett Syndrome, Regulates Neuronal Morphogenesis via Rac1 Signaling. J. Neurosci. 2010, 30, 12777–12786. [Google Scholar] [CrossRef] [PubMed]
  124. Barbiero, I.; Peroni, D.; Tramarin, M.; Chandola, C.; Rusconi, L.; Landsberger, N.; Kilstrup-Nielsen, C. The Neurosteroid Pregnenolone Reverts Microtubule Derangement Induced by the Loss of a Functional CDKL5-IQGAP1 Complex. Hum. Mol. Genet. 2017, 26, 3520–3530. [Google Scholar] [CrossRef] [PubMed]
  125. Fuchs, C.; Trazzi, S.; Torricella, R.; Viggiano, R.; De Franceschi, M.; Amendola, E.; Gross, C.; Calzà, L.; Bartesaghi, R.; Ciani, E. Loss of CDKL5 Impairs Survival and Dendritic Growth of Newborn Neurons by Altering AKT/GSK-3β Signaling. Neurobiol. Dis. 2014, 70, 53–68. [Google Scholar] [CrossRef] [PubMed]
  126. Tang, S.; Wang, I.-T.J.; Yue, C.; Takano, H.; Terzic, B.; Pance, K.; Lee, J.Y.; Cui, Y.; Coulter, D.A.; Zhou, Z. Loss of CDKL5 in Glutamatergic Neurons Disrupts Hippocampal Microcircuitry and Leads to Memory Impairment in Mice. J. Neurosci. 2017, 37, 7420–7437. [Google Scholar] [CrossRef]
  127. Demarest, S.T.; Olson, H.E.; Moss, A.; Pestana-Knight, E.; Zhang, X.; Parikh, S.; Swanson, L.C.; Riley, K.D.; Bazin, G.A.; Angione, K.; et al. CDKL5 Deficiency Disorder: Relationship between Genotype, Epilepsy, Cortical Visual Impairment, and Development. Epilepsia 2019, 60, 1733–1742. [Google Scholar] [CrossRef]
  128. Olson, H.E.; Demarest, S.T.; Pestana-Knight, E.M.; Swanson, L.C.; Iqbal, S.; Lal, D.; Leonard, H.; Cross, J.H.; Devinsky, O.; Benke, T.A. Cyclin-Dependent Kinase-Like 5 Deficiency Disorder: Clinical Review. Pediatr. Neurol. 2019, 97, 18–25. [Google Scholar] [CrossRef]
  129. Fehr, S.; Downs, J.; Ho, G.; de Klerk, N.; Forbes, D.; Christodoulou, J.; Williams, S.; Leonard, H. Functional Abilities in Children and Adults with the CDKL5 Disorder. Am. J. Med. Genet. Part A 2016, 170, 2860–2869. [Google Scholar] [CrossRef]
  130. Amendola, E.; Zhan, Y.; Mattucci, C.; Castroflorio, E.; Calcagno, E.; Fuchs, C.; Lonetti, G.; Silingardi, D.; Vyssotski, A.L.; Farley, D.; et al. Mapping Pathological Phenotypes in a Mouse Model of CDKL5 Disorder. PLoS ONE 2014, 9, e91613. [Google Scholar] [CrossRef]
  131. Wang, I.-T.J.; Allen, M.; Goffin, D.; Zhu, X.; Fairless, A.H.; Brodkin, E.S.; Siegel, S.J.; Marsh, E.D.; Blendy, J.A.; Zhou, Z. Loss of CDKL5 Disrupts Kinome Profile and Event-Related Potentials Leading to Autistic-like Phenotypes in Mice. Proc. Natl. Acad. Sci. USA 2012, 109, 21516–21521. [Google Scholar] [CrossRef]
  132. Okuda, K.; Takao, K.; Watanabe, A.; Miyakawa, T.; Mizuguchi, M.; Tanaka, T. Comprehensive Behavioral Analysis of the Cdkl5 Knockout Mice Revealed Significant Enhancement in Anxiety- and Fear-Related Behaviors and Impairment in Both Acquisition and Long-Term Retention of Spatial Reference Memory. PLoS ONE 2018, 13, e0196587. [Google Scholar] [CrossRef] [PubMed]
  133. Miller, T.H. Bipolar Disorder. Prim. Care Clin. Off. Pract. 2016, 43, 269–284. [Google Scholar] [CrossRef] [PubMed]
  134. Grande, I.; Berk, M.; Birmaher, B.; Vieta, E. Bipolar Disorder. Lancet 2016, 387, 1561–1572. [Google Scholar] [CrossRef] [PubMed]
  135. Van Rheenen, T.E.; Lewandowski, K.E.; Bauer, I.E.; Kapczinski, F.; Miskowiak, K.; Burdick, K.E.; Balanzá-Martínez, V. Current Understandings of the Trajectory and Emerging Correlates of Cognitive Impairment in Bipolar Disorder: An Overview of Evidence. Bipolar Disord. 2020, 22, 13–27. [Google Scholar] [CrossRef] [PubMed]
  136. Birmaher, B.; Axelson, D.; Goldstein, B.; Strober, M.; Gill, M.K.; Hunt, J.; Houck, P.; Ha, W.; Iyengar, S.; Kim, E.; et al. Four-Year Longitudinal Course of Children and Adolescents with Bipolar Spectrum Disorders: The Course and Outcome of Bipolar Youth (COBY) Study. Am. J. Psychiatry 2009, 166, 795–804. [Google Scholar] [CrossRef] [PubMed]
  137. Kawa, I.; Carter, J.D.; Joyce, P.R.; Doughty, C.J.; Frampton, C.M.; Elisabeth Wells, J.; Walsh, A.E.; Olds, R.J. Gender Differences in Bipolar Disorder: Age of Onset, Course, Comorbidity, and Symptom Presentation. Bipolar Disord. 2005, 7, 119–125. [Google Scholar] [CrossRef]
  138. Cunningham, R.; Crowe, M.; Stanley, J.; Haitana, T.; Pitama, S.; Porter, R.; Baxter, J.; Huria, T.; Mulder, R.; Clark, M.T.R.; et al. Gender and Mental Health Service Use in Bipolar Disorder: National Cohort Study. BJPsych Open 2020, 6, e138. [Google Scholar] [CrossRef]
  139. Sato, T.; Bottlender, R.; Schröter, A.; Möller, H.-J. Frequency of Manic Symptoms during a Depressive Episode and Unipolar ‘Depressive Mixed State’ as Bipolar Spectrum. Acta Psychiatr. Scand. 2003, 107, 268–274. [Google Scholar] [CrossRef]
  140. Xu, X.; Zhang, C.; Denton, D.T.; O’Connell, D.; Drolet, D.W.; Geisbrecht, B.V. Inhibition of the Complement Alternative Pathway by Chemically Modified DNA Aptamers That Bind with Picomolar Affinity to Factor B. J. Immunol. 2021, 206, 861–873. [Google Scholar] [CrossRef]
  141. Solé, B.; Varo, C.; Torrent, C.; Montejo, L.; Jiménez, E.; Bonnin, C. del M.; Clougher, D.; Verdolini, N.; Amoretti, S.; Piazza, F.; et al. Sex Differences in Neurocognitive and Psychosocial Functioning in Bipolar Disorder. J. Affect. Disord. 2022, 296, 208–215. [Google Scholar] [CrossRef]
  142. Gogos, A.; Son, J.; Rossell, S.L.; Karantonis, J.; Furlong, L.S.; Felmingham, K.; Rheenen, T.E.V. Characterisation of Deficits and Sex Differences in Verbal and Visual Memory/Learning in Bipolar Disorder. J. Int. Neuropsychol. Soc. 2023, 29, 12–23. [Google Scholar] [CrossRef] [PubMed]
  143. Gogos, A.; Joshua, N.; Rossell, S.L. Use of the Repeatable Battery for the Assessment of Neuropsychological Status (RBANS) to Investigate Group and Gender Differences in Schizophrenia and Bipolar Disorder. Aust. N. Z. J. Psychiatry 2010, 44, 220–229. [Google Scholar] [CrossRef] [PubMed]
  144. Carrus, D.; Christodoulou, T.; Hadjulis, M.; Haldane, M.; Galea, A.; Koukopoulos, A.; Kumari, V.; Frangou, S. Gender Differences in Immediate Memory in Bipolar Disorder. Psychol. Med. 2010, 40, 1349–1355. [Google Scholar] [CrossRef]
  145. Gogos, A.; van den Buuse, M.; Rossell, S. Gender Differences in Prepulse Inhibition (PPI) in Bipolar Disorder: Men Have Reduced PPI, Women Have Increased PPI. Int. J. Neuropsychopharmacol. 2009, 12, 1249–1259. [Google Scholar] [CrossRef]
  146. Bücker, J.; Popuri, S.; Muralidharan, K.; Kozicky, J.-M.; Baitz, H.A.; Honer, W.G.; Torres, I.J.; Yatham, L.N. Sex Differences in Cognitive Functioning in Patients with Bipolar Disorder Who Recently Recovered from a First Episode of Mania: Data from the Systematic Treatment Optimization Program for Early Mania (STOP-EM). J. Affect. Disord. 2014, 155, 162–168. [Google Scholar] [CrossRef] [PubMed]
  147. Fellendorf, F.T.; Kainzbauer, N.; Platzer, M.; Dalkner, N.; Bengesser, S.A.; Birner, A.; Queissner, R.; Rauch, P.; Hamm, C.; Pilz, R.; et al. Gender Differences in the Association between Physical Activity and Cognitive Function in Individuals with Bipolar Disorder. J. Affect. Disord. 2017, 221, 232–237. [Google Scholar] [CrossRef]
  148. Shi, J.; Guo, H.; Fan, F.; Fan, H.; An, H.; Wang, Z.; Tan, S.; Yang, F.; Tan, Y. Sex Differences of Hippocampal Structure in Bipolar Disorder. Psychiatry Res. Neuroimaging 2018, 273, 35–41. [Google Scholar] [CrossRef]
  149. Mitchell, R.H.; Metcalfe, A.W.; Islam, A.H.; Toma, S.; Patel, R.; Fiksenbaum, L.; Korczak, D.; MacIntosh, B.J.; Goldstein, B.I. Sex Differences in Brain Structure among Adolescents with Bipolar Disorder. Bipolar Disord. 2018, 20, 448–458. [Google Scholar] [CrossRef]
  150. Frey, B.N.; Dias, R.S. Sex Hormones and Biomarkers of Neuroprotection and Neurodegeneration: Implications for Female Reproductive Events in Bipolar Disorder. Bipolar Disord. 2014, 16, 48–57. [Google Scholar] [CrossRef]
  151. Gogos, A.; Ney, L.J.; Seymour, N.; Van Rheenen, T.E.; Felmingham, K.L. Sex Differences in Schizophrenia, Bipolar Disorder, and Post-Traumatic Stress Disorder: Are Gonadal Hormones the Link? British J. Pharmacol. 2019, 176, 4119–4135. [Google Scholar] [CrossRef]
  152. Rainville, J.R.; Hodes, G.E. Inflaming Sex Differences in Mood Disorders. Neuropsychopharmacology 2019, 44, 184–199. [Google Scholar] [CrossRef] [PubMed]
  153. Kulkarni, J.; Berk, M.; Wang, W.; Mu, L.; Scarr, E.; Van Rheenen, T.E.; Worsley, R.; Gurvich, C.; Gavrilidis, E.; de Castella, A.; et al. A Four Week Randomised Control Trial of Adjunctive Medroxyprogesterone and Tamoxifen in Women with Mania. Psychoneuroendocrinology 2014, 43, 52–61. [Google Scholar] [CrossRef] [PubMed]
  154. Meinhard, N.; Kessing, L.V.; Vinberg, M. The Role of Estrogen in Bipolar Disorder, a Review. Nord. J. Psychiatry 2014, 68, 81–87. [Google Scholar] [CrossRef] [PubMed]
  155. Rosenblat, J.D.; McIntyre, R.S. Bipolar Disorder and Immune Dysfunction: Epidemiological Findings, Proposed Pathophysiology and Clinical Implications. Brain Sci. 2017, 7, 144. [Google Scholar] [CrossRef]
  156. O’Brien, S.M.; Scully, P.; Scott, L.V.; Dinan, T.G. Cytokine Profiles in Bipolar Affective Disorder: Focus on Acutely Ill Patients. J. Affect. Disord. 2006, 90, 263–267. [Google Scholar] [CrossRef]
  157. Ortiz-Domínguez, A.; Hernández, M.E.; Berlanga, C.; Gutiérrez-Mora, D.; Moreno, J.; Heinze, G.; Pavón, L. Immune Variations in Bipolar Disorder: Phasic Differences. Bipolar Disord. 2007, 9, 596–602. [Google Scholar] [CrossRef]
  158. Keshavarzi, A.; Eftekharian, M.M.; Komaki, A.; Omrani, M.D.; Kholghi Oskooei, V.; Taheri, M.; Ghafouri-Fard, S. Sexual Dimorphism in Up-Regulation of Suppressors of Cytokine Signaling Genes in Patients with Bipolar Disorder. BMC Psychiatry 2019, 19, 402. [Google Scholar] [CrossRef]
  159. Fusar-Poli, L.; Amerio, A.; Cimpoesu, P.; Grimaldi Filioli, P.; Natale, A.; Zappa, G.; Aguglia, E.; Amore, M.; Serafini, G.; Aguglia, A. Gender Differences in Complete Blood Count and Inflammatory Ratios among Patients with Bipolar Disorder. Brain Sci. 2021, 11, 363. [Google Scholar] [CrossRef]
  160. Thijs, R.D.; Surges, R.; O’Brien, T.J.; Sander, J.W. Epilepsy in Adults. Lancet 2019, 393, 689–701. [Google Scholar] [CrossRef]
  161. Perucca, P.; Camfield, P.; Camfield, C. Does Gender Influence Susceptibility and Consequences of Acquired Epilepsies? Neurobiol. Dis. 2014, 72, 125–130. [Google Scholar] [CrossRef]
  162. Herzog, A.G. Hormonal Therapies: Progesterone. Neurotherapeutics 2009, 6, 383–391. [Google Scholar] [CrossRef] [PubMed]
  163. Kotsopoulos, I.A.W.; de Krom, M.C.T.F.M.; Kessels, F.G.H.; Lodder, J.; Troost, J.; Twellaar, M.; van Merode, T.; Knottnerus, A.J. The Diagnosis of Epileptic and Non-Epileptic Seizures. Epilepsy Res. 2003, 57, 59–67. [Google Scholar] [CrossRef] [PubMed]
  164. Reddy, D.S.; Thompson, W.; Calderara, G. Molecular Mechanisms of Sex Differences in Epilepsy and Seizure Susceptibility in Chemical, Genetic and Acquired Epileptogenesis. Neurosci. Lett. 2021, 750, 135753. [Google Scholar] [CrossRef] [PubMed]
  165. Savic, I. Sex Differences in Human Epilepsy. Exp. Neurol. 2014, 259, 38–43. [Google Scholar] [CrossRef]
  166. Janszky, J.; Schulz, R.; Janszky, I.; Ebner, A. Medial Temporal Lobe Epilepsy: Gender Differences. J. Neurol. Neurosurg. Psychiatry 2004, 75, 773–775. [Google Scholar] [CrossRef]
  167. Reddy, D.S.; Rogawski, M.A. Neurosteroids as Endogenous Regulators of Seizure Susceptibility and Use in the Treatment of Epilepsy. Epilepsia 2010, 51, 84. [Google Scholar] [CrossRef]
  168. Reddy, D.S. Pharmacology of Endogenous Neuroactive Steroids. Crit. Rev. Neurobiol. 2003, 15, 38. [Google Scholar] [CrossRef]
  169. Reddy, D.S. Neurosteroids and Their Role in Sex-Specific Epilepsies. Neurobiol. Dis. 2014, 72, 198–209. [Google Scholar] [CrossRef]
  170. Giatti, S.; Garcia-Segura, L.M.; Barreto, G.E.; Melcangi, R.C. Neuroactive Steroids, Neurosteroidogenesis and Sex. Prog. Neurobiol. 2019, 176, 1–17. [Google Scholar] [CrossRef]
  171. Rogawski, M.A. The Intrinsic Severity Hypothesis of Pharmacoresistance to Antiepileptic Drugs. Epilepsia 2013, 54 (Suppl. S2), 33–40. [Google Scholar] [CrossRef]
  172. Reddy, D.S.; Matthew Carver, C.; Clossen, B.; Wu, X. Extrasynaptic GABA-A Receptor-Mediated Sex Differences in the Antiseizure Activity of Neurosteroids in Status Epilepticus and Complex Partial Seizures. Epilepsia 2019, 60, 730–743. [Google Scholar] [CrossRef] [PubMed]
  173. Reddy, D.S. Neuroendocrine Aspects of Catamenial Epilepsy. Horm. Behav. 2013, 63, 254–266. [Google Scholar] [CrossRef] [PubMed]
  174. Oberlander, J.G.; Woolley, C.S. 17β-Estradiol Acutely Potentiates Glutamatergic Synaptic Transmission in the Hippocampus through Distinct Mechanisms in Males and Females. J. Neurosci. 2016, 36, 2677–2690. [Google Scholar] [CrossRef]
  175. Del Río, J.P.; Alliende, M.I.; Molina, N.; Serrano, F.G.; Molina, S.; Vigil, P. Steroid Hormones and Their Action in Women’s Brains: The Importance of Hormonal Balance. Front. Public Health 2018, 6, 141. [Google Scholar] [CrossRef]
  176. Bäckström, T.; Zetterlund, B.; Blom, S.; Romano, M. Effects of Intravenous Progesterone Infusions on the Epileptic Discharge Frequency in Women with Partial Epilepsy. Acta Neurol. Scand. 1984, 69, 240–248. [Google Scholar] [CrossRef] [PubMed]
  177. Scharfman, H.E.; MacLusky, N.J. The Influence of Gonadal Hormones on Neuronal Excitability, Seizures, and Epilepsy in the Female. Epilepsia 2006, 47, 1423–1440. [Google Scholar] [CrossRef]
  178. Velíšková, J. Estrogens and Epilepsy: Why Are We So Excited? Neuroscientist 2007, 13, 77–88. [Google Scholar] [CrossRef]
  179. Velíšková, J.; Velíšek, L. β-Estradiol Increases Dentate Gyrus Inhibition in Female Rats via Augmentation of Hilar Neuropeptide Y. J. Neurosci. 2007, 27, 6054–6063. [Google Scholar] [CrossRef]
  180. Reddy, D.S. The Role of Neurosteroids in the Pathophysiology and Treatment of Catamenial Epilepsy. Epilepsy Res. 2009, 85, 1–30. [Google Scholar] [CrossRef]
  181. Reddy, D.S. Anticonvulsant Activity of the Testosterone-Derived Neurosteroid 3α-Androstanediol. NeuroReport 2004, 15, 515. [Google Scholar] [CrossRef]
  182. Reddy, D.S.; Jian, K. The Testosterone-Derived Neurosteroid Androstanediol Is a Positive Allosteric Modulator of GABAA Receptors. J. Pharmacol. Exp. Ther. 2010, 334, 1031–1041. [Google Scholar] [CrossRef] [PubMed]
  183. Tandon, R.; Nasrallah, H.A.; Keshavan, M.S. Schizophrenia, “Just the Facts” 4. Clinical Features and Conceptualization. Schizophr. Res. 2009, 110, 1–23. [Google Scholar] [CrossRef] [PubMed]
  184. Rund, B.R.; Borg, N.E. Cognitive Deficits and Cognitive Training in Schizophrenic Patients: A Review. Acta Psychiatr. Scand. 1999, 100, 85–95. [Google Scholar] [CrossRef] [PubMed]
  185. Green, M.F. Cognitive Impairment and Functional Outcome in Schizophrenia and Bipolar Disorder. J. Clin. Psychiatry 2006, 67 (Suppl. S9), 3–8, discussion 36–42. [Google Scholar] [CrossRef]
  186. Aquila, R.; Citrome, L. Cognitive Impairment in Schizophrenia: The Great Unmet Need. CNS Spectr. 2015, 20, 32–40. [Google Scholar] [CrossRef]
  187. Howes, O.D.; Murray, R.M. Schizophrenia: An Integrated Sociodevelopmental-Cognitive Model. Lancet 2014, 383, 1677–1687. [Google Scholar] [CrossRef]
  188. Abel, K.M.; Drake, R.; Goldstein, J.M. Sex Differences in Schizophrenia. Int. Rev. Psychiatry 2010, 22, 417–428. [Google Scholar] [CrossRef]
  189. van der Werf, M.; Hanssen, M.; Köhler, S.; Verkaaik, M.; Verhey, F.R.; Investigators, R.; van Winkel, R.; van Os, J.; Allardyce, J. Systematic Review and Collaborative Recalculation of 133,693 Incident Cases of Schizophrenia. Psychol. Med. 2014, 44, 9–16. [Google Scholar] [CrossRef]
  190. Ferrara, M.; Srihari, V.H. Early Intervention for Psychosis in the United States: Tailoring Services to Improve Care for Women. Psychiatr. Serv. 2021, 72, 5–6. [Google Scholar] [CrossRef]
  191. Orrico-Sánchez, A.; López-Lacort, M.; Muñoz-Quiles, C.; Sanfélix-Gimeno, G.; Díez-Domingo, J. Epidemiology of Schizophrenia and Its Management over 8-Years Period Using Real-World Data in Spain. BMC Psychiatry 2020, 20, 149. [Google Scholar] [CrossRef]
  192. Sevilla-Llewellyn-Jones, J.; Cano-Domínguez, P.; de-Luis-Matilla, A.; Peñuelas-Calvo, I.; Espina-Eizaguirre, A.; Moreno-Kustner, B.; Ochoa, S. Personality Traits and Psychotic Symptoms in Recent Onset of Psychosis Patients. Compr. Psychiatry 2017, 74, 109–117. [Google Scholar] [CrossRef] [PubMed]
  193. Leung, A.; Chue, P. Sex Differences in Schizophrenia, a Review of the Literature. Acta Psychiatr. Scand. 2000, 101, 3–38. [Google Scholar] [CrossRef] [PubMed]
  194. Gur, R.E.; Turetsky, B.I.; Cowell, P.E.; Finkelman, C.; Maany, V.; Grossman, R.I.; Arnold, S.E.; Bilker, W.B.; Gur, R.C. Temporolimbic Volume Reductions in Schizophrenia. Arch. Gen. Psychiatry 2000, 57, 769–775. [Google Scholar] [CrossRef] [PubMed]
  195. Choleris, E.; Galea, L.A.M.; Sohrabji, F.; Frick, K.M. Sex Differences in the Brain: Implications for Behavioral and Biomedical Research. Neurosci. Biobehav. Rev. 2018, 85, 126–145. [Google Scholar] [CrossRef] [PubMed]
  196. Mendrek, A.; Stip, E. Sexual Dimorphism in Schizophrenia: Is There a Need for Gender-Based Protocols? Expert Rev. Neurother. 2011, 11, 951–959. [Google Scholar] [CrossRef] [PubMed]
  197. Kulkarni, J.; Hayes, E.; Gavrilidis, E. Hormones and Schizophrenia. Curr. Opin. Psychiatry 2012, 25, 89. [Google Scholar] [CrossRef]
  198. Markham, J.A. Sex Steroids and Schizophrenia. Rev. Endocr. Metab. Disord. 2012, 13, 187–207. [Google Scholar] [CrossRef]
  199. Weickert, T.W.; Weinberg, D.; Lenroot, R.; Catts, S.V.; Wells, R.; Vercammen, A.; O’Donnell, M.; Galletly, C.; Liu, D.; Balzan, R.; et al. Adjunctive Raloxifene Treatment Improves Attention and Memory in Men and Women with Schizophrenia. Mol. Psychiatry 2015, 20, 685–694. [Google Scholar] [CrossRef]
  200. Sun, J.; Walker, A.J.; Dean, B.; van den Buuse, M.; Gogos, A. Progesterone: The Neglected Hormone in Schizophrenia? A Focus on Progesterone-Dopamine Interactions. Psychoneuroendocrinology 2016, 74, 126–140. [Google Scholar] [CrossRef]
  201. McGregor, C.; Riordan, A.; Thornton, J. Estrogens and the Cognitive Symptoms of Schizophrenia: Possible Neuroprotective Mechanisms. Front. Neuroendocrinol. 2017, 47, 19–33. [Google Scholar] [CrossRef]
  202. Begemann, M.J.H.; Dekker, C.F.; van Lunenburg, M.; Sommer, I.E. Estrogen Augmentation in Schizophrenia: A Quantitative Review of Current Evidence. Schizophr. Res. 2012, 141, 179–184. [Google Scholar] [CrossRef] [PubMed]
  203. Gogos, A.; Sbisa, A.M.; Sun, J.; Gibbons, A.; Udawela, M.; Dean, B. A Role for Estrogen in Schizophrenia: Clinical and Preclinical Findings. Int. J. Endocrinol. 2015, 2015, e615356. [Google Scholar] [CrossRef] [PubMed]
  204. Kaneda, Y.; Ohmori, T. Relation Between Estradiol and Negative Symptoms in Men with Schizophrenia. J. Neuropsychiatry Clin. Neurosci. 2005, 17, 239–242. [Google Scholar] [CrossRef] [PubMed]
  205. Brand, B.A.; de Boer, J.N.; Sommer, I.E.C. Estrogens in Schizophrenia: Progress, Current Challenges and Opportunities. Curr. Opin. Psychiatry 2021, 34, 228–237. [Google Scholar] [CrossRef] [PubMed]
  206. Busatto, G.F.; Kerwin, R.W. Perspectives on the Role of Serotonergic Mechanisms in the Pharmacology of Schizophrenia. J. Psychopharmacol. 1997, 11, 3–12. [Google Scholar] [CrossRef]
  207. Abi-Dargham, A.; Laruelle, M.; Aghajanian, G.K.; Charney, D.; Krystal, J. The Role of Serotonin in the Pathophysiology and Treatment of Schizophrenia. J. Neuropsychiatry Clin. Neurosci. 1997, 9, 1–17. [Google Scholar] [CrossRef]
  208. Halari, R.; Kumari, V.; Mehrotra, R.; Wheeler, M.; Hines, M.; Sharma, T. The Relationship of Sex Hormones and Cortisol with Cognitive Functioning in Schizophrenia. J. Psychopharmacol. 2004, 18, 366–374. [Google Scholar] [CrossRef]
  209. Rubin, L.H.; Carter, C.S.; Drogos, L.; Pournajafi-Nazarloo, H.; Sweeney, J.A.; Maki, P.M. Peripheral Oxytocin Is Associated with Reduced Symptom Severity in Schizophrenia. Schizophr. Res. 2010, 124, 13–21. [Google Scholar] [CrossRef]
  210. Ritsner, M.S.; Ratner, Y. The Long-Term Changes in Coping Strategies in Schizophrenia: Temporal Coping Types. J. Nerv. Ment. Dis. 2006, 194, 261. [Google Scholar] [CrossRef]
  211. Ko, Y.-H.; Jung, S.-W.; Joe, S.-H.; Lee, C.-H.; Jung, H.-G.; Jung, I.-K.; Kim, S.-H.; Lee, M.-S. Association between Serum Testosterone Levels and the Severity of Negative Symptoms in Male Patients with Chronic Schizophrenia. Psychoneuroendocrinology 2007, 32, 385–391. [Google Scholar] [CrossRef]
  212. Sisek-Šprem, M.; Križaj, A.; Jukić, V.; Milošević, M.; Petrović, Z.; Herceg, M. Testosterone Levels and Clinical Features of Schizophrenia with Emphasis on Negative Symptoms and Aggression. Nord. J. Psychiatry 2015, 69, 102–109. [Google Scholar] [CrossRef] [PubMed]
  213. Mendrek, A. Reversal of Normal Cerebral Sexual Dimorphism in Schizophrenia: Evidence and Speculations. Med. Hypotheses 2007, 69, 896–902. [Google Scholar] [CrossRef]
  214. Moffat, S.D. Effects of Testosterone on Cognitive and Brain Aging in Elderly Men. Ann. N. Y. Acad. Sci. 2005, 1055, 80–92. [Google Scholar] [CrossRef] [PubMed]
  215. Akhondzadeh, S.; Rezaei, F.; Larijani, B.; Nejatisafa, A.-A.; Kashani, L.; Abbasi, S.H. Correlation between Testosterone, Gonadotropins and Prolactin and Severity of Negative Symptoms in Male Patients with Chronic Schizophrenia. Schizophr. Res. 2006, 84, 405–410. [Google Scholar] [CrossRef] [PubMed]
  216. Sasayama, D.; Hattori, K.; Teraishi, T.; Hori, H.; Ota, M.; Yoshida, S.; Arima, K.; Higuchi, T.; Amano, N.; Kunugi, H. Negative Correlation between Cerebrospinal Fluid Oxytocin Levels and Negative Symptoms of Male Patients with Schizophrenia. Schizophr. Res. 2012, 139, 201–206. [Google Scholar] [CrossRef] [PubMed]
  217. Cochran, D.; Fallon, D.; Hill, M.; Frazier, J.A. The Role of Oxytocin in Psychiatric Disorders: A Review of Biological and Therapeutic Research Findings. Harv. Rev. Psychiatry 2013, 21, 219–247. [Google Scholar] [CrossRef]
  218. Frost, K.; Keller, W.; Buchanan, R.; Gold, J.; Koenig, J.; Ossenfort, K.; Katz, A.; Strauss, G. C-14 Plasma Oxytocin Levels Are Associated with Impaired Social Cognition and Neurocognition in Schizophrenia. Arch. Clin. Neuropsychol. 2014, 29, 577–578. [Google Scholar] [CrossRef]
  219. Feifel, D. Is Oxytocin a Promising Treatment for Schizophrenia? Expert Rev. Neurother. 2011, 11, 157–159. [Google Scholar] [CrossRef]
  220. Borsboom, D.; Cramer, A.O.J.; Kalis, A. Brain Disorders? Not Really: Why Network Structures Block Reductionism in Psychopathology Research. Behav. Brain Sci. 2019, 42, e2. [Google Scholar] [CrossRef]
  221. Yang, L.; Zhao, H.; Cui, H. Treatment and New Progress of Neonatal Hypoxic-Ischemic Brain Damage. Histol. Histopathol. 2020, 35, 929–936. [Google Scholar] [CrossRef]
  222. Bristow, G.C.; Bostrom, J.A.; Haroutunian, V.; Sodhi, M.S. Sex Differences in GABAergic Gene Expression Occur in the Anterior Cingulate Cortex in Schizophrenia. Schizophr. Res. 2015, 167, 57–63. [Google Scholar] [CrossRef] [PubMed]
  223. Sun, X.; Zhang, J.; Niu, W.; Guo, W.; Song, H.; Li, H.; Fan, H.; Zhao, L.; Zhong, A.; Dai, Y.; et al. A Preliminary Analysis of MicroRNA as Potential Clinical Biomarker for Schizophrenia. Am. J. Med. Genet. Part B Neuropsychiatr. Genet. 2015, 168, 170–178. [Google Scholar] [CrossRef] [PubMed]
  224. Chang, H.; Xiao, X.; Li, M. The Schizophrenia Risk Gene ZNF804A: Clinical Associations, Biological Mechanisms and Neuronal Functions. Mol. Psychiatry 2017, 22, 944–953. [Google Scholar] [CrossRef] [PubMed]
  225. Huang, Y.; Huang, J.; Zhou, Q.-X.; Yang, C.-X.; Yang, C.-P.; Mei, W.-Y.; Zhang, L.; Zhang, Q.; Hu, L.; Hu, Y.-Q.; et al. ZFP804A Mutant Mice Display Sex-Dependent Schizophrenia-like Behaviors. Mol. Psychiatry 2021, 26, 2514–2532. [Google Scholar] [CrossRef] [PubMed]
  226. de Castro-Catala, M.; Mora-Solano, A.; Kwapil, T.R.; Cristóbal-Narváez, P.; Sheinbaum, T.; Racioppi, A.; Barrantes-Vidal, N.; Rosa, A. The Genome-Wide Associated Candidate Gene ZNF804A and Psychosis-Proneness: Evidence of Sex-Modulated Association. PLoS ONE 2017, 12, e0185072. [Google Scholar] [CrossRef]
  227. Perez-Becerril, C.; Morris, A.G.; Mortimer, A.; McKenna, P.J.; de Belleroche, J. Common Variants in the Chromosome 2p23 Region Containing the SLC30A3 (ZnT3) Gene Are Associated with Schizophrenia in Female but Not Male Individuals in a Large Collection of European Samples. Psychiatry Res. 2016, 246, 335–340. [Google Scholar] [CrossRef]
  228. Arnold, A.P. The Organizational–Activational Hypothesis as the Foundation for a Unified Theory of Sexual Differentiation of All Mammalian Tissues. Horm. Behav. 2009, 55, 570–578. [Google Scholar] [CrossRef]
  229. 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, 959–992. [Google Scholar] [CrossRef]
  230. Christiansen, D.M.; Hansen, M. Accounting for Sex Differences in PTSD: A Multi-Variable Mediation Model. Eur. J. Psychotraumatol. 2015, 6, 26068. [Google Scholar] [CrossRef]
  231. Rubinow, D.R.; Schmidt, P.J. Sex Differences and the Neurobiology of Affective Disorders. Neuropsychopharmacology 2019, 44, 111–128. [Google Scholar] [CrossRef]
  232. Pitman, R.K.; Rasmusson, A.M.; Koenen, K.C.; Shin, L.M.; Orr, S.P.; Gilbertson, M.W.; Milad, M.R.; Liberzon, I. Biological Studies of Post-Traumatic Stress Disorder. Nat. Rev. Neurosci. 2012, 13, 769–787. [Google Scholar] [CrossRef] [PubMed]
  233. Ney, L.J.; Matthews, A.; Bruno, R.; Felmingham, K.L. Modulation of the Endocannabinoid System by Sex Hormones: Implications for Posttraumatic Stress Disorder. Neurosci. Biobehav. Rev. 2018, 94, 302–320. [Google Scholar] [CrossRef] [PubMed]
  234. LeGates, T.A.; Kvarta, M.D.; Thompson, S.M. Sex Differences in Antidepressant Efficacy. Neuropsychopharmacology 2019, 44, 140–154. [Google Scholar] [CrossRef]
  235. Angold, A.; Costello, E.J.; Worthman, C.M. Puberty and Depression: The Roles of Age, Pubertal Status and Pubertal Timing. Psychol. Med. 1998, 28, 51–61. [Google Scholar] [CrossRef] [PubMed]
  236. Frokjaer, V.G. Pharmacological Sex Hormone Manipulation as a Risk Model for Depression. J. Neurosci. Res. 2020, 98, 1283–1292. [Google Scholar] [CrossRef] [PubMed]
  237. Rasmusson, A.M.; Marx, C.E.; Pineles, S.L.; Locci, A.; Scioli-Salter, E.R.; Nillni, Y.I.; Liang, J.J.; Pinna, G. Neuroactive Steroids and PTSD Treatment. Neurosci. Lett. 2017, 649, 156–163. [Google Scholar] [CrossRef]
  238. Glover, E.M.; Jovanovic, T.; Norrholm, S.D. Estrogen and Extinction of Fear Memories:Implications for Posttraumatic Stress Disorder Treatment. Biol. Psychiatry 2015, 78, 178–185. [Google Scholar] [CrossRef]
  239. Li, S.; Graham, B.M. Estradiol Is Associated with Altered Cognitive and Physiological Responses during Fear Conditioning and Extinction in Healthy and Spider Phobic Women. Behav. Neurosci. 2016, 130, 614–623. [Google Scholar] [CrossRef]
  240. White, E.C.; Graham, B.M. Estradiol Levels in Women Predict Skin Conductance Response but Not Valence and Expectancy Ratings in Conditioned Fear Extinction. Neurobiol. Learn. Mem. 2016, 134, 339–348. [Google Scholar] [CrossRef]
  241. Valentino, R.J.; Bangasser, D.; Bockstaele, E.V. Sex Biased Stress Signaling: The corticotropin-releasing factor receptor as a model. Mol. Pharmacol. 2013, 83, 737–745. [Google Scholar] [CrossRef]
  242. Doyle, H.H.; Eidson, L.N.; Sinkiewicz, D.M.; Murphy, A.Z. Sex Differences in Microglia Activity within the Periaqueductal Gray of the Rat: A Potential Mechanism Driving the Dimorphic Effects of Morphine. J. Neurosci. 2017, 37, 3202–3214. [Google Scholar] [CrossRef] [PubMed]
  243. Torrisi, S.A.; Rizzo, S.; Laudani, S.; Ieraci, A.; Drago, F.; Leggio, G.M. Acute Stress Alters Recognition Memory and AMPA/NMDA Receptor Subunits in a Sex-Dependent Manner. Neurobiol. Stress 2023, 25, 100545. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Gender bias in neuropsychiatric disorders.
Figure 1. Gender bias in neuropsychiatric disorders.
Ijms 24 14571 g001
Table 1. Sex-related differences in neuropsychiatric disorders.
Table 1. Sex-related differences in neuropsychiatric disorders.
Etiology/Molecular MechanismFemaleMale
Autism spectrum disorder (ASD)Prenatal and perinatal altered brain development and neural reorganization (e.g., abnormal prenatal exposure to testosterone correlates positively with several ASD features) [1,2]
  • Less restricted interest and activities and more significant social communication difficulties [3,4,5].
  • More severely affected [6,7,8,9].
Down syndrome (DS)Genetic-trisomy of chromosome 21
  • Higher risk of developing Alzheimer-like dementia [10,11].
  • Greater behavioral problems (attention deficit, hyperactivity-like behaviors in childhood, and depression in adulthood) [12]
  • Higher prevalence of sleep disorder breathing [13]
Rett syndrome (RTT)Genetic-mutations in the chromosome X-linked methyl-CpG-binding protein 2 (MECP2) gene
  • Intellectual disability, neurological regression, loss of speech, learning deficits, epilepsy (60–80% of RTT cases) [14]
  • RTT is rare (boys generally do not survive past infancy) [15,16,17,18,19]
Fragile X Syndrome (FXS)Genetic->200 repeats of the CGG motif in the chromosome X-linked FMR1 gene and loss of its product, fragile X mental retardation 1 protein (FMRP)
  • Less severe symptoms than males (due to the compensation by the X chromosome) [20]
  • Mild intellectual disability or normal intelligence [21,22]
  • Intellectual impairment and autistic-like behaviors [23]
Bipolar Disorder (BD)Genetic, neuroanatomic, neurochemical and environmental factors
E.g., dysregulation of several genes and pathways associated with ubiquitin pathway, synaptic functions [24], intracellular protein trafficking, protein metabolic processes [25] and neuroinflammation [26]
  • A higher rate of rapid cycles [27]
  • higher number of depressive episodes [28,29]
  • more frequent suicide attempts [30,31]
  • A higher rate of manic episodes and unipolar mania [28]
EpilepsyDysfunction of the normal connectivity of neural networks or/and imbalance of inhibitory and excitatory neurotransmitters (genetic, structural, infectious, metabolic, and immune factors)
  • Less frequent incidence of epilepsy than males [32,33,34,35]
  • Higher rate of excitability episodes and the occurrence of epileptic seizures [32,33,34,35].
Schizophrenia (SZ)Structural and functional brain abnormalities, neurochemical disturbance (in dopamine and NMDA receptor function) and genetic factors. Estrogen has a neuroprotective effect on neurotransmitter systems: dopaminergic [36,37] and glutamatergic [38,39,40].
  • More affective symptoms (depression, impulsivity, self-harm, and suicide attempts) [41]
  • More severe negative and cognitive symptoms (social withdrawal, lack of motivation, poverty of speech) [42,43,44]
Stress-related depressive disorder (MDD, PTSD)deregulation of neurotransmitters; neuroinflammation, imbalance in the hypothalamic–pituitary–adrenal axis after extreme stress [45,46]
  • Lower threshold for manifestation of depressive disorders [45,47]
  • estradiol is a possible risk factor in the prevalence and severity of depressive disorder [48]
  • Lower risk of depressive episodes in men than in women [49]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Ziemka-Nalecz, M.; Pawelec, P.; Ziabska, K.; Zalewska, T. Sex Differences in Brain Disorders. Int. J. Mol. Sci. 2023, 24, 14571. https://doi.org/10.3390/ijms241914571

AMA Style

Ziemka-Nalecz M, Pawelec P, Ziabska K, Zalewska T. Sex Differences in Brain Disorders. International Journal of Molecular Sciences. 2023; 24(19):14571. https://doi.org/10.3390/ijms241914571

Chicago/Turabian Style

Ziemka-Nalecz, Malgorzata, Paulina Pawelec, Karolina Ziabska, and Teresa Zalewska. 2023. "Sex Differences in Brain Disorders" International Journal of Molecular Sciences 24, no. 19: 14571. https://doi.org/10.3390/ijms241914571

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

Article Metrics

Back to TopTop